Scolaris Content Display Scolaris Content Display

Tratamientos sistémicos para la prevención o el tratamiento de los síntomas musculoesqueléticos inducidos por los inhibidores de la aromatasa en el cáncer de mama temprano

Contraer todo Desplegar todo

Antecedentes

Los inhibidores de la aromatasa (IA) adyuvantes mejoran la supervivencia en comparación con el tamoxifeno en mujeres posmenopáusicas con cáncer de mama con sobreexpresión de receptores hormonales en estadio I a III. En aproximadamente la mitad de estas mujeres, los IA se asocian con síntomas musculoesqueléticos inducidos por los inhibidores de la aromatasa (SMEIA), a menudo descritos como dolor y molestias simétricos en las articulaciones, dolor musculoesquelético y rigidez articular. Los SMEIA podrían tener un efecto significativo y prolongado en la calidad de vida de las mujeres. Los SMEIA reducen la adherencia al tratamiento con IA en hasta la mitad de las mujeres, lo que puede comprometer los desenlaces del cáncer de mama. Se han investigado diferentes tratamientos sistémicos para la prevención y el tratamiento de los SMEIA, pero su efectividad aún no está clara.

Objetivos

Evaluar los efectos de los tratamientos sistémicos en la prevención o el tratamiento de los SMEIA en mujeres con cáncer de mama con sobreexpresión de receptores hormonales en estadio I a III.

Métodos de búsqueda

Se realizaron búsquedas en CENTRAL, MEDLINE, Embase, en los registros de la Plataforma de registros internacionales de ensayos clínicos (ICTRP) de la OMS y Clinicaltrials.gov hasta septiembre de 2020 y en el registro especializado del Grupo Cochrane de Cáncer de mama (Cochrane Breast Cancer Group [CBCG]) hasta marzo de 2021.

Criterios de selección

Se incluyeron todos los ensayos controlados aleatorizados que compararon los tratamientos sistémicos con un grupo de comparación. Las intervenciones con tratamientos sistémicos incluyeron todos los tratamientos farmacológicos, los suplementos dietéticos y las medicinas complementarias y alternativas (MCA). Se permitieron todos los grupos de comparación, incluido el placebo o la atención estándar (o ambos) con analgesia sola. Fueron elegibles los estudios publicados no revisados por pares.

Obtención y análisis de los datos

Dos autores de la revisión examinaron de forma independiente los estudios, extrajeron los datos y evaluaron el riesgo de sesgo y la certeza de la evidencia mediante el método GRADE. Los desenlaces evaluados fueron el dolor, la rigidez, la fuerza de prensión, los datos de seguridad, la interrupción del IA, la calidad de vida relacionada con la salud (CdVRS), la calidad de vida específica del cáncer de mama (CdVECM), la incidencia de SMEIA, la supervivencia específica del cáncer de mama (SECM) y la supervivencia general (SG). Para los desenlaces continuos se utilizó el recuento de votos informando cuántos estudios comunicaron un beneficio clínicamente significativo dentro de los intervalos de confianza (IC) de la diferencia de medias (DM) entre los grupos de tratamiento, según lo determinado por la diferencia mínima cínicamente importante (DMCI) para esa escala de desenlaces. Para los desenlaces dicotómicos se proporcionaron los desenlaces como una razón de riesgos (RR) con IC del 95%.

Resultados principales

Se incluyeron 17 estudios con 2034 participantes asignados al azar. Cuatro estudios evaluaron los tratamientos sistémicos para la prevención de los SMEIA y 13 estudios investigaron el tratamiento de los SMEIA. Debido a la variación de los estudios de tratamientos sistémicos, incluidos los farmacológicos, y los de medicina complementaria y alternativa, o a los datos no disponibles, el metanálisis fue limitado, y sólo se combinaron dos ensayos. La certeza de la evidencia para todos los desenlaces fue baja o muy baja.

Estudios de prevención

La evidencia sobre el efecto de los tratamientos sistémicos en el dolor (desde el inicio hasta el final de la intervención; dos estudios, 183 mujeres) es muy incierta. Los dos estudios, que investigaron la vitamina D y los ácidos grasos omega‐3, mostraron un efecto del tratamiento con IC del 95% que no incluyeron una DMCI para el dolor. Los tratamientos sistémicos podrían tener un efecto escaso o nulo sobre la fuerza de prensión (RR 1,08; IC del 95%: 0,37 a 3,17; un estudio, 137 mujeres) o sobre el hecho de que las mujeres continúen tomando el IA (RR 0,16; IC del 95%: 0,01 a 2,99; un estudio, 147 mujeres). La evidencia indica un efecto escaso o nulo sobre la CdVRS y la CdVECM desde el inicio hasta el final de la intervención (el mismo estudio; 44 mujeres, ambos desenlaces de calidad de vida mostraron un efecto del tratamiento con IC del 95% que incluyeron una DMCI).

La evidencia con respecto a los desenlaces que evalúan la incidencia de SMEIA (RR 0,82, IC del 95%: 0,63 a 1,06; dos estudios, 240 mujeres) y la seguridad de los tratamientos sistémicos (cuatro estudios, 344 mujeres; evidencia de certeza muy baja) es muy incierta. En un estudio se emitió una alerta de la Food and Drug Administration de EE.UU. para la intervención (inhibidor de la ciclooxigenasa‐2) durante el estudio, pero no hubo eventos adversos graves en este ni en ningún otro estudio.

No hubo datos sobre la rigidez, la SECM ni la SG.

Estudios de tratamientos

La evidencia sobre el efecto de los tratamientos sistémicos sobre el dolor desde el inicio hasta el final de la intervención en el tratamiento de los SMEIA (diez estudios, 1099 mujeres) es muy incierta. Cuatro estudios mostraron una DMCI en las puntuaciones de dolor que se encontraba dentro del IC del 95% del efecto medido (vitamina D, hueso de tigre artificial, granulado de Yi Shen Jian Gu, calcitonina). Seis estudios mostraron un efecto del tratamiento con un IC del 95% que no incluyó una DMCI (vitamina D, testosterona, ácidos grasos omega‐3, duloxetina, aceite de emú, uña de gato).

La evidencia para los desenlaces de cambio en la rigidez (cuatro estudios, 295 mujeres), la CdVRS (tres estudios, 208 mujeres) y la CdVECM (dos estudios, 147 mujeres) desde el inicio hasta el final de la intervención, fue muy incierta. La evidencia indica que los tratamientos sistémicos podrían tener poco o ningún efecto sobre la fuerza de prensión (un estudio, 107 mujeres). La evidencia sobre la seguridad de los tratamientos sistémicos (diez estudios, 1250 mujeres) es muy incierta. No se notificaron eventos adversos de grado 4‐5 en ninguno de los estudios. El estudio de la duloxetina informó de que hubo más eventos adversos de todo tipo en este grupo de tratamiento que en el grupo de comparación.

No hubo datos sobre la incidencia de SMEIA, el número de mujeres que seguían tomando IA, la SECM ni la SG a partir de los estudios de tratamiento.

Conclusiones de los autores

Los SMEIA son síntomas crónicos y complejos con un impacto significativo en las mujeres con cáncer de mama en estadio inicial que reciben un IA. Hasta la fecha, la evidencia de la seguridad y efectividad de los tratamientos sistémicos para la prevención o el tratamiento de los SMEIA ha sido mínima. Aunque esta revisión identificó 17 estudios con 2034 participantes asignadas al azar, la revisión fue difícil debido a la heterogeneidad de las intervenciones con tratamientos sistémicos y las metodologías de estudio, así como a la falta de disponibilidad de ciertos datos de los ensayos. Por lo tanto, el metanálisis fue limitado y los resultados de la revisión no fueron concluyentes. Se recomienda la realización de más estudios de investigación sobre los tratamientos sistémicos para los SMEIA, que incluyan ECA de calidad alta con poder estadístico suficiente, descripciones completas de la intervención/placebo y definiciones sólidas de la enfermedad y de los desenlaces estudiados.

PICO

Population
Intervention
Comparison
Outcome

El uso y la enseñanza del modelo PICO están muy extendidos en el ámbito de la atención sanitaria basada en la evidencia para formular preguntas y estrategias de búsqueda y para caracterizar estudios o metanálisis clínicos. PICO son las siglas en inglés de cuatro posibles componentes de una pregunta de investigación: paciente, población o problema; intervención; comparación; desenlace (outcome).

Para saber más sobre el uso del modelo PICO, puede consultar el Manual Cochrane.

Tratamientos sistémicos para la prevención o el tratamiento de los síntomas musculoesqueléticos inducidos por los inhibidores de la aromatasa en el cáncer de mama temprano

¿Cuál era el objetivo de esta revisión?

La terapia hormonal con inhibidores de la aromatasa se utiliza para tratar un tipo de cáncer de mama incipiente (con sobreexpresión de receptores hormonales) en mujeres después de la menopausia. Los IA causan efectos secundarios como dolores y rigidez articulares y musculares (síntomas musculoesqueléticos de los inhibidores de la aromatasa, o los llamados SMEIA), que podrían hacer que algunas mujeres dejen de tomarlos, y potencialmente empeorar la supervivencia. El objetivo de esta revisión Cochrane fue examinar si los tratamientos sistémicos (tratamientos que llegan a las células de todo el cuerpo viajando por la sangre) pueden prevenir o tratar los SMEIA. Los autores de la revisión recopilaron y analizaron todos los estudios relevantes para responder esta pregunta.

Mensajes clave

No está muy claro si los tratamientos sistémicos mejoran, empeoran o no suponen una diferencia en el dolor o la calidad de vida de las mujeres que toman inhibidores de la aromatasa. La mayoría de la evidencia fue de calidad muy baja. No estuvo muy claro si los tratamientos sistémicos para los SMEIA fueron seguros.

¿Qué estudió la revisión?

Se examinaron los estudios de investigación sobre tratamientos sistémicos, que incluyeron medicamentos, vitaminas y medicinas complementarias y alternativas, para ver si podían prevenir o tratar los dolores y la rigidez articular y muscular de las mujeres que tomaban inhibidores de la aromatasa. Se incluyeron ensayos de tratamientos sistémicos comparados con placebo (tratamiento falso), o con tratamientos estándar. Se incluyeron mujeres tratadas con inhibidores de la aromatasa para el cáncer de mama incipiente con sobreexpresión de receptores hormonales. La mayoría de los estudios eran para el tratamiento de los SMEIA.

Los desenlaces que se estudiaron fueron los cambios en el dolor, la rigidez, la fuerza de la mano (fuerza de prensión), la seguridad y los efectos secundarios de los tratamientos de estudio, el número de mujeres que siguieron tomando los inhibidores de la aromatasa, la calidad de vida de las mujeres, el número de mujeres que presentaron dolores musculares y articulares debido a los inhibidores de la aromatasa y la supervivencia.

¿Cuáles son los principales resultados de esta revisión?

Después de recopilar y analizar todos los estudios relevantes, se encontraron 17 estudios con 2034 mujeres incluidas. En estos estudios participaron diferentes cantidades de mujeres, entre 37 y 299. Cuatro estudios analizaron los tratamientos sistémicos para prevenir los dolores articulares y musculares de los inhibidores de la aromatasa; 13 estudios investigaron los tratamientos sistémicos para tratar estos síntomas. Diez estudios se realizaron en EE.UU., tres en China, dos en Australia, uno en Italia y uno en Brasil. Muchos de los estudios contaron con un escaso número de mujeres, lo que puede haber dificultado la detección de pequeñas diferencias. Hubo problemas con algunos estudios con riesgo de sesgo. Otros problemas se debieron a que varios estudios no habían publicado completamente la información sobre los ingredientes del tratamiento o sus resultados, por lo que algunos datos no estaban disponibles para su revisión o análisis. Además, los estudios utilizaron muchos tipos distintos de tratamiento, y no fue apropiado combinar sus resultados en el análisis.

Estudios de prevención de los SMEIA

No está claro si alguno de estos estudios encontró un efecto positivo o negativo sobre el dolor, y sobre el número de mujeres que desarrollaron SMEIA debido a la calidad muy baja de la evidencia. Los tratamientos sistémicos podrían tener un efecto escaso o nulo sobre la fuerza de prensión, la calidad de vida o el hecho de que las mujeres continuaran tomando los inhibidores de la aromatasa (evidencia de calidad baja). Ninguno de los estudios analizó la rigidez.

Estudios de tratamiento de los SMEIA

No está claro si alguno de estos estudios encontró un efecto positivo o negativo sobre el dolor, la rigidez y la calidad de vida de las mujeres debido a la calidad muy baja de la evidencia. Es probable que los tratamientos sistémicos produzcan poco o ningún cambio en la fuerza de prensión en las mujeres con SMEIA (evidencia de calidad baja). Ninguno de los estudios analizó el número de mujeres que continuaron tomando los inhibidores de la aromatasa o que desarrollaron SMEIA, ni la supervivencia.

Seguridad

No se sabe si los tratamientos sistémicos para los SMEIA son seguros, ya que la evidencia es muy incierta. No hubo efectos secundarios graves. Un tratamiento, la duloxetina, provocó un aumento de los efectos secundarios en las mujeres, y un tratamiento, el etoricoxib, tuvo una alerta de seguridad durante el ensayo. La duración del seguimiento de las mujeres en muchos estudios fue corta. Los datos de seguridad se deben interpretar con precaución.

¿Cuál es el grado de actualización de esta revisión?

La última búsqueda de estudios (publicados y en curso) para esta revisión se realizó en septiembre de 2020 dentro de las bases de datos especificadas y en marzo de 2021 en el Registro especializado de grupo Cochrane de Cáncer de mama.

Authors' conclusions

Implications for practice

Aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS) is a chronic symptom complex with a significant impact on women with early breast cancer taking aromatase inhibitors (AI). Poor adherence to AI therapy due to AIMSS impacts breast cancer survival. To date, evidence for safe and effective systemic therapies for prevention or treatment of AIMSS has been minimal. 

This review has demonstrated that there is overall very low‐certainty evidence for systemic therapies for prevention or management of AIMSS in postmenopausal women with early breast cancer for most outcomes. Studies were clinically and methodologically heterogeneous and investigated diverse pharmacological and complementary and alternative medicines (CAM) therapies, which were often not appropriate to combine for meta‐analysis. Certain data were not available, which also limited meta‐analysis. Consequent to this, we were very uncertain of the effect of systemic therapies for either prevention or treatment of AIMSS on many important AIMSS outcomes including change in pain, health‐related quality of life and cancer‐specific quality of life. There was also limited information on long term benefits and harms of studied interventions. Certain studies had limited information available about the ingredients of the intervention and/or the placebo. Based on the very low certainty of evidence across multiple outcomes, no definitive recommendations for systemic therapies for either prevention or treatment of AIMSS on the basis of this review can be made.

An evidence‐based summary on management of AIMSS suggested that duloxetine (as per the study by Henry 2018) is one of the treatment options for women with AIMSS among other options, particularly if women have comorbid anxiety, depression or chronic osteoarthritis (Gupta 2020). The trial by Henry 2018 demonstrated the mean joint pain score was 0.82 points lower for women who received duloxetine compared with those who received placebo by 12 weeks (95% CI −1.24 to −0.40; P = 0.0002). However, this result did not indicate a clinically significant benefit, with the clinically significant change in mean pain defined as a decrease from baseline of at least two points. Within 12 weeks of discontinuing either duloxetine or placebo, pain scores were equivalent between the two arms. There were higher adverse event rates with duloxetine over placebo, which potentially led to unblinding. A subsequent retrospective exploratory analysis suggested women with obese range body mass index (BMI) of 30 kg/m2 of greater received more benefit from duloxetine (Henry 2019), however the original study was not designed to examine differences in pain scores by BMI. Given the short‐term duration of the study (Henry 2018), no evidence is available for the effectiveness of duloxetine for long‐term treatment of AIMSS. Similarly, no evidence is available that indicates whether duloxetine will improve AI adherence. The implications for practice of the evidence for duloxetine for AIMSS treatment are potentially limited based on our Cochrane Review. Comorbid conditions that may be treated by duloxetine or alter its efficacy in the setting of AIMSS have not been addressed in this Cochrane Review. 

Vitamin D was investigated in both the prevention (Khan 2017Niravath 2019) and treatment (Rastelli 2011Shapiro 2016) of AIMSS. Clinical and methodological heterogeneity was present in these studies, and overall evidence was very uncertain on the effect of vitamin D across multiple outcomes. Hence, measurement of vitamin D concentration and routine vitamin D oral supplementation cannot be recommended for prevention or management of AIMSS on the basis of our review; however, it may be indicated for bone health management in early breast cancer and for documented vitamin D insufficiency/deficiency. Similar recommendations with respect to vitamin D have been made by Gupta 2020.

Further research into optimal systemic therapies to prevent and treat AIMSS are recommended to improve outcomes and quality of life for women with early breast cancer.

Implications for research

This Cochrane Review has highlighted many of the research priorities for postmenopausal women with early breast cancer experiencing AIMSS. Further adequately powered, high‐quality, randomised phase three trials are required to definitively answer research questions on whether systemic therapies can prevent or treat AIMSS in postmenopausal women with breast cancer. AIMSS remains a poorly understood symptom complex with an unclear aetiology (Hershman 2015bNiravath 2013). Without a clear understanding of the aetiology, researchers are yet to design targeted interventions or drug modifications with systemic therapies that comprehensively address the various aspects of AIMSS.

Ongoing clinical trials of systemic therapy would likely be enhanced by further research into the aetiology of AIMSS. However, as noted by Hershman 2015b, it may be the lowering of oestradiol caused by AI, the desirable effect for breast cancer efficacy, is also the inciting factor for AIMSS. Careful attention would then be needed with any future trials directed against this potential mechanism of action of AIMSS as these could affect the efficacy of AI (Hershman 2015b).

Similar to the Cochrane Review of exercise for AIMSS (Roberts 2020) and as noted by Hershman 2015b, research has been limited by heterogeneous populations. Prior chemotherapy, prior hormone replacement therapy and increased bodyweight are associated with an increased risk of AIMSS (Hershman 2015bNiravath 2013). One large observational study demonstrated that CAM use prior to the initiation of letrozole did not prevent or improve the development of AIMSS; however, the CAM group had higher pain levels throughout the observation period (Hack 2020). Exploratory subgroup analyses of trials of omega‐3 fatty acid (Hershman 2015aShen 2018) and duloxetine (Henry 2018Henry 2019) suggested obese participants gained more analgesic benefit than non‐obese participants. Understanding the impact of host factors on personalised risk of AIMSS and targeting research interventions to those most at risk are research priorities for future clinical trials. Stratification for risk factors, particularly obesity, should similarly be considered for future trials (Henry 2019).

Future research into AIMSS would also be improved by enhancing methodological rigor and reducing bias. All trials should ideally be prospectively registered and published in peer‐reviewed journals on completion, regardless of size. Of particular focus, research would be improved by standardisation of measurement of outcomes in AIMSS. Importantly, there remains no consensus on the constellation of symptoms and signs that define AIMSS (Hershman 2015bNiravath 2013). Studies have used multiple different outcome measures, predominantly patient‐reported outcomes (PROs) (Hershman 2015b). PROs measure subjective experiences such as symptoms from the patient's perspective, and considered the gold standard for quantifying symptomatic effects of treatments (Basch 2010Patrick 2007) as physicians under‐report treatment‐emergent toxicities in people with cancer (Basch 2016Basch 2017). As PROs are the most reliable tools for quality of life research (Deshpande 2011), consensus on the definition of the syndrome and the most appropriate outcome measures utilising PROs remains a research priority. Development of more reliable instruments is underway (Castel 2015); however, further research is required. The systematic incorporation of PROs, including electronic platforms, with better standardisation, will improve research quality, and improve patient and carers' research experience (Brandt 2019Rocque 2018). Standardisation of outcome measures would be required to reduce the heterogeneity that has thus far hampered meta‐analysis in research efforts into AIMSS. In addition, the interpretation of the clinical importance or meaningfulness of results for many of the PROs utilised in AIMSS research has not been specifically established in AIMSS. Interpretation of meaningfulness of trial data has been extrapolated from other chronic pain, rheumatological or cancer pain syndromes (Arnold 2005Dworkin 2009Farrar 2010Mease 2011Olsen 2018). Ongoing research is hence required to continue to develop and evaluate the appropriate outcome instruments in AIMSS.

Research questions should also address the time points most appropriate for measurement of outcomes. AIMSS have variable and fluctuating clinical progress (Hershman 2015b), and studies thus far have included variable assessment timing, making comparison and meta‐analysis challenging. Future research studies of systemic therapies should also include outcome measurements beyond the completion of the intervention to quantify if the effects of the intervention on AIMSS are sustained. Trials of supportive systemic therapies for AIMSS thus far have been predominantly of 6 months' duration or less and do not address pertinent research questions of establishing safe and effective long‐term therapies. Research challenges will be significant to design systemic therapy trials that continue for the entire duration of AI adjuvant therapy; in reality these may not be feasible. However, it is a research priority to design interventions of longer duration, as only then is it likely that knowledge gaps can be addressed – for instance, whether a systemic therapy for AIMSS can affect AI compliance.

