Scolaris Content Display Scolaris Content Display

Tratamiento dirigido al factor de crecimiento endotelial vascular (VEGF) para el cáncer cervical persistente, recidivante o metastásico

Collapse all Expand all

Antecedentes

El cáncer de cuello uterino es la cuarta causa de muerte por cáncer en las mujeres. Históricamente, las mujeres con cáncer de cuello de útero metastásico o recidivante han tenido opcionesterapéuticas limitadas. Los nuevos tratamientos contra la angiogénesis, como los agentes dirigidos al factor de crecimiento endotelial vascular (VEGF), ofrecen una estrategia alternativa a la quimioterapia convencional, ya que actúan al inhibir el crecimiento de nuevos vasos sanguíneos, y limitan así el crecimiento del tumor al bloquear el suministro de sangre.

Objetivos

Evaluar los efectos beneficiosos y perjudiciales de los agentes dirigidos al VEGF en el tratamiento del cáncer cervical persistente, recidivante o metastásico.

Métodos de búsqueda

Se realizaron búsquedas en el Registro Cochrane central de ensayos controlados (Cochrane Central Register of Controlled Trials, CENTRAL), MEDLINE, Embase, registros en línea de ensayos clínicos y resúmenes de reuniones científicas hasta el 27 de mayo de 2020.

Criterios de selección

Se examinaron los ensayos controlados aleatorizados (ECA) que evaluaron la administración de agentes dirigidos al VEGF solos o en combinación con quimioterapia convencional u otros agentes dirigidos al VEGF.

Obtención y análisis de los datos

Dos autores de la revisión analizaron de forma independiente los resultados de las estrategias de búsqueda, extrajeron los datos, evaluaron el riesgo de sesgo y analizaron los datos según los métodos estándar previstos por Cochrane. La certeza de la evidencia se evaluó con el método GRADE.

Resultados principales

Se identificaron 1634 registros. De ellos, se identificaron cuatro estudios con 808 participantes para inclusión. También se identificaron dos estudios que estaban pendientes de clasificación y nueve estudios en curso.

Bevacizumab más quimioterapia versus quimioterapia

El tratamiento con bevacizumab más quimioterapia podría dar lugar a un menor riesgo de muerte en comparación con la quimioterapia sola (cociente de riesgos instantáneos [CRI] 0,77; intervalo de confianza [IC] del 95%: 0,62 a 0,95; un estudio, 452 participantes; evidencia de certeza baja). Sin embargo, es probable que haya más eventos adversos específicos en comparación con la quimioterapia sola, como perforaciones o fístulas gastrointestinales (razón de riesgos [RR] 18,00; IC del 95%: 2,42 a 133,67; un estudio, 440 participantes; evidencia de certeza moderada); eventos tromboembólicos graves (RR 4,5; IC del 95%: 1,55 a 13,08; un estudio, 440 participantes; evidencia de certeza moderada); e hipertensión (RR 13,75; IC del 95%: 5,07 a 37,29; un estudio, 440 participantes; evidencia de certeza moderada). También podría haber una mayor incidencia de hemorragias graves (RR 5,00; IC del 95%: 1,11 a 22,56; un estudio, 440 participantes; evidencia de certeza baja). Además, probablemente es mayor la incidencia de eventos adversos graves (RR 1,44; IC del 95%: 1,16 a 1,79; un estudio, 439 participantes; evidencia de certeza moderada). La relación coste‐efectividad incremental fue 295,164 dólares por año de vida ajustado por la calidad (un estudio, 452 participantes; evidencia de certeza baja).

Cediranib más quimioterapia versus quimioterapia

El tratamiento con cediranib más quimioterapia podría o no dar lugar a un riesgo similar de muerte en comparación con la quimioterapia sola (CRI 0,94; IC del 95%: 0,53 a 1,65; un estudio, 69 participantes; evidencia de certeza baja). Se encontraron resultados muy inciertos para las incidencias de eventos adversos específicos, que incluyen perforaciones o fístulas gastrointestinales (RR 3,27; IC del 95%: 0,14 a 77,57; un estudio, 67 participantes; evidencia de certeza muy baja); hemorragia grave (RR 5,45; IC del 95%: 0,27 a 109,49; un estudio, 67 participantes; evidencia de certeza muy baja); eventos tromboembólicos graves (RR 3,41; IC del 95%: 0,14 a 80,59; un estudio, 60 participantes; evidencia de certeza muy baja); e hipertensión grave (RR 0,36; IC del 95%: 0,02 a 8,62; un estudio, 67 participantes; evidencia de certeza muy baja). Además, podría haber o no una incidencia similar de eventos adversos graves en comparación con la quimioterapia sola (RR 1,15; IC del 95%: 0,75 a 1,78; un estudio, 67 participantes; evidencia de certeza baja).

Apatinib más quimioterapia o quimioterapia/braquiterapia versus quimioterapia o quimioterapia/braquiterapia

El tratamiento con apatinib más quimioterapia o quimioterapia/braquiterapia podría o no dar lugar a un riesgo similar de muerte en comparación con la quimioterapia sola o la quimioterapia/braquiterapia sola (CRI 0,90; IC del 95%: 0,51 a 1,60; un estudio, 52 participantes; evidencia de certeza baja). Sin embargo, los eventos de hipertensión podrían ocurrir con una mayor incidencia en comparación con la quimioterapia sola o la quimioterapia/braquiterapia sola (RR 5,14; IC del 95%: 1,28 a 20,73; un estudio, 52 participantes; evidencia de certeza baja).

Pazopanib más lapatinib versus lapatinib

El tratamiento con pazopanib más lapatinib podría dar lugar a un mayor riesgo de muerte en comparación con lapatinib solo (CRI 2,71; IC del 95%: 1,16 a 6,31; un estudio, 117 participantes; evidencia de certeza baja). Se encontraron resultados muy inciertos para las incidencias de eventos adversos específicos, que incluyen perforaciones o fístulas gastrointestinales (RR 2,00; IC del 95%: 0,19 a 21,59; un estudio, 152 participantes; evidencia de certeza muy baja); hemorragia (RR 2,00; IC del 95%: 0,72 a 5,58; un estudio, 152 participantes; evidencia de certeza muy baja); y eventos tromboembólicos (RR 3,00; IC del 95%: 0,12 a 72,50; un estudio, 152 participantes; evidencia de certeza muy baja). Además, la incidencia de eventos de hipertensión es probablemente mayor (RR 12,00; IC del 95%: 2,94 a 49,01; un estudio, 152 participantes; evidencia de certeza moderada). Podría haber o no una incidencia similar de eventos adversos graves en comparación con lapatinib solo (RR 1,45; IC del 95%: 0,94 a 2,26; un estudio, 152 participantes; evidencia de certeza baja).

Pazopanib versus lapatinib

El tratamiento con pazopanib podría o no dar lugar a un riesgo similar de muerte en comparación con lapatinib (CRI 0,96; IC del 95%: 0,67 a 1,38; un estudio, 152 participantes; evidencia de certeza baja). Se encontraron resultados muy inciertos para las incidencias de eventos adversos específicos, que incluyen perforaciones o fístulas gastrointestinales (RR 1,03; IC del 95%: 0,07 a 16,12; un estudio, 150 participantes; evidencia de certeza muy baja); hemorragia (RR 1,03; IC del 95%: 0,31 a 3,40; un estudio, 150 participantes; evidencia de certeza muy baja); y eventos tromboembólicos (RR 3,08; IC del 95%: 0,13 a 74,42; un estudio, 150 participantes; evidencia de certeza muy baja). Además, la incidencia de eventos de hipertensión es probablemente mayor (RR 11,81; IC del 95%: 2,89 a 48,33; un estudio, 150 participantes; evidencia de certeza moderada). El riesgo de eventos adversos graves podría o no ser similar en comparación con lapatinib (RR 1,31; IC del 95%: 0,83 a 2,07; un estudio, 150 participantes; evidencia de certeza baja).

Conclusiones de los autores

Se encontró evidencia de certeza baja a favor de la administración de bevacizumab más quimioterapia. Sin embargo, el bevacizumab probablemente aumenta los eventos adversos específicos (perforaciones o fístulas gastrointestinales, eventos tromboembólicos, hipertensión) y los eventos adversos graves. Se encontró evidencia de certeza baja que no apoya la administración de cediranib más quimioterapia, apatinib más quimioterapia, apatinib más quimioterapia/braquiterapia o monoterapia con pazopanib. Se encontró evidencia de certeza baja que indica que pazopanib más lapatinib empeora los desenlaces. Los inhibidores del VEGF apatinib y pazopanib podrían aumentar la probabilidad de eventos de hipertensión.

PICOs

Population
Intervention
Comparison
Outcome

The PICO model is widely used and taught in evidence-based health care as a strategy for formulating questions and search strategies and for characterizing clinical studies or meta-analyses. PICO stands for four different potential components of a clinical question: Patient, Population or Problem; Intervention; Comparison; Outcome.

See more on using PICO in the Cochrane Handbook.

Inhibición de la formación de vasos sanguíneos en el cáncer de cuello de útero avanzado

¿Cuál es el objetivo de esta revisión?
El objetivo de esta revisión Cochrane es determinar si los fármacos dirigidos al factor de crecimiento endotelial vascular (VEGF, por sus siglas en inglés), que inhiben la formación de nuevos vasos sanguíneos, pueden mejorar la supervivencia de las mujeres con cáncer de cuello uterino que se ha extendido a lugares distantes (metástasis) o que no ha respondido o ha reaparecido (recidiva) después del tratamiento inicial. Estos fármacos incluyen bevacizumab, cediranib, apatinib, pazopanib, anlotinib y nintedanib.

Mensajes clave
El bevacizumab más quimioterapia podría mejorar la supervivencia y probablemente aumenta los episodios adversos específicos y graves como las perforaciones gastrointestinales a través de la pared intestinal, la formación de coágulos de sangre (episodios tromboembólicos) en los vasos sanguíneos, la hipertensión (aumento de la presión arterial) y el sangrado (hemorragia).

Cediranib o apatinib más quimioterapia, o pazopanib solo, podrían suponer una diferencia mínima o nula en la supervivencia. Pazopanib más lapatinib podría reducir la supervivencia.

¿Cuáles son los principales resultados de la revisión?
Se encontraron cuatro ensayos controlados aleatorizados (ECA) que cumplieron los criterios de inclusión y reclutaron a 808 mujeres.

Se encontró un estudio que incluyó 452 mujeres y evaluó la administración de bevacizumab más quimioterapia versus quimioterapia sola. La inclusión de bevacizumab podría mejorar la supervivencia general y probablemente aumenta la incidencia de episodios adversos específicos y graves.

Un segundo estudio analizó 69 mujeres tratadas con cediranib más quimioterapia versus quimioterapia sola. El cediranib podría suponer una diferencia mínima o nula en la supervivencia, y no se sabe con certeza si aumenta la incidencia de episodios adversos específicos o graves.

Otro estudio con 59 mujeres aportó datos sobre la administración de apatinib más quimioterapia o quimiorradioterapia (quimioterapia y radioterapia concurrentes) versus quimioterapia o quimiorradioterapia solas. El apatinib podría suponer una diferencia mínima o nula en la supervivencia, pero mostró una eficacia prometedora en la supervivencia sin progresión.

Se encontró un estudio con 228 mujeres que comparó pazopanib más lapatinib versus lapatinib solo, o pazopanib versus lapatinib. Pazopanib más lapatinib podría reducir la supervivencia y probablemente aumenta la incidencia de hipertensión; pazopanib solo podría dar lugar a poca o ninguna diferencia en la supervivencia y probablemente aumenta la incidencia de hipertensión.

En general, la calidad (certeza) de la evidencia fue baja, ya que cada comparación solo incluyó un estudio y la mayoría de los estudios fueron pequeños.

Authors' conclusions

Implications for practice

Low‐certainty evidence suggests that the use of bevacizumab plus chemotherapy for women with persistent, recurrent, or metastatic cervical cancer prolonged median overall survival (OS) by 3.5 months as compared to chemotherapy alone. This benefit showed some heterogeneity and appeared to be greater in previously untreated women with pelvic disease, with cancers of the squamous histological type, and at medium/high risk by the Moore criteria (Moore 2010). Meanwhile, women should weigh any potential benefit versus increased serious and specific adverse events and should determine whether they would be able to afford the incremental costs.

Low‐certainty evidence does not support the use of cediranib plus chemotherapy, as it did not improve OS as compared to chemotherapy alone. The effects of other tyrosine kinase inhibitors (TKIs) of vascular endothelial growth factor (VEGF) receptor (apatinib, anlotinib, nintedanib) plus chemotherapy are being assessed in ongoing studies.

Low‐certainty evidence does not support the use of apatinib plus chemotherapy or chemotherapy/brachytherapy, as this treatment did not improve OS as compared to chemotherapy or chemotherapy/ brachytherapy alone.

Low‐certainty evidence does not support the use of pazopanib plus lapatinib, which reduced OS as compared to lapatinib monotherapy.

Low‐certainty certainty evidence does not support the use of pazopanib monotherapy, as it did not improve OS as compared to lapatinib monotherapy.

No certain conclusions can be drawn as yet from Ongoing studies looking at apatinib plus anti‐programmed death 1 antibody plus chemoradiotherapy, or apatinib monotherapy versus supportive care.

Implications for research

Persistent, recurrent, or metastatic cervical cancer is a heterogeneous disease with different patient or disease factors. Future studies should assess the drug responses of specific subgroups and should search for predictive biomarkers of response to VEGF targeting agents.

Most participants included in clinical trials discontinued treatment due to disease progression or toxicity. These women needed additional salvage therapy or supportive care until death. Future studies should assess the effects of subsequent line treatments on OS for these women and should fully evaluate quality of life outcomes for this group when treatment is largely provided with palliative intent.

Summary of findings

Open in table viewer
Summary of findings 1. Bevacizumab plus chemotherapy compared to chemotherapy

Bevacizumab plus chemotherapy compared to chemotherapy for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Settings: hospital

Intervention: bevacizumab plus chemotherapy

Comparison: chemotherapy only

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with the chemotherapy only

Risk with bevacizumab plus chemotherapy

Overall survival

791 per 1000

701 per 1000
(621 to 774)

HR 0.77
(0.62 to 0.95)

452
(1 RCT)

⊕⊕⊝⊝
lowa,b

Maximum follow‐up was longer than 50 months. Median overall survival in the bevacizumab groups was 16.8 months (mortality rate 170/227 participants). Median overall survival in the control groups was 13.3 months (mortality rate 178/225 participants)

Specificadverse events: gastrointestinal perforations or fistulae

5 per 1,000

82 per 1,000
(11 to 608)

RR 18.00
(2.42 to 133.67)

440
(1 RCT)

⊕⊕⊕⊝
moderatea

At least 18 women (a maximum of 23) had fistulae events (N = 18) or perforation events (N = 5) in the bevacizumab group

Specific adverse events: serious haemorrhage

9 per 1,000

45 per 1,000
(10 to 205)

RR 5.00
(1.11 to 22.56)

440
(1 RCT)

⊕⊕⊝⊝
lowa,b

Specific adverse events: serious thromboembolic events

18 per 1,000

82 per 1,000
(28 to 238)

RR 4.50
(1.55 to 13.08)

440
(1 RCT)

⊕⊕⊕⊝
moderatea

Specific adverse events: hypertension

18 per 1,000

250 per 1,000
(92 to 678)

RR 13.75
(5.07 to 37.29)

440
(1 RCT)

⊕⊕⊕⊝
moderatea

Serious adverse events

361 per 1,000

519 per 1,000
(418 to 646)

RR 1.44
(1.16 to 1.79)

439
(1 RCT)

⊕⊕⊕⊝
moderatea

Economic evaluation

see comment

see comment

452
(1 RCT)

⊕⊕⊝⊝
lowa,c

The incremental cost‐effectiveness ratio (ICER) was USD 295,164 per quality‐adjusted life‐year (QALY), or USD 24,597 per quality‐adjusted life‐month

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious heterogeneity. Outcomes might be different in different subgroups in the only included study.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by one for serious risk of bias. This analysis was not planned in the study protocol.

Open in table viewer
Summary of findings 2. Cediranib plus chemotherapy compared to placebo plus chemotherapy

Cediranib plus chemotherapy compared to placebo plus chemotherapy for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Settings: hospital

Intervention: cediranib plus chemotherapy

Comparison: placebo plus chemotherapy

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with placebo plus chemotherapy

Risk with cediranib plus chemotherapy

Overall survival

771 per 1000

750 per 1000
(543 to 912)

HR 0.94
(0.53 to 1.65)

69
(1 RCT)

⊕⊕⊝⊝
lowa,b

Median follow‐up was 24.2 months. Median overall survival in the cediranib group was 13.6 months (mortality rate 25/34 participants). Median overall survival in the placebo group was 14.8 months (mortality rate 27/35 participants)

Specific adverse events: gastrointestinal perforations or fistulae

0 per 1000

0 per 1000
(0 to 0)

RR 3.27
(0.14 to 77.57)

67
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: serious haemorrhage

0 per 1000

0 per 1000
(0 to 0)

RR 5.45
(0.27 to 109.49)

67
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: serious thromboembolic events

0 per 1000

0 per 1000
(0 to 0)

RR 3.41
(0.14 to 80.59)

60
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: serioushypertension

29 per 1000

10 per 1000
(1 to 246)

RR 0.36
(0.02 to 8.62)

67
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Serious adverse events

514 per 1000

591 per 1000
(386 to 915)

RR 1.15
(0.75 to 1.78)

67
(1 RCT)

⊕⊕⊝⊝
lowa,b

Economic evaluation ‐ not measured

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious heterogeneity. Outcomes might be different in different subgroups in the only included study.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by two for very serious imprecision. There were few events and the confidence interval included appreciable benefit and harm.

Open in table viewer
Summary of findings 3. Apatinib plus chemotherapy or chemotherapy/brachytherapy compared to chemotherapy or chemotherapy/brachytherapy

Apatinib plus chemotherapy or chemotherapy/brachytherapy compared with chemotherapy or chemotherapy/brachytherapy for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with recurrent or FIGO stage IVB cervical cancer

Settings: hospital

Intervention: apatinib plus chemotherapy or chemotherapy/brachytherapy

Comparison: chemotherapy or chemotherapy/brachytherapy

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with chemotherapy or chemotherapy/brachytherapy

Risk with apatinibplus chemotherapy or chemotherapy/brachytherapy

Overall survival

667 per 1000

628 per 1000
(161 to 429)

HR 0.90
(0.51 to 1.60)

52
(1 RCT)

⊕⊕⊝⊝
lowa,b

Median follow‐up was 14 months. Median overall survival in the apatinib group was 14.7 months (mortality rate 11/28 participants). Median overall survival in the control group was 12.8 months (mortality rate 16/24 participants)

Specific adverse events: gastrointestinal perforations or fistulae ‐ not reported

Specific adverse events: haemorrhage ‐ not reported

Specific adverse events: thromboembolic events ‐ not reported

Specific adverse events: hypertension

83 per 1000

428 per 1000
(107 to 1000)

RR 5.14
(1.28 to 20.73)

52
(1 RCT)

⊕⊕⊝⊝
lowa,c

Serious adverse events ‐ not reported

Economic evaluation ‐ not measured

*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; FIGO: International Federation of Obstetrics and Gynaecology; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious risk of bias. This single small study was funded by the drug manufacturer.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by one for serious risk of bias. Trial registration occurred later than trial completion.

Open in table viewer
Summary of findings 4. Pazopanib plus lapatinib compared to lapatinib

Pazopanib plus lapatinib compared to lapatinib for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Setting: clinic

Intervention: pazopanib plus lapatinib

Comparison: lapatinib only

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with lapatinib only

Risk with pazopanib plus lapatinib

Overall survival

172 per 1000

401 per 1000
(197 to 697)

HR 2.71
(1.16 to 6.31)

117
(1 RCT)

⊕⊕⊝⊝
lowa,b

Maximum follow‐up was longer than 40 weeks. Median overall survival in the pazopanib plus lapatinib group was 25.7 weeks (mortality rate 14/59 participants). Median overall survival in the lapatinib group was 35.0 weeks (mortality rate 10/58 participants)

Specific adverse events: gastrointestinal perforations or fistulae

13 per 1000

26 per 1000
(3 to 284)

RR 2.00
(0.19 to 21.59)

152
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: haemorrhage

66 per 1000

132 per 1000
(47 to 367)

RR 2.00
(0.72 to 5.58)

152
(1 RCT)

⊕⊝⊝⊝
verylowa,c

At least 10 women (maximum of 29) with 29 bleeding events were included in the pazopanib plus lapatinib group; at least 5 women (maximum of 11) with 11 bleeding events were included in the lapatinib group

Specific adverse events: thromboembolic events

0 per 1000

0 per 1000
(0 to 0)

RR 3.00
(0.12 to 72.50)

152
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: hypertension

26 per 1000

316 per 1000
(77 to 1000)

RR 12.00
(2.94 to 49.01)

152
(1 RCT)

⊕⊕⊕⊝
moderatea

Serious adverse events

289 per 1000

420 per 1000
(272 to 654)

RR 1.45
(0.94 to 2.26)

152
(1 RCT)

⊕⊕⊝⊝
lowa,b

A total of 5 women In the pazopanib plus lapatinib group had fatal adverse events

Economic evaluation ‐ not measured

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious risk of bias. This single small study was funded by the drug manufacturer.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by two for very serious imprecision. There were few events and the confidence interval included appreciable benefit and harm.

Open in table viewer
Summary of findings 5. Pazopanib compared to lapatinib

Pazopanib compared to lapatinib for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Setting: clinic

Intervention: pazopanib only

Comparison: lapatinib only

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with lapatinib only

Risk with pazopanib only

Overall survival

795 per 1000

781 per 1000
(654 to 888)

HR 0.96
(0.67 to 1.38)

152
(1 RCT)

⊕⊕⊝⊝
lowa,b

Maximum follow‐up was longer than 150 weeks. Median overall survival in the pazopanib group was 49.7 weeks (mortality rate 56/74 participants). Median overall survival in the lapatinib group was 44.1 weeks (mortality rate 62/78 participants)

Specific adverse events: gastrointestinal perforations or fistulae

13 per 1000

14 per 1000
(1 to 212)

RR 1.03
(0.07 to 16.12)

150
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: haemorrhage

66 per 1000

68 per 1000
(20 to 224)

RR 1.03
(0.31 to 3.40)

150
(1 RCT)

⊕⊝⊝⊝
verylowa,c

At least 5 women (maximum of 13) in the pazopanib group had 13 bleeding events; at least 5 women (maximum of 11) in the lapatinib group had 11 bleeding events

Specific adverse events: thromboembolic events

0 per 1000

0 per 1000
(0 to 0)

RR 3.08
(0.13 to 74.42)

150
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: hypertension

26 per 1000

311 per 1000
(76 to 1,000)

RR 11.81
(2.89 to 48.33)

150
(1 RCT)

⊕⊕⊕⊝
moderatea

Serious adverse events

289 per 1000

379 per 1000
(240 to 599)

RR 1.31
(0.83 to 2.07)

150
(1 RCT)

⊕⊕⊝⊝
lowa,b

Economic evaluation ‐ not measured

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious risk of bias. This single small study was funded by the drug manufacturer.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by two for very serious imprecision. There were few events and the confidence interval included appreciable benefit and harm.

Background

Description of the condition

Cervical cancer is the second most commonly diagnosed cancer affecting women worldwide and the third leading cause of death from cancer among women in low‐income countries (Bray 2018; Ferlay 2019; Siegel 2019). Almost 70% of the global burden falls on low‐ and middle‐income countries, in which 9 out of 10 cervical cancer deaths occur (Ferlay 2019). A woman's risk of developing cervical cancer before the age of 74 years is estimated to be 1.36% worldwide, ranging from 0.9% in more developed to 1.6% in less developed areas (Ferlay 2019; Torre 2015). The World Health Organization (WHO) recognises three pathological types of cervical cancer, including squamous cell carcinoma (70% to 80%), adenocarcinoma (20% to 25%), and other types (adenosquamous carcinoma, neuroendocrine tumour, and undifferentiated carcinoma) (Marth 2017). Since the introduction of cytology‐based cervical screening, the incidence and mortality rates of cervical cancer have been reduced by approximately 75% in countries with co‐ordinated population vaccination and screening programmes (Cancer Research UK 2014; ISD Scotland 2011; Siegel 2019; Welsh Cancer Intelligence and Surveillance 2011). A majority of cervical cancer cases (60%) are associated with inadequate screening and therefore are potentially preventable, or cervical cancer may be diagnosed at a very early stage, remaining easily curable with surgery (Alldredge 2016; Eskander 2014; Rebolj 2019). However, even in women diagnosed at a relatively early stage who have undergone treatment with surgery or radical chemoradiotherapy, the risk of recurrence is 10% to 20% for stages IB to IIA, and 28% to 64% for locally advanced FIGO stages IIB to IV, as per the classification of the International Federation of Obstetrics and Gynaecology (FIGO) (see Figure 1 for FIGO cervical cancer staging 2018) (Bhatla 2018; Bhatla 2019; Bray 2018; Cancer Research UK 2014).


FIGO stage 2018 (with correction Bhatla 2018; Bhatla 2019).

FIGO stage 2018 (with correction Bhatla 2018; Bhatla 2019).

Early‐stage cancer (FIGO stages IA, IB1, IB2, and IIA1) can be treated by surgery or chemoradiotherapy (Bhatla 2018; Bhatla 2019; Marth 2017; NCCN 2018). For locally advanced disease (FIGO stages IB3 to IVA), standard treatment is concurrent platinum‐based chemoradiotherapy, such as weekly cisplatin, during radiotherapy (Bhatla 2018; Bhatla 2019; Marth 2017; NCCN 2018). In general, five‐year survival rates are 99% for stage IA1 and 98% for stage IA2; for stages IB to IIA, five‐year survival rates are 88% to 95% without lymph node metastasis, and 51% to 78% with lymph node metastasis; for stage IIB, the rate is 65%, for stage III 40%, and for stage IV 10% to 20% (Cao 2014; Kim 2000). Very advanced cervical cancer (FIGO stage IVB), in which the disease has spread to areas outside of the pelvis upon diagnosis (distant metastatic disease), has a very poor prognosis. Treatment options are limited and are unlikely to be curative; therefore management is aimed at prolonging life and managing symptoms. Similarly, in recurrent disease, unless exenterative surgery is possible, women usually are treated with palliative chemotherapy. Chemotherapy aims to prolong survival, provide symptom control, and improve quality of life. For women not medically fit enough for chemotherapy, treatment options in both metastatic and recurrent disease may be limited to at best supportive care, with the option of palliative radiotherapy, if appropriate (Eleje 2019; Fanfani 2016; Greer 2010; Matsuo 2015; Waggoner 2003).