Many of the trials in this review, 10/17 (Birrell 2009Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aKhan 2017Liu 2014Niravath 2019Rastelli 2011Shenouda 2019), were funded by industry sources, or had researchers with disclosed conflicts of interests with industry of varying relevance. In three trials, information was insufficient to make a full determination of relationships (Massimino 2011Peng 2018Rosati 2011). Several pharmaceutical company‐ or industry‐sponsored trials had design biases – the main example being four trials with inclusion criteria restricting entry to participants being treated with the sponsor's AI (Birrell 2009Khan 2017Liu 2014Rastelli 2011). Rosati 2011 was similarly restricted to the sponsor's AI as an entry criterion but AIMSS was not the primary outcome of the study. For‐profit funded trials have a higher chance of a favourable outcome than not‐for‐profit or mixed funding sponsored trials (Bhandari 2004Delgado 2017), although we acknowledge that research interventions in this review were designed to address a treatment‐emergent toxicity. Future research should preferentially access not‐for‐profit or mixed funding sources. This will require recognition and prioritisation of trials to address cancer treatment‐emergent toxicities by institutional and governmental bodies (as per the American Society of Clinical Oncology (ASCO) statement (Markham 2020)). 

Investigators designing trials of systemic therapy for AIMSS need to adequately acknowledge and account for the 'placebo effect'. Improvement due to the placebo effect has been noted in multiple trials in this review (Birrell 2009Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aPeng 2018Shenouda 2019), and has been previously documented in trials of other systemic therapies for cancer treatment emergent toxicities to be up to 30% to 50% (Cruciani 2012Loprinzi 2009Smith 2013). Better understanding of the aetiology of the placebo effect may potentially improve outcomes for women with AIMSS, particularly if the effect could be harnessed. Failure to account for the placebo effect will likely result in statistically underpowered trials that do not detect small differences between interventions, and thus may not answer the pertinent research questions (Henry 2015). The ethical implications of the placebo effect as prevention or treatment for AIMSS also requires further research.

Ideally, further research into systemic therapy interventions for AIMSS would involve translational research aiming to understand the pharmacology of, and adequately document and study all potential active ingredients of, the intervention. This is of particular importance with trials of CAM with many potentially active compounds (e.g. Chan 2017Massimino 2011Peng 2018Shenouda 2019Sordi 2019). Such research would allow any potentially effective traditional or CAM for AIMSS to then align with World Health Organization traditional and complementary medicine policies which highlight the role and importance of product regulation (WHO 2013). To avoid contamination, the choice of the placebo itself remains an important issue for research into systemic therapy for AIMSS. The placebo arm(s) of future trials should ideally contain no ingredients that potentially have activity (Wan 2013) in prevention or treatment of AIMSS in order to reduce research heterogeneity and uncertainty. As examples, in the trial by Hershman 2015a, possible concerns were raised about contamination from ingredients in the placebo which contained soybean oil and therefore potentially plant phyto‐oestrogens (Henry 2018Hershman 2015a). A trial of TCM had a placebo containing compounds that may have had uncertain activity, with the placebo granules were made from dextrin (95%) and Herba pogostemonis (5%) (Peng 2018). Herba pogostemonis may have anti‐inflammatory effects in in vitro models (Xian 2011). Careful reporting of all placebo and excipient details of any placebo will be important for any ongoing or further research into systemic therapy which includes a placebo arm. The challenges of matching the placebo to achieve blinding need to continue to be addressed in future research of systemic therapies in AIMSS, especially as most primary outcomes are PROS. Attention needs to be paid to matching all sensory aspects of the placebo to the investigational product (Wan 2013).

Future research should necessarily include rigorous monitoring for evidence of harm. Trials thus far have often included minimal long‐term adverse event monitoring. There has been conflicting evidence on the effect of supplement/antioxidant use on breast cancer recurrence and mortality in women with early breast cancer (Ambrosone 2020Greenlee 2012Poole 2013). Hence, monitoring safety and tolerability is thus of utmost importance for future trials of systemic therapy for AIMSS. However, the authors acknowledge the potential research challenges due to studies being potentially underpowered to detect possible small survival differences in studies of systemic therapies for prevention or treatment of AIMSS in women who are being treated with AI for early breast cancer.

Summary of findings

Open in table viewer
Summary of findings 1. Summary of findings table ‐ Systemic therapy compared to control for treating aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Systemic therapy compared to control for treating aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Patient or population: women with early breast cancer
Setting:
Intervention: systemic therapy
Comparison: control

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with control

Risk with systemic therapy

Change in pain from baseline to end of intervention
assessed with: Brief Pain Inventory (BPI) worst pain; BPI severity
follow‐up: 24 weeks

There were 2 studies (omega‐3 fatty acids, vitamin D) that showed a treatment effect with 95% CI that did not include a minimal clinically important difference (MCID) for BPI pain scale.

183
(2 RCTs)

⊕⊝⊝⊝
Very lowa,b,c

The evidence is very uncertain about the effect of systemic therapies on change in pain from baseline to end of intervention.

Change in grip strength from baseline to end of intervention
follow‐up: 24 weeks

85 per 1000

91 per 1000
(31 to 268)

RR 1.08
(0.37 to 3.17)

137
(1 RCT)

⊕⊕⊝⊝
Lowb,c

The evidence suggests that systemic therapies results in little to no difference in grip strength from baseline to end of intervention.

Safety of systemic therapies in AIMSS

1 study had a US Food and Drug Administration alert issued for the class of drug which the study drug belonged to (cyclo‐oxygenase‐2 inhibitors), but the study reported no serious adverse effects. No serious adverse events noted in any study.

344
(4 RCTs)

⊕⊝⊝⊝
Very lowc,d,e

The evidence is very uncertain about the effect of systemic therapies on safety of systemic therapies in AIMSS.

Effect on discontinuation of aromatase inhibitors (AI)
follow‐up: 24 weeks

39 per 1000

6 per 1000
(0 to 116)

RR 0.16
(0.01 to 2.99)

147
(1 RCT)

⊕⊕⊝⊝
Lowb,c

The evidence suggests that systemic therapies results in little to no difference in effect on discontinuation of AIs.

Effect on breast cancer‐specific quality of life (BCS‐QoL)
assessed with: Functional Assessment of Cancer Therapy – Breast (FACT‐B)
follow‐up: 24 weeks

1 study (omega‐3 fatty acids) showed a treatment effect with 95% CI that did include an MCID for this outcome measure.

44
(1 RCT)

⊕⊕⊝⊝
Lowa,c

The evidence suggests that systemic therapies results in little to no difference in effect on BCS‐QoL.

Health‐related quality of life (HRQoL) ‐ HRQoL: Total Functional Assessment of Cancer Therapy – General(FACT‐G) score
assessed with: Functional Assessment of Cancer Therapy – General (FACT‐G)
follow‐up: 24 weeks

A single study (omega 3 fatty acids) showed a treatment effect with 95% CI which did include a MCID for this outcome measure.

(1 RCT)

⊕⊕⊝⊝
Lowa,c

The evidence suggests that systemic therapies results in little to no difference in effect on HRQoL.

Incidence of AIMSS
follow‐up: range 24 weeks to 52 weeks

537 per 1000

440 per 1000
(338 to 569)

RR 0.82
(0.63 to 1.06)

240
(2 RCTs)

⊕⊝⊝⊝
Very lowb,c,d

The evidence is very uncertain about the effect of systemic therapies on change in Incidence of AIMSS.

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference; RR: risk ratio

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

See interactive version of this table: https://gdt.gradepro.org/presentations/#/isof/isof_question_revman_web_424110183232321857.

a High risk of reporting bias and attrition bias in single study. Downgraded one level.
b Downgraded one level for indirectness (restricted population dependent on vitamin D level).
c Downgraded one level for imprecision (small sample sizes).
d Downgraded one level for risk of bias (high risk of bias across multiple domains).
e High suspicion of publication bias (too few studies for funnel plot; one study in abstract form only).

Open in table viewer
Summary of findings 2. Summary of findings table ‐ Systemic therapy compared to control for preventing aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Systemic therapy compared to control for preventing aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Patient or population: health problem or population
Setting:
Intervention: Systemic therapy
Comparison: Control

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with Control

Risk with Systemic therapy

Change in pain from baseline to end of intervention
follow‐up: range 30 days to 6 months

4 studies showed a minimal clinically important difference (MCID) in pain scores that fell within the 95% CIs of the measured effect (calcitonin, bionic tiger bone, Yi Shen Jian Gu granules and vitamin D). 6 studies showed a treatment effect with 95% CIs that did not include an MCID (testosterone, vitamin D, duloxetine, omega‐3 fatty acids, emu oil, Cat's claw). Due to the variation in systemic therapies, including pharmacological, complementary and alternative medicines, the studies could not be combined for meta‐analysis.

1099
(10 RCTs)

⊕⊝⊝⊝
Very lowa,b,c,d

The evidence is very uncertain about the effect of systemic therapies on change in pain from baseline to the end of intervention in the treatment of AIMSS.

Change in stiffness from baseline to end of intervention

2 studies showed a minimal clinically important difference (MCID) in stiffness scores that fell within the 95% CIs of the measured effect (bionic tiger bone, Yi Shen Jian Gu granules). 2 studies that showed a treatment effect with 95% CIs that did not include an MCID (vitamin D, emu oil).

295
(4 RCTs)

⊕⊝⊝⊝
Very lowb,c,e,f

The evidence is very uncertain about the effect of systemic therapies on change in stiffness from baseline to the end of the intervention in the treatment of AIMSS.

Grip strength ‐ Vitamin D

There was a single study which did not show a MCID for grip strength which falls within the 95% CI for the measured effect (vitamin D).

(1 RCT)

⊕⊕⊝⊝
Lowc,f

The evidence is uncertain about the effect of systemic therapies on change in grip strength from baseline to the end of the intervention in the treatment of AIMSS 'little to no effect'

Effect on breast cancer‐specific quality of life (BCS‐QoL)

2 studies investigated the effect on BCS‐QoL (bionic tiger bone, Yi Shen Jian Gu granules). All subscales of the same quality of life tool utilised in both of these studies (Functional Assessment of Cancer Therapy – Breast (FACT‐B)) showed an MCID for this tool that falls within the 95% CIs of the measured effect.

147
(2 RCTs)

⊕⊝⊝⊝
Very lowa,g,h

The evidence is very uncertain about the effect of systemic therapies on effect on BCS‐QoL from baseline to the end of the intervention in the treatment of AIMSS.

Effect on health‐related quality of life (HRQoL)

2 studies investigated the effect of BCS‐QoL (bionic tiger bone, Yi Shen Jian Gu granules) using the Functional Assessment of Cancer Therapy – General (FACT‐G) tool. All subscales of this quality of life tool showed an MCID for this tool that fell within the 95% CIs of the measured effect. 1 study used the 36‐item Short Form (SF‐36) and showed most individual subscales in this outcome showing effect size that did not include MCID for this tool within the 95% CIs of the measured effect.

208
(3 RCTs)

⊕⊝⊝⊝
Very lowa,b,g,h

The evidence is very uncertain about the effect of systemic therapies on HRQoL from baseline to the end of the intervention in the treatment of AIMSS.

Safety of systemic therapies for the treatment of AIMSS

There were no grade 4/5 adverse events reported in any studies. 1 study investigating duloxetine reported significantly more all‐grade adverse events in the systemic therapy arm (78%) than the control arm (50%) (P < 0.001).

1250
(10 RCTs)

⊕⊝⊝⊝
Very lowa,c,i

The evidence is very uncertain about the safety data in the use of systemic therapies for the treatment of AIMSS.

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

See interactive version of this table: https://gdt.gradepro.org/presentations/#/isof/isof_question_revman_web_424113354545582340.

a Downgraded one level for risk of bias (high risk of bias across multiple domains).
b Downgraded one level for inconsistency (heterogeneity in interventions).
c Downgraded one level for imprecision (small sample sizes and unable to be combined for meta‐analysis).
d Downgraded one level for publication bias (funnel plot displayed asymmetry; multiple studies only written in abstract form).
e Downgraded one level for risk of bias (one study had high risk of both performance and detection bias).
f Downgraded one level for indirectness (one study with poorly defined AIMSS).
g Downgraded one level for indirectness (no criteria for an AIMSS in one study and exclusion of bisphosphonates, which are frequently utilised in the target population).
h Downgraded for imprecision (small sample size).
i Downgraded one level for publication bias (multiple studies published in abstract form only).

Background

Description of the condition

Despite advances in screening and treatment, breast cancer continues to significantly impact the global community. There was an estimated 1.67 million new cases diagnosed in 2012 making breast cancer the most common non‐skin cancer in women, and it is the fifth most common cause of cancer death globally (Ferlay 2012). In women in high‐income regions of the world, breast cancer is second to lung cancer as the leading cause of death, and in lower income regions, breast cancer remains the leading cause of death (Ferlay 2012). Eighty percent of breast cancer is hormone receptor (i.e. oestrogen receptor or progesterone receptor, or both)‐positive, which is often described as 'endocrine‐sensitive' breast cancer (Nadji 2005). Treatment of postmenopausal women with hormone receptor‐positive breast cancer with aromatase inhibitor (AI) medications is effective. When compared to treatment with another hormonal therapy, tamoxifen, five years of AI therapy in early breast cancer improves disease‐free‐survival (DFS) and breast cancer‐specific survival (BCSS) (EBCTCG 2015). An AI medication called exemestane, used in combination with ovarian suppression in women with higher risk premenopausal breast cancer has also shown an improvement in DFS compared with tamoxifen (Francis 2015Francis 2018). These results may see the adoption of AI therapy in a greater proportion of women with early breast cancer.

However, AIs are commonly associated with joint and muscular symptoms, commonly referred to as aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS) (Lintermans 2013). Approximately half of all women treated with AIs experience these musculoskeletal effects (Beckwee 2017), which can significantly impact on their quality of life. AIMSS usually presents as symmetrical pain or soreness in multiple joints within the first two to three months of initiating the AI (Burstein 2007). Women may also experience early‐morning stiffness and difficulty sleeping (Burstein 2007). Despite the survival advantage of AIs, between a quarter and a half of all women on this treatment choose to discontinue therapy (Chim 2013Henry 2012Kadakia 2016). AIMSS is the most common reason for women to discontinue AI therapy (Kwan 2017).

Description of the intervention

We aimed to investigate the impact of systemic therapies on the prevention or management of AIMSS. Systemic therapies range from prescription medications to dietary supplements such as vitamins. We also included complementary and alternative medicines (CAM) in our review. The National Centre for Complementary and Integrative Health defines CAM as "a group of diverse medical and health care systems, practices and products that are not presently considered to be part of conventional medicine" (NCCIH 2016).

How the intervention might work

There is a body of research investigating the mechanisms of action and the effectiveness of systemic therapies on other musculoskeletal conditions such as osteoarthritis, but quality evidence is still lacking for the optimal systemic therapy management of AIMSS. Examples of research into the management of other musculoskeletal conditions include reviews on glucosamine therapy for osteoarthritis (Towheed 2005), opioids for osteoarthritis of the knee or hip (Da Costa 2014), and muscle relaxants for pain management in rheumatoid arthritis (Richards 2012). Reviews such as these have enabled evidence‐based guidelines to be developed for the management of conditions such as osteoarthritis (NICE 2014). Systemic therapy approaches for AIMSS investigated by researchers have included interventions previously studied in osteoarthritis, rheumatological arthritis and chronic pain conditions (Hershman 2015b).

The exact cause of AIMSS remains unknown, but has been hypothesised to be related to multiple factors, including oestrogen deprivation, vitamin D insufficiency and the activation of molecules within the body that promote inflammation (Borrie 2017Hershman 2015b). Of these, oestrogen deprivation appears to be the key mediator. Oestrogen has an effect on both inflammation and the neural processing of a painful stimulus, which could result in heightened pain in the setting of oestrogen depletion (Felson 2005). Researchers have also identified an association between AIMSS and various genetic variations in women, which may result in women being more susceptible to experiencing musculoskeletal toxicity from AIs (Borrie 2017Lintermans 2016).

Risk factors that have been attributed to developing AIMSS include previous hormone replacement therapy (Sestak 2008), previous taxane chemotherapy (Crew 2007Lintermans 2014), younger age (Kanematsu 2011Lintermans 2014), fewer years since last menstrual period (Kanematsu 2011Mao 2011), body mass index less than 25 or greater than 30 kg/m2 (Beckwee 2017Crew 2007), severity of menopausal symptoms (Castel 2013), presence of joint‐related comorbidity at baseline (Castel 2013Lintermans 2014), stage two breast cancer (Beckwee 2017), and fear of recurrence (Lopez 2015). However, clinicopathological associations have been inconsistent across studies (Beckwee 2017), except for time since last menstrual period (Kanematsu 2011Mao 2009).

With only postulated causes of AIMSS, the interventions that have been investigated to date vary widely. Research interventions with systemic therapies have attempted to address various postulated causes of AIMSS, sometimes potentially via multiple complex and postulated pharmacological targets or pathways (or both).

Omega‐3 fatty acids (O3‐FA), as found in fish oil (particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have anti‐inflammatory properties (Hershman 2015aHershman 2015b), and have shown effect on pain and stiffness in inflammatory joint pain (Goldberg 2007). EPA and DHA inhibit conversion of arachidonic acid to prostaglandin and leukotrienes, reducing inflammation (Cleland 1988Sundrarjun 2004). O3‐FA supplements have thus been investigated in AIMSS (Hershman 2015aLustberg 2015).

Other anti‐inflammatory systemic therapies have been investigated, including a non‐steroidal anti‐inflammatory drug (NSAID), etoricoxib (Rosati 2011) and prednisolone (Kubo 2012). NSAIDS are utilised as part of the management strategy for osteoarthritis, with traditional NSAIDS inhibiting the cyclo‐oxygenase‐1 (COX‐1) enzyme (Song 2016). Etoricoxib is selective for the cyclo‐oxygenase‐2 (COX‐2) inducible form of cyclo‐oxygenase, potentially improving the central hyperalgesic action (Song 2016). Rosati 2011 examined the effect of AI on musculoskeletal events as a secondary endpoint in a trial where the primary outcome was to investigate whether etoricoxib or placebo improved DFS in addition to adjuvant anastrozole in early breast cancer. Similarly, glucosamine with chondroitin has been investigated in AIMSS (Greenlee 2013). Chondroitin with glucosamine slightly improves pain in osteoarthritis (Singh 2015), with multiple postulated mechanisms of action in preclinical research, including anti‐inflammatory effects in joints and glucosamine increasing the synthesis of proteoglycans in articular cartilage (Greenlee 2013Reginster 2012).

Oestrogen is thought to play a role in the modulation of nociceptive pain pathways (Felson 2005). With a higher incidence of autoimmune disease in women than men, hormonally active androgens are believed to be anti‐inflammatory and oestrogens are pro‐inflammatory (Schmidt 2006). The balance between oestrogen and androgen mediated by the AI enzyme is thought important in joint health (Schmidt 2006). Hence, testosterone has been investigated as it was hypothesised that this could improve AIMSS (Birrell 2009Cathcart‐Rake 2020).

Duloxetine, as investigated by Henry 2018, is a serotonin‐noradrenaline reuptake inhibitor (SNRI) with antidepressant and analgesic properties (Bellingham 2010). The analgesic effect of duloxetine is postulated to be achieved by augmentation of the tone of the descending pain inhibitory pathways from the central nervous system (Bellingham 2010). Duloxetine is indicated for treatment of fibromyalgia, diabetic peripheral neuropathic pain and chronic musculoskeletal pain (Lilly 2020), and has been investigated for management of AIMSS (Henry 2018).

Low vitamin D levels have been associated with higher levels of joint pain in postmenopausal women (Chlebowski 2011). Vitamin D deficiency has been found to be prevalent in women with breast cancer undergoing adjuvant chemotherapy (Crew 2009). Oestrogen activates both vitamin D and it's receptor (Gallagher 1980), with AIs causing a subsequent functional deficiency of vitamin D. As vitamin D deficiency and insufficiency can contribute to musculoskeletal symptoms (Hershman 2015bHolick 2007), vitamin D supplementation for prevention and management of AIMSS has thus been investigated in randomised controlled trials (RCTs) (Khan 2017Niravath 2019Rastelli 2011Shapiro 2016).

Calcitonin acts with parathyroid hormone and 1,25 dihydroxycholecalciferol to regulate short‐term calcium homeostasis, mediated in part by inhibiting bone resorption by an effect on osteoclasts (Chesnut 2008). Salmon calcitonin has thus been used as an anti‐resorptive therapy in osteoporosis and other bone‐associated pain conditions (Chesnut 2008), and has been studied by Liu 2014.

Use of CAM is anecdotally widely noted by care providers and, therefore, reviewing the evidence of effect and toxicities is important (Boon 2000Kremser 2008). Women with AIMSS may be unwilling to take medications with side effects to treat side effects, and hence may seek CAM as an alternative potential management strategy (Hershman 2015b).

Traditional Chinese medicine (TCM) has been widely used in care of people with cancer in China (Li 2013), and Chinese patients have sought out TCM practitioners due to lack of effective therapies for AIMSS (Peng 2018). Investigators have trialled various TCM for AIMSS (bionic tiger bone, Li 2017Yi ShenJian Gu (YSJG) granules, Peng 2018; Blue Citrus capsules, Massimino 2011).

Tiger bone, whose "main ingredients are calcium and collagen", has been utilised in TCM for proposed strengthening of muscles and bones (Li 2017). However, as tigers are protected animals, "scientists adopted bionic research method to develop bionic tiger bone powder, which has similar ingredients to natural tiger bone"(Li 2017). "Bionic TB [tiger bone] powder has more than 20 kinds of amino‐acid and microelements essential to human. Besides, calcium to phosphorus ratio of TB makes it suitable for body to absorb, whereas it also contains various organic components, such as collagen, analgesic peptide, bone morphogenetic protein, bone growth factors, and polyose" (Li 2017). Li 2017 investigated bionic tiger bone powder for management of AIMSS because of proposed anti‐inflammatory, analgesic and anti‐osteoporotic effects. 

YSJG granules, patented by the Beijing Hospital of Traditional Chinese Medicine, are composed of an empiric formula of 12 herbs, including Radix rehmanniae Preparata (ShuDiHuang), Fructus Corni (ShanZhuYu), Semen cuscutae (TuSiZi), Radix Achyranthis Bidentatae (NiuXi), Rhizoma cyperi (XiangFu), Radix Angelicae Sinensis (Dang‐Gui), Poria (FuLing), Radix Paeoniae Alba (BaiShao), Rhizoma chuanxiong (ChuanXiong), Rhizoma corydalis (YanHuSuo), Phryma leptostachya (TouGuCao) and Caulis trachelospermi (LuoShiTeng) (Peng 2014). YSGJ granules have been used in TCM treatment of musculoskeletal symptoms of postmenopausal women with osteoporosis and arthrosis based on TCM principles, and have been investigated for management of AIMSS (Peng 2018). "Because the formula for YSJG is currently being patented, the ingredients in YSJG cannot be published at this time" (Peng 2014), and therefore it is not possible to postulate on the proposed action of this intervention in AIMSS due to the large number and complexity of the ingredients, and the uncertainty arising from the lack of information on the ingredients with the pending patent.

Similarly, the TCM Blue Citrus has been investigated for AIMSS, due to anecdotal reports of improvements of AIMSS (Massimino 2011). According to the National Cancer Institute drug dictionary (NCI 2021), Blue Citrus is "An oral capsule formulation of a traditional Chinese herbal medicine with potential analgesic activity. In addition to other herbs, seeds and fruits, blue citrus‐based herbal capsule contains the Chinese herb blue citrus (qing pi), which is produced from the dried immature green peel of the tangerine Citrus reticulata Blanco. Blue citrus contains large amounts of limonene, citral and synephrine, which may attribute to its analgesic activity. However, due to the complexity of its chemical components, the exact mechanism of action of this agent remains to be determined".

Emu oil, obtained from the fat of emus, a large bird indigenous to the Australian continent, has been used by Australian Aboriginal people as a traditional medicine (Turner 2015Whitehouse 1998). The liquid fat was applied topically by Aboriginal people for musculoskeletal disorders and to assist wound healing (Turner 2015Whitehouse 1998) with transdermal absorption considered to be anti‐inflammatory in rat models of arthritis (Whitehouse 1998). There were anecdotal evidence of effectiveness in osteoarthritis (Power 2004), and hence topical application for treatment of AIMSS has been investigated (Chan 2017). Emu oil "contains several fatty acids (myristic, palmitic, stearic, palmitoleic, oleic, linoleic, and linolenic), and it is not known which specific component provides symptomatic relief" (Chan 2017).

Uncaria tomentosa (Cat's claw) is a plant species found in the Amazon (Sordi 2019), and has been used for centuries by the Incas as a traditional medicine to treat arthritis, arthrosis and other inflammatory conditions. The active metabolites (including pentacyclic and indole oxindole alkaloids and quinovic acid glycosides) have reported antioxidant, immunomodulatory, antineoplastic, anti‐inflammatory and antiviral activities (Aguilar 2002Aquino 1989Sordi 2019). Sordi 2019 investigated the use of the dry extract of Cat's claw for women with AIMSS.

Cherries contain many bioactive compounds with reported health benefits (Kelley 2018). Flavonoids and anthocyanins in tart cherry extract reportedly reduce inflammation, and some clinical trials suggest improvement in joint pain in osteoarthritis and gout (Kelley 2018Shenouda 2019). Shenouda 2019 investigated tart cherry extract in AIMSS.

As the cause of AIMSS remains unknown, it is important to further review the evidence for systemic prevention and management of musculoskeletal symptoms specific to AIMSS, rather than extrapolating evidence from other non‐AIMSS musculoskeletal conditions.

Why it is important to do this review

The high prevalence of musculoskeletal symptoms secondary to AIs can result in detrimental outcomes for patients. Several studies have shown poor participant adherence to AI therapy (Brier 2017Hadji 2014Henry 2012Hershman 2011Partridge 2008Presant 2007). This is particularly concerning in the setting of early breast cancer, where non‐compliance with hormonal therapies such as AIs, used in the curative setting, have been shown to be detrimental to patient survival (Hershman 2011). Given the prevalence of breast cancer in the community, the implications of the potential impact of non‐adherence on breast cancer outcomes are important. 

While there has been a few reviews to assist clinicians with the management of AIMSS (Roberts 2017Yang 2017), there have been no reviews dedicated solely to the systemic management of AIMSS. Several of the authors from this review have conducted a separate Cochrane Review entitled "Exercise therapies for preventing or treating aromatase inhibitor‐induced musculoskeletal symptoms in early breast cancer" (Roberts 2020). There has been a meta‐analysis on acupuncture for AIMSS (Chen 2017). Identifying current evidence and potential areas for further research in this field is required for the optimisation of management of women with endocrine‐sensitive breast cancer.

Objectives

To assess the effects of systemic therapies on the prevention or management of AIMSS in women with stage I to III hormone receptor‐positive breast cancer.

Methods

Criteria for considering studies for this review

Types of studies

RCTs examining the prevention or management of AIMSS in women with stage I to III hormone receptor‐positive breast cancer. AIMSS was defined by the study authors of each trial. We excluded animal and in vitro studies. We considered studies in all languages for inclusion.

Types of participants

Women with stage I to III oestrogen‐receptor (ER) or progesterone‐receptor (PR) (or both)‐positive breast cancer, being treated adjuvantly with AIs.

Types of interventions

Intervention: all systemic therapy interventions, including pharmacological therapies, dietary supplements, and CAM. We excluded acupuncture administered as a sole intervention as it was not considered a systemic therapy.

Comparator: all comparator groups were allowed, including placebo or standard of care (or both) with analgesia alone.

Types of outcome measures

Primary outcomes

  • Symptoms of AIMSS (specifically pain, stiffness and grip strength) from baseline. These were preferably assessed by validated questionnaires, such as the Visual Analogue Scale (VAS), Brief Pain Inventory (BPI), Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), Functional Assessment of Cancer Therapy – General (FACT‐G), Medical Outcome Study Short Form 36 (SF‐36) and the Modified Score for the Assessment of Chronic Rheumatoid Affections of the Hands (M‐SACRAH). These questionnaires identified participant symptoms including, but not limited to, pain (e.g. severity of pain, change in pain scores), physical function (e.g. using stairs, sitting up, performing domestic duties) and stiffness. These outcomes were assessed for both the impact on AIMSS immediately after the intervention, and in the long‐term, where available.

  • Safety of the intervention, including adverse effects. All grade of adverse events were considered from each intervention.

Secondary outcomes

  • Persistence and adherence of participants continuing to take their AI medication due to the intervention.

  • Patient health‐related quality of life (HRQoL), also preferably assessed by validated patient‐reported outcome (PRO) questionnaires. Where available, this was analysed for the impact on participants during the intervention, immediately after the intervention and in the long term.

  • Incidence of AIMSS.

  • Breast cancer‐specific survival (BCSS).

  • Overall survival (OS).

Search methods for identification of studies

Electronic searches

The Information Specialist (KR) designed and conducted systematic searches in the selected databases and trial registries without language, publication year or publication status restrictions on 10 September 2020. Cochrane Breast Cancer's Information Specialist conducted the search of the Specialised Register on 27 March 2021. Where appropriate, the search strategies also included adaptations of the Highly Sensitive Search Strategy designed by the Cochrane Collaboration (Lefebvre 2011), and the search filter for CINAHL (EBSCO) created by Mark Clowes at SIGN for identifying RCTs and controlled clinical trials.

We searched the following databases and trial registries up to 10 September 2020.

The following was searched up to 27 March 2021.

  • The Cochrane Breast Cancer Group's (CBCG's) Specialised Register. Trials with the keywords “breast cancer” and related terms, “aromatase inhibitors”, “exemestane”, “anastrozole”, “letrozole”, were extracted and considered for inclusion in the review.

Searching other resources

Adverse effects

We did not perform a separate search for adverse effects of interventions used for the treatment of AIMSS. We considered adverse effects in included studies only.

Searching within other reviews

We scanned the reference lists of existing systematic reviews relevant to this systematic review that the search identified, or reference/citation lists, for additional trials and to check robustness of search strategy.

Bibliographic searching

We identified further studies from reference and citation lists of identified relevant trials or reviews. We obtained a copy of the full article for each reference reporting a potentially eligible trial for those studies selected for full‐text review. Where this was not possible, such as with the inclusion of conference abstracts, we contacted the authors for additional information and reviewed additional information from clinical trial databases.

Grey searching

The San Antonio Breast Cancer Symposium (SABCS) and American Society of Clinical Oncology (ASCO) conference proceedings are included in the Embase database and, therefore, were not searched separately. See Electronic searches for searching of clinical trials databases.

Data collection and analysis

Selection of studies

Two review authors (KER and NW) screened and retrieved abstracts from the literature search to assess whether they met the specified selection criteria. Subsequently, two review authors (KER and NW) reviewed full‐texts of all remaining studies, ensuring they still met the selection criteria. Any disagreements on study selection were resolved by a separate review author (IA or SC). The selection process was recorded in a PRISMA flow diagram (Figure 1). We documented the reason for excluding studies in the Characteristics of excluded studies table. Clinical trials that were terminated are detailed as Excluded studies, and those that are ongoing and awaiting publication, are included in the Studies awaiting classification table.


Study flow diagram.

Study flow diagram.

No studies required translation from other languages.

Data extraction and management

Three review authors (IA, SC, NW) used a standardised data extraction form and collected the following information. A fourth review author (KER) resolved disagreements.

Characteristics of the study

  • Study sponsors and author affiliations.

  • Study funding.

  • Methods, including study design, method of sequence generation, allocation concealment, blinding of outcome, participant attrition and exclusions, and selective outcome reporting.

  • Full‐text available versus abstract only.

Characteristics of the study population

  • Country of enrolment.

  • Inclusion/exclusion criteria.

  • Study definition of AIMSS.

  • Number of participants in each treatment arm.

  • Mean and range of participant ages.

  • Number of participants aged less than 40 years; aged 40 to 60 years; and aged greater than 60 years.

  • Menopausal status (i.e. requirement for biochemical ovarian suppression versus no requirement).

  • Previous use of taxane (yes/no).

  • Type of AI prescribed, and time since commencement of AI.

Characteristics of the intervention

  • Description of the intervention.

  • Details of control group.

  • Ingredients of placebo, if applicable.

  • Compliance with intervention.

  • Safety.

Characteristics of the outcomes

  • Scoring systems used (and documentation of PRO versus investigator‐reported outcomes).

  • Outcomes of pain, stiffness, functioning and HRQoL.

  • Timing of outcome data collection, including length of time between intervention and last collected outcome measurement.

  • Follow‐up period.

Two review authors (IA and NW) entered the data into Review Manager Web (Review Manager 2014). Where there was more than one publication for a study, the data was extracted from all publications as required, but the most recent version of the study was considered the primary reference. Where possible, records relating to the same study were combined under an overall trial name or study.

Assessment of risk of bias in included studies

Two review authors (of NW, SC or IA) independently assessed risk of bias for all RCTs using the Cochrane's risk of bias assessment tool (Higgins 2011a). A third review author (KR) resolved any disagreements. This tool included seven specific domains: random sequence generation; allocation concealment; blinding of outcome assessment; blinding of participant and personnel; incomplete outcome data; selective reporting and other sources of bias. Each risk of bias domain was assessed as high risk, low risk or unclear risk. For risk of bias for cross‐over RCTs, we referred to guidance in Chapter 23 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011b).

Where there was incomplete reporting of a study's conduct, we attempted to contact the study authors to clarify such uncertainties. Risk of bias tables for each study were presented in the Characteristics of included studies table. Risk of bias domain‐level judgements were presented in forest plots, and a summary table listing the risk of bias judgement for all studies is presented in Figure 2 and Figure 3


Risk of bias graph: review authors' judgements about each risk of bias domain, for each included trial. 

Risk of bias graph: review authors' judgements about each risk of bias domain, for each included trial. 


Risk of bias summary: review authors' judgements about each risk of bias item, presented as percentages across all included trials.

Risk of bias summary: review authors' judgements about each risk of bias item, presented as percentages across all included trials.

Measures of treatment effect

It was expected that studies would use a variety of different tools to measure the outcomes of interest (pain, stiffness, grip strength and HRQoL) and mostly report continuous outcomes. Because of the clinical heterogeneity of the included studies, a random‐effects model was considered. 

The measurement of the treatment effect was performed using a mean difference (MD) analysis. This is different from our protocol, which we had intended to measure the treatment effect by performing a standardised mean difference (SMD) analysis and the random‐effects model to combine data from different scoring systems measuring the same outcome of interest. But, due to inconsistent reporting of standard deviations (SD), with some studies reporting only 'end‐of‐treatment' SD and others reporting only 'change score' SD, we were unable to combine these results for the calculation of SMD. Multiple studies did not report SD of change scores and only provided SD from baseline or end‐of‐treatment SD. If change score means with SD were not available, we reported end‐of‐treatment means and SD for both groups. As discussed in Chapter 10 of the Cochrane Handbook for Systematic Reviews of Interventions, "in a randomized study, MD based on changes from baseline can usually be assumed to be addressing exactly the same underlying intervention effects as analyses based on post‐intervention measurements" (Deeks 2021). If end‐of‐treatment means and SD were used, this was highlighted in the analysis.

If SDs could not be obtained for studies, imputing the SD was attempted as per Chapter 6 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011a). In studies where only CIs were available, we used the following formula to determine the SD: SD = √n × (upper limit 95% CI − lower limit 95% CI)/2 × T value calculated by the T distribution), where n was the sample size and CI was the confidence interval. We estimated appropriate T values for smaller sample sizes using the TINV function (TINV(1‐0.95,n‐1)) in Excel. If the standard error (SE) was reported instead of SD, then SD was calculated with the following formula, SE = SD × √n, as per guidance from Chapter 6 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2021). One study reported the use of mean and SD, but the data were more consistent with the reporting of median values and interquartile ranges (IQR) (Liu 2014). Reporting the median is often an indicator that the data are skewed, so it should be incorporated into a meta‐analysis with caution (Higgins 2021). To calculate SD from IQR, we used the following formula: SD = (q3‐q1)/1.35, where q3 is quartile 3 and q1 is quartile 1 (Higgins 2021). 

If studies reported dichotomous outcomes (e.g. incidence rates), the treatment outcome was measured by the risk ratio (RR), in combination with a 95% CI. We reported the ratio of treatment effect so that RR less than 1.0 favoured the intervention group for relief of AIMSS symptoms and RR greater than 1.0 favoured the control group.

The most appropriate time point across all studies for each outcome depended on the availability of suitable data. As recommended in Section 16.7.1 of the Cochrane Handbook for Systematic Reviews of Interventions, we avoided multiple testing of the treatment effect at each time point (Higgins 2011c). We collected data on the treatment effect at baseline, immediately following the intervention and long‐term data, if available. The only study that we did not use end‐of‐treatment outcomes was Rastelli 2011. Instead, we utilised outcome data at two months to eliminate any differences in the stratums in the intervention arm, which received differing doses of intervention between two and four months of the study. 

Unit of analysis issues

There were two studies that would have created unit of analysis issues, including one study with a cross‐over trial design (Massimino 2011), and one study with multiple treatment arms (Birrell 2009). However, both studies were only published in abstract form without pursuing full publication. We were unable to obtain information or data from either of the study authors by correspondence, and, therefore, did not have adequate data to analyse the results. Our planned approach for unit of analysis issues that were not utilised in this review can be found in our protocol (Roberts 2018). 

Dealing with missing data

In the case of missing data, we attempted to contact the study authors to source additional information through clinical trial registries or data repositories. If the required data were still not available, we contacted original investigators via email and gave three weeks to reply to the request. If the corresponding authors did not reply, we attempted further contact with the original investigators and either the first or last author of each paper (if not the primary corresponding author). 

Assessment of heterogeneity

We assessed clinical heterogeneity using the I2 statistic, Chi2 test and visual inspection of forest plots, as recommended in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011a). An I2 statistic of 30% to 60% may represent moderate heterogeneity, a result of 50% to 90% may represent substantial heterogeneity and a result of 75% to 100% may represent considerable heterogeneity (Higgins 2011a). The importance of the I2 statistic depended on the magnitude and direction of effects and the strength of evidence for heterogeneity. For the Chi2 test, we used P < 0.1 to indicate significant heterogeneity. We planned to use both a random‐effects model and a fixed‐effect model for analysis of results of meta‐analysis, but there were only two studies combined for meta‐analysis of one outcome, and there was no evidence of statistical heterogeneity; therefore, fixed‐effect and random‐effects models did not need to be compared.

Assessment of reporting biases

We included one funnel plot in the assessment of reporting biases for the outcome with the largest number of studies. We could not undertake any further assessments due to the small number of studies contributing data to each outcome (fewer than 10 studies). 

Data synthesis

We used Review Manager Web to perform statistical analysis (Review Manager Web 2021). We assessed analyses for clinical heterogeneity (see Assessment of heterogeneity). We performed a fixed‐effect meta‐analysis using the inverse variance method to combine data results for one outcome where at least two studies were appropriate to be combined for meta‐analysis. We reported the meta‐analysis using a forest plot and in the summary of findings table.

There needed to be sufficient data available for meta‐analysis between studies. If there were insufficient outcome data, we attempted to contact authors for additional data. If there were insufficient studies, or where the data could not be combined due to insufficient data for comparison, we presented the findings in a narrative manner.

To enable synthesis of our outcomes where meta‐analysis could not be done for the majority of outcomes, we applied the vote‐counting method as detailed by McKenzie 2021. We used vote‐counting by reporting continuous outcomes as the number of studies that showed a treatment effect that included a clinically significant benefit within the CIs of the MD between studies as determined by reported minimal clinically importance difference (MCID) for that outcome scale, compared to the number of studies that did not include an MCID within the CIs of the treatment effect. The MCID is a change in score that has been determined to represent a meaningful change in quality of life.

For binary outcomes, we reported RR as per our initial protocol. There were no trials with binary outcomes that reported zero events in both arms.

Subgroup analysis and investigation of heterogeneity

We did not undertake any subgroup analyses, as there were insufficient studies and participants to undertake any meaningful subgroup analysis within this review. Our planned subgroup analyses can be found in our protocol (Roberts 2018). 

Sensitivity analysis

There were not enough studies of each intervention in our review to undertake meaningful sensitivity analyses. Our planned sensitivity analyses can be found in our protocol (Roberts 2018).

Summary of findings and assessment of the certainty of the evidence

We used GRADEpro GDT software to present summary of findings tables to illustrate the main outcomes and grade the certainty of the evidence. We assessed the level of evidence using the five GRADE considerations (study limitations, consistency of effect, imprecision, indirectness and publication bias) as per the Cochrane Handbook of Systematic Reviews of Interventions (Schünemann 2021). At least two review authors (KR, IA) independently assessed the evidence using the GRADE technique, and a third review author (NW) resolved any disputes.

Main outcomes in the summary of findings tables: prevention studies

  • Change in pain from baseline to end of intervention (symptoms of AIMSS)

  • Adverse effects secondary to the intervention (safety data)

  • Change in grip strength from baseline to end of intervention (symptoms of AIMSS)

  • Effect on discontinuation of AIs

  • Effect on BCS‐QoL

  • Effect on HRQoL

  • Change in incidence of AIMSS.

Main outcomes in the summary of findings table: treatment studies

  • Change in pain from baseline to end of intervention (symptoms of AIMSS)

  • Change in stiffness from baseline to end of intervention (symptoms of AIMSS)

  • Overall change in grip strength (symptoms of AIMSS)

  • Adverse effects secondary to the intervention (safety data)

  • Effect on BCS‐QoL

  • Effect on HRQoL

Results

Description of studies

Results of the search

The search retrieved 3693 records from four databases (3350 records; see Appendix 1Appendix 2Appendix 3Appendix 4 in Electronic searches), the Specialised Register (307 records), clinical trial registries and reference lists of included studies (36 records). Once duplicates were removed, there were 2919 records. We excluded 2861 records during title and abstract screening, and obtained the full text (where possible) of the remaining 58 records. At full‐text review, we excluded 10 studies (see Characteristics of excluded studies table). We identified four ongoing studies (see Characteristics of ongoing studies table).

We included 17 studies reported in 44 references (see Characteristics of included studies table) and four ongoing studies (see Characteristics of ongoing studies table). The full details of our screening process is detailed in the study flow diagram (Figure 1).

Included studies

The review included 17 studies (see Characteristics of included studies table). 

Thirteen studies investigated the treatment of AIMSS (Birrell 2009Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aLi 2017Liu 2014Massimino 2011Peng 2018Rastelli 2011Shapiro 2016Shenouda 2019Sordi 2019), and all of these studies investigated at least one symptom of AIMSS as a primary outcome. 

Four studies investigated the prevention of AIMSS (Khan 2017Lustberg 2018Niravath 2019Rosati 2011). One of these studies investigated the five‐year event‐free survival (EFS) as a result of adding adjuvant etoricoxib versus placebo to adjuvant anastrozole, and examined the musculoskeletal events as a secondary outcome (Rosati 2011); another trial investigated adherence and tolerability as a primary outcome, and investigated pain as a secondary outcome (Lustberg 2018); the other two studies investigated prevention of AIMSS as a primary outcome (Khan 2017Niravath 2019).

Ten studies enrolled participants in the USA (Cathcart‐Rake 2020Henry 2018Hershman 2015aKhan 2017Lustberg 2018Massimino 2011Niravath 2019Rastelli 2011Shapiro 2016Shenouda 2019), three studies in China (Li 2017Liu 2014Peng 2018), two in Australia (Birrell 2009Chan 2017), and one each in Italy (Rosati 2011) and Brazil (Sordi 2019). One trial used a multi‐arm study design (Birrell 2009), and another trial employed a cross‐over design (Massimino 2011). 