Persistent, recurrent, and metastatic diseases are the main causes of death among women with cervical cancer (Scatchard 2012). In persistent disease, visible lesions appear within six months of primary surgery, or within three to six months of completion of primary radiotherapy; in recurrent disease, visible lesions appear within an interval of more than six months after primary surgery, or within three to six months of completion of primary chemoradiotherapy (Cao 2011). More than 60% of recurrent diseases are detected within two years of primary treatment, and about 10% are detected after five years (Cao 2011). Persistent or recurrent cancers occur in about 35% of women with stages IB to IIA disease, and in 50% to 70% of patients with stages IIB to IVa disease; if cancer recurs, five‐year survival rates drop to 5% to 10% (Arbyn 2020; Cao 2017Cao 2019). More than 60% of recurrent disease occurs in the pelvic cavity, including the central pelvic cavity (uterine cervix, vagina, and uterine body), parametrium, and pelvic side wall (Cao 2014). Women who experience distant metastases (i.e. cancer that has spread from the original (primary) tumour location to distant organs or distant lymph nodes, outside of the pelvis), either upon initial diagnosis (FIGO stage IVB) or at the time of recurrence, will have a poor prognosis. In recurrent disease, metastatic rates (16% to 31% for IB and IIA, 26% for IIB, 39% for III, and 75% for IVA) increase with FIGO stage (Cao 2017). Sites of distant metastatic disease often include the lungs (21%), bone (16%), the para‐aortic region (11%), the abdominal cavity (8%), and the supraclavicular region (7%) (Cao 2014). Three phase 3 randomised trials (GOG‐204, GOG‐240, and Japanese Clinical Oncology Group 0505 (JCOG‐0505)) have examined chemotherapy regimens for women with advanced and recurrent cervical cancer (Monk 2009a; Saito 2010; Tewari 2014). The main concern was acquired resistance to platinum‐based chemotherapy in individuals with recurrent cervical cancer who were previously treated with chemotherapy. However, regimens that did not include a platinum agent were not found to be superior to cisplatin‐based treatments; therefore cisplatin plus paclitaxel remains the chemotherapy standard for recurrent and metastatic disease (Eleje 2019; Monk 2014).

Angiogenesis inhibitors are a class of promising biological agents that target the process by which a tumour forms new blood vessels to supply it with oxygen and nutrients. This process is key to the growth of cervical cancer, both locally and in metastatic deposits (Willmott 2009). In 1971, Folkman proposed a tumour angiogenesis‐dependent doctrine, which suggested that the development of tumours was due to the ability of individual cancer cells to grow without normal controls but was also dependent on the nutrient and oxygen supply via blood vessels (Folkman 1971). The transition of a tumour from an avascular to a vascular phase was termed the 'angiogenic switch' and was thought to be an important factor in the clinical progression from tumour dormancy to cancerous invasion (Hanahan 1996). The angiogenic switch occurs through a dynamic process involving pro‐angiogenic and anti‐angiogenic factors that are normally maintained in balance (physiological homeostasis). Cancer is characterised by up‐regulation of pro‐angiogenic factors, such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet‐derived growth factor (PDGF), and angiopoietins, and down‐regulation of anti‐angiogenic factors, such as thrombospondin, angiostatin, and endostatin, leading to the development of new blood vessels (neovascularisation) and tumour progression (Hicklin 2005). Neovascularisation is therefore a vital process in metastatic spread, which allows malignant cells to enter into the circulation (Falzone 2019; Folkman 1971; Folkman 1990; Pfaendler 2016). Activation of VEGF promotes the proliferation and migration of endothelial cells, which form the lining of blood vessels, and the development of new blood vessels. Moreover, VEGF activation enhances the leakiness (permeability) of existing blood vessels and consequently causes increased leakage of multiple plasma proteins that are involved in the angiogenesis process (Nagy 2007; Turajlic 2018). These proteins are ligands to three VEGF receptors (VEGF‐R1 to ‐R3) and are responsible for initiating a cascade of reactions that promote the migration, proliferation, and survival of endothelial cells, leading to the formation of new blood vessels. VEGF‐R1 and VEGF‐R2 are responsible for the regulation of angiogenesis, whereas VEGF‐R3 plays a secondary role in angiogenesis but is essential for lymphangiogenesis (development of new lymphatic vessels) and therefore is involved in the production of ascites and lymphatic dissemination of metastatic factors (Ellis 2008). Angiogenesis is present in pre‐malignant conditions of the cervix, as well as in invasive disease, which can be seen at colposcopy with the occurrence of vascular changes. Studies have shown that microvessel (small vessel) density increases progressively with the grades of cervical intraepithelial neoplasia (CIN), suggesting that angiogenesis may be an early event in the development of cervical pre‐cancer and cancer (Dobbs 1997; Guidi 1994; Krill 2015; Smith‐McCune 1994). Cervical cancer has been shown to be more dependent on angiogenesis than most other solid cancers because human papillomavirus (the cause of almost all cervical cancers) and hypoxia, with increased hypoxia‐inducible factor 1 alpha, are associated with higher levels of VEGF (Monk 2009b). In addition, increased intratumoural microvessel density during histological examination and enhanced expression of proteins, such as the endothelial antigen CD31, have been associated with a poor prognosis (Dellas 1997; Dobbs 1997; Tjalma 1999). Targeting the cancer microenvironment, specifically, new blood vessel formation, using VEGF pathway inhibition is the main goal of inhibition of angiogenesis.

Description of the intervention

Angiogenesis, the development of new blood vessels, is essential for initiation of tumours, their growth, and metastasis (Tan 2010). Tumours require angiogenesis to receive enough nutrients and oxygen to grow beyond 1 to 2 mm in diameter and to facilitate metastasis (Wagner 2009). The process of tumour‐related angiogenesis is regulated by various pro‐angiogenic factors, such as VEGF, and their cognate receptors, which are the dominant regulators of the proliferation of endothelial cells and the formation of new blood vessels (Tan 2010). VEGF acts by binding to its receptor, which then signals within the cell via tyrosine kinase enzyme activity (tyrosine kinases). VEGF‐R1 (Flt‐1 or fms‐like‐tyrosine kinase) and VEGF‐R2 (KDR/flk‐1) belong to the best characterised angiogenesis signalling pathway, and they can be blocked by inhibitors, such as antibodies (bevacizumab) or soluble receptors, which bind to VEGF and prevent it from binding to its receptors on cells. VEGF‐R2 is thought to be the dominant receptor responsible for translating and regulating angiogenesis (Bellone 2007).

Unlike chemotherapy agents that attack tumour cells, angiogenesis inhibitors are developed to block VEGF signalling pathways (Jayson 2016). This was first shown in 1993, when anti‐VEGF monoclonal antibodies were found to exert a potent inhibitory effect on the growth of three tumour cell lines injected subcutaneously into mice (Kim 1993). It is interesting to note that the antibody had no effects on cell lines in vitro. However, several parallel studies confirmed in vivo growth inhibition, which correlated with decreased tumour microvessel density and inhibition of tumour angiogenesis (Borgstrom 1996; Borgstrom 1998; Borgstrom 1999). It is now known that VEGF targeting agents ‐ angiogenesis inhibitors ‐ starve the tumour by preventing the formation of blood vessels and inhibit the growth and spread of tumours. VEGF targeting agents have been tested in some solid tumours, including metastatic colorectal cancer, metastatic thyroid cancer, ovarian cancer, high‐grade glioblastoma, and endocrine refractory or resistant metastatic breast cancer (Gaitskell 2011; Khasraw 2014; Tan 2010; Wagner 2009; Wagner 2012).

Phase 1 to 3 clinical trials of some VEGF targeting agents (bevacizumab, cediranib, pazopanib, sunitinib, and sorafenib) have been completed in women with persistent, recurrent, metastatic, and locally advanced cervical cancer (Mackay 2010; Milosevic 2016; Monk 2009a; Monk 2010; Schefter 2014; Symonds 2015; Tewari 2014; Zighelboim 2013). Bevacizumab is a humanised monoclonal antibody directed against all major isoforms of VEGF (VEGF‐A, VEGF‐B, VEGF‐C, VEGF‐D, placental growth factor (PIGF), VEGF‐E, and VEGF‐F members). It is used extensively in the USA and is the most studied biological agent in gynaecological cancer (Grothey 2009; Tewari 2014). Several published randomised controlled trials (RCTs) and non‐randomised trials have reported binding and inactivation of VEGF through the clinical use of bevacizumab as a single agent (Monk 2009a; Monk 2009b; Schefter 2012; Schefter 2014; Takano 2013; Tan 2010; Tewari 2005; Tewari 2014; Tewari 2014b; Wright 2006). It has now been approved in more than 60 countries for the treatment of persistent, recurrent, or metastatic disease, in combination with paclitaxel and cisplatin, or paclitaxel and topotecan, based on improved overall survival (OS) with typical adverse events (hypertension, thromboembolic events, and gastrointestinal fistulae) in the Gynecologic Oncology Group (GOG)‐240 trial (Tewari 2017).

The treatment plan for persistent, recurrent, or metastatic cervical cancer depends on the performance status of the person with cancer (i.e. his or her general well‐being), the disease site, the extent of disease, and previous treatment (Bhatla 2018; Bhatla 2019). For women with isolated distant metastasis or locoregional recurrence (i.e. original microscopic cancer cells that have resisted primary therapy and become visible at the original site), treatment includes individualised radiotherapy (external radiation or brachytherapy (intracavity radiotherapy), or both), with or without cisplatin chemotherapy; surgical resection can be used for control of local disease and symptoms (NCCN 2018). For women with central pelvic recurrence after primary radiotherapy, pelvic exenteration (i.e. removal of part or all of the pelvic organs), with or without intraoperative radiotherapy, may be considered. Surgical mortality rates of less than 5% and long‐term survival rates of approximately 50% have been reported (Morley 1989). Radical hysterectomy or brachytherapy may also be considered, if the central pelvic lesion is small (< 2 cm) and isolated (NCCN 2018). For women with extrapelvic metastases or recurrences who are unsuitable to undergo surgery or radiotherapy, combination cisplatin‐based chemotherapy regimens are often recommended (i.e. paclitaxel and cisplatin combined with bevacizumab) over the use of single agents (NCCN 2018). However, overall response rates to chemotherapy are about 20% (15% to 46%), with median OS of six to eight months and median progression‐free survival (PFS) of three months (Marth 2017; Moore 2006; Tewari 2005).

How the intervention might work

Over‐expression of VEGF has been found in most human cancers compared with normal tissues (Jayson 2016). VEGF is released by tumour cells and interacts with its receptor (VEGF‐R), a transmembrane protein, on the nearby endothelial cells of normal blood vessels (see Gaitskell 2011 for figures describing receptor function). VEGF binds to the extracellular domain of VEGF‐R, stimulates tyrosine kinase activity in the intracellular domain of VEGF‐R, and triggers the downstream network of cell signalling pathways that promote the proliferation of endothelial cells and the formation of new blood vessels (Kerbel 2008). VEGF and VEGF‐R have already become the dominant targets for clinical angiogenesis inhibitors (Jayson 2016). VEGF signalling pathways can be blocked by several mechanisms, including VEGF blockades (e.g. anti‐VEGF antibodies (bevacizumab), VEGF‐trap (aflibercept)), VEGF‐R blockades (e.g. anti‐VEGF‐R antibodies (ramucirumab)), and interruption of intracellular signalling pathways (e.g. tyrosine kinase inhibitors (TKIs) cediranib, pazopanib, sorafenib, sunitinib) (Alldredge 2016). Results of xenograft models of human tumours in mice have shown that VEGF targeting agents cause regression of microvascular tissues and reduction in metastatic disease progression (Genentech 2020; Morton 2012).