Thirteen studies were published as full texts (Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aKhan 2017Li 2017Liu 2014Lustberg 2018Niravath 2019Peng 2018Rastelli 2011Shapiro 2016Sordi 2019), whereas four studies were published as an abstract or in poster form only (Birrell 2009Massimino 2011Rosati 2011Shenouda 2019). Author correspondence resulted in additional data information from four studies (Chan 2017Khan 2017Niravath 2019Sordi 2019). 

Population

This review included 2034 randomised participants in 17 studies. The sample sizes ranged from 37 to 299 participants. 

Participant age ranges were from 27 to 83 years. Eight studies reported the mean age of participants, which ranged from 59 to 61.5 years (Cathcart‐Rake 2020Khan 2017Liu 2014Lustberg 2018Peng 2018Rastelli 2011Shapiro 2016Sordi 2019). Six studies reported the median age of the participants, which ranged from 56.9 to 64 years (Chan 2017Henry 2018Hershman 2015aLi 2017Niravath 2019Rosati 2011). Nine studies reported age ranges (Chan 2017Henry 2018Khan 2017Li 2017Lustberg 2018Niravath 2019Peng 2018Rosati 2011Sordi 2019). Three studies reported only in abstract/poster format provided no participant baseline characteristics data (Birrell 2009Massimino 2011Shenouda 2019). 

Most participants were already receiving AI treatment at enrolment into the studies, except for the three prevention studies, which were investigating women commencing AI (Khan 2017Niravath 2019Rosati 2011). The prevention study by Lustberg 2018 included women with short duration (less than 21 days) of AI exposure. The treatment study by Li 2017 included women with a duration of AI treatment of less than one month, despite being designed to investigate the treatment of AIMSS. Six treatment studies reported the duration of AI therapy at baseline, with mean durations ranging from 47.9 weeks to 20.6 months (Henry 2018Rastelli 2011Shapiro 2016Sordi 2019), and median durations reported as 1.2 years (Hershman 2015a) and 13.3 months (emu oil group) to 16.9 months (placebo group; Chan 2017). Inclusion criteria of note related to two of the vitamin D studies that required participants to have specific serum 25‐hydroxyvitamin D (25‐OHD) levels at baseline (Khan 2017Rastelli 2011), particularly 25‐OHD levels of 40 ng/mL or less for Khan 2017 and between 10 ng/mL and 29 ng/mL for Rastelli 2011. Mean baseline 25‐OHD levels in the studies investigating vitamin D supplementation ranged from 22.5 ng/mL to 36.6 ng/mL (Khan 2017Niravath 2019Rastelli 2011Shapiro 2016).

Several studies excluded women with potentially confounding comorbid musculoskeletal conditions such as rheumatoid arthritis, fibromyalgia and connective tissue disorders (Chan 2017Henry 2018Li 2017Lustberg 2018Massimino 2011Peng 2018Rastelli 2011Shapiro 2016Sordi 2019), or specifically the use of COX‐2 inhibitors for arthritis (Rosati 2011). All studies excluded women with metastatic disease; however, one woman with metastatic breast cancer was randomised and included in the study by Liu 2014. Inclusion criteria for Lustberg 2018 was stage I to III breast cancer; however, one women with ductal carcinoma in situ (DCIS) only was randomised; however, DCIS was allowed in the inclusion criteria in the study by Sordi 2019 but only one participant with DCIS was enrolled.

Definition of aromatase inhibitor‐induced musculoskeletal symptoms 

Studies that included participants with AIMSS at baseline varied in their definitions of AIMSS. Most studies specified the requirement of arthralgia/myalgias or musculoskeletal symptoms to be related to the AI as an inclusion criterion, although the specific definition of AIMSS and the determination of the relationship to AI therapy varied markedly (Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aLi 2017Liu 2014Peng 2018Rastelli 2011Shapiro 2016Shenouda 2019Sordi 2019). Several of these studies specified a minimum pain score to qualify for inclusion (Cathcart‐Rake 2020Henry 2018Hershman 2015aPeng 2018Shapiro 2016Sordi 2019). Two other studies included women experiencing any joint symptoms while taking an AI (Birrell 2009Massimino 2011), and one of these stipulated a minimum pain score to qualify for inclusion (Birrell 2009); however, both studies were reported in abstract form with minimal details. 

Interventions

The interventions investigated for systemic therapy for AIMSS varied widely. These were:

Thirteen of the 17 studies were placebo controlled (Birrell 2009Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aKhan 2017Lustberg 2018Massimino 2011Peng 2018Rastelli 2011Rosati 2011Shenouda 2019Sordi 2019). Seven studies described the placebo, in varying detail (Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aLustberg 2018Peng 2018Sordi 2019). The control arm of the other studies included calcium carbonate daily orally (Li 2017), oral caltrate D 600 mg/day (Liu 2014), oral vitamin D3 800 IU daily (Niravath 2019), and oral vitamin D3 600 IU daily (Shapiro 2016). The duration of the intervention varied, from four weeks to two years. 

The duration of follow‐up varied between four weeks and five years.

Excluded studies

The reasons for excluding studies are listed in the Characteristics of excluded studies table. Ten studies were excluded from the analysis. Most studies were not RCTs (four studies) or the studies were withdrawn/terminated early (three studies). One study had an incorrect participant population (women with chemotherapy‐induced arthralgia) and one had an incorrect intervention (local rather than systemic treatment).

Studies awaiting classification

There are no studies awaiting classification.

Ongoing studies

There are four ongoing studies (NCT02831582NCT03865992NCT04205786UMIN000027481). See Characteristics of ongoing studies table.

Risk of bias in included studies

Details of the risk of bias are available in the risk of bias tables in the Characteristics of included studies table. 

We requested further information to clarify unclear risk of bias from 11 studies, which authors of four studies provided (Chan 2017Khan 2017Niravath 2019Sordi 2019). We also requested further information from another five studies to clarify potential bias including those at high risk of bias from selective reporting; however, we received no responses. The risk of bias summary can be viewed in Figure 2 and risk of bias graph in Figure 3

Allocation

Random sequence generation and allocation concealment

We judged six studies at unclear risk of selection bias because there was insufficient information to permit judgement about the adequacy of methods of random sequence generation or allocation concealment (Birrell 2009Liu 2014Massimino 2011Rosati 2011Shenouda 2019Sordi 2019). Four of these studies were published as abstract alone or in poster format and there was no further information (Birrell 2009Massimino 2011Rosati 2011Shenouda 2019). One study had insufficient information available in the publication to permit judgement and no further information could be obtained by author correspondence (Liu 2014). One study had insufficient information available to permit judgement on methods of random sequence generation or allocation concealment, although author correspondence did provide additional information for this study (Sordi 2019).

Eleven studies were at low risk of selection bias for both methods of random sequence generation and allocation concealment (Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aKhan 2017Li 2017Lustberg 2018Niravath 2019Peng 2018Rastelli 2011Shapiro 2016). These 11 studies all described in sufficient detail methods to generate randomisation sequences that produced comparable groups, and adequate methods to conceal allocation.

Blinding

Blinding of participants and personnel

We judged three studies at high risk of performance bias (Henry 2018Li 2017Niravath 2019). One study was not blinded, with participants randomised to receive either oral vitamin D3 at 50,000 International Units (IU) per week for 12 weeks followed by 2000 IU daily for 40 weeks, or vitamin D3 at 800 IU daily for 52 weeks (Niravath 2019). A second study had differences in the dosing between the intervention (bionic tiger bone) and the control which could foreseeably have led to unblinding of participants and personnel (Li 2017). The third study examining duloxetine versus placebo found that more participants in the duloxetine group experienced adverse effects (78% with duloxetine versus 50% with placebo; Henry 2018). More participants in the duloxetine group compared with the placebo arm believed they were receiving duloxetine (79% with duloxetine versus 50% with placebo; P < 0.001). Likely due to adverse events experienced, it was foreseeable that participants and personnel were at risk of unblinding.

Five studies were at unclear risk of performance bias due to insufficient information available to permit judgement on measures used to blind participants and personnel (Birrell 2009Liu 2014Massimino 2011Rosati 2011Shenouda 2019). We judged nine studies at low risk of performance bias due to effective blinding of participants and personnel (Cathcart‐Rake 2020Chan 2017Hershman 2015aKhan 2017Lustberg 2018Peng 2018Rastelli 2011Shapiro 2016Sordi 2019).

Blinding of outcome assessment

Most outcomes were PROs. We judged three studies at high risk of detection bias (Henry 2018Li 2017Niravath 2019). The primary outcomes for all three of these studies were PROs where participants were the outcome assessors. One of these studies had lack of blinding with outcome assessors, that is, participants, having knowledge of the assigned intervention (Niravath 2019). In two other studies, it was highly likely that participants, and therefore the outcome assessors, had potential knowledge of the assigned intervention (Henry 2018Li 2017). Five studies were at unclear risk of detection bias (Birrell 2009Liu 2014Massimino 2011Rosati 2011Shenouda 2019). There was insufficient information to permit judgement on measures used to blind the participants who were the outcome assessors in four of these studies (Birrell 2009Liu 2014Massimino 2011Shenouda 2019), and insufficient information on blinding procedures related to personnel who were the outcome assessors in the remaining study (Rosati 2011). We judged nine studies at low risk of detection bias due to effective blinding of outcome assessors (Cathcart‐Rake 2020Chan 2017Hershman 2015aKhan 2017Lustberg 2018Peng 2018Rastelli 2011Shapiro 2016Sordi 2019).

Incomplete outcome data

We assessed seven studies at high risk of attrition bias (Cathcart‐Rake 2020Liu 2014Lustberg 2018Niravath 2019Rastelli 2011Rosati 2011Shenouda 2019). We based these judgements on high drop‐out rates of 20% or greater (Cathcart‐Rake 2020Lustberg 2018Rosati 2011Shenouda 2019Niravath 2019), high proportional rates of exclusion from analysis with disparities between groups (Liu 2014), and a high drop‐out rate of 20% of greater and disparity in dropout rates between the intervention and control groups (Rastelli 2011). Two studies published in abstract/poster only were at unclear risk of attrition bias due to insufficient information to permit judgement (Birrell 2009Massimino 2011).

We judged eight studies at low risk of attrition bias as handling of incomplete outcome data was adequately described and unlikely to have produced bias (Chan 2017Henry 2018Hershman 2015aKhan 2017Li 2017Peng 2018Shapiro 2016Sordi 2019).

Selective reporting

We judged two studies at high risk of selective reporting (Lustberg 2018Rastelli 2011). One study of high‐ versus standard‐dose vitamin D reported an improvement in two‐month pain scores for the high‐dose group; however, the two‐month PRO pain outcome data scores did not appear to be a prespecified outcome on the trial registry (Rastelli 2011). The six‐month PRO outcomes were the registered primary outcomes. The positive effect of high‐dose vitamin D supplementation was not maintained at six months. The second study of O3‐FA compared to placebo reported safety and tolerability as the primary outcomes (Lustberg 2018); however, the trial registration listed pain score change after six months based on the Functional Assessment of Cancer Therapy – Breast (FACT‐B) and Functional Assessment of Cancer Treatment – Endocrine Symptoms (FACT‐ES) instrument as the primary outcome, and did not specify specifically safety and tolerability as secondary outcomes on the trial registry. No further information could be obtained by author correspondence.

Nine studies were at unclear risk of selective reporting (Birrell 2009Khan 2017Li 2017Liu 2014Massimino 2011Peng 2018Rosati 2011Shenouda 2019Sordi 2019). The reasons for judgement were as follows: insufficient information to permit judgement as study published only in abstract/poster form and no further information available (Birrell 2009Massimino 2011Rosati 2011Shenouda 2019), or insufficient information available in the publication to permit judgement and no further information able to be obtained (Liu 2014); at least one relevant missing unreported outcome among a very high number of planned outcomes in the protocol (Khan 2017); insufficient information whether certain outcomes had been analysed in accordance with a prespecified statistical plan (Li 2017); uncertainties about the time points for the primary outcomes with insufficient information available to permit judgement (Peng 2018); and inability to access the trial registry or statistical analysis plan to permit judgement (Sordi 2019). 

We assessed six studies at low risk of selective reporting because these studies reported all of their proposed outcomes (Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aNiravath 2019), or only secondary/exploratory outcomes included in the initial trial registration were not reported in the study and these outcomes were not of relevance to our review (Shapiro 2016). 

Other potential sources of bias

We judged two studies at high risk of additional bias due to authors holding patents for either the intervention itself (Birrell 2009), or for patents relevant to the intervention (Cathcart‐Rake 2020). Birrell 2009 was published only in abstract/poster form and no further information could be obtained by author correspondence.

Eight studies were at unclear risk of additional sources of bias (Hershman 2015aLiu 2014Lustberg 2018Massimino 2011Peng 2018Rosati 2011Shapiro 2016Shenouda 2019). Four of these studies were deemed to be unclear risk due to insufficient information to permit judgement (Liu 2014Massimino 2011Rosati 2011Shenouda 2019). Two of these studies were considered at unclear risk of contamination, due to the unclear exposure to O3‐FA in the placebo arm (Hershman 2015aLustberg 2018). Two studies had run‐in designs, whereby participants were provided with vitamin D (Shapiro 2016) or calcium and vitamin D supplementation (Peng 2018) prior to randomisation. The authors in one study stated the study was designed to mimic the high prevalence of the use of supplemental vitamin D among women with AIMSS (Shapiro 2016). As it was possible that vitamin D had a role in altering AIMSS in women with low 25(OH)D (less than 30 ng/mL), and this may have enhanced or diminished the effect of subsequent randomised intervention, these studies were judged at unclear risk of other bias (Peng 2018Shapiro 2016).

We considered the remaining seven studies at low risk of additional sources of bias, as the introduction of possible bias was thought to be adequately assessed through the primary domains of bias consideration (Chan 2017Henry 2018Khan 2017Li 2017Niravath 2019Rastelli 2011Sordi 2019).

Effects of interventions

See: Summary of findings 1 Summary of findings table ‐ Systemic therapy compared to control for treating aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer; Summary of findings 2 Summary of findings table ‐ Systemic therapy compared to control for preventing aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Prevention of aromatase inhibitor‐induced musculoskeletal symptoms

See summary of findings Table 1.

Pain (from baseline to the end of the intervention)

See Table 1Analysis 1.1Figure 4.

Open in table viewer
Table 1. Prevention studies: pain

Study

Intervention vs control

Treatment duration

Intervention

Control

Baseline pain, mean (SD)

Change from baseline, mean (SD)

n

Change from baseline, mean (SD)

n

Outcome measure (scale)

Khan 2017

Vitamin D3 30,000 IU weekly vs placebo

24 weeks

2.39 (2.34)

0.8 

67

0.86 

72

BPI worst pain

(0–10)

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of USA diet

24 weeks

1.1 (1.55)

0.11 (1.88*)

22

−0.70 (1.88*)

22

BPI severity 

(0–10)

*Calculated from standard error (SE). SE of change scores in intervention arm 0.40; SE of change scores in control arm 0.41.
BPI: Brief Pain Inventory; IU: international units; n: number of participants; SD: standard deviation.


Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Two studies reported sufficient data (vitamin D3Khan 2017; O3‐FA, Lustberg 2018) and showed no clinically meaningful difference in the change in mean pain scores between treatment arms (Analysis 1.1). A clinically meaningful change in BPI pain scales is considered to be two points (Mease 2011Williams 2011). 

PROs for pain included:

Omega‐3 fatty acids

In Lustberg 2018, there was no clinically meaningful improvement in pain scores over the course of the study in either the O3‐FA arm or the control arm (Table 1). The change in pain scores between arms was MD 0.81 (95% CI −0.30 to 1.92; Analysis 1.1). 

Vitamin D3

Author correspondence from Khan 2017 provided us with unpublished mean pain scores for BPI subscales. The difference in the change in mean pain scores between arms was MD −0.31 (95% CI −1.23 to 0.61; Analysis 1.1). 

We rated the certainty of evidence for pain as very low due to concerns with risk of bias, including high risk of attrition and reporting bias in one study. One study only included women with 25‐OHD levels of 40 ng/mL or less, which resulted in concerns with indirectness, and the small sample sizes in the study raised concerns for imprecision. See summary of findings Table 1.

Two other studies only reported the incidence of pain, rather than the mean pain scores for patients overall (Niravath 2019Rosati 2011), despite Niravath 2019 utilising a VAS to record pain scores. We were unable to obtain the mean pain scores from the authors for this study.

Stiffness (from baseline to the end of the intervention)

None of the prevention studies investigated stiffness. 

Grip strength

See Analysis 1.2.

Vitamin D

Two studies investigated grip strength with the initiation of vitamin D plus an AI (Khan 2017Niravath 2019); however, there were limited results for both studies, which prohibited these studies from being combined for meta‐analysis. Khan 2017 reported the incidence of worsening grip strength (−6.2 kg) of about 9% in the intervention and treatment arms (RR 1.08, 95% CI 0.37 to 3.17; 1 study, 147 participants; Analysis 1.2; low‐certainty evidence). The evidence was downgraded due to concerns with indirectness, as one study only included women with 25‐OHD levels of 40 ng/mL or less. The small sample size also raised concerns for imprecision. See summary of findings Table 1.

Niravath 2019 tested participants' grip strengths at baseline, week 12 and week 52, but only reported the mean change in grip strength in the group who had shown symptoms of AIMSS (mean change −1.3 mmHg) compared to the group without symptoms of AIMSS (mean change −3.5 mmHg) (P = 0.37). It was not possible to incorporated this study in the summary of findings table for this outcome due to lack of information. 

Safety

See Table 2.

Open in table viewer
Table 2. Prevention: safety

Studies

Intervention vs control

Treatment duration

Intervention

Control

Safety reporting

n

Safety reporting

n

Khan 2017

Vitamin D3 30,000 IU weekly vs placebo

24 weeks

No AEs attributed to Vitamin D

70

1 case of hypercalcaemia. No discussion of other AEs

77

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of the American diet

24 weeks

1 grade 3 AE (diarrhoea)

No grade 4/5 AEs

 

22

No grade 3/4/5 AEs

 

22

Niravath 2019

Vitamin D3 50,000 IU weekly for 12 weeks then 2000 daily for 40 weeks vs 800 IU daily

52 weeks

No grade 4/5 events

8 grade 3 events, deemed unrelated to study drugs

44

No grade 4/5 events

4 grade 3 events, deemed unrelated to study drugs

43

Rosati 2011

Etoricoxib (COX‐2 inhibitor) 60 mg/day vs placebo

2 years

FDA alert on COX‐2 inhibitors

Study reported 0 serious AE

16 

No report of safety data in control arm
 

50
 

AE: adverse event; COX: cyclo‐oxygenase; FDA: Food and Drug Administration; IU: international unit; n: number of participants.

Four studies reported safety data (Khan 2017Niravath 2019Lustberg 2018Rosati 2011). No study reported serious adverse events. Niravath 2019 included 87 participants and reported no grade four or five events. There were 12 grade three adverse events, with eight of these occurring in the high‐dose vitamin D arm, and the remaining four occurring in the standard‐dose vitamin D arm. All of these events were deemed unrelated to the study drugs. Grade three events included arthralgia, peripheral neuropathy, hypertension, hyperglycaemia and skin infection. Rosati 2011 reported "none of the patients in the treatment arm developed serious adverse event", but did not give any further details. During the course of the study, there was an FDA alert for COX‐2 inhibitors (FDA 2018), resulting in 38% of study participants withdrawing from the trial. Lustberg 2018 investigating O3‐FA reported a detailed list of adverse events. There was only one grade three event, which was in the intervention arm, due to diarrhoea. There were no grade four events. 

The evidence for this outcome was very low certainty, due to risk of bias concerns, with high risk of bias in multiple studies, including one study with high risk of attrition bias following a significant number of participants dropping out following an FDA warning for the intervention (FDA 2018). Two other studies had high risk of attrition bias, and one study had high risk of both performance and detection bias. We downgraded for imprecision, due to small study samples, and also for indirectness due to one study only including women with 25‐OHD levels of 40 ng/mL or less. See summary of findings Table 1

Discontinuation of aromatase inhibitors

See Analysis 1.3.

Vitamin D

Two studies investigated discontinuation of AIs with vitamin D for AIMSS (Khan 2017Niravath 2019). Khan 2017 reported 3/77 (4%) participants in the placebo arm versus 0/70 (0%) participants in the intervention arm (high‐dose vitamin D3 weekly) discontinued AI due to adverse events (RR 0.16, 95% CI 0.01 to 2.99; 1 study, 147 participants; Analysis 1.3; low‐certainty evidence). We downgraded the evidence due to concerns with indirectness, as one study only included women with 25‐OHD levels of 40 ng/mL or less. The small sample size raised concerns for imprecision. See summary of findings Table 1.

Niravath 2019 attempted to investigate patient compliance with AI therapy using tablet counting, but only had data available for 14/93 (13%) randomised participants. Of the available data, the participants in the standard‐dose vitamin D arm had taken 96.5% of their AI therapy, and the participants in the high‐dose vitamin D arm had taken 98.1% of their AI therapy. As this study did not provide data on the number of participants who discontinued AI, it was not included in the summary of findings table for this outcome. 

The other two prevention studies did not investigate adherence (Lustberg 2018Rosati 2011). 

Breast cancer‐specific quality of life

See Analysis 1.5Table 3.

Open in table viewer
Table 3. Prevention: breast cancer‐specific quality of life (BCS‐QoL)

Study

Intervention vs control

Treatment duration

Intervention

Control

MD (95% CI)

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline to end of intervention, mean (SD)

n

Change from baseline to end of intervention, mean (SE)

n

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of the USA diet

24 weeks

119

(12.66)

 

2.5 (10.79)

22

0.95 (10.79)

22

1.55 (−4.83 to 7.93)

FACT‐B

(0–148) 

SD calculated from SE. Baseline intervention arm SE = 2.7; change score SE = 2.3.
FACT‐B: Functional Assessment of Cancer Therapy – Breast; MD: mean difference; n: number of participants; SD: standard deviation; SE: standard error.

Two studies investigated BCS‐QoL (vitamin D3Khan 2017; O3‐FA, Lustberg 2018), but one study did not report values for this outcome (Khan 2017). In Lustberg 2018, the MD total FACT‐B change scores from baseline until the end of intervention between arms was 1.55 (95% CI −4.83 to 7.93; 1 study, 44 participants; Analysis 1.5; low‐certainty evidence). For the FACT tools, a high score is equivalent to a higher quality of life. There was also no clinically meaningful improvement in mean scores in either group from baseline until the end of the intervention (Table 3). The MCID for FACT‐B total score is 7 to 8 points (Eton 2004). We downgraded the evidence due to concern with risk of bias, high risk of attrition and reporting bias in one study, and concern with imprecision due to small study sample. See summary of findings Table 1.

The other studies did not collect BCS‐QoL data (Niravath 2019Rosati 2011). 

Health‐related quality of life

See Analysis 1.6Table 4.

Open in table viewer
Table 4. Prevention: health‐related quality of life (HRQoL)

Study

Intervention vs control

Treatment duration

FACT‐G subscale

Intervention

Control

Difference in change scores between arms, MD (95% CI)

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline to end of intervention, mean (SD)

n

Change from baseline to end of intervention, mean (SD)

n

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of the American diet

24 weeks

FACT‐G overall score

92 (10.32) 

1.9 (7.97)

22 

0.047 (8.44)

22 

1.85 (−3.00 to 6.70)

FACT‐G overall score 

(0–108) 

Physical Well‐being 

25 (2.77) 

−0.027 (3.24) 

22 

0.015 (3.33)

22 

−0.04 (−1.98 to 1.90)

FACT‐G physical well‐being subscale 

(0–28)

Social Well‐being

24 (4.5) 

0.39 (5.16)

22 

−0.96 (5.16)

22 

1.35 (−1.70 to 4.40)

FACT‐G social well‐being subscale 

(0–28)

Emotional Well‐being

20 (2.53) 

0.76 (2.81)

 22

 0.36 (2.86)

 22

0.40 (−1.28 to 2.08)

FACT‐G emotional well‐being subscale

 (0–24)

Functional Well‐being

 23 (5.16)

1.1 (3.19)

22 

 0.72 (3.28)

22

0.38 (−1.53 to 2.29)

FACT‐G functional well‐being subscale 

(0–28)

SD calculated from standard error (SE).
CI: confidence interval; FACT‐B: Functional Assessment of Cancer Therapy – Breast; FACT‐G: Functional Assessment of Cancer Therapy – General (higher scores equate to better quality of life); SD: standard deviation.

Two studies investigated HRQoL (vitamin D, Khan 2017; O3‐FA, Lustberg 2018), but one study did not report values for this outcome (Khan 2017). In Lustberg 2018, the MD in FACT‐G overall scores from baseline until the end of intervention between arms was 1.85 (95% CI −3.00 to 6.70; 1 study, 44 participants; Analysis 1.6; low‐certainty evidence). The MCID for a FACT‐G overall score is 5 to 6 points (Eton 2004). We downgraded the evidence due to concern with risk of bias, high risk of attrition and reporting bias in one study, and concern with imprecision due to small study sample. See summary of findings Table 1.

The other studies did not collect data on overall HR‐QoL (Niravath 2019Rosati 2011). 

Incidence of aromatase inhibitor‐induced musculoskeletal symptoms

See Analysis 1.4Table 5

Open in table viewer
Table 5. Prevention: incidence of aromatase inhibitor‐induced musculoskeletal symptoms

Study

Intervention vs control

Treatment duration

Intervention

Control

Incidence of AIMSS (%)

n

Incidence of AIMSS (%)

n

Khan 2017

Vitamin D3 30,000 IU weekly vs placebo

24 weeks

(37%)

70

(51%) 

77 

Niravath 2019

Vitamin D3 50,000 IU weekly for 12 weeks then 2000 IU daily for 40 weeks vs 800 IU daily

52 weeks

25 (54%) 

47 

27 (57%) 

46 

Rosati 2011

Etoricoxib 60 mg/day vs placebo

2 years

(31%)

 16

(76%)

 50

AIMSS: aromatase inhibitor‐induced musculoskeletal symptoms; IU: international units; n: number of participants.

Three studies reported the incidence of AIMSS (Khan 2017Niravath 2019Rosati 2011Table 5). Two of these studies, which both investigated the use of high‐dose vitamin D in the prevention of AIMSS, were combined for meta‐analysis (Khan 2017Niravath 2019). The third study was not included in the meta‐analysis as it investigated a different systemic therapy, etoricoxib (Rosati 2011).

Vitamin D

The RR for the incidence of AIMSS between the vitamin D and control arms was 0.82 (95% CI 0.63 to 1.06; I2 = 0%; 2 studies, 240 participants; Analysis 1.4; very low‐certainty evidence). There was no statistical heterogeneity among the studies involved in the meta‐analysis. We downgraded the certainty of the evidence due to high risk of performance bias, detection bias and attrition bias. We downgraded for indirectness, due to one study only including women with 25‐OHD levels of 40 ng/mL or less, and downgraded for imprecision due to small study samples. See summary of findings Table 1.

Etoricoxib

Rosati 2011 used the investigator‐reported outcome of the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) v3.0 to record musculoskeletal pain incidence (Table 5). Limited information is available for this trial as it has not progressed to the publication of a manuscript. There were no details regarding the breakdown of severity of pain experienced by participants in each arm. The study had a high risk of attrition bias due to a high number of participants withdrawing from the study due to a US Food and Drug Administration (FDA) alert on COX‐2 inhibitors (FDA 2018). There was insufficient data on the time point of the data collection for musculoskeletal symptoms.

Breast cancer‐specific survival and overall survival

None of the prevention studies reported BCSS or OS. One study investigating etoricoxib collected data on survival in the form of EFS (Rosati 2011), but the definition of EFS was unclear as the study was only presented at a conference without pursuing full publication. Rosati 2011 reported five‐year EFS of 83% in the etoricoxib arm versus 71% in the placebo arm (hazard ratio 1.9, 95% CI 1.03 to 3.59; P = 0.03). This was after nearly 40% of participants (37/93 in the intervention arm versus 33/89 in the control arm) had dropped out of the study due to an FDA alert regarding COX‐2 inhibitor safety concerns (FDA 2018). Due to the lack of information from this study, this outcome was not presented in the summary of findings table. 

Treatment of aromatase inhibitor‐induced musculoskeletal symptoms

See summary of findings Table 2

Pain (from baseline to end of intervention due to aromatase inhibitor‐induced musculoskeletal symptoms) 

See Analysis 2.1Table 6

Open in table viewer
Table 6. Treatment: pain

 Studies

Intervention vs control

Treatment duration

Intervention

Control

Difference in change scores between arm, mean

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline, Mean (SD)

Change from baseline, mean (SD)

n

Cathcart‐Rake 2020

Testosterone: 120 mg subcutaneous pellets, changed to topical daily gel due to slow accrual vs placebo

6 months

5.4 (1.7)

−1.9 (2.2)

80

−2.2 (2.7)

77

0.3

BPI‐AIA

mean pain

(0–10)

Shapiro 2016

Vitamin D3 4000 IU daily vs 600 IU daily

6 months

10.8 (3)

−1.2 (3.8)

39

−0.6 (3.8)

36

−0.6

WOMAC pain

(0–20)

Hershman 2015a

Omega‐3 fatty acids 3.3 g daily vs soybean/cornoil placebo

24 weeks

 7.06 (1.53a)

−2.23 

94

−1.81

98

−0.42 

BPI worst pain

(0–10)

Peng 2018 

Yi Shen Jian Gu granules twice daily vs placebo
 

12 weeks

6.18 (1.58)

−3.10

40

−1.65 

37

−1.47

BPI‐SF

worst pain

(0–10)

Chan 2017
 

Emu oil topically 3 times daily vs placebo

8 weeks

N/A

−0.82 (1.9a)

36

−0.93 (1.7a)

37

0.11 

BPI severity (0–10)

Sordi 2019

Cat's claw (Uncaria tomentosa

3 times daily vs placebo

 30 days

 8 (2.1)

0

32

−3

29

3

VAS

(0–10)

 Li 2017

Bionic tiger bone capsules 3 times daily vs calcium carbonate 600 mg daily

12 weeks

6.59 (2.11)

−2.72 

35

0.87

35

−3.59

BPI worst pain 

(0–10)

Henry 2018

Duloxetine 30 mg daily for 1 week then 60 mg daily for 11 weeks vs placebo
 

12 weeks

5.53 (N/A)

−2.6 

127

−1.97 

128

−0.63

BPI mean pain 

(0–10)

Rastelli 2011

Vitamin D2: either 50,000 IU weekly for 8 weeks if vitamin D insufficiency then monthly; or 50,000 IU weekly for 16 weeks if vitamin D deficiency then monthly; or control, 400 IU daily

6 months

5.2 (2.4)

−1.5 

28

−0.9

29

−0.4

BPI worst pain

(0–10)

Liu 2014

Calcitonin 200 IU + 600 mg daily caltrate D vs 600 mg daily caltrate D

 3 months

 5.0 (0.74)

−3.00 (1.48)

42

−1.00 (1.11)

40

−2

VAS scale 

(0–10)

Where SD is not available, this was due to lack of change score SD reported in the study. For these studies, end of treatment MD were used in the analysis.
Rastelli 2011: 2 months of BPI data used to eliminate differences between dosing in Stratum A and B.
Liu 2014: although study reported mean (SD) being utilised, data more consistent with median and interquartile ranges. Data were treated as such.
aSD calculated from provided confidence intervals.
BPI: Brief Pain Inventory; BPI‐AIA: Brief Pain Inventory for Aromatase Inhibitor Arthralgia; BPI‐SF: Brief Pain Inventory – Short Form; IU: international unit; MD: mean difference; n: number of participants; N/A: not available; SD: standard deviation; VAS: Visual Analogue Scale.

Thirteen studies investigated the treatment of AIMSS (Birrell 2009Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aLi 2017Liu 2014Massimino 2011Peng 2018Rastelli 2011Shapiro 2016Shenouda 2019Sordi 2019). A summary of the pain outcomes and tools used to measure pain is shown in Table 6. Three studies were not included in this table or in the assessment of this outcome due to lack of information, as each study was only published as an abstract (Birrell 2009Massimino 2011Shenouda 2019). The authors were either unable to be contacted or unable to provide further information. None of the studies investigating pain could be combined for meta‐analysis due either to the wide‐ranging interventions among studies, or because for the testosterone studies (Birrell 2009Cathcart‐Rake 2020), there was insufficient information from the Birrell 2009 to allow meta‐analysis. 

PROs for pain included:

Testosterone

Two studies investigated testosterone (Birrell 2009Cathcart‐Rake 2020). In Cathcart‐Rake 2020, the MD between arms for the change in mean joint pain from baseline to the end of intervention was 0.30 (95% CI −0.47 to 1.07; Analysis 2.1; very low‐certainty evidence). There were limited data available from Birrell 2009, which was published as an abstract only; the study reported decreased VAS scores of 35% in the placebo arm compared with a decrease of 43% (P = 0.06) with testosterone undecanoate 40 mg daily and 70% (P = 0.04) with testosterone undecanoate 80 mg daily. Due to the lack of data, testosterone study results were not combined for meta‐analysis, and the study by Birrell 2009 was not included in the summary of findings Table 2 for this outcome. 

Vitamin D

Two studies investigated vitamin D (Rastelli 2011Shapiro 2016). Rastelli 2011 used high‐dose vitamin D2 supplementation, and the difference in the change in pain scores from baseline to two months between arms was MD −1.50 (95% CI −2.59 to −0.41; P = 0.041; Analysis 2.1; very low‐certainty evidence; Table 6). Shapiro 2016 investigated the use of high‐dose (4000 IU) vitamin D3 supplementation versus a usual‐dose (600 IU) vitamin D3. Using the WOMAC v3.1 tool, which is commonly used to investigate symptoms of arthritis in the lower limbs, the MD in pain scores from baseline to the end of the intervention between arms was −0.60 (95% CI −2.32 to 1.12; Analysis 2.1; very low‐certainty evidence; Table 6). There were similar results using the AUSCAN tool, which was developed to investigate osteoarthritis of the hand with the change in mean pain scores between baseline and the end‐of‐intervention in the control group of −0.2 (SD 3.3) compared to −0.9 (SD 3.3) in the intervention group (P = 0.21). These two vitamin D studies were not combined for meta‐analysis, due to the differing interventions and patient populations, with one study only including women with vitamin D deficiency and then stratifying women into different treatment doses based on their baseline vitamin D result (Rastelli 2011), and the other comparing a high‐ and low‐dose vitamin D supplementation in a patient population that was not selected based on baseline vitamin D levels (Shapiro 2016). 

Duloxetine

One study investigated duloxetine to treat AIMSS pain (Henry 2018). Using the data we had available, including baseline SDs only, we calculated the MD between groups at the end of treatment (12 weeks) using BPI mean pain as −0.63 (95% CI −0.97 to −0.29; Analysis 2.1; very low‐certainty evidence; Table 6). This was similar to the effect reported by the study in a supplemental table for outcomes at 12 weeks of therapy (MD −0.56, 95% CI −1.11 to −0.01) using a multivariate linear regression including covariates for baseline mean pain and prior taxane use. The study's statistical plan was powered to test a difference by arm at 12 weeks only. A clinically significant change in mean pain in this study was defined as a decrease from baseline of at least two points, which is consistent with the literature (Mease 2011Williams 2011). A decrease in pain scores of at least two points was seen in a higher proportion of participants in the duloxetine arm at six weeks (68% with duloxetine versus 49% with placebo; P = 0.003); however, this benefit was not sustained by the end of study intervention period at 12 weeks (68% with duloxetine versus 59% with placebo; P = 0.18). Change in pain score subscales were not provided for the WOMAC tool.

Omega‐3 fatty acids

One study on O3‐FA which gave the control group a placebo capsule made of soybean and corn oil, measured pain with the BPI‐SF and GRCS (Hershman 2015a). The difference in pain change scores between the two groups from baseline to the end of intervention was MD −0.24 (95% CI −1.0 to −0.52; Analysis 2.1; very low‐certainty evidence; Table 6). The study authors reported similar change in MD between arms at the end of intervention using a linear regression adjusting for baseline BPI score and stratification factors (MD −0.36, 95% CI −1.08 to 0.38).

Bionic tiger bone

One study investigated the use of bionic tiger bone capsules versus placebo (calcium carbonate tablets 600 mg) for 12 weeks (Li 2017). There was a reduction in mean pain scores between arms measured by BPI worst pain (MD −4.09, 95% CI −5.25 to −2.93; 70 participants; Analysis 2.1; very low‐certainty evidence; Table 6). This is consistent with an MCID for BPI pain in this study (Mease 2011Williams 2011). There were similar results for the change in mean VAS scores at 12 weeks (−3.31 with bionic tiger bone vs 0.21 with placebo; P < 0.001) and BPI mean pain scores at 12 weeks (−2.62 with bionic tiger bone vs 0.75 with placebo; P < 0.001). This was listed as a blinded study, yet the intervention group was given bionic tiger bone capsules three times a day and the control group was given calcium carbonate tablets once daily. 

Yi Shen Jian Gu

One study examined the use of YSJG granules (Peng 2018). Worst pain mean scores decreased between arms, with the CIs including the MCID for a BPI scale (MD −1.34, 95% CI −2.10 to −0.58; Analysis 2.1; very low‐certainty evidence; Table 6). Pain severity and pain‐related interference scores also decreased at 12 weeks (−2.41 with YSJG versus −1.22 with placebo; P = 0.003) and 24 weeks (−2.6 with YSJG versus −1.34 with placebo; P = 0.001). The decrease in the mean pain score in the intervention arm remained clinically meaningful at 24‐week follow‐up, even though the duration of intervention was only 12 weeks. Pain scores from both the WOMAC and M‐SACRAH pain subscales reflected similar findings.

Emu oil

One study investigated the use of emu oil drops applied topically to affected joints versus a placebo oil (Chan 2017). Between baseline and eight weeks of treatment, the MD in BPI severity scores between arms was 0.11 (95% CI −0.72 to 0.94; P = 0.78; Analysis 2.1; very low‐certainty evidence; Table 6). There were similar results with the use of the VAS tool. VAS scores improved by −1.46 in the control arm (95% CI −2.12 to −0.81) compared to −−1.09 in the emu oil arm (95% CI −1.83 to −0.35) (P = 0.45). 

Calcitonin

One study investigated the use of calcitonin (Liu 2014). Although the study authors documented the reporting of mean and SD for results, the results were more consistent with median and IQR. The study reported a change in VAS scores of −1.00 (IQR (probably) −1.50 to 0.00) in the control group compared to a change in VAS score of −3.00 (IQR (probably) −4.00 to −2.00) in the calcitonin arm (P < 0.001). Assuming that the data were indeed reported using median/IQR, our analysis showed a difference between groups at the end of the intervention of MD −2.0 (95% CI −2.56 to −1.44; Analysis 2.1; very low‐certainty evidence). The Cochrane Handbook for Systematic Reviews of Interventions warns that the reporting of median values and IQR is often associated with skewed data (Higgins 2021).

Cat's claw

One study investigated the use of cat's claw (Uncaria tomentosa) for four weeks versus placebo (Sordi 2019). The change in mean VAS score in the control arm was −3 compared to no change in the intervention arm (MD 3.00, 95% CI 1.66 to 4.34; P = 0.02), which means that the placebo arm experienced an improvement in pain scores measured with the VAS tool and the intervention arm experienced no change in pain scores. This study also reported BPI severity scores; however, the baseline mean pain score in the control arm had an SD of 8.3 and the intervention arm end‐of‐treatment pain score had an SD of 7.3, using a pain scale of 0 to 10 with the BPI severity tool, as confirmed in the BPI user guide (Cleeland 2009). These SDs were not plausible for this pain scale, therefore the BPI data were not used. Further clarification could not be obtained from the study author. 

Three studies had not been published as a manuscript and there was only limited information available (Birrell 2009Massimino 2011Shenouda 2019). We were unable to obtain further information from study authors and, therefore, these studies were not included in summary of findings Table 2 for pain. 

Overall, we were unable to determine the effect of systemic therapies on the treatment of pain from AIMSS, because we rated the evidence as very uncertain due to a number of concerns. There were concerns with risk of bias in multiple studies, the sample sizes of studies were small with serious concerns of imprecision, and significant heterogeneity in interventions used in these studies, therefore, concerns regarding inconsistency. A funnel plot showed an asymmetrical scatter of small studies with more studies showing a positive result than those showing a negative result (Figure 5), which raises suspicion of publication bias. In an extensive literature search, there were multiple studies in this outcome which have only been published in abstract form, without pursuing full/peer‐reviewed publication. See summary of findings Table 2


Funnel plot of comparison: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.1 Pain.

Funnel plot of comparison: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.1 Pain.

Stiffness (from baseline to end of intervention) 

See Analysis 2.2Table 7.

Open in table viewer
Table 7. Treatment: stiffness

Study

Intervention vs control

Treatment duration

Intervention

Control

Difference in change scores between arms, MD (95% CI)

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline to end of intervention, mean (SD)

n

Change from baseline to end of intervention, mean (SD)

n

Shapiro 2016

Vitamin D3 4000 IU daily vs 600 IU daily

6 months

5.5 (1.5)

−0.5 (1.7)

39

−0.5 (2.0)

36

0 (−0.82 to 0.82) 

 WOMAC

(0–8)

Hershman 2015a
 

Omega‐3 fatty acids 3.3 g daily vs soybean/cornoil placebo

24 weeks

N/Aa

0.59 (1.25)b

109

0.46 (1.25)b

114

0.13 (−0.20 to 0.46)

GRCS at 6 weeks

Peng 2018
 

Yi Shen Jian Gu granules twice daily vs placebo
 

12 weeks

70.43 (43.49)

−37.53 

40

−20.82 

37

−16.71 
 

WOMAC

(0–200)

Li 2017
 

Bionic tiger bone capsules 3 times daily vs calcium carbonate 600 mg daily

12 weeks

5.49 (1.58)

−3.08 

35

1.87 

35

 −4.95 

Modified BPI with stiffness 

(0–10)

Henry 2018
 

Duloxetine 30 mg daily for 1 week then 60 mg daily for 11 weeks

12 weeks

N/Aa

OR 3.38 (1.85 to 6.18; P = 0.0001)c

Chan 2017
 

Emu oil topically 3 times daily vs placebo

8 weeks

1.9 (0.67)

−0.31 (0.82)

36

−0.41 (0.82)

37

 0.10 (−0.28 to 0.48)

VAS scale 

(0–3)

a Evaluates changes in symptoms since last visit, so no baseline scores were collected.

bSome standard deviations (SD) not available, due to lack of reported change score SD. For these studies, end of treatment means (SD) were used in the analysis of effect size. See Analysis 2.2.
cReported as a binary outcome, OR. 
Henry 2018Hershman 2015a could not be assessed for the effect of stiffness on the treatment of AIMSS, including confidence intervals and, therefore, were not included in the final assessment of this outcome.
BPI: Brief Pain Inventory; CI: confidence interval; GRCS: Global Rating of Change Scale; IU: international unit; MD: mean difference; n: number of participants; N/A: not available/applicable; OR: odds ratio; SD: standard deviation; VAS: Visual Analogue Scale; WOMAC: Western Ontario and McMaster Universities Osteoarthritis scale.