Bevacizumab (Avastin) is a VEGF‐directed antibody that has been given as an intravenous infusion as monotherapy or with other therapies to treat cervical cancer in clinical trials. In a phase 2 study of 46 women with persistent or recurrent squamous cell carcinoma, single‐agent bevacizumab was active and well tolerated. Median OS was 7.29 months, and median PFS was 3.40 months, with common severe adverse events of hypertension, thromboembolic events, and gastrointestinal disorders (Monk 2009a). In a phase 2 study of 27 women with persistent or recurrent disease (66.6% were squamous), bevacizumab was used with cisplatin and topotecan, median OS was 13.2 months, and median PFS was 7.1 months. However, severe haematologic toxicities were common, including thrombocytopenia, leukopenia, anaemia, and neutropenia. Most of the women required at least one unexpected hospital admission for supportive care (Zighelboim 2013). In a phase 2 study of 49 women with locally advanced disease (63% were IIB, 80% were squamous), bevacizumab (10 mg/kg every 14 days for three cycles) was used with concurrent platinum‐based chemoradiotherapy; three‐year OS and PFS rates were 81.3% and 68.7%, respectively, and no significant treatment‐related adverse events occurred (Schefter 2014). In a GOG‐240 phase 3 study of 452 women with persistent, recurrent, or metastatic cervical cancer (68.6% were squamous), participants were randomised to receive bevacizumab with chemotherapy (paclitaxel and cisplatin, or paclitaxel and topotecan) or chemotherapy alone. The addition of bevacizumab to chemotherapy improved median OS to 17.0 months versus 13.3 months, and median PFS to 8.2 months versus 5.9 months, compared with chemotherapy alone. However, adverse events, including hypertension, severe thromboembolic events, and severe gastrointestinal fistulae, were increased (Tewari 2014).

Cediranib (Recentin) is a small molecule TKI of VEGF‐R. In a phase 2 study of 69 women with metastatic or recurrent disease, participants were randomised to receive cediranib with chemotherapy (paclitaxel and carboplatin) or chemotherapy alone. The addition of cediranib to chemotherapy improved median PFS as compared with chemotherapy alone (8.1 months versus 6.7 months) but did not improve median OS (13.6 months versus 14.8 months); adverse events, including hypertension, febrile neutropenia, and diarrhoea, were increased (Symonds 2015).

Pazopanib (Votrient) is an oral multi‐targeted TKI of VEGF‐R, a platelet‐derived growth factor receptor (PDGF‐R), and c‐kit. Lapatinib is an oral dual TKI of epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2/neu). In a phase 2 study of 228 women with persistent, recurrent, or metastatic cervical cancer, participants were randomised to receive single‐agent pazopanib (800 mg once daily), single‐agent lapatinib (1500 mg once daily), or pazopanib plus lapatinib (this arm was terminated due to toxicity). Pazopanib improved median OS (50.7 weeks versus 39.1 weeks) and PFS (18.1 weeks versus 17.1 weeks) as compared with lapatinib; the most common adverse events were similar in both groups: diarrhoea, nausea, hypertension, and anorexia (Monk 2010).

Sunitinib (Sutent) is an oral multi‐targeted TKI of VEGF‐R, PDGF‐R, and c‐kit. In a phase 2 study of 19 women with incurable locally advanced or metastatic disease, single‐agent sunitinib was administered in a six‐week cycle for up to a maximum of six cycles; no tumour response was observed during follow‐up (median 4.4 months), and five cases of fistula formation were observed (Mackay 2010).

Sorafenib (Nexavar) is an oral multi‐targeted inhibitor that targets both VEGF‐R and PDGF‐R tyrosine kinase and Raf serine/threonine kinase. In a phase 1 study of 13 women with locally advanced disease, sunitinib was administered before and during concurrent platinum‐based chemoradiotherapy, but the study was terminated after a preliminary analysis because sorafenib increased tumour hypoxia, which is a strong predictor of disease progression (Milosevic 2016).

Why it is important to do this review

Increasing knowledge about the molecular biology of gynaecological cancers has led to the development of biological therapies. Treatment options for persistent, recurrent, and metastatic cervical cancer are limited, and the purpose of current treatment is to relieve symptoms and improve quality of life (Bhatla 2018; Marth 2017; NCCN 2018). Other agents have not shown a survival advantage compared with standard therapy (Marth 2017; NCCN 2018). However, a phase 3 clinical trial showed that the addition of bevacizumab to chemotherapy improved survival from 13.3 months to 16.8 months (Penson 2015; Tewari 2014; Tewari 2017). Since publication of the early results of Tewari 2014, bevacizumab has become the standard of care in combination with chemotherapy for individuals with metastatic and recurrent cervical cancer in some high‐income countries (Genentech 2020). Other anti‐angiogenic agents have been studied to a lesser degree than bevacizumab.

Cochrane Reviews of VEGF targeting therapy have been produced for glioma and for thyroid, ovarian, colorectal, and breast cancers (Gaitskell 2011; Khasraw 2014; Tan 2010; Wagner 2009; Wagner 2012). Clinical trials have shown that VEGF targeting agents improved OS or PFS but increased tolerable toxicities among women with persistent, recurrent, or metastatic cervical cancer (Monk 2010; Symonds 2015; Tewari 2014). This Cochrane Review aims to assess the benefits and harms of VEGF inhibitors alone or in the adjuvant setting for treatment of persistent, recurrent, or metastatic cervical cancer, with the goal of assisting clinicians and women involved in the decision‐making process.

Objectives

To assess the benefits and harms of VEGF targeting agents in the management of persistent, recurrent, or metastatic cervical cancer.

Methods

Criteria for considering studies for this review

Types of studies

We included only randomised controlled trials (RCTs) that evaluated use of anti‐angiogenesis therapy for persistent, recurrent, or metastatic cervical cancer. We included RCTs reported as full text, those published as abstract only, and unpublished data (no abstract only or unpublished data were found). We excluded observational studies such as case reports, case series, case‐control studies, and cohort studies. We excluded quasi‐randomised trials, cluster‐randomised trials, and cross‐over trials.

To evaluate economics, full or partial economic evaluation studies and single effectiveness studies were considered for inclusion. This review considered only health economics studies conducted alongside effectiveness studies included in the effectiveness component of the review.

Types of participants

We included adult women (aged 18 years or older) with a diagnosis of persistent, recurrent, or metastatic cervical cancer of any stage. We did not include women with any other concurrent malignancy.

Types of interventions

  • Vascular endothelial growth factor (VEGF) targeting agents plus other treatments versus other treatments only

  • VEGF targeting agents versus other treatments

  • VEGF targeting agents versus placebo or no treatment

  • VEGF targeting agents versus other VEGF targeting agents

Other treatments included standard chemotherapy, radiotherapy, other types of molecular targeted therapy, and surgery.

Types of outcome measures

Primary outcomes

  • Overall survival (OS), defined as the time interval from randomisation to death from any cause, or to last follow‐up

  • Specific adverse events, including gastrointestinal perforations or fistulae, haemorrhage, thromboembolic events, and hypertension

Secondary outcomes

  • Progression‐free survival (PFS), defined as the time interval from randomisation to progression of disease or death

  • Quality of life (QoL), while on treatment and post treatment, when reported as measured on a scale that has been validated through reporting of norms in a peer‐reviewed publication (i.e. European Organisation for Research and Treatment of Cancer (EORTC) QLQ‐CX24 cervical cancer‐specific quality of life questionnaire (Greimel 2006); Functional Assessment of Cancer Therapy‐Cervix Trial Outcome Index (FACT‐Cx TOI) (www.facit.org/facitorg/); EORTC Quality Questionnaire Core 30 items (QLQ‐C30); Functional Assessment of Chronic Illness Therapy (FACIT) Measurement System; Rotterdam Symptom Checklist (RSCL); and Symptom Distress Scale (SDS)) (Tamburini 2001)

  • Adverse events, classified according to CTCAE 2017

  • Economic evaluation, as defined by individual studies, including full economic evaluation (cost‐benefit analysis, cost‐effectiveness analysis, and cost‐utility analysis), partial economic evaluation (cost analysis, cost‐description study, and cost‐outcome description), or single effectiveness evaluation (description, measurement, or valuation of resource use associated with an intervention) (Shemilt 2011)

We presented a 'Summary of findings' table reporting the following outcomes listed in order of priority.

  • Overall survival.

  • Adverse events: gastrointestinal perforations or fistulae.

  • Adverse events: haemorrhage.

  • Adverse events: thromboembolic events.

  • Adverse events: hypertension.

  • Serious adverse events.

  • Economic evaluation.

Search methods for identification of studies

We applied no language or date restrictions during our searches. We searched for papers in all languages and had papers translated as necessary.

Electronic searches

We searched the following electronic databases up to 27 May 2020.

  • the Cochrane Central Register of Controlled Trials (CENTRAL; Issue 5, 2020), in the Cochrane Library (Appendix 1);

  • MEDLINE via Ovid (1946 to May week 3, 2020), (Appendix 2);

  • Embase via Ovid (1980 to 2020 week 21), (Appendix 3).

Economics search filters were not used because most of the economics studies to be considered would be retrieved using the study design search filter (Shemilt 2011).

Searching other resources

All relevant articles were identified on PubMed, and we conducted a further search for newly published articles using the 'Related articles' feature.

Unpublished and grey literature

We searched the following for ongoing trials.

If ongoing trials that had not been published were identified through these searches, we approached the principal investigators and major co‐operative groups active in this area to ask for relevant data.

We also searched the reports of conferences in the Conference Proceedings Citation Index‐Science (CPCI-S), along with the bibliography and abstracts in the National Health Service (NHS) Economic Evaluation Database (NHS EED).

Handsearching

We handsearched the citation lists of included studies, key textbooks, and previous systematic reviews, and we contacted experts in the field, to identify further reports of trials. We also handsearched the reports of conferences from the following sources.

  • Gynecologic Oncology (Annual Meeting of the American Society of Gynecologic Oncology).

  • International Journal of Gynecological Cancer (Annual Meeting of the International Gynecologic Cancer Society).

  • British Journal of Cancer.

  • British Cancer Research Meeting.

  • Annual Meeting of the European Society of Medical Oncology (ESMO).

  • Annual Meeting of the American Society of Clinical Oncology (ASCO).

Data collection and analysis

Selection of studies

We downloaded all titles and abstracts retrieved by electronic searching to a reference management database (Endnote), and we removed duplicates. Three review authors (YC, XZ, and IR) examined the remaining references independently. We excluded studies that clearly did not meet the inclusion criteria, and we obtained copies of the full text of potentially relevant references. Three review authors (YC, XZ, and IR) independently assessed the eligibility of retrieved reports/publications. We resolved any disagreement through discussion; if required, we consulted another person (GD). We identified and excluded duplicate reports and collated multiple reports of the same study, so that each study rather than each report was the unit of interest in the review. We recorded the selection process in sufficient detail to complete a PRISMA flow diagram and a Characteristics of excluded studies table (Liberati 2009).

Data extraction and management

Two review authors (YC and XZ) independently extracted study characteristics and outcome data from included studies to a piloted data collection form. We noted in the Characteristics of included studies table if outcome data were not reported in a usable way. We resolved disagreements by reaching consensus or by involving a third person (GD). One review author (GD) transferred data into Review Manager 2014. We double‐checked that data had been entered correctly by comparing data presented in the systematic review with data provided in study reports. A second review author (YL) 'spot‐checked' the accuracy of study characteristics against the trial report.

For included studies, we extracted the following data.

  • Author, year of publication, and journal citation (including language).

  • Country where the trial was performed.

  • Study design and method.

  • Inclusion and exclusion criteria.

  • Study population.

    • Total number enrolled.

    • Participant characteristics.

    • Age.

    • Comorbidities (e.g. gastrointestinal disease, cardiovascular disease).

    • Previous therapy.

  • Cervical cancer characteristics.

    • FIGO stage, tumour grade, histological type of cervical cancer.

    • Details about recurrent and metastatic disease.

  • Total number of intervention groups.

  • Intervention details.

    • Type of VEGF targeting agent.

    • Dose of VEGF targeting agent.

    • Treatment regimen of VEGF targeting agent.

    • Frequency and number of cycles.

  • Comparison.

    • Type of control.

    • Dose, regimen, frequency and number of cycles.

  • Risk of bias in study (see below).

  • Duration of follow‐up.

  • Outcomes such as OS, PFS, QoL, symptom control, and adverse events.

  • Number of participants allocated to each intervention group, total number analysed for each outcome, and missing participants.

Results were extracted as follows.

  • For time‐to‐event data (OS and PFS), we extracted the hazard ratio (HR), log of the hazard ratio [log(HR)], and its standard error (SE) from trial reports. If these were not reported, we attempted to estimate the log(HR) and its SE using the methods of Parmar 1998 (e.g. number of events in each arm, log‐rank P value comparing relevant outcomes in each arm).

  • For dichotomous outcomes (e.g. adverse events, deaths), we extracted the number of participants in each treatment arm who experienced the outcome of interest and the number of participants assessed at end point, in order to estimate a risk ratio (RR).

  • For continuous outcomes (e.g. QoL measures), we extracted the final value and the standard deviation (SD) for the outcome of interest and the number of participants assessed at end point in each treatment arm at the end of follow‐up, in order to estimate the mean difference between treatment arms and its SE.

If reported, we extracted both unadjusted and adjusted statistics. If available, we would use adjusted results; otherwise we would use unadjusted results.

When possible, all data extracted would be those relevant to an intention‐to‐treat analysis, in which participants would be analysed in the groups to which they were assigned.

We noted the time points at which outcomes were collected and reported.

In the event of duplicate publications, companion documents, or multiple reports of a primary study, we maximised the yield of information by collating all available data and using the most complete data set aggregated across all known publications. In case of doubt, we gave priority to the publication reporting the longest follow‐up associated with our primary or secondary outcomes.

Assessment of risk of bias in included studies

We assessed and reported on the methodological risk of bias of included studies as specified in Chapter 8 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2020), which recommends explicit reporting of the following individual elements for RCTs.