Seven studies investigating the treatment of AIMSS reported stiffness (Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aLi 2017Peng 2018Shapiro 2016), but none could be combined for meta‐analysis due to vastly differing interventions (Table 7). Cathcart‐Rake 2020 included BPI‐AIA which has a question on stiffness, but did not publish these data. They did report that "testosterone supplementation did not improve the average pain or joint stiffness, compared with placebo". As the CIs could not be assessed, this study was not included in the assessment for this outcome in summary of findings Table 2

PROs for stiffness in the treatment studies included:

Vitamin D

In Shapiro 2016, using the WOMAC tool, there was no difference in change scores between arms from baseline until the end of the six months' intervention (MD 0.00, 95% CI −0.84 to 0.84; Analysis 2.2; very low‐certainty evidence; Table 7). Results were similar using the AUSCAN tool, which investigates arthritic symptoms of the hand, with a change in stiffness scores from baseline to the end of intervention in the control group of −0.1 (SD 1.0) compared to −0.1 (SD 0.9) in the vitamin D group (P = 0.91). 

Duloxetine

Henry 2018 used the GRCS for Stiffness and due to the nature of the tool, the effect of stiffness from baseline until the end of the intervention could not be measured. The GRCS for Stiffness requires a patient to recall their symptoms at a previous time point and compare this with their current symptoms, therefore no baseline recording is required (Kamper 2009). The study reported an improvement in stiffness scores with an odds ratio of 3.38 (95% CI 1.85 to 6.18; P = 0.0001) at two weeks. It is not clear what the criteria for 'improvement in GRCS for stiffness' were for the calculation of this as a binary outcome, and if this was consistent with the literature (Kamper 2009). Because this study could not be assessed for its effect on stiffness from baseline until the end of intervention, it was not included in summary of findings Table 2.

Omega‐3 fatty acids

Hershman 2015a used the GRCS for Stiffness and recorded data at week six, week 12 and week 24. The study reported between‐group MDs at six weeks of 0.13 (95% CI −0.20 to 0.46; P = 0.44); at 12 weeks of 0.28 (95% CI −0.07 to 0.62; P = 0.12); and at 24 weeks of −0.003 (95% CI −0.41 to 0.41; P = 0.99). Stiffness was also measured using WOMAC and M‐SACRAH tools, but subscales were not provided. As this study could not be assessed for its effect on stiffness from baseline until the end of intervention, it was not included in summary of findings Table 2

Bionic tiger bone

Using the modified BPI scale, Li 2017 reported an MD in stiffness scores of −4.53 (95% CI −5.69 to −3.37; Analysis 2.2; very low‐certainty evidence; Table 7). It is unclear what the MCID would be for this modified BPI with stiffness tool, but it is likely that the 95% CIs for this treatment effect includes a clinically important benefit. 

Yi Shen Jian Gu 

Using the WOMAC stiffness subscale, Peng 2018 reported an MD in stiffness scores between arms of −28.82 (95% CI −47.13 to −10.51; Analysis 2.2; very low‐certainty evidence; Table 7). This was consistent with findings from the M‐SACRAH subscale, showing a change in mean scores of −47.28 in the treatment arm at 12 weeks, compared to −31.75 in the control arm (P = 0.011). The change in mean stiffness scores between treatment arms persisted at the 24‐week follow‐up. There has been wide‐ranging estimates of the MCID for stiffness using the WOMAC tool, from 12.9 to 25 (Maredupaka 2020), with the results of this study showing a clinically meaningful benefit in stiffness.

Emu oil

Chan 2017 used a VAS scale measuring 0 to 3 points. The results were not published, but we obtained data via author correspondence. The MD in stiffness scores between arms was 0.10 (95% CI −0.28 to 0.48; Analysis 2.2; very low‐certainty evidence; Table 7). 

Overall, we rated the evidence for this outcome of stiffness as very low certainty due to concern with risk of bias; indirectness as one study had poorly defined criteria for AIMSS which means that participants may have experienced musculoskeletal symptoms from causes other than AIMSS; inconsistency due to heterogeneity in interventions used between studies; and imprecision due to the small sample sizes and wide CIs. See summary of findings Table 2

Grip strength

See Analysis 2.3.

One study investigated the effect of vitamin D on grip strength (Shapiro 2016). The study reported no difference in the mean change in grip strength between the control and intervention arms over the course of the study using a Jamar hydraulic hand dynamometer (MD 0.80, 95% CI −2.69 to 4.29; P = 0.3; Analysis 2.3; low‐certainty evidence). An MCID for grip strength is estimated to be 5 kg to 6.5 kg (Bohannon 2019). We downgraded the certainty of the evidence due to imprecision and indirectness as the single study included women experiencing musculoskeletal symptoms regardless of temporal association with the start of the AI, which may have resulted in women without AIMSS being enrolled. See summary of findings Table 2

Safety

Eleven studies reported safety data (Birrell 2009Cathcart‐Rake 2020Chan 2017Henry 2018Hershman 2015aLi 2017Liu 2014Peng 2018Rastelli 2011Shapiro 2016Sordi 2019). Shenouda 2019 and Massimino 2011 did not report safety data. Overall, there were no grade four or five adverse events in any of the treatment studies.

Testosterone

Cathcart‐Rake 2020 reported "no significant differences" between treatment arms in specific symptoms associated with testosterone toxicity as assessed on the Symptom Experience Diary, but incidence and P values were not provided. The testosterone study by Birrell 2009 is published in abstract only; however, they stated "no significant androgenic side effects (hirsutism, alopecia and acne) were observed and serum lipids remained unaltered". No values were given. 

Vitamin D

In Shapiro 2016, the most frequent adverse events were musculoskeletal symptoms (18%) and gastrointestinal symptoms (17%) and the authors reported that these symptoms "did not differ in incidence between arms". No P values were given. In Rastelli 2011, they reported no toxicities in the vitamin D group. However, at two months, four participants in the high‐dose vitamin D group and one participant in the placebo group were removed from the study due to asymptomatic hypercalciuria. 

Duloxetine

Henry 2018 reported no grade four or five adverse events. Grade three adverse events occurred in 12 (9%) participants in the duloxetine arm versus five (4%) participants in the placebo arm (P = 0.08). Adverse events of any grade occurred in 78% of the duloxetine group versus 50% of the placebo group (P < 0.001). The most common adverse events in the duloxetine arm were fatigue, nausea, dry mouth and headache. The toxicity profile of duloxetine may have unmasked the blinding of participants in this study, as more participants in the duloxetine arm compared with the placebo arm thought they were receiving duloxetine (79% with duloxetine versus 50% with placebo; P < 0.001). 

Omega‐3 fatty acids

Hershman 2015a reported no grade four or grade five adverse events. With regard to grade three adverse events, there was one case each of diarrhoea, dyspepsia and pain in the extremity in the O3‐FA arm and one case each of arthralgia, pain, peripheral motor neuropathy and rash in the placebo arm. 

Bionic tiger bone

In Li 2017, the only adverse event reported was 'stomach discomfort' in six participants overall (two in the treatment arm, four in the control arm). The grade of this adverse event was not listed. The study stated there were no other adverse events. 

Yi Shen Jian Gu

Peng 2018 reported that 33% of participants in the intervention arm experienced all‐grade toxicities compared with 39% of participants in the placebo arm (P = 0.589). One participant in the intervention arm experienced an increase in CA‐125 levels during the study (27.4 U/mL at baseline versus 40.78 U/mL). Follicle‐stimulating hormone and oestradiol levels remained in the postmenopausal range throughout the study. 

Emu oil

Chan 2017 reported no treatment‐emergent adverse events associated with emu oil and no serious adverse events in the total patient population. 

Calcitonin

In Liu 2014, there was limited safety data. The study authors reported no difference in biochemistry parameters between the treatment and control arms over the course of the study (including serum calcium, P = 0.8758; serum phosphorus, P = 0.5128; serum osteocalcin, P = 0.9224; and alkaline phosphatase, P = 0.6528), but did not discuss other adverse events.

Cat's claw

In Sordi 2019, there were no grade three or grade four adverse events reported. Two participants in the experimental arm and three participants in the placebo arm discontinued treatment because of adverse events, but the adverse events were not detailed further. 

We downgraded the certainty of the evidence for this outcome to very low, due to concerns with risk of bias in multiple studies, imprecision due to small sample sizes, and publication bias as multiple studies were published in abstract form only without pursuing full publication. See summary of findings Table 2.

Persistence and adherence to aromatase inhibitor medication

None of the treatment studies reported persistence or adherence to AI medication. 

Breast cancer‐specific quality of life

See Analysis 2.4Table 8

Open in table viewer
Table 8. Treatment: breast cancer‐specific quality of life (BCS‐QoL)

Studies

Intervention vs control

Treatment duration

FACT‐B subscale 

(scale)

Intervention

Control

Difference in change scores between arms, MD 

Baseline BCS‐QoL, mean (SD)

Change from baseline, mean (SD)

n

Change from baseline, mean (SD)

n

Peng 2018

Yi Shen Jian Gu granules twice daily vs placebo

 12 weeks

Physical Well‐being (0–28)

18.85 (4.53)

4.77 

 40

 3.41

 37

2.46

Social/Family Well‐being (0–24)

16.95 (4.62)

3.1 

 2.54

2.27

Emotional Well‐being (0–24)

21.78 (4.31)

21.78 

 1.40

1.54

Functional Well‐being (0–28)

17.15 (4.42)

4.26 

 3 

4.27

Additional Concerns (0–36)

23.58 (4.93)

2.93 

 1.84 

2.51

Li 2017
 

Bionic tiger bone capsules 3 times daily vs calcium carbonate 600 mg daily

 12 weeks

 Physical Well‐being (0–28)

19.23 (5.02)

35

 −0.1 

35

2.30

Social/Family Well‐being (0–24)

19.81 (3.98)

0.93 

−0.97 

0.90

Emotional Well‐being (0–24)

15.34 (4.41)

0.49 

 0.12 

0.81

Functional Well‐being (0–28)

15.26 (4.67)

0.71 

 −0.09 

0.10

Additional Concerns (0–36)

26.73 (4.61)

0.73 

 0.75 

−0.40

Where SD are not available for change scores, end of treatment means (SD) were used in the analysis, see Analysis 2.4.
In FACT scales, higher scores equate to better quality of life.
BCS‐QoL: breast cancer‐specific quality of life; FACT‐B: Functional Assessment of Cancer Therapy – Breast; n: number of participants; SD: standard deviation.

Two studies investigated quality of life using FACT‐B (Li 2017Peng 2018). These studies could not be combined for meta‐analysis due to using different interventions (Table 8). 

Bionic tiger bone

In Li 2017, the Physical Well‐being subscale showed an MD between arms of 2.30 (95% CI 0.05 to 4.55; Analysis 2.4; very low‐certainty evidence; Table 8). For all the other FACT‐B subscales, the effect sizes between groups reported CIs that crossed zero, but all had CIs that included a potentially clinically meaningful benefit according to the MCID for FACT‐B (Social/Family Well‐being MD 0.90, 95% CI −1.37 to 3.17; Emotional Well‐being MD 0.81, 95% CI −1.66 to 3.28; Functional Well‐being MD 0.10, 95% CI −2.36 to 2.56; Additional Concerns MD −0.40, 95% CI −2.88 to 2.08; Analysis 2.4; very low‐certainty evidence). The MCID estimate for the overall FACT‐B tool is 7 to 8 points, and FACT‐B subscales is 2 to 3 points (Eton 2004Yost 2014). For the FACT‐B tool, increased scores equate to better quality of life.

Yi Shen Jian Gu

In Peng 2018, all the FACT‐B subscales showed effect size between arms at the end of the intervention with CIs that included the MCID for FACT‐B subscales (Physical Well‐being MD 2.46, 95% CI 0.73 to 4.19; Social/Family Well‐being MD 2.27, 95% CI 0.07 to 4.47; Emotional Well‐being MD 1.54, 95% CI 0.06 to 3.02; Functional Well‐being MD 4.27, 95% CI 2.54 to 6.00; Additional Concerns MD 2.51, 95% CI −0.14 to 5.16; Analysis 2.4; very low‐certainty evidence; Table 8). 

We downgraded the certainty of the evidence for BCS‐QoL due to high risk of both performance and detection bias in one study and unclear reporting bias. We also downgraded the certainty of the evidence for indirectness, as one study did not include any criteria for an AIMSS diagnosis and excluded use of agents influencing bone metabolism, including bisphosphonate, which are frequently utilised in the target population. We also downgraded for imprecision, due to small sample sizes. See summary of findings Table 2

Breast cancer‐specific quality of life

None of the other studies assessed BCS‐QoL.

Health‐related quality of life

See Analysis 2.5Table 8Figure 6


Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.5: Health‐related quality of life.

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.5: Health‐related quality of life.

HR‐QoL results are represented in Figure 6 though none of the studies could be combined for meta‐analysis. Overall, HR‐QoL was not investigated in the studies by Birrell 2009Chan 2017Cathcart‐Rake 2020Liu 2014Massimino 2011, or Shenouda 2019

Bionic tiger bone

Li 2017 utilised FACT‐B as a quality of life measure, which incorporates the overall HR‐QoL tool FACT‐G plus a BCS‐QoL specific subscale (Additional Concerns). The FACT subscales have been reported above in BCS‐QoL. 

Yi Shen Jian Gu

Peng 2018 also used the FACT‐B tool. The FACT subscales have been reported above in BCS‐QoL. 

Cat's claw

Sordi 2019 used the 36‐Item Short Form Survey (SF‐36) to assess HRQoL. In our review of the data, it appears that either incorrect data points were published for the SF‐36 (Physical Limitations) or the recorded data was not mean (SD), despite confirmation by study authors, as baseline placebo mean scores were published as 0 (SD 35.8). See Analysis 2.5 for the results for the eight SF‐36 domains (very low‐certainty evidence). 

Two other studies did not report FACT‐G total score of subscales. 

We downgraded the certainty of the evidence due to concerns with risk of bias, imprecision due to small sample sizes, inconsistency due to heterogeneity in intervention between studies and indirectness because one study did not include any criteria for an AIMSS diagnosis and excluded use of agents influencing bone metabolism, including bisphosphonates which are frequently utilised in the target population. See summary of findings Table 2

Incidence of aromatase inhibitor‐induced musculoskeletal symptoms

None of the treatment studies reported change in incidence of AIMSS.

Breast cancer‐specific survival and overall survival

None of the treatment studies reported BCSS or OS.

Discussion

Summary of main results

This Cochrane Review investigated systemic therapies for the prevention and treatment of AIMSS in early breast cancer, and included 17 RCTs with 2034 randomised participants. Systemic therapies investigated were diverse and included 'conventional' pharmacological therapies and CAM, and also included studies of vitamin D in the setting of insufficiency. There was limited information about certain study interventions or placebo ingredients (or both). In most studies, the comparator arm was a placebo, however, in some studies, the comparator was a vitamin supplement. Meta‐analysis was limited in this review. Due to the clinical and methodological heterogeneity of the studies and the insufficient data available from several studies, only one outcome had data that could be combined for meta‐analysis. Overall, the certainty of the evidence was very low for multiple outcomes for systemic therapies assessing prevention and treatment of AIMSS.

For preventing AIMSS, we found that the evidence for the effect of systemic therapies on change in pain from baseline to the end of the intervention was very uncertain. The evidence suggests systemic therapies for preventing AIMSS may have little to no effect on grip strength, little to no effect on change in HRQoL and BCS‐QoL from baseline to the end of the intervention, and little to no effect on women continuing to take their AI, although the certainty of this evidence was low. The evidence was very uncertain on the outcome of change in incidence of AIMSS. The evidence is very uncertain for the safety of systemic therapies for the prevention of AIMSS. One study of a COX‐2 inhibitor had an FDA safety alert issued for this class of medications during the study leading to significant participant withdrawal/discontinuation; however, there were no serious adverse events in this study, or any other prevention study. There were no data to assess the impact of systemic therapies for prevention of AIMSS on stiffness, BCSS or OS from the prevention studies.

For treating AIMSS, the evidence was very uncertain about the effect of systemic therapies on the effect of change in pain, stiffness, HRQoL and BCS‐QoL from baseline to the end of the intervention. The evidence suggests systemic therapies may have little to no effect on grip strength, although the certainty of this evidence was low. The evidence is very uncertain about the safety data for the use of systemic therapies for the treatment of AIMSS. There were no grade four or five adverse events in any of the treatment studies. One study reported significantly more all‐grade adverse events with duloxetine than with control (78% with duloxetine versus 50% with control; P < 0.001). There were no data on the incidence of AIMSS, number of women continuing to take AI, or BCS‐QoL or OS from the treatment studies. 

The follow‐up interval was relatively short for most studies, and hence long‐term safety data are not available. Safety results should thus be interpreted with caution for both prevention and treatment studies.

Overall completeness and applicability of evidence

This review included 17 trials on 2034 participants. Insufficient data was a difficulty in this review, although additional data/information was obtained by author correspondence for four studies (Chan 2017Khan 2017Niravath 2019Sordi 2019). The studies in this review were diverse in terms of clinical and methodological heterogeneity. There is considerable heterogeneity in the type of systemic therapy studied. These included 'conventional' pharmacological therapies and CAM, with the vitamin D studies not all fitting the Cochrane definition of CAM (Wieland 2011). There was also heterogeneity in the route of administration of the studied systemic therapy with most being oral therapy (16 trials) and only one trial of topical therapy with possible or postulated systemic absorption and effects (Chan 2017); the proposed mechanism of action of the systemic therapy; and the duration of intervention and the follow‐up. Several studies had limited details about the intervention, including the active compounds or the comparator arms (or both) (YSJG granules (Peng 2018), bionic tiger bone (Li 2017), Blue Citrus (Massimino 2011), tart cherry (Shenouda 2019), Cat's claw (Sordi 2019)). There were no known systemic interventions investigating the treatment of AIMSS missed in this review, but given the limited information from some studies and the incomplete understanding of the aetiology of AIMSS (Hershman 2015bNiravath 2013), it is, therefore, not possible to make definitive conclusions about whether all relevant systemic or potentially effective therapy studies have been completed. We identified no studies that required translation into English, which raises the possibility that this review may have missed potential studies investigating a complementary or alternative medicine reported in a language other than English. Although non‐English papers were not excluded in this review, we searched only Western databases. There are ongoing studies of other systemic therapies with differing proposed mechanisms of action (see Characteristics of ongoing studies table). This review only included the investigation of systemic therapies for the treatment of AIMSS and, therefore, excluded other interventions that have been trialled for AIMSS that were not considered to be a systemic therapy, such as acupuncture and exercise. Most studies included in this analysis were conducted in hospital/oncology outpatient clinic settings. The limited information provided by some studies may limit the applicability to certain clinical settings.

The studies of vitamin D were methodologically and clinically heterogeneous (Khan 2017Niravath 2019Rastelli 2011Shapiro 2016), and were performed either to prevent or treat AIMSS. Inclusion criteria in two of the vitamin D studies required participants to have specific 25‐OHD levels at baseline (Khan 2017Rastelli 2011), particularly 25‐OHD levels of 40 ng/mL or less for the prevention trial of Khan 2017 and between 10 ng/mL and 29 ng/mL for the treatment trial of Rastelli 2011. Vitamin D deficiency is defined by most experts as a serum 25‐OHD level less than 20 ng/mL, and relative insufficiency of vitamin D as a level of 25‐OHD of 21 ng/mL to 29 ng/mL (Holick 2007). All women included in Rastelli 2011 had vitamin D deficiency or insufficiency, and many women in Khan 2017 also met these criteria. "Vitamin D deficiency in adults can precipitate or exacerbate osteopenia and osteoporosis, cause osteomalacia and muscle weakness, and increase the risk of fracture" (Holick 2007) and pain associated with these conditions could potentially be confused with AIMSS. Both studies were performed in the USA in predominantly white women (Khan 2017Rastelli 2011). A nested case‐control correlative study has suggested vitamin D levels were not significantly associated with development of AI arthralgia (Niravath 2018). The studies that selected women with inclusion criteria related to serum vitamin D levels may therefore have uncertain generalisability and applicability to the entire population of women with AIMSS, and limit the overall completeness of the evidence related to vitamin D.

Patient populations included in this review had ethnic and racial diversity; however, diversity was sometimes related to particular characteristics of certain studied systemic therapy interventions, notably CAM, and to the ethnic, racial and cultural characteristics of the study populations (e.g. studies of TCM conducted in China) (Peng 2018Li 2017); study of traditional Incan medicine, Cat's claw, conducted in Brazil (Sordi 2019). However, this was not consistent across all studies (e.g. study of TCM Blue Citrus conducted in the USA) (Massimino 2011). For studies examining TCM, choice of this intervention for AIMSS research in Chinese populations may relate to beliefs that TCMs for cancer care based on "deep cultural grounding", and beliefs that TCM is safe and effective (Xu 2006). Chinese herbal medicine was used by 76.8% (Chen 2008) and 86.7 % (Cui 2004) of Chinese women with breast cancer surveyed in Shanghai. In contrast, in a Canadian survey, although more than 80% of women with breast cancer reported using CAM (41% specifically to manage their breast cancer), only 2.2% of women with breast cancer consulted a TCM practitioner (Boon 2007). CAM use in ethnically and racially diverse populations has been found to be complex and nuanced; however, ethnicity has an independent role in the type of CAM used (Kronenberg 2006). One systematic review has highlighted the importance of traditional and CAM to indigenous people with cancer of Australia, Canada, New Zealand and the USA (Gall 2018). Ethnic, racial, cultural and linguistic diversity may therefore have relevance in limiting applicability, acceptability and generalisability of certain studied systemic therapy interventions (particularly traditional medicines and CAM) to other diverse populations with diverse cultural belief systems and corresponding traditional and CAM preferences. In addition, certain ethnic and racial groups were not adequately represented (or represented at all) in other studies of certain systemic therapies. As examples, in the treatment studies by Henry 2018 of duloxetine, Hershman 2015a of O3FA, Khan 2017 and Rastelli 2011 of vitamin D, greater than 85% of participants were white. However, in the prevention study of vitamin D by Niravath 2019 conducted in the USA, most participants enrolled were from minorities with 44% Latina people and 18% African American people enrolled.

The incidence of AIMSS has generally been reported to be approximately 50% (Beckwee 2017Crew 2007Henry 2008); however, one Chinese cohort reported an incidence of 72% (Xu 2014). A degree of current uncertainty about racial and ethnic variability in the incidence of AIMSS may affect data applicability and generalisability from this review. One placebo‐controlled trial of letrozole after five years of tamoxifen demonstrated in an unplanned subgroup analysis that women from minority ethnic and racial groups receiving letrozole had less arthritis however compliance was poorer than in Caucasian women receiving letrozole, and highlighted the low number of minority participants enrolled in this trial (Moy 2006). Pharmacogenetic predictors of AI toxicity have been reported but have not been adequately validated (Hertz 2017). Therefore, there is insufficient evidence from this review about differences in adverse events and other outcomes with the clinically heterogeneous systemic therapy interventions studied between different ethnic and racial participant groups and this may also limit generalisability and applicability.

The median age of women diagnosed with breast cancer in the USA is 62 years (Howlader 2019), similar to this review, and also similar to postmenopausal women diagnosed with breast cancer in other high‐income countries. However, similar to the Cochrane Review "Exercise therapies for preventing or treating aromatase inhibitor‐induced musculoskeletal symptoms in early breast cancer"(Roberts 2020), very few postmenopausal women older (greater than 75 years) or younger (less than 35 years) were included. Younger women at higher risk of recurrence are increasingly being treated with AI in combination with ovarian function suppression rather than with tamoxifen alone, due to improvements in DFS reported by landmark trials (Francis 2015Francis 2018). There were high rates of musculoskeletal symptoms reported in these trials (88.7% for ovarian suppression with exemestane treatment versus 69% with tamoxifen treatment alone). The studies in this review included very few young women rendered postmenopausal and treated with AI, likely as a result of when the landmark trials were published (Francis 2015Francis 2018). Young women treated with AI and ovarian function suppression or ablation represent an area of unmet clinical need, and an increasing population with AIMSS whose baseline incidence may be different to women included in this review. AIMSS may be inversely correlated with time since menopause (Mao 2009), and younger women with abrupt oestrogen withdrawal may be at higher risk of symptoms. Systemic therapy interventions for AIMSS may therefore have different applicability or even effectiveness in younger women.

Data were available for the primary outcome of pain for many of the studies. However, unfortunately data for certain important missing outcomes was unable to be obtained (e.g. Birrell 2009) to allow meta‐analysis (e.g. for testosterone in treatment – Birrell 2009Cathcart‐Rake 2020). Many studies did not assess BCS‐QoL or HRQoL outcomes. Women experiencing pain from taking AI experience changes in QoL, with the degree of change in QoL depending on the type of pain experienced (Laroche 2017). The minimal QoL evidence available for this review raises concerns with completeness of the evidence. Many studies did not report important AIMSS PROs such as joint stiffness. There was no evidence available on the effect of systemic therapies for treatment of AIMSS on the incidence of AIMSS. There was no evidence available on the effect of systemic therapies for AIMSS on OS. 

Studies that included participants with AIMSS at baseline varied in their definitions of AIMSS, and the comorbid conditions included. As noted in the Cochrane Review "Exercise therapies for preventing or treating aromatase inhibitor‐induced musculoskeletal symptoms in early breast cancer" (Roberts 2020), the lack of consensus on standardised definitions for AIMSS, the absence of objective outcome measures, and the multiple PROs reported in different studies meant it was not possible to make conclusions on completeness of the evidence of outcome measures in this review. These research barriers have been noted previously by Hershman 2015b and Niravath 2013. As the trials assessed multiple varied outcomes, and interventions were heterogeneous, it was neither possible nor appropriate to combine the majority of the outcomes for meta‐analysis. Long‐term safety data were lacking for many of the studies. Due to small sample sizes and heterogeneity of the trials, planned subgroup analysis was not possible, which further limits applicability of the findings.

There was considerable heterogeneity in the timing of the outcome measures of the included trials, which also limited comparability. The duration of the intervention in the included trials ranged between four weeks and two years. AIMSS are chronic symptoms with an unpredictable course. There was also considerable heterogeneity in follow‐up in the included studies. Evidence for long‐term benefits and harms is of increasing importance given the guideline recommendations for extended duration of AI therapy in certain women with breast cancer (Burstein 2019); however, limited evidence was available from this review in this setting. Two of the prevention studies reported adherence rates to AI therapy (Khan 2017Niravath 2019), and in none of the treatment studies. Adherence to AI therapy is an important outcome of an intervention for AIMSS, as AI adherence impacts breast cancer survival (Chirgwin 2016). Therefore, evidence is incomplete given the very low‐certainty evidence from the small number of studies reporting AI adherence as an outcome.

There were notable limitations in the data obtained from this review. Many studies were of small size and either had some concerns or were at high risk of bias, and evidence was of very low certainty across multiple outcomes. As a result, caution needs to be taken in the interpretation of the outcome data from this review, and in assessment of the long‐term risks and harms of systemic therapies for AIMSS. These considerations also limit the generalisability and applicability of the studied systemic therapy interventions.

The outcomes analysed in this review were chosen based on which outcome domains the studies researching AIMSS used at the time of our protocol development, in addition to guidance from working groups such as Outcome Measures in Rheumatology (OMERACT 2021). In updates of this review, we would consider utilising Cochrane Musculoskeletal Group's core outcome list (CMSG 2021), although outcomes would have to extrapolated from other predefined musculoskeletal conditions, such as osteoarthritis or rheumatoid arthritis. 

Quality of the evidence

Very low‐certainty evidence does not support the use of systemic therapy to prevent or treat AIMSS. Studies were generally of small size with the number of participants ranging from 37 to 299. Clinically heterogeneous systemic therapies or the clinically heterogenous settings in which they were studied (or both) meant that it was not appropriate to combine most outcomes for meta‐analysis. Hence, outcome assessments had low numbers of participants included, which led to downgrading of the evidence due to concerns with imprecision. 

Many studies were either at moderate risk of bias, or had some concerns. A few of the studies were not blinded. One study had higher rates of adverse events with the intervention (duloxetine) than with the placebo, leading to higher numbers of participants successfully guessing which intervention they were receiving, and potentially unblinding these participants (Henry 2018). Similarly, another study used an intervention dosed three times daily versus a comparator with single daily dosing, which posed a high risk of unblinding the participants and assessors in this 'double‐blinded' trial (Li 2017). For these studies, there was a high risk of performance bias. There was also a high risk of detection bias as most outcomes were PROs, and the remaining outcomes were not blinded to outcome assessors. Several of the studies had inadequate outcome data and selective reporting of outcomes. There was high risk of attrition bias in several studies, which had a high risk of affecting outcome data, particularly in such small studies. We downgraded the evidence by at least one level for all our outcomes related to these studies due to serious concerns with the risk of bias. 

Several studies had restrictive entry criteria which meant that the evidence obtained may not directly answer our review question. Particularly several of the vitamin D studies had criteria that specified low vitamin D levels for enrolment, specifically 25‐OHD levels at baseline (Khan 2017Rastelli 2011), particularly 25‐OHD levels of 40 ng/mL or less for Khan 2017 and between 10 ng/mL and 29 ng/mL for Rastelli 2011. We downgraded the evidence by at least one level for all of our outcomes related to these studies due to indirectness, as these studies investigated a patient population that is not representative of all people with AIMSS. A number of outcomes were also downgraded for indirectness due to studies including patient populations that were not well‐defined for AIMSS. For example, one study included people experiencing musculoskeletal symptoms regardless of temporal association with the start of the AI, which resulted in a patient population that may have had pre‐existing musculoskeletal conditions rather than AIMSS.

There was no statistical heterogeneity in the single meta‐analysis performed in this review. However, there was significant clinical heterogeneity between interventions investigated in the review, which resulted in downgrading of the evidence for inconsistency for outcomes that incorporated multiple clinically heterogeneous interventions. 

Four of the 17 studies have been published only in abstract form (Birrell 2009Massimino 2011Rosati 2011Shenouda 2019). A considerable period of time has elapsed since the publication of three of these studies (Birrell 2009Massimino 2011Rosati 2011), and we were unable to obtain data. Some of these outcomes were of significant interest to this review; however, these were unable to be obtained. One study of etoricoxib had an FDA safety alert issued for the class of drug (COX‐2 inhibitors) during the study leading to high rates of participant withdrawal (Rosati 2011). Missing outcomes raise concerns about the overall quality of the evidence, and the potential for influence of publication bias on our outcomes. The only outcome with enough studies to warrant investigation with a funnel plot for evidence of publication bias confirmed our suspicions of the presence of publication bias. Concerns around publication bias led to downgrading of the evidence by one level.

Potential biases in the review process

This review undertook a comprehensive search strategy led by an Information Specialist (KR), with no language limitation. This is a strength of this review. Several authors (KER, SC, IA and NW) independently screened reference lists of all included studies and any systematic reviews (two authors for all references). We handsearched conference and meeting abstracts for the relevant organisations. Two review authors (IA, SC, KER, NW) independently performed data extraction and risk of bias assessments on each study. The search strategy should have identified most studies, however all studies identified were published in English language journals. Although several studies were conducted in non‐English speaking countries (e.g. China (Li 2017Liu 2014Peng 2018) and Brazil (Sordi 2019)), and were published in English, this observation does raise the possibility of a potential bias of missed publications in languages other than English.

The inconsistent definition of AIMSS (Hershman 2015bNiravath 2013), and the potentially wide definition of systemic therapies may have been potential sources of bias in the search strategy. A wide search strategy was employed to attempt to account for these potential biases. All potential generally accepted definitions of systemic therapies were included in the search strategy, as were all AI in current clinical practice.

We contacted 15 study authors for further information. We received replies from four authors. A limitation of this review is that certain data were unavailable, which introduced selection bias. Certain outcomes were not available for meta‐analysis, due to our inability to obtain further data on request.

We did not systematically evaluate the measurement instruments for AIMSS in this review due to resource constraints. This is a potential limitation of our review; however, there is significant variability in the outcome assessments for AIMSS and no consensus (Hershman 2015bNiravath 2013). Assessment of outcome measurements would therefore need to be interpreted within this clinical and research heterogeneity.

Agreements and disagreements with other studies or reviews

This Cochrane Review is the first of systemic therapies for prevention or treatment of AIMSS in postmenopausal women with early breast cancer. We searched other studies and reviews (systematic reviews and meta‐analyses) for comparison with this review. A broad search strategy identified the references, and we handsearched the identified studies for systematic reviews and meta‐analyses on the research question. The same reviews for treatment of AIMSS (which included systemic therapies and exercise) (Kim 2018Nahm 2018Roberts 2017Yang 2017) were also identified by several of the current authorship team involved in a previous Cochrane Review of exercise for the prevention and management of AIMSS in early breast cancer (Roberts 2020), albeit the search date was earlier. One systematic review of systematic reviews (Kim 2018) identified the same three systematic reviews that included systemic therapies (Nahm 2018Roberts 2017Yang 2017). We excluded systematic reviews of acupuncture alone for AIMSS.

Roberts 2017 published a review entitled "Management of aromatase inhibitor induced musculoskeletal symptoms in postmenopausal early breast cancer: a systematic review and meta‐analysis". Several of the authors from the current Cochrane Review were also involved in this review. The author's search strategy was designed to include both prospective and retrospective clinical trials, including RCTs, cohort and case‐control studies and preventive trials of interventions for AIMSS including pharmacological, non‐pharmacological (including exercise), and CAM interventions. Meta‐analysis was conducted when there were sufficient data. Publications in English language only were considered. Studies of systemic therapy were grouped narratively into pharmacological therapies and CAM, and AIMSS outcomes reported. In contrast to our review, no meta‐analysis of any AIMSS outcomes was undertaken for systemic therapies. There was significant between‐study heterogeneity. Non‐randomised data was included in the overall systematic review of Roberts 2017 and the scope of studies included was broader, and hence this review is not directly comparable to our current Cochrane Review. In agreement with our review, evidence quality was determined to be poor overall, and there was limited evidence found to recommend systemic therapies for management of AIMSS. In contrast to our current Cochrane Review, Roberts 2017 undertook no reporting or analysis of stiffness, QoL outcomes or adverse event reporting.

The review by Yang 2017 entitled "Interventions for the treatment of aromatase inhibitor associated arthralgia in breast cancer survivors. A systematic review and meta‐analysis" included all studies that were RCTs and "quasi‐experimental design". The primary outcome was pain mean score as assessed by BPI at the end time point of the intervention. Yang 2017 separated the studies into "pharmacological approaches" and "nutritional supplementation". Four non‐randomised studies were said to be included in pharmacological approaches in the meta‐analysis (Briot 2010Henry 2011Kubo 2012Zhang 2010), with pharmacological approaches reported to show "a very large effect in improving pain" (SMD −1.186, 95% CI −2.312 to −0.060). This subgroup meta‐analysis, with the inclusion of non‐randomised data, is different to our Cochrane Review. Three studies of diverse nutritional supplements were included together in meta‐analysis including a single‐arm non‐randomised open‐label phase two trial of glucosamine with chondroitin (Greenlee 2013), and RCTs of O3‐FA (Hershman 2015a) and vitamin D (Rastelli 2011). Nutritional supplementation showed no significant effect on joint pain, although there was "a tendency to decrease joint pain" (SMD −0.124, 95% CI −0.341 to 0.092). The review by Yang 2017 differs from our Cochrane Review as we have not analysed such diverse systemic therapies together due their postulated differing mechanisms of action, and we have not included non‐randomised data. In addition, in contrast to our current Cochrane Review, Yang 2017 did not undertake adverse event reporting.

In contrast to our current review, the systematic review by Nahm 2018 performed no meta‐analyses. This systematic review is not directly comparable to our current Cochrane Review as Nahm 2018 included all management interventions for AIMSS, and included non‐ randomised studies. In agreement with our review, the authors concluded, "While several trials had positive findings, the major methodological limitations of the studies meant that no definitive evidence could be found supporting any of the interventions".

Kim 2018 performed the most recent review with a search date of June 2018. Kim 2018 systematically reviewed all eligible systematic reviews and then subjected these to evidence mapping. The RCTs included in the reviews were handsearched for network meta‐analysis. The search strategy, statistical methodology and included studies therefore are different to our current Cochrane Review. Mean and SD of BPI was the outcome measure. Kim 2018 identified six trials that provided information on BPI for inclusion in their network meta‐analysis. Two of the six RCT included in the network meta‐analysis were systemic therapy studies, which were an RCT of O3‐FA (Lustberg 2018), and vitamin D (Rastelli 2011). These RCTs were not combined but compared separately to wait list control. Kim 2018 concluded "omega‐3 fatty acids (MD −2.10, 95% CI −3.23 to −0.97) showed statistically significant improvement in pain severity scores". However, a larger RCT of O3‐FA by Hershman 2015a of 262 participants which had BPI‐SF as a primary endpoint was not included in this network meta‐analysis, which is in contrast to our current Cochrane Review which included Hershman 2015a. For network meta‐analysis, the authors concluded "all interventions had a better effect than vitamin D" with only the trial by Rastelli 2011 included in their network meta‐analysis. The review by Kim 2018 is not comparable to our Cochrane Review as in contrast to our current review, it did not include vitamin D studies by Khan 2017Shapiro 2016 (did not use BPI as an endpoint) and Niravath 2019 (published subsequently). The conclusions of Kim 2018 are not in agreement with our Cochrane Review findings. Adverse event reporting was concluded to be poor, and as a result of this, Kim 2018 did not perform network meta‐analysis of adverse events. We found that adverse event reporting was overall of very low‐certainty evidence, and data were not available from certain studies.

Study flow diagram.

Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias domain, for each included trial. 

Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias domain, for each included trial. 

Risk of bias summary: review authors' judgements about each risk of bias item, presented as percentages across all included trials.

Figuras y tablas -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item, presented as percentages across all included trials.

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Figuras y tablas -
Figure 4

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Funnel plot of comparison: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.1 Pain.

Figuras y tablas -
Figure 5

Funnel plot of comparison: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.1 Pain.

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.5: Health‐related quality of life.

Figuras y tablas -
Figure 6

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2.5: Health‐related quality of life.

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Figuras y tablas -
Analysis 1.1

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2: Grip strength

Figuras y tablas -
Analysis 1.2

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2: Grip strength

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 3: Discontinuation of aromatase inhibitors

Figuras y tablas -
Analysis 1.3

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 3: Discontinuation of aromatase inhibitors

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 4: Incidence of AIMSS

Figuras y tablas -
Analysis 1.4

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 4: Incidence of AIMSS

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 5: Breast cancer‐specific quality of life

Figuras y tablas -
Analysis 1.5

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 5: Breast cancer‐specific quality of life

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 6: Health‐related quality of life (HRQoL)

Figuras y tablas -
Analysis 1.6

Comparison 1: Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 6: Health‐related quality of life (HRQoL)

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Figuras y tablas -
Analysis 2.1

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 1: Pain

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2: Stiffness

Figuras y tablas -
Analysis 2.2

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 2: Stiffness

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 3: Grip strength

Figuras y tablas -
Analysis 2.3

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 3: Grip strength

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 4: Breast cancer‐specific quality of life

Figuras y tablas -
Analysis 2.4

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 4: Breast cancer‐specific quality of life

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 5: Health‐related quality of life

Figuras y tablas -
Analysis 2.5

Comparison 2: Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS), Outcome 5: Health‐related quality of life

Summary of findings 1. Summary of findings table ‐ Systemic therapy compared to control for treating aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Systemic therapy compared to control for treating aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Patient or population: women with early breast cancer
Setting:
Intervention: systemic therapy
Comparison: control

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with control

Risk with systemic therapy

Change in pain from baseline to end of intervention
assessed with: Brief Pain Inventory (BPI) worst pain; BPI severity
follow‐up: 24 weeks

There were 2 studies (omega‐3 fatty acids, vitamin D) that showed a treatment effect with 95% CI that did not include a minimal clinically important difference (MCID) for BPI pain scale.

183
(2 RCTs)

⊕⊝⊝⊝
Very lowa,b,c

The evidence is very uncertain about the effect of systemic therapies on change in pain from baseline to end of intervention.

Change in grip strength from baseline to end of intervention
follow‐up: 24 weeks

85 per 1000

91 per 1000
(31 to 268)

RR 1.08
(0.37 to 3.17)

137
(1 RCT)

⊕⊕⊝⊝
Lowb,c

The evidence suggests that systemic therapies results in little to no difference in grip strength from baseline to end of intervention.

Safety of systemic therapies in AIMSS

1 study had a US Food and Drug Administration alert issued for the class of drug which the study drug belonged to (cyclo‐oxygenase‐2 inhibitors), but the study reported no serious adverse effects. No serious adverse events noted in any study.

344
(4 RCTs)

⊕⊝⊝⊝
Very lowc,d,e

The evidence is very uncertain about the effect of systemic therapies on safety of systemic therapies in AIMSS.

Effect on discontinuation of aromatase inhibitors (AI)
follow‐up: 24 weeks

39 per 1000

6 per 1000
(0 to 116)

RR 0.16
(0.01 to 2.99)

147
(1 RCT)

⊕⊕⊝⊝
Lowb,c

The evidence suggests that systemic therapies results in little to no difference in effect on discontinuation of AIs.

Effect on breast cancer‐specific quality of life (BCS‐QoL)
assessed with: Functional Assessment of Cancer Therapy – Breast (FACT‐B)
follow‐up: 24 weeks

1 study (omega‐3 fatty acids) showed a treatment effect with 95% CI that did include an MCID for this outcome measure.

44
(1 RCT)

⊕⊕⊝⊝
Lowa,c

The evidence suggests that systemic therapies results in little to no difference in effect on BCS‐QoL.

Health‐related quality of life (HRQoL) ‐ HRQoL: Total Functional Assessment of Cancer Therapy – General(FACT‐G) score
assessed with: Functional Assessment of Cancer Therapy – General (FACT‐G)
follow‐up: 24 weeks

A single study (omega 3 fatty acids) showed a treatment effect with 95% CI which did include a MCID for this outcome measure.

(1 RCT)

⊕⊕⊝⊝
Lowa,c

The evidence suggests that systemic therapies results in little to no difference in effect on HRQoL.

Incidence of AIMSS
follow‐up: range 24 weeks to 52 weeks

537 per 1000

440 per 1000
(338 to 569)

RR 0.82
(0.63 to 1.06)

240
(2 RCTs)

⊕⊝⊝⊝
Very lowb,c,d

The evidence is very uncertain about the effect of systemic therapies on change in Incidence of AIMSS.

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference; RR: risk ratio

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

See interactive version of this table: https://gdt.gradepro.org/presentations/#/isof/isof_question_revman_web_424110183232321857.

a High risk of reporting bias and attrition bias in single study. Downgraded one level.
b Downgraded one level for indirectness (restricted population dependent on vitamin D level).
c Downgraded one level for imprecision (small sample sizes).
d Downgraded one level for risk of bias (high risk of bias across multiple domains).
e High suspicion of publication bias (too few studies for funnel plot; one study in abstract form only).