  • Selection bias: random sequence generation and allocation concealment.

  • Performance bias: blinding of participants and personnel.

  • Detection bias: blinding of outcome assessment.

  • Attrition bias: incomplete outcome data.

  • Reporting bias: selective reporting of outcomes.

  • Other possible sources of bias.

Two review authors (YC and XZ) applied the 'Risk of bias' tool independently and resolved differences by discussion with a third review author (GD). We judged each item as being at high, low, or unclear risk of bias as set out in the criteria provided by Higgins 2020, and we provided a quote from the study report or a statement as justification for the judgement for each item in the 'Risk of bias' table, or both. We summarised results in both a 'Risk of bias' graph and a 'Risk of bias' summary. When interpreting treatment effects, we took into account the risk of bias for studies that contributed to that outcome. When information on risk of bias related to unpublished data or correspondence with a trialist, we noted this in the 'Risk of bias' table. See 'Risk of bias' items (Appendix 4).

We defined the following end points as subjective outcomes: PFS, QoL.

We defined the following end points as objective outcomes: OS, adverse events (gastrointestinal perforations and fistulae, haemorrhage, thromboembolic events, hypertension), economic evaluation.

Measures of treatment effect

We used the following measures of the effect of treatment.

  • For time‐to‐event data, we used the HR, if possible.

  • For dichotomous outcomes, we analysed data based on the number of events and the number of women assessed in intervention and comparison groups. We used these to calculate the RR and the 95% confidence interval (CI).

  • For continuous outcomes, we analysed data based on the mean, the standard deviation (SD), and the number of women assessed for both intervention and comparison groups to calculate the mean difference (MD) between treatment arms with a 95% CI. If the MD was reported without individual group data, we would use this to report the study results. If more than one study measures the same outcome using different tools, we would calculate the standardised mean difference (SMD) and the 95% CI using the inverse variance method in Review Manager 5 (Review Manager 2014).

We planned to perform meta‐analysis only when this was meaningful (i.e. if treatments, participants, and the underlying clinical question were similar enough for pooling to make sense). We narratively described skewed data that were reported as medians and interquartile ranges. When multiple trial arms were reported in a single trial, we included only the relevant arms.

Unit of analysis issues

We did not anticipate unit of analysis issues, and we noted no unit of analysis issues within the included studies.

Dealing with missing data

For missing outcome or summary data, we did not impute missing data, and we reported in the review any assumptions that we made. We attempted to contact study authors to obtain missing data (participants, outcomes, or summary data). We reported on levels of loss to follow‐up and assessed this information as a source of potential bias.

Assessment of heterogeneity

We did not perform an assessment of heterogeneity, as the review included only four completed trials (only one trial for each comparison), which we did not meta‐analyse. We had specified in the protocol that we planned to combine data using meta‐analysis if the studies were considered similar enough. We had intended to assess the degree of heterogeneity by visually inspecting forest plots; by estimating the percentage of heterogeneity (I² measurement) between trials that cannot be ascribed to sampling variation (Higgins 2003); by performing a formal statistical test of the significance of the heterogeneity (Chi²) (Deeks 2001); and, if possible, by conducting subgroup analyses. We had intended to regard heterogeneity to be substantial if I² was greater than 30% and either T² was greater than zero or the P value was low (< 0.10) in the Chi² test for heterogeneity.

Assessment of reporting biases

We did not perform a formal assessment of potential publication bias, as the review included only four trials. We had specified in the protocol that we planned to address reporting biases according to the recommendations provided in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2020).

Data synthesis

We planned to carry out meta‐analyses by using Review Manager software (Review Manager 2014), according to recommendations provided in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2020), if data were sufficiently similar to be combined (in terms of participants, settings, interventions, comparisons, and outcome measures).

Meta‐analysis was not feasible, as the review included only four completed trials (only one trial for each comparison). We noted a high degree of heterogeneity in population characteristics across trials on this topic. In future updates when meta‐analysis is feasible, we will use the random‐effects model with inverse variance weighting for all meta‐analyses (DeSimonian 1986).

Subgroup analysis and investigation of heterogeneity

We had considered performing subgroup analyses for the following characteristics.

  • Different VEGF targeting agents (anti‐VEGF antibodies, VEGF trap, anti‐VEGF‐receptor antibodies, TKIs, and others).

  • Different histological types (squamous, adenocarcinoma, and others).

However, we did not perform the intended subgroup analyses, as only four studies were included (only one trial for each comparison). Although different histological types had similar survival in persistent, recurrent, or metastatic disease (Seamon 2018), we considered this factor when interpreting the OS.

Sensitivity analysis

We had planned to re‐analyse data using a random‐effects model instead of a fixed‐effect model. We had planned to re‐analyse data by including/excluding studies on the basis of differences in risk of bias, quality components (full‐text publications versus abstracts and preliminary results versus mature results), participant dropout, and missing data. However, only four studies (one trial for each comparison) were included, so sensitivity analysis was not possible.

Summary of findings and assessment of the certainty of the evidence

We presented the overall certainty of the evidence for prespecified outcomes (Types of outcome measures), including overall survival, adverse events, and economic evaluation, according to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach, which takes into account issues not only related to internal validity (risk of bias, inconsistency, imprecision, publication bias) but also to external validity such as directness of results (Langendam 2013). We created 'Summary of findings' tables (summary of findings Table 1, summary of findings Table 2, summary of findings Table 3, summary of findings Table 4, summary of findings Table 5) based on the methods described in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2020) and using GRADEpro GDT. We used the GRADE checklist and GRADE Working Group certainty of evidence definitions (Meader 2014; Gyatt 2011). We downgraded the evidence from 'high' certainty by one level for serious (or by two for very serious) concerns for each limitation for the outcomes detailed under Types of outcome measures:

  • 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.

Results

Description of studies

See Characteristics of included studies,Characteristics of excluded studies,Characteristics of studies awaiting classification, and Characteristics of ongoing studies.

Results of the search

The literature search was designed to find all relevant references about vascular endothelial growth factor (VEGF) targeting therapy for persistent, recurrent, or metastatic cervical cancer. Through the main database searches, we identified 1752 records; we identified an additional 43 records by searching other sources. After removing duplicates, we screened 1634 unique records for eligibility and excluded 1576 records during the initial screening process; we assessed the full text or abstract of the remaining 58 records. We identified 28 references related to four studies that fulfilled the inclusion criteria and were included in the review. The study flow diagram in Figure 2 provides an overview of the study selection process.


Study flow diagram.

Study flow diagram.

Included studies

We included four studies that fulfilled the inclusion criteria of this review (Guo 2020; Monk 2010Symonds 2015; Tewari 2014). Tewari 2014 and Symonds 2015 used the VEGF targeting agents anti‐VEGF antibody and tyrosine kinase inhibitor (TKI) of VEGF receptor (VEGF‐R), respectively, plus cisplatin/paclitaxel chemotherapy versus chemotherapy only (Tewari 2014), or versus placebo (Symonds 2015). Guo 2020 used VEGF targeting agent (TKI of VEGF‐R) plus chemotherapy (carboplatin and paclitaxel) or chemoradiotherapy versus chemotherapy or chemoradiotherapy only. Monk 2010 used VEGF targeting agent (TKI of VEGF‐R) alone or another molecular targeted therapy agent (TKI of epidermal growth factor receptor and human epidermal growth factor receptor 2) versus a combination of both targeted agents. See Characteristics of included studies for full details of each study.

Study design and methods

Tewari 2014 was an open‐label study reporting that participants were randomly assigned to receive cisplatin/paclitaxel chemotherapy with or without bevacizumab, or topotecan/paclitaxel chemotherapy with or without bevacizumab. Symonds 2015 was a double‐blind study reporting that participants were randomly assigned 1:1 to receive carboplatin/paclitaxel chemotherapy with cediranib or placebo. Guo 2020 was an open‐label study reporting that participants were randomly assigned 1:1 to receive chemotherapy or chemoradiotherapy with or without apatinib. Monk 2010 was an open‐label study reporting that participants were randomly assigned 1:1:1 to receive pazopanib or lapatinib, or both.

Sample sizes and participants

Tewari 2014 included 452 participants: 227 in the intervention arm and 225 in the control arm; 452 were randomised and 440 received study treatment; median age was 48 years (range 20 to 85 years); histological types included squamous (N = 310), adenocarcinoma (N = 86), and other (N = 56); disease sites and extents included pelvic disease (N = 242) and extra‐pelvic disease (N = 210).

Symonds 2015 included 69 participants: 69 randomised and 67 receiving study treatment; median age was 43.5 years and interquartile range (IQR) 37 to 60 years in the intervention arm; median age was 44 years and IQR 34 to 53 years in the control arm; histological types included squamous (N = 47), adenocarcinoma (N = 14), and other (N = 8); disease sites and extents included local relapse only (N = 9), extra‐pelvic metastases only (N = 21), and both (N = 39).

Guo 2020 included 59 participants with recurrent and advanced cervical cancer who were randomised to the apatinib group (N = 30) and the control group (N = 29); median age was 49.5 years (range 28 to 62 years) in the intervention arm and 51 years (range 30 to 69 years) in the control arm; histological types included squamous (N = 44) and adenocarcinoma (N = 8); 34 of 52 participants had more than two extra‐pelvic disease sites (59 were randomised; 52 were included in the analysis).

Monk 2010 included 228 participants: 228 were randomised and 226 received study treatment; median age was 49.5 years (range 29 to 73 years) in the pazopanib arm, 49 years (range 23 to 81 years) in the lapatinib arm, and 48.5 years (range 28 to 82 years) in the pazopanib plus lapatinib arm; histological types included squamous (N = 151), adenocarcinoma (N = 42), and other (N = 35); 213 received prior anti‐cancer therapies.

Setting

Tewari 2014 was conducted at 81 sites in the USA, Canada, and Spain. Symonds 2015 was conducted at 17 sites in the UK. Guo 2020 was conducted at a single centre in China. Monk 2010 was conducted at 50 sites in the USA, Europe, South America, and Asia.

Interventions

In Tewari 2014, participants were randomised to receive cisplatin (50 mg/m²) plus paclitaxel (135 or 175 mg/m²) with or without bevacizumab (15 mg/kg) intravenously, or topotecan (0.75 mg/m², days 1 to 3) plus paclitaxel (175 mg/m²) with or without bevacizumab (15 mg/kg) intravenously, for each 21‐day chemotherapy cycle until disease progression or unacceptable toxicity.

In Symonds 2015, participants were randomised to receive carboplatin (area under the curve (AUC) of free carboplatin plasma concentration versus time (Calvert 1989), AUC 5 mg/mL/min) plus paclitaxel (175 mg/m²) with cediranib (20 mg) or placebo orally once daily, for each 21‐day chemotherapy cycle and for a maximum of six cycles, plus cediranib or placebo 20 mg orally once daily until disease progression or unacceptable toxicity.

In Guo 2020, participants with recurrent disease were randomised to carboplatin (AUC 5 mg/mL/min) plus paclitaxel (135 to 175 mg/m²) with or without apatinib (500 mg orally once daily) for each 21‐day chemotherapy cycle and for a maximum of six cycles until disease progression or unacceptable toxicity; participants with stage IVB disease were randomised to carboplatin (AUC 5 mg/mL/min) plus paclitaxel (135 to 175 mg/m²) plus brachytherapy (intracavity radiotherapy) (total 50 to 60 Gy) with or without apatinib (500 mg orally once daily) for each 21‐day chemotherapy cycle and for a maximum of six cycles until disease progression or unacceptable toxicity.

In Monk 2010, participants were randomised to receive pazopanib (800 mg) or lapatinib (1500 mg), or both, orally daily until disease progression or unacceptable toxicity.

Outcomes

Tewari 2014 reported overall survival (OS), progression‐free survival (PFS), quality of life (QoL), adverse events, and economic evaluation, with maximum follow‐up longer than 50 months for OS. Moore criteria were used to assess the level of risk of death (Moore 2010). These criteria include five negative factors: ethnicity, performance status 1, pelvic disease, prior cisplatin, and progression‐free interval less than 365 days. Risk categories included low risk (zero to one factor), medium risk (two to three factors), and high risk (four to five factors). QoL was measured using the Trial Outcome of Index of the Functional Assessment of Cancer Therapy (FACT‐Cx‐TOI) (Cella 1993), the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group‐Neurotoxicity (FACT/GOG‐NTX) (Huang 2007), and the Brief Pain Inventory (BPI) (Cleeland 1994). Specific adverse events included gastrointestinal perforations or fistulae, serious haemorrhage, serious thromboembolic events, and hypertension.

Symonds 2015 reported OS, PFS, QoL, and adverse events, with median follow‐up of 24.2 months for OS. QoL was measured using the European Organisation for Research and Treatment of Cancer (EORTC) QLQ‐C30 questionnaire (Aaronson 1993). Specific adverse events included gastrointestinal perforations or fistulae, serious haemorrhage, serious thromboembolic events, and serious hypertension.

Guo 2020 reported OS, PFS, and hypertension events, with median follow‐up of 14 months for OS.

Monk 2010 reported OS, PFS, and adverse events, with maximum follow‐up longer than 150 weeks for OS. Specific adverse events included gastrointestinal perforations or fistulae, haemorrhage, thromboembolic events, and hypertension.

Excluded studies

We excluded seven studies (Godoy‐Ortiz 2018; Grau 2020; ISRCTN10841582; JPRN‐jRCTs031180007; NCT03635567; NCT03912415; Phippen 2015). Five studies were not relevant comparisons (VEGF targeting agents alone or in combination with other treatments versus placebo, no treatment, other treatments, or another VEGF targeting agent) (Grau 2020; ISRCTN10841582; JPRN‐jRCTs031180007; NCT03635567; NCT03912415). Two studies were not RCTs (Godoy‐Ortiz 2018; Phippen 2015).

Studies awaiting classification

The review authors found one completed study comparing TKI of VEGF‐R (apatinib) plus chemoradiotherapy with chemoradiotherapy alone for women (40 participants) with FIGO stage IIB to IV cervical cancer (Rao 2018). Rao 2018 reported the median OS in the apatinib group was 19.88 months versus 10.68 in the control group (P = 0.022). The review authors found a second completed study that compared bevacizumab plus chemoradiotherapy with chemoradiotherapy alone for women (75 participants) with advanced cervical adenocarcinoma but did not address OS (Xie 2016). However, trial registration details were not found for the two studies, and contacting the principal Investigators did not result in the receipt of any additional data.

The review authors found nine ongoing studies (ChiCTR1800017291; ChiCTR1800017846; ChiCTR1800018232; ChiCTR1900020912; ChiCTR1900021190; ChiCTR1900025465; ChiCTR1900025992; NCT02009579; Zhou 2018) for TKI of VEGF‐R. These studies have not to date reported any outcomes.

  • TKI of VEGF‐R plus chemotherapy versus chemotherapy: apatinib (ChiCTR1800017291; ChiCTR1800017846; ChiCTR1800018232; ChiCTR1900025992), anlotinib (ChiCTR1900020912; ChiCTR1900021190), nintedanib (NCT02009579): expected recruitment 698 participants.

  • TKI of VEGF‐R plus chemotherapy versus anti‐programmed death 1 (PD‐1) antibody plus chemotherapy: apatinib (ChiCTR1900025992): expected recruitment 72 participants.

  • TKI of VEGF‐R plus chemoradiotherapy versus anti‐PD‐1 antibody plus chemoradiotherapy versus TKI of VEGF‐R plus anti‐PD‐1 antibody plus chemoradiotherapy: apatinib (ChiCTR1900025465): expected recruitment 60 participants.

  • TKI of VEGF‐R versus supportive care: apatinib (Zhou 2018): expected recruitment 60 participants.

Risk of bias in included studies

The risk of bias of included studies is summarised in Figure 3 and Figure 4 (Guo 2020; Monk 2010; Symonds 2015; Tewari 2014). See the 'Risk of bias' tables within Characteristics of included studies for assessment details.


Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.


Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Allocation

Monk 2010, Symonds 2015, and Tewari 2014 were judged as having adequate allocation concealment. Participants were randomly assigned by validated randomisation systems.

In Guo 2020, information about random sequence generation or allocation concealment was not available. We assessed this study to be at unclear risk of selection bias.

Blinding

Guo 2020, Monk 2010, and Tewari 2014 were reported as 'open‐label'; we assessed their risk of bias arising from lack of blinding of participants, investigators, and outcomes assessors as low for objective outcomes (i.e. OS, adverse events, and economic evaluation), and we assessed their risk as high for subjective outcomes (i.e. PFS and QoL). Symonds 2015 was reported as 'double‐blind'; we assessed this study to be at low risk of bias for both objective and subjective outcomes.

Incomplete outcome data

Monk 2010, Symonds 2015, and Tewari 2014 included all participants who were randomised and received study medications for the analyses. In Guo 2020, 88% of women were assessed in the analysis (59 participants were randomised, and 52 were included in the analysis). We assessed these studies to be at low risk of attrition bias.

Selective reporting

Monk 2010, Symonds 2015, and Tewari 2014 protocols were available and reported primary outcomes. Outcomes in the study protocols were the same as those finally reported in the publications. We assessed these studies to be at low risk of selective reporting bias.

For Guo 2020, trial registration occurred later than study completion. We assessed this study to be at high risk of selective reporting bias.

Other potential sources of bias

The study on apatinib was sponsored by HengRui Medicine, the manufacturer of apatinib (Guo 2020). The study on pazopanib was sponsored by GlaxoSmithKline, the manufacturer of pazopanib (Monk 2010). Economic evaluation was not a planned analysis in the study protocol for Tewari 2014.

Effects of interventions

See: Summary of findings 1 Bevacizumab plus chemotherapy compared to chemotherapy; Summary of findings 2 Cediranib plus chemotherapy compared to placebo plus chemotherapy; Summary of findings 3 Apatinib plus chemotherapy or chemotherapy/brachytherapy compared to chemotherapy or chemotherapy/brachytherapy; Summary of findings 4 Pazopanib plus lapatinib compared to lapatinib; Summary of findings 5 Pazopanib compared to lapatinib

Bevacizumab plus chemotherapy versus chemotherapy only

Overall survival

In Tewari 2014, treatment with bevacizumab plus chemotherapy may have resulted in lower risk of death compared to chemotherapy alone (hazard ratio (HR) 0.77, 95% confidence interval (CI) 0.62 to 0.95; 1 study, 452 participants; low‐certainty evidence; Analysis 1.1summary of findings Table 1). Tewari 2014 reported median overall survival (OS) in the bevacizumab groups as 16.8 months (mortality rate 170/227 participants) and median OS in the control groups as 13.3 months (mortality rate 178/225 participants).

Two hundred seventy‐one participants died (131/227 in the bevacizumab groups, 140/225 in the control groups).

  • Bevacizumab reduced risk of death in the pelvic disease subgroup (N = 242) but had no effect and appreciable benefit for the extra‐pelvic disease subgroup (N = 210) (details of HR and CI were not provided).

  • Bevacizumab reduced risk of death in the squamous subgroup (N = 310) but had appreciable benefit and harm for non‐squamous subgroups (adenocarcinoma N = 86, adenosquamous N = 44, others N = 12) (details of HR and CI were not provided).

Moore criteria included the following.

  • Low‐risk population: for risk of death, HR was 0.96 (95% CI 0.51 to 1.83); median OS was 22.9 months in the bevacizumab groups (mortality rate 17/40 participants) and 21.8 months in the control groups (mortality rate 21/44 participants).

  • Medium‐risk population: treatment with bevacizumab plus chemotherapy may result in lower risk of death compared to chemotherapy alone (HR 0.67, 95% CI 0.51 to 0.91); median OS was 17.9 months in the bevacizumab groups (mortality rate 84/152 participants) and 12.1 months in the control groups (mortality rate 92/151 participants).

  • High‐risk population: treatment with bevacizumab plus chemotherapy may result in lower risk of death compared to chemotherapy alone (HR 0.54, 95% CI 0.32 to 0.91); median OS was 12.1 months in the bevacizumab groups (mortality rate 30/35 participants) and 6.3 months in the control groups (mortality rate 27/30 participants).

Specific adverse events
Gastrointestinal perforations or fistulae

Tewari 2014 reported a higher incidence of gastrointestinal perforations or fistulae in 18 of 220 participants (at least 18) receiving bevacizumab plus chemotherapy compared to 1 of 220 participants receiving chemotherapy alone (risk ratio (RR) 18.00, 95% CI 2.42 to 133.67; 1 study, 440 participants; moderate‐certainty evidence; Analysis 1.2).

Haemorrhage

Tewari 2014 reported a higher incidence of serious haemorrhage in 10 of 220 participants receiving bevacizumab plus chemotherapy compared to 2 of 220 participants receiving chemotherapy alone (RR 5.00, 95% CI 1.11 to 22.56; 1 study, 440 participants; low‐certainty evidence; Analysis 1.3).

Thromboembolic events

Tewari 2014 reported a higher incidence of serious thromboembolic events in 18 of 220 participants receiving bevacizumab plus chemotherapy compared to 4 of 220 participants receiving chemotherapy alone (RR 4.5, 95% CI 1.55 to 13.08; 1 study, 440 participants; moderate‐certainty evidence; Analysis 1.4).

Hypertension

Tewari 2014 reported a higher incidence of hypertension in 55 of 220 participants receiving bevacizumab plus chemotherapy compared to 4 of 220 participants receiving chemotherapy alone (RR 13.75, 95% CI 5.07 to 37.29; 1 study, 440 participants; moderate‐certainty evidence; Analysis 1.5).

Progression‐free survival

In Tewari 2014, treatment with bevacizumab plus chemotherapy resulted in lower risk of disease progression compared to chemotherapy alone (HR 0.68, 95% CI 0.56 to 0.83; 1 study, 452 participants; Analysis 1.6). Tewari 2014 reported median progression‐free survival (PFS) as 8.2 months in the bevacizumab groups (morbidity rate 199/227 participants) and as 6.0 months in the control groups (morbidity rate 206/225 participants).

Quality of life

In Tewari 2014, participants were assessed at five time points with the Trial Outcome of Index of the Functional Assessment of Cancer Therapy (FACT‐Cx‐TOI) (Cella 1993), the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group‐Neurotoxicity (FACT/GOG‐NTX) (Huang 2007), and the Brief Pain Inventory (BPI) (Cleeland 1994). There was no difference in quality of life (QoL) (FACT‐Cx‐TOI scores) with or without bevacizumab (mean difference (MD) ‐1.2, 98.75% CI ‐4.1 to 1.7; 390 participants). There may or not have been a difference in neurotoxicity (FACT/GOG‐NTX scores) with or without bevacizumab (MD 0.23, 98.75% CI ‐1.19 to 1.64; 390 participants). There may or may not have been a difference in pain (BPI) with or without bevacizumab (MD 0.5, 95% CI ‐0.14 to 1.14; 390 participants).

Adverse events

Tewari 2014 reported total adverse events in 218 of 220 participants receiving bevacizumab plus chemotherapy and in 219 of 219 participants receiving chemotherapy alone (RR 0.99, 95% CI 0.98 to 1.01; 1 study, 439 participants; Analysis 1.8). However, treatment with bevacizumab plus chemotherapy (114/220) probably resulted in a higher incidence of serious adverse events compared to chemotherapy alone (79/219) (RR 1.44, 95% CI 1.16 to 1.79; 1 study, 439 participants; moderate‐certainty evidence; Analysis 1.9).

Economic evaluation

In Tewari 2014, study authors evaluated the cost‐effectiveness of bevacizumab using a Markov model based on the GOG‐240 study. The total cost of bevacizumab plus chemotherapy was about 13.2 times that of chemotherapy alone (USD 79,844 versus USD 6053), and the incremental cost‐effectiveness ratio (ICER) was USD 295,164 per quality‐adjusted life‐year (QALY) or USD 24,597 per quality‐adjusted life‐month (1 study, 452 participants; low‐certainty evidence).

Cediranib plus chemotherapy versus chemotherapy only

Overall survival

Symonds 2015 reported risk of death (HR 0.94, 95% CI 0.53 to 1.65; 1 study, 69 participants; low‐certainty evidence; Analysis 2.1summary of findings Table 2); median OS was 13.6 months in the cediranib group (mortality rate 25/34 participants) and 14.8 months in the placebo group (mortality rate 27/35 participants). The effect of cediranib on overall survival might depend on disease site.

  • The 95% CI for the extra‐pelvic metastases only subgroup (N = 21) showed greater benefit compared to the 95% CI for those with local relapse (N = 48) but showed no effects on age, histological subtype, length of disease‐free survival, number of previous treatments, or performance status (details of HR and CI were not provided).

  • The 95% CIs for both the squamous subgroup (N = 47) and the non‐squamous subgroup (N = 22) showed appreciable benefit and harm (details of HR and CI were not provided).

Specific adverse events
Gastrointestinal perforations or fistulae

Symonds 2015 reported gastrointestinal perforations or fistulae in 1 of 32 participants receiving cediranib plus chemotherapy and in 0 of 35 participants receiving placebo plus chemotherapy (RR 3.27, 95% CI 0.14 to 77.57; 1 study, 67 participants; very low‐certainty evidence; Analysis 2.2).

Haemorrhage

Symonds 2015 reported serious haemorrhage in 2 of 32 participants receiving cediranib plus chemotherapy and in 0 of 35 participants receiving placebo plus chemotherapy (RR 5.45, 95% CI 0.27 to 109.49; 1 study, 67 participants; very low‐certainty evidence; Analysis 2.3).

Thromboembolic events

Symonds 2015 reported serious thromboembolic events in 1 of 28 participants receiving cediranib plus chemotherapy and in 0 of 32 participants receiving placebo plus chemotherapy (RR 3.41, 95% CI 0.14 to 80.59; 1 study, 60 participants; very low‐certainty evidence; Analysis 2.4).

Hypertension

Symonds 2015 reported serious hypertension in 0 of 32 participants receiving cediranib plus chemotherapy and in 1 of 35 participants receiving placebo plus chemotherapy (RR 0.36, 95% CI 0.02 to 8.62; 1 study, 67 participants; very low‐certainty evidence; Analysis 2.5).