Figuras y tablas -
Summary of findings 1. Summary of findings table ‐ Systemic therapy compared to control for treating aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer
Summary of findings 2. Summary of findings table ‐ Systemic therapy compared to control for preventing aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Systemic therapy compared to control for preventing aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer

Patient or population: health problem or population
Setting:
Intervention: Systemic therapy
Comparison: Control

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with Control

Risk with Systemic therapy

Change in pain from baseline to end of intervention
follow‐up: range 30 days to 6 months

4 studies showed a minimal clinically important difference (MCID) in pain scores that fell within the 95% CIs of the measured effect (calcitonin, bionic tiger bone, Yi Shen Jian Gu granules and vitamin D). 6 studies showed a treatment effect with 95% CIs that did not include an MCID (testosterone, vitamin D, duloxetine, omega‐3 fatty acids, emu oil, Cat's claw). Due to the variation in systemic therapies, including pharmacological, complementary and alternative medicines, the studies could not be combined for meta‐analysis.

1099
(10 RCTs)

⊕⊝⊝⊝
Very lowa,b,c,d

The evidence is very uncertain about the effect of systemic therapies on change in pain from baseline to the end of intervention in the treatment of AIMSS.

Change in stiffness from baseline to end of intervention

2 studies showed a minimal clinically important difference (MCID) in stiffness scores that fell within the 95% CIs of the measured effect (bionic tiger bone, Yi Shen Jian Gu granules). 2 studies that showed a treatment effect with 95% CIs that did not include an MCID (vitamin D, emu oil).

295
(4 RCTs)

⊕⊝⊝⊝
Very lowb,c,e,f

The evidence is very uncertain about the effect of systemic therapies on change in stiffness from baseline to the end of the intervention in the treatment of AIMSS.

Grip strength ‐ Vitamin D

There was a single study which did not show a MCID for grip strength which falls within the 95% CI for the measured effect (vitamin D).

(1 RCT)

⊕⊕⊝⊝
Lowc,f

The evidence is uncertain about the effect of systemic therapies on change in grip strength from baseline to the end of the intervention in the treatment of AIMSS 'little to no effect'

Effect on breast cancer‐specific quality of life (BCS‐QoL)

2 studies investigated the effect on BCS‐QoL (bionic tiger bone, Yi Shen Jian Gu granules). All subscales of the same quality of life tool utilised in both of these studies (Functional Assessment of Cancer Therapy – Breast (FACT‐B)) showed an MCID for this tool that falls within the 95% CIs of the measured effect.

147
(2 RCTs)

⊕⊝⊝⊝
Very lowa,g,h

The evidence is very uncertain about the effect of systemic therapies on effect on BCS‐QoL from baseline to the end of the intervention in the treatment of AIMSS.

Effect on health‐related quality of life (HRQoL)

2 studies investigated the effect of BCS‐QoL (bionic tiger bone, Yi Shen Jian Gu granules) using the Functional Assessment of Cancer Therapy – General (FACT‐G) tool. All subscales of this quality of life tool showed an MCID for this tool that fell within the 95% CIs of the measured effect. 1 study used the 36‐item Short Form (SF‐36) and showed most individual subscales in this outcome showing effect size that did not include MCID for this tool within the 95% CIs of the measured effect.

208
(3 RCTs)

⊕⊝⊝⊝
Very lowa,b,g,h

The evidence is very uncertain about the effect of systemic therapies on HRQoL from baseline to the end of the intervention in the treatment of AIMSS.

Safety of systemic therapies for the treatment of AIMSS

There were no grade 4/5 adverse events reported in any studies. 1 study investigating duloxetine reported significantly more all‐grade adverse events in the systemic therapy arm (78%) than the control arm (50%) (P < 0.001).

1250
(10 RCTs)

⊕⊝⊝⊝
Very lowa,c,i

The evidence is very uncertain about the safety data in the use of systemic therapies for the treatment of AIMSS.

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

See interactive version of this table: https://gdt.gradepro.org/presentations/#/isof/isof_question_revman_web_424113354545582340.

a Downgraded one level for risk of bias (high risk of bias across multiple domains).
b Downgraded one level for inconsistency (heterogeneity in interventions).
c Downgraded one level for imprecision (small sample sizes and unable to be combined for meta‐analysis).
d Downgraded one level for publication bias (funnel plot displayed asymmetry; multiple studies only written in abstract form).
e Downgraded one level for risk of bias (one study had high risk of both performance and detection bias).
f Downgraded one level for indirectness (one study with poorly defined AIMSS).
g Downgraded one level for indirectness (no criteria for an AIMSS in one study and exclusion of bisphosphonates, which are frequently utilised in the target population).
h Downgraded for imprecision (small sample size).
i Downgraded one level for publication bias (multiple studies published in abstract form only).

Figuras y tablas -
Summary of findings 2. Summary of findings table ‐ Systemic therapy compared to control for preventing aromatase inhibitor‐induced musculoskeletal symptoms in women with early breast cancer
Table 1. Prevention studies: pain

Study

Intervention vs control

Treatment duration

Intervention

Control

Baseline pain, mean (SD)

Change from baseline, mean (SD)

n

Change from baseline, mean (SD)

n

Outcome measure (scale)

Khan 2017

Vitamin D3 30,000 IU weekly vs placebo

24 weeks

2.39 (2.34)

0.8 

67

0.86 

72

BPI worst pain

(0–10)

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of USA diet

24 weeks

1.1 (1.55)

0.11 (1.88*)

22

−0.70 (1.88*)

22

BPI severity 

(0–10)

*Calculated from standard error (SE). SE of change scores in intervention arm 0.40; SE of change scores in control arm 0.41.
BPI: Brief Pain Inventory; IU: international units; n: number of participants; SD: standard deviation.

Figuras y tablas -
Table 1. Prevention studies: pain
Table 2. Prevention: safety

Studies

Intervention vs control

Treatment duration

Intervention

Control

Safety reporting

n

Safety reporting

n

Khan 2017

Vitamin D3 30,000 IU weekly vs placebo

24 weeks

No AEs attributed to Vitamin D

70

1 case of hypercalcaemia. No discussion of other AEs

77

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of the American diet

24 weeks

1 grade 3 AE (diarrhoea)

No grade 4/5 AEs

 

22

No grade 3/4/5 AEs

 

22

Niravath 2019

Vitamin D3 50,000 IU weekly for 12 weeks then 2000 daily for 40 weeks vs 800 IU daily

52 weeks

No grade 4/5 events

8 grade 3 events, deemed unrelated to study drugs

44

No grade 4/5 events

4 grade 3 events, deemed unrelated to study drugs

43

Rosati 2011

Etoricoxib (COX‐2 inhibitor) 60 mg/day vs placebo

2 years

FDA alert on COX‐2 inhibitors

Study reported 0 serious AE

16 

No report of safety data in control arm
 

50
 

AE: adverse event; COX: cyclo‐oxygenase; FDA: Food and Drug Administration; IU: international unit; n: number of participants.

Figuras y tablas -
Table 2. Prevention: safety
Table 3. Prevention: breast cancer‐specific quality of life (BCS‐QoL)

Study

Intervention vs control

Treatment duration

Intervention

Control

MD (95% CI)

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline to end of intervention, mean (SD)

n

Change from baseline to end of intervention, mean (SE)

n

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of the USA diet

24 weeks

119

(12.66)

 

2.5 (10.79)

22

0.95 (10.79)

22

1.55 (−4.83 to 7.93)

FACT‐B

(0–148) 

SD calculated from SE. Baseline intervention arm SE = 2.7; change score SE = 2.3.
FACT‐B: Functional Assessment of Cancer Therapy – Breast; MD: mean difference; n: number of participants; SD: standard deviation; SE: standard error.

Figuras y tablas -
Table 3. Prevention: breast cancer‐specific quality of life (BCS‐QoL)
Table 4. Prevention: health‐related quality of life (HRQoL)

Study

Intervention vs control

Treatment duration

FACT‐G subscale

Intervention

Control

Difference in change scores between arms, MD (95% CI)

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline to end of intervention, mean (SD)

n

Change from baseline to end of intervention, mean (SD)

n

Lustberg 2018

Omega‐3 fatty acids vs placebo containing mixture of fats and oils typical of the American diet

24 weeks

FACT‐G overall score

92 (10.32) 

1.9 (7.97)

22 

0.047 (8.44)

22 

1.85 (−3.00 to 6.70)

FACT‐G overall score 

(0–108) 

Physical Well‐being 

25 (2.77) 

−0.027 (3.24) 

22 

0.015 (3.33)

22 

−0.04 (−1.98 to 1.90)

FACT‐G physical well‐being subscale 

(0–28)

Social Well‐being

24 (4.5) 

0.39 (5.16)

22 

−0.96 (5.16)

22 

1.35 (−1.70 to 4.40)

FACT‐G social well‐being subscale 

(0–28)

Emotional Well‐being

20 (2.53) 

0.76 (2.81)

 22

 0.36 (2.86)

 22

0.40 (−1.28 to 2.08)

FACT‐G emotional well‐being subscale

 (0–24)

Functional Well‐being

 23 (5.16)

1.1 (3.19)

22 

 0.72 (3.28)

22

0.38 (−1.53 to 2.29)

FACT‐G functional well‐being subscale 

(0–28)

SD calculated from standard error (SE).
CI: confidence interval; FACT‐B: Functional Assessment of Cancer Therapy – Breast; FACT‐G: Functional Assessment of Cancer Therapy – General (higher scores equate to better quality of life); SD: standard deviation.

Figuras y tablas -
Table 4. Prevention: health‐related quality of life (HRQoL)
Table 5. Prevention: incidence of aromatase inhibitor‐induced musculoskeletal symptoms

Study

Intervention vs control

Treatment duration

Intervention

Control

Incidence of AIMSS (%)

n

Incidence of AIMSS (%)

n

Khan 2017

Vitamin D3 30,000 IU weekly vs placebo

24 weeks

(37%)

70

(51%) 

77 

Niravath 2019

Vitamin D3 50,000 IU weekly for 12 weeks then 2000 IU daily for 40 weeks vs 800 IU daily

52 weeks

25 (54%) 

47 

27 (57%) 

46 

Rosati 2011

Etoricoxib 60 mg/day vs placebo

2 years

(31%)

 16

(76%)

 50

AIMSS: aromatase inhibitor‐induced musculoskeletal symptoms; IU: international units; n: number of participants.

Figuras y tablas -
Table 5. Prevention: incidence of aromatase inhibitor‐induced musculoskeletal symptoms
Table 6. Treatment: pain

 Studies

Intervention vs control

Treatment duration

Intervention

Control

Difference in change scores between arm, mean

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline, Mean (SD)

Change from baseline, mean (SD)

n

Cathcart‐Rake 2020

Testosterone: 120 mg subcutaneous pellets, changed to topical daily gel due to slow accrual vs placebo

6 months

5.4 (1.7)

−1.9 (2.2)

80

−2.2 (2.7)

77

0.3

BPI‐AIA

mean pain

(0–10)

Shapiro 2016

Vitamin D3 4000 IU daily vs 600 IU daily

6 months

10.8 (3)

−1.2 (3.8)

39

−0.6 (3.8)

36

−0.6

WOMAC pain

(0–20)

Hershman 2015a

Omega‐3 fatty acids 3.3 g daily vs soybean/cornoil placebo

24 weeks

 7.06 (1.53a)

−2.23 

94

−1.81

98

−0.42 

BPI worst pain

(0–10)

Peng 2018 

Yi Shen Jian Gu granules twice daily vs placebo
 

12 weeks

6.18 (1.58)

−3.10

40

−1.65 

37

−1.47

BPI‐SF

worst pain

(0–10)

Chan 2017
 

Emu oil topically 3 times daily vs placebo

8 weeks

N/A

−0.82 (1.9a)

36

−0.93 (1.7a)

37

0.11 

BPI severity (0–10)

Sordi 2019

Cat's claw (Uncaria tomentosa

3 times daily vs placebo

 30 days

 8 (2.1)

0

32

−3

29

3

VAS

(0–10)

 Li 2017

Bionic tiger bone capsules 3 times daily vs calcium carbonate 600 mg daily

12 weeks

6.59 (2.11)

−2.72 

35

0.87

35

−3.59

BPI worst pain 

(0–10)

Henry 2018

Duloxetine 30 mg daily for 1 week then 60 mg daily for 11 weeks vs placebo
 

12 weeks

5.53 (N/A)

−2.6 

127

−1.97 

128

−0.63

BPI mean pain 

(0–10)

Rastelli 2011

Vitamin D2: either 50,000 IU weekly for 8 weeks if vitamin D insufficiency then monthly; or 50,000 IU weekly for 16 weeks if vitamin D deficiency then monthly; or control, 400 IU daily

6 months

5.2 (2.4)

−1.5 

28

−0.9

29

−0.4

BPI worst pain

(0–10)

Liu 2014

Calcitonin 200 IU + 600 mg daily caltrate D vs 600 mg daily caltrate D

 3 months

 5.0 (0.74)

−3.00 (1.48)

42

−1.00 (1.11)

40

−2

VAS scale 

(0–10)

Where SD is not available, this was due to lack of change score SD reported in the study. For these studies, end of treatment MD were used in the analysis.
Rastelli 2011: 2 months of BPI data used to eliminate differences between dosing in Stratum A and B.
Liu 2014: although study reported mean (SD) being utilised, data more consistent with median and interquartile ranges. Data were treated as such.
aSD calculated from provided confidence intervals.
BPI: Brief Pain Inventory; BPI‐AIA: Brief Pain Inventory for Aromatase Inhibitor Arthralgia; BPI‐SF: Brief Pain Inventory – Short Form; IU: international unit; MD: mean difference; n: number of participants; N/A: not available; SD: standard deviation; VAS: Visual Analogue Scale.

Figuras y tablas -
Table 6. Treatment: pain
Table 7. Treatment: stiffness

Study

Intervention vs control

Treatment duration

Intervention

Control

Difference in change scores between arms, MD (95% CI)

Outcome measure

(scale)

Baseline pain, mean (SD)

Change from baseline to end of intervention, mean (SD)

n

Change from baseline to end of intervention, mean (SD)

n

Shapiro 2016

Vitamin D3 4000 IU daily vs 600 IU daily

6 months

5.5 (1.5)

−0.5 (1.7)

39

−0.5 (2.0)

36

0 (−0.82 to 0.82) 

 WOMAC

(0–8)

Hershman 2015a
 

Omega‐3 fatty acids 3.3 g daily vs soybean/cornoil placebo

24 weeks

N/Aa

0.59 (1.25)b

109

0.46 (1.25)b

114

0.13 (−0.20 to 0.46)

GRCS at 6 weeks

Peng 2018
 

Yi Shen Jian Gu granules twice daily vs placebo
 

12 weeks

70.43 (43.49)

−37.53 

40

−20.82 

37

−16.71 
 

WOMAC

(0–200)

Li 2017
 

Bionic tiger bone capsules 3 times daily vs calcium carbonate 600 mg daily

12 weeks

5.49 (1.58)

−3.08 

35

1.87 

35

 −4.95 

Modified BPI with stiffness 

(0–10)

Henry 2018
 

Duloxetine 30 mg daily for 1 week then 60 mg daily for 11 weeks

12 weeks

N/Aa

OR 3.38 (1.85 to 6.18; P = 0.0001)c

Chan 2017
 

Emu oil topically 3 times daily vs placebo

8 weeks

1.9 (0.67)

−0.31 (0.82)

36

−0.41 (0.82)

37

 0.10 (−0.28 to 0.48)

VAS scale 

(0–3)

a Evaluates changes in symptoms since last visit, so no baseline scores were collected.

bSome standard deviations (SD) not available, due to lack of reported change score SD. For these studies, end of treatment means (SD) were used in the analysis of effect size. See Analysis 2.2.
cReported as a binary outcome, OR. 
Henry 2018Hershman 2015a could not be assessed for the effect of stiffness on the treatment of AIMSS, including confidence intervals and, therefore, were not included in the final assessment of this outcome.
BPI: Brief Pain Inventory; CI: confidence interval; GRCS: Global Rating of Change Scale; IU: international unit; MD: mean difference; n: number of participants; N/A: not available/applicable; OR: odds ratio; SD: standard deviation; VAS: Visual Analogue Scale; WOMAC: Western Ontario and McMaster Universities Osteoarthritis scale.

Figuras y tablas -
Table 7. Treatment: stiffness
Table 8. Treatment: breast cancer‐specific quality of life (BCS‐QoL)

Studies

Intervention vs control

Treatment duration

FACT‐B subscale 

(scale)

Intervention

Control

Difference in change scores between arms, MD 

Baseline BCS‐QoL, mean (SD)

Change from baseline, mean (SD)

n

Change from baseline, mean (SD)

n

Peng 2018

Yi Shen Jian Gu granules twice daily vs placebo

 12 weeks

Physical Well‐being (0–28)

18.85 (4.53)

4.77 

 40

 3.41

 37

2.46

Social/Family Well‐being (0–24)

16.95 (4.62)

3.1 

 2.54

2.27

Emotional Well‐being (0–24)

21.78 (4.31)

21.78 

 1.40

1.54

Functional Well‐being (0–28)

17.15 (4.42)

4.26 

 3 

4.27

Additional Concerns (0–36)

23.58 (4.93)

2.93 

 1.84 

2.51

Li 2017
 

Bionic tiger bone capsules 3 times daily vs calcium carbonate 600 mg daily

 12 weeks

 Physical Well‐being (0–28)

19.23 (5.02)

35

 −0.1 

35

2.30

Social/Family Well‐being (0–24)

19.81 (3.98)

0.93 

−0.97 

0.90

Emotional Well‐being (0–24)

15.34 (4.41)

0.49 

 0.12 

0.81

Functional Well‐being (0–28)

15.26 (4.67)

0.71 

 −0.09 

0.10

Additional Concerns (0–36)

26.73 (4.61)

0.73 

 0.75 

−0.40

Where SD are not available for change scores, end of treatment means (SD) were used in the analysis, see Analysis 2.4.
In FACT scales, higher scores equate to better quality of life.
BCS‐QoL: breast cancer‐specific quality of life; FACT‐B: Functional Assessment of Cancer Therapy – Breast; n: number of participants; SD: standard deviation.

Figuras y tablas -
Table 8. Treatment: breast cancer‐specific quality of life (BCS‐QoL)
Comparison 1. Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Pain Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.2 Grip strength Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.3 Discontinuation of aromatase inhibitors Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.4 Incidence of AIMSS Show forest plot

2

240

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.63, 1.06]

1.5 Breast cancer‐specific quality of life Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6 Health‐related quality of life (HRQoL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6.1 HRQoL: Total Functional Assessment of Cancer Therapy – General (FACT‐G) score

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6.2 FACT‐G Physical Well‐being subscale

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6.3 FACT‐G Social Well‐being subscale

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6.4 FACT‐G Emotional Well‐being subscale

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.6.5 FACT‐G Functional Well‐being subscale

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 1. Prevention of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS)
Comparison 2. Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Pain Show forest plot

10

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

2.1.1 Testosterone

1

157

Mean Difference (IV, Fixed, 95% CI)

0.30 [‐0.47, 1.07]

2.1.2 Vitamin D

2

132

Mean Difference (IV, Fixed, 95% CI)

‐1.24 [‐2.16, ‐0.32]

2.1.3 Duloxetine

1

255

Mean Difference (IV, Fixed, 95% CI)

‐0.63 [‐0.97, ‐0.29]

2.1.4 Calcitonin

1

82

Mean Difference (IV, Fixed, 95% CI)

‐2.00 [‐2.56, ‐1.44]

2.1.5 Omega‐3 fatty acids

1

192

Mean Difference (IV, Fixed, 95% CI)

‐0.24 [‐1.00, 0.52]

2.1.6 Bionic tiger bone

1

70

Mean Difference (IV, Fixed, 95% CI)

‐4.09 [‐5.25, ‐2.93]

2.1.7 Emu oil

1

73

Mean Difference (IV, Fixed, 95% CI)

0.11 [‐0.72, 0.94]

2.1.8 Yi Shen Jian Gu granules

1

77

Mean Difference (IV, Fixed, 95% CI)

‐1.34 [‐2.10, ‐0.58]

2.1.9 Cat's claw (Uncaria tomentosa)

1

61

Mean Difference (IV, Fixed, 95% CI)

3.00 [1.53, 4.47]

2.2 Stiffness Show forest plot

5

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.2.1 Vitamin D

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.2.2 Omega‐3 fatty acids

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.2.3 Yi Shen Jian Gu granules

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.2.4 Bionic tiger bone

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.2.5 Emu oil

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.3 Grip strength Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.3.1 Vitamin D

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.4 Breast cancer‐specific quality of life Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.4.1 Physical Well‐being

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.4.2 Social/ Family Well‐being

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.4.3 Emotional Well‐being

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.4.4 Functional Well‐being

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.4.5 Additional Concerns

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

2.5 Health‐related quality of life Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.1 FACT Physical Well‐being

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.2 FACT Social/Family Well‐being

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.3 FACT Emotional Well‐being

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.4 FACT Functional Well‐being

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.5 SF36 Functional Capacity

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.6 SF36 Physical Limitations

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.7 SF36 Pain

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.8 SF36 Overall Health Status

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.9 SF36 Vitality

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.10 SF36 Social Aspects

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.11 SF‐36 Emotional Aspects

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.12 SF‐36 Mental Health

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 2. Treatment of aromatase inhibitor‐induced musculoskeletal symptoms (AIMSS)