Progression‐free survival

Symonds 2015 reported risk of disease progression (HR 0.58, 95% CI 0.33 to 1.03; 1 study, 69 participants; Analysis 2.6); median PFS was 8.1 months in the cediranib group (morbidity rate 26/34 participants) and 6.7 months in the placebo group (morbidity rate 29/35 participants).

Quality of life

In Symonds 2015, participants were assessed with the European Organisation for Research and Treatment of Cancer (EORTC) QLQ‐C30 questionnaire (Aaronson 1993). There may or may not be a difference in QoL (median area under the curve values, AUC values from analysis of EORTC QLQ‐C30 by time spent) at 13 time points between cediranib (median AUC ‐5.4, 95% CI ‐13.1 to ‐1.0) and placebo (median AUC ‐11.1, 95% CI ‐20.8 to ‐7.4). QoL associated with diarrhoea was probably worse in women with cediranib (median AUC difference 18, 95% CI 5 to 37).

Adverse events

Symonds 2015 reported total adverse events in 32 of 32 participants receiving cediranib plus chemotherapy and in 35 of 35 participants receiving placebo plus chemotherapy (RR 1.00, 95% CI 0.94 to 1.06; 1 study, 67 participants; Analysis 2.8) and serious adverse events in 19 of 32 participants receiving cediranib plus chemotherapy and in 18 of 35 participants receiving placebo plus chemotherapy (RR 1.15, 95% CI 0.75 to 1.78; 1 study, 67 participants; low‐certainty evidence; Analysis 2.9).

Economic evaluation

This was not measured.

Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only

Overall survival

Guo 2020 reported risk of death (HR 0.90, 95% CI 0.51 to 1.60; 1 study, 52 participants; low‐certainty evidence; Analysis 3.1; summary of findings Table 3); median OS was 14.7 months in the apatinib group (mortality rate 11/28 participants) and 12.8 months in the control group (mortality rate 16/24 participants).

Summary of adverse events

Apatinib required dose reduction in three participants due to severe treatment‐associated toxicity, including hypertension and hand‐foot syndrome. Severe hypertension and hand‐foot syndrome were adequately relieved when the drug dose was reduced, or when participants were given symptomatic treatment to support medication adherence and apatinib tolerance. Apatinib did not significantly increase the incidence of neutropenia compared with control (75% versus 75%), nor the incidence of anaemia (75% versus 66.7%); diarrhoea (57.1% versus 41.6%); nausea and vomiting (57.1% versus 37.5%); fatigue (50% versus 25%); dysuria, urinary urgency, and urinary frequency (28.6% versus 25%); alopecia (18.6% versus 25%); thrombocytopenia (25% versus 16.7%); and liver toxicity (14.3% versus 12.5%). However, proteinuria (53.6% versus 16.7%), hand‐foot syndrome (50% versus 16.7%), mucositis (46.4% versus 12.5%), and hypertension (42.8% versus 8.3%) at all severities were more common in the apatinib group than in the control group.

Specific adverse events
Gastrointestinal perforations or fistulae

This was not reported.

Haemorrhage

This was not reported.

Thromboembolic events

This was not reported.

Hypertension

Guo 2020 reported a higher incidence of hypertension in 12 of 28 participants receiving apatinib plus chemotherapy or chemotherapy/brachytherapy compared to 2 of 24 participants receiving chemotherapy or chemotherapy/brachytherapy (RR 5.14, 95% CI 1.28 to 20.73; 1 study, 52 participants; low‐certainty evidence; Analysis 3.2).

Progression‐free survival

In Guo 2020, treatment with apatinib plus chemotherapy or chemotherapy/brachytherapy resulted in lower risk of disease progression compared to chemotherapy or chemotherapy/brachytherapy alone (HR 0.44, 95% CI 0.25 to 0.78; 1 study, 52 participants; Analysis 3.3). Guo 2020 reported median PFS as 10.1 months in the apatinib group and 6.4 months in the control group.

Quality of life

This was not measured.

Economic evaluation

This was not measured.

Pazopanib plus lapatinib versus lapatinib only

Overall survival

In Monk 2010, treatment with pazopanib plus lapatinib probably resulted in higher risk of death compared to treatment with lapatinib alone (HR 2.71, 95% CI 1.16 to 6.31; 2 arms from 1 study, 117 participants; low‐certainty evidence; Analysis 4.1; summary of findings Table 4). Monk 2010 reported median OS in the pazopanib plus lapatinib group as 25.7 weeks (mortality rate 14/59 participants) and in the lapatinib group as 35.0 weeks (mortality rate 10/58 participants).

Specific adverse events
Gastrointestinal perforations or fistulae

Monk 2010 reported gastrointestinal perforations or fistulae in 2 of 76 participants receiving pazopanib plus lapatinib and in 1 of 76 participants receiving lapatinib alone (RR 2.00, 95% CI 0.19 to 21.59; 2 arms from 1 study, 152 participants; very low‐certainty evidence; Analysis 4.2).

Haemorrhage

Monk 2010 reported haemorrhage in 10 of 76 (at least 10) participants receiving pazopanib plus lapatinib and in 5 of 76 (at least five) participants receiving lapatinib alone (RR 2.00, 95% CI 0.72 to 5.58; 2 arms from 1 study, 152 participants; very low‐certainty evidence; Analysis 4.3).

Thromboembolic events

Monk 2010 reported thromboembolic events in 1 of 76 participants receiving pazopanib plus lapatinib and in 0 of 76 participants receiving lapatinib alone (RR 3.00, 95% CI 0.12 to 72.50; 2 arms from 1 study, 152 participants; very low‐certainty evidence; Analysis 4.4).

Hypertension

Monk 2010 reported a higher incidence of hypertension in 24 of 76 participants receiving pazopanib plus lapatinib compared to 2 of 76 participants receiving lapatinib alone (RR 12.00, 95% CI 2.94 to 49.01; 2 arms from 1 study, 152 participants; moderate‐certainty evidence; Analysis 4.5).

Progression‐free survival

Monk 2010 reported risk of disease progression (HR 1.05, 95% CI 0.59 to 1.86; 2 arms from 1 study, 117 participants; Analysis 4.6); median PFS was 12.6 months in the pazopanib plus lapatinib group (morbidity rate 20/59 participants) and 12.6 months in the lapatinib alone group (morbidity rate 28/58 participants).

Quality of life

This was not measured.

Adverse events

Monk 2010 reported total adverse events in 72 of 76 participants receiving pazopanib plus lapatinib and in 71 of 76 participants receiving lapatinib (RR 1.01, 95% CI 0.94 to 1.10; 2 arms from 1 study, 152 participants; Analysis 4.7) and serious adverse events in 32 of 76 participants receiving pazopanib plus lapatinib and in 22 of 76 participants receiving lapatinib alone (RR 1.45, 95% CI 0.94 to 2.26; 2 arms from 1 study, 152 participants; low‐certainty evidence; Analysis 4.8).

Economic evaluation

This was not measured.

Pazopanib versus lapatinib

Overall survival

Monk 2010 reported risk of death (HR 0.96, 95% CI 0.67 to 1.38; 2 arms from 1 study, 152 participants; low‐certainty evidence; Analysis 5.1; summary of findings Table 5); median OS was 49.7 weeks in the pazopanib group (mortality rate 56/74 participants) and 44.1 weeks in the lapatinib group (mortality rate 62/78 participants).

Specific adverse events
Gastrointestinal perforations or fistulae

Monk 2010 reported gastrointestinal perforations or fistulae in 1 of 74 participants receiving pazopanib monotherapy and in 1 of 76 participants receiving lapatinib monotherapy (RR 1.03, 95% CI 0.07 to 16.12; 2 arms from 1 study, 150 participants; very low‐certainty evidence; Analysis 5.2).

Haemorrhage

Monk 2010 reported haemorrhage in 5 of 74 (at least five) participants receiving pazopanib monotherapy and in 5 of 76 (at least five) participants receiving lapatinib monotherapy (RR 1.03, 95% CI 0.31 to 3.40; 2 arms from 1 study, 150 participants; very low‐certainty evidence; Analysis 5.3).

Thromboembolic events

Monk 2010 reported thromboembolic events in 1 of 74 participants receiving pazopanib monotherapy and in 0 of 76 participants receiving lapatinib monotherapy (RR 3.08, 95% CI 0.13 to 74.42; 2 arms from 1 study, 150 participants; very low‐certainty evidence; Analysis 5.4).

Hypertension

Monk 2010 reported a higher incidence of hypertension in 23 of 74 participants receiving pazopanib monotherapy compared to 2 of 76 participants receiving lapatinib monotherapy (RR 11.81, 95% CI 2.89 to 48.33; 2 arms from 1 study, 150 participants; moderate‐certainty evidence; Analysis 5.5).

Progression‐free survival

In Monk 2010, treatment with pazopanib monotherapy probably resulted in lower risk of disease progression compared to lapatinib monotherapy (HR 0.66, 95% CI 0.45 to 0.97; 2 arms from 1 study, 152 participants; Analysis 5.6); Monk 2010 reported median PFS as 18.1 weeks in the pazopanib group (morbidity rate 46/74 participants) and as 17.1 weeks in the lapatinib group (morbidity rate 59/78 participants).

Quality of life

This was not measured.

Adverse events

Monk 2010 reported total adverse events in 72 of 74 participants receiving pazopanib monotherapy and in 71 of 76 participants receiving lapatinib monotherapy (RR 1.04, 95% CI 0.97 to 1.12; 2 arms from 1 study, 150 participants; Analysis 5.7) and serious adverse events in 28 of 74 participants receiving pazopanib monotherapy and in 22 of 76 participants receiving lapatinib monotherapy (RR 1.31, 95% CI 0.83 to 2.07; 2 arms from 1 study, 150 participants; low‐certainty evidence; Analysis 5.8).

Economic evaluation

This was not measured.

Discussion

Summary of main results

The review authors identified four studies with a total of 808 participants. Tewari 2014 and Symonds 2015 were funded by government agencies. Guo 2020 and Monk 2010 were funded by manufacturers.

One study included 452 women and compared bevacizumab plus chemotherapy versus chemotherapy alone (Tewari 2014). Bevacizumab improved overall survival (OS) (low‐certainty evidence); however, it increased specific adverse events including gastrointestinal perforations or fistulae (moderate‐certainty evidence), serious haemorrhage (low‐certainty evidence), serious thromboembolic events (moderate‐certainty evidence), and hypertension (moderate‐certainty evidence); it also increased serious adverse events (moderate‐certainty evidence) and costs (low‐certainty evidence).

A second study included 69 women and compared cediranib plus chemotherapy versus placebo plus chemotherapy (Symonds 2015). Cediranib made no difference in OS (low‐certainty evidence) nor in specific adverse events including gastrointestinal perforations or fistulae (very low‐certainty evidence), serious haemorrhage (very low quality of evidence), serious thromboembolic events (very low‐certainty evidence), and serious hypertension (very low‐certainty evidence); it also made no difference in serious adverse events (low‐certainty evidence).

A third study included 59 women and compared apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy alone (Guo 2020). Apatinib made no difference in OS (low‐certainty evidence) but increased hypertension (low‐certainty evidence).

A fourth trial included 228 women (Monk 2010). This study compared (1) pazopanib plus lapatinib versus lapatinib alone, and (2) pazopanib versus lapatinib.

  • Pazopanib plus lapatinib versus lapatinib alone: pazopanib plus lapatinib reduced OS (low‐certainty evidence); made no difference in some specific adverse events including gastrointestinal perforations or fistulae (very low‐certainty evidence), haemorrhage (very low‐certainty evidence), thromboembolic events (very low‐certainty evidence), and increased hypertension (moderate‐certainty evidence); and made no difference in serious adverse events (low‐certainty evidence).

  • Pazopanib versus lapatinib: pazopanib made no difference in OS (low‐certainty evidence); made no difference in some specific adverse events including gastrointestinal perforations or fistulae (very low‐certainty evidence), haemorrhage (very low‐certainty evidence), thromboembolic events (very low‐certainty evidence), and increased hypertension (moderate‐certainty evidence); and made no difference in serious adverse events (low‐certainty evidence).

The review authors found two completed studies with a total of 115 women and added these to Studies awaiting classification. One study included 75 women and compared bevacizumab plus chemoradiotherapy versus chemoradiotherapy alone for women with advanced cervical adenocarcinoma and did not address OS (Xie 2016). A second study included 40 women, compared apatinib plus chemoradiotherapy versus chemoradiotherapy alone for women with FIGO (International Federation of Obstetrics and Gynaecology) stage IIB to IV cervical cancer, and addressed OS (Rao 2018). Both studies were not registered online (Rao 2018Xie 2016); although we contacted their principal investigators, no additional data were provided.

The review authors also identified nine ongoing studies (expected recruitment 854 participants) for three tyrosine kinase inhibitors (TKIs) of vascular endothelial growth factor (VEGF) receptor (VEGF‐R) (apatinib, anlotinib, nintedanib). These studies have not reported any outcomes to date (Ongoing studies).

Overall completeness and applicability of evidence

The four included studies were insufficient to address all of the objectives of this review. Types of interventions were limited, including bevacizumab plus chemotherapy, cediranib plus chemotherapy, apatinib plus chemotherapy or chemotherapy/brachytherapy, pazopanib plus lapatinib, and pazopanib monotherapy. Overall completeness and applicability of evidence were low, as each comparison included only one study, and most studies were small.

Factors such as disease spread, histological type, and risk category should be considered in the applicability of main results. Tumour biology may be different between pelvic site and extra‐pelvic metastasis, between measurable and unmeasurable, between squamous and non‐squamous, and between low risk and medium/high risk. The benefit of bevacizumab plus chemotherapy appears to be greater in women untreated with pelvic disease, cancers of squamous histological type, and medium/high risk. The effect of cediranib plus chemotherapy appears to be invalid in untreated women, especially for those with extra‐pelvic disease.

Apatinib may make little to no difference in survival among untreated women; however, Guo 2020 mixed two types of interventions: apatinib plus chemotherapy in women with recurrent disease, and apatinib plus chemotherapy/brachytherapy in women with stage IVB disease. No independent survival results were provided for chemotherapy or chemotherapy/brachytherapy.

Pazopanib plus lapatinib reduced OS, and data do not support this treatment for women who have received prior anti‐cancer therapies. Pazopanib monotherapy may not be better (except for progression‐free survival) than lapatinib monotherapy for women who have received prior anti‐cancer therapies.

These studies reported substantial discontinuation of treatment or toxicity (participants did not complete the intervention). Regardless of the intention‐to‐treat population, 95%, 99%, and 94% of participants discontinued the intervention in Monk 2010, Tewari 2014, and Symonds 2015, respectively. Discontinuation implied that most women needed additional salvage therapy or supportive care.

Bevacizumab improved OS by 3.5 months (versus median 13.3 months) with a higher incidence (52%, 114/220 versus 36%, 79/219) of serious adverse events and higher incremental costs in the bevacizumab plus chemotherapy groups. Women with a high preference for survival prolongation and with limited aversion to serious adverse events who do not consider bevacizumab therapy a burden may opt to use bevacizumab; others may not.

Quality of the evidence

Risk of selection bias was low in Monk 2010, Symonds 2015, and Tewari 2014, as participants were randomly assigned by validated randomisation systems. Risk of selection bias was unclear in Guo 2020, as information about random sequence generation or allocation concealment was not available. Risk of performance and detection bias was low in Symonds 2015, as participants, investigators, and outcomes assessors were blinded. Risk of performance and detection bias was high for subjective outcomes (i.e. progression‐free survival, quality of life) in Guo 2020, Monk 2010, and Tewari 2014 as 'open‐label' but was low for objective outcomes (i.e. OS, adverse events, economic evaluation). Risk of attrition was low in all four studies. Risk of reporting bias was low in Monk 2010, Symonds 2015, and Tewari 2014, as outcomes in the protocols were the same in these publications. Risk of reporting bias was high in Guo 2020, as study registration occurred later than study completion. Risk of other bias was high in Guo 2020 and Monk 2010, as these studies were supported and sponsored by the manufacturer. Risk of other bias was unclear in Tewari 2014, as economic evaluation was not a planned analysis in the study protocol.

The level of certainty of evidence for bevacizumab plus chemotherapy compared to chemotherapy was low to moderate (summary of findings Table 1). We downgraded all outcomes by one for serious heterogeneity, as outcomes might be different for different subgroups in the only included study if the sample were larger. In addition, we downgraded 'OS and serious haemorrhage' by one for serious imprecision, as optimal information size was not met; we downgraded 'economic evaluation' by one for serious risk of bias, as this was not a planned analysis in the study protocol.

The level of certainty of evidence for 'cediranib plus chemotherapy compared to placebo plus chemotherapy' was low to very low (summary of findings Table 2). We downgraded all outcomes by one for serious heterogeneity, as outcomes might be different for different subgroups in the only included study and with a large sample. In addition, we downgraded 'OS' by one for serious imprecision, as optimal information size was not met; we downgraded 'gastrointestinal perforations or fistulae, serious haemorrhage, serious thromboembolic events, and serious hypertension' by two for very serious imprecision, as there were few events and the confidence interval included appreciable benefit and harm; we downgraded 'serious adverse events' by one for serious imprecision, as optimal information size was not met.

The level of certainty of evidence for 'apatinib plus chemotherapy or chemotherapy/brachytherapy compared to chemotherapy or chemotherapy/brachytherapy' was low (summary of findings Table 3). We downgraded all outcomes by one for serious risk of bias, as the only included study was funded by the drug manufacturer. In addition, we downgraded 'OS' by one for serious imprecision, as optimal information size was not met; we downgraded 'hypertension' by one for serious risk of bias, as trial registration occurred later than study completion.

The level of certainty of evidence for 'pazopanib plus lapatinib compared to lapatinib' was moderate to very low (summary of findings Table 4). We downgraded all outcomes by one for serious risk of bias, as the only included study was funded by the drug manufacturer and this may have contributed to bias in the final results. In addition, we downgraded 'OS' by one for serious imprecision, as optimal information size was not met; we downgraded 'gastrointestinal perforations or fistulae, haemorrhage, and thromboembolic events' by two for very serious imprecision, as there were few events and the confidence interval included appreciable benefit and harm; we downgraded 'serious adverse events' by one for serious imprecision, as optimal information size was not met.

The level of certainty of evidence for 'pazopanib compared to lapatinib' was moderate to very low (summary of findings Table 5). We downgraded all outcomes by one for serious risk of bias, as the only included study was funded by the drug manufacturer and this may have biased the final outcomes. In addition, we downgraded 'OS' by one for serious imprecision, as optimal information size was not met; we downgraded 'gastrointestinal perforations or fistulae, haemorrhage, and thromboembolic events' by two for very serious imprecision, as there were few events and the confidence interval included appreciable benefit and harm; we downgraded 'serious adverse events' by one for serious imprecision, as optimal information size was not met.

Potential biases in the review process

To prevent potential biases in the review process, we performed a comprehensive search, including a thorough search of multiple databases, online registers of clinical trials, and conference proceedings, with assistance from the Cochrane Gynaecological, Neuro‐oncology and Orphan Cancer Group. Three review authors independently performed all relevant processes (search, data collection, and analysis). We were unable to assess publication bias, as the review included only four studies.

Two awaiting classification studies addressed the effects of agents (apatinib or bevacizumab) plus chemoradiotherapy. Even with data, the two small studies (115 total participants) may not be able to answer the main question of this review.

The effects of other agents (apatinib, anlotinib, or nintedanib) plus chemotherapy are being assessed in seven ongoing studies.

Agreements and disagreements with other studies or reviews

The results of this Cochrane Review on the efficacy of anti‐VEGF antibody (bevacizumab) were consistent with those of other published systematic reviews (Rosen 2017; Zagouri 2012), as well as a multitude of narrative reviews (Alldredge 2016; Eskander 2014; Gadducci 2015; Jayson 2016; Krill 2015; Luvero 2017; Minion 2017; Pfaendler 2018; Tomao 2014; Wei 2013). Zagouri 2012 was a systematic review that included 23 phase 1 and 2 trials and seven observational studies to narrate molecular targeted therapy for cervical cancer (not limited to persistent, recurrent, or metastatic disease). These molecular targeted agents included anti‐VEGF antibody (bevacizumab), TKIs of VEGF‐R, TKIs of epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2/neu), mammalian target of rapamycin inhibitors, cyclo‐oxygenase‐2 inhibitors, and histone deacetylase inhibitors, among others. No phase 3 trials were found. The results for bevacizumab from five non‐randomised studies were promising. Rosen 2017 was a systematic review and network meta‐analysis that included five randomised controlled trials (RCTs) (Kitagawa 2015; Long 2005; Monk 2009b; Symonds 2015; Tewari 2014); this review showed that bevacizumab plus chemotherapy (paclitaxel/topotecan and paclitaxel/cisplatin) was likely to prolong OS over other non‐bevacizumab chemotherapy regimens, which were not included in Tewari 2014. Our review provides the most recent information from RCTs about TKIs of VEGF‐R. This review aimed to provide evidence from multiple comparisons to evaluate the effectiveness and safety of different VEGF targeting agents alone or in the adjuvant setting for treatment of persistent, recurrent, or metastatic cervical cancer.

FIGO stage 2018 (with correction Bhatla 2018; Bhatla 2019).

Figures and Tables -
Figure 1

FIGO stage 2018 (with correction Bhatla 2018; Bhatla 2019).

Study flow diagram.

Figures and Tables -
Figure 2

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Figures and Tables -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Figures and Tables -
Figure 4

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 1: Overall survival

Figures and Tables -
Analysis 1.1

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 1: Overall survival

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Figures and Tables -
Analysis 1.2

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 3: Specific adverse events: serious haemorrhage

Figures and Tables -
Analysis 1.3

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 3: Specific adverse events: serious haemorrhage

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 4: Specific adverse events: serious thromboembolic events

Figures and Tables -
Analysis 1.4

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 4: Specific adverse events: serious thromboembolic events

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 5: Specific adverse events: hypertension

Figures and Tables -
Analysis 1.5

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 5: Specific adverse events: hypertension

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 6: Progression‐free survival

Figures and Tables -
Analysis 1.6

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 6: Progression‐free survival

Quality of life

Study

Heading 1

Tewari 2014

There was no difference in QoL (FACT‐Cx‐TOI scores) with or without bevacizumab (MD ‐1.2, 98.75% CI ‐4.1 to 1.7; participants = 390).

There was no difference in neurotoxicity (FACT/GOG‐NTX scores) with or without bevacizumab (MD 0.23, 98.75% CI ‐1.19 to 1.64; participants = 390).

There was no difference in pain (BPI) with or without bevacizumab (MD 0.5, 95% CI ‐0.14 to 1.14; participants = 390).

Figures and Tables -
Analysis 1.7

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 7: Quality of life

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 8: Total adverse events

Figures and Tables -
Analysis 1.8

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 8: Total adverse events

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 9: Serious adverse events

Figures and Tables -
Analysis 1.9

Comparison 1: Bevacizumab plus chemotherapy versus chemotherapy only, Outcome 9: Serious adverse events

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 1: Overall survival

Figures and Tables -
Analysis 2.1

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 1: Overall survival

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Figures and Tables -
Analysis 2.2

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 3: Specific adverse events: serious haemorrhage

Figures and Tables -
Analysis 2.3

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 3: Specific adverse events: serious haemorrhage

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 4: Specific adverse events: serious thromboembolic events

Figures and Tables -
Analysis 2.4

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 4: Specific adverse events: serious thromboembolic events

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 5: Specific adverse events: serious hypertension

Figures and Tables -
Analysis 2.5

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 5: Specific adverse events: serious hypertension

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 6: Progression‐free survival

Figures and Tables -
Analysis 2.6

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 6: Progression‐free survival

Quality of life

Study

Heading 1

Symonds 2015

There was no difference in QoL (median AUC values, analysis of EORTC QLQ‐C30 by the time spent) at 13 time points between cediranib (median AUC ‐5.4, 95% CI ‐13.1 to ‐1.0) and placebo (median AUC ‐11.1, 95% CI ‐20.8 to ‐7.4). QoL associated with diarrhoea was worse in patients with cediranib (median AUC difference 18, 95% CI 5 to 37).

Figures and Tables -
Analysis 2.7

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 7: Quality of life

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 8: Total adverse events

Figures and Tables -
Analysis 2.8

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 8: Total adverse events

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 9: Serious adverse events

Figures and Tables -
Analysis 2.9

Comparison 2: Cediranib plus chemotherapy versus chemotherapy only, Outcome 9: Serious adverse events

Comparison 3: Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only, Outcome 1: Overall survival

Figures and Tables -
Analysis 3.1

Comparison 3: Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only, Outcome 1: Overall survival

Comparison 3: Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only, Outcome 2: Specific adverse events: hypertension

Figures and Tables -
Analysis 3.2

Comparison 3: Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only, Outcome 2: Specific adverse events: hypertension

Comparison 3: Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only, Outcome 3: Progression‐free survival

Figures and Tables -
Analysis 3.3

Comparison 3: Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only, Outcome 3: Progression‐free survival

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 1: Overall survival

Figures and Tables -
Analysis 4.1

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 1: Overall survival

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Figures and Tables -
Analysis 4.2

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 3: Specific adverse events: haemorrhage

Figures and Tables -
Analysis 4.3

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 3: Specific adverse events: haemorrhage

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 4: Specific adverse events: thromboembolic events

Figures and Tables -
Analysis 4.4

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 4: Specific adverse events: thromboembolic events

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 5: Specific adverse events: hypertension

Figures and Tables -
Analysis 4.5

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 5: Specific adverse events: hypertension

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 6: Progression‐free survival

Figures and Tables -
Analysis 4.6

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 6: Progression‐free survival

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 7: Total adverse events

Figures and Tables -
Analysis 4.7

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 7: Total adverse events

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 8: Serious adverse events

Figures and Tables -
Analysis 4.8

Comparison 4: Pazopanib plus lapatinib versus lapatinib only, Outcome 8: Serious adverse events

Comparison 5: Pazopanib versus lapatinib, Outcome 1: Overall survival

Figures and Tables -
Analysis 5.1

Comparison 5: Pazopanib versus lapatinib, Outcome 1: Overall survival

Comparison 5: Pazopanib versus lapatinib, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Figures and Tables -
Analysis 5.2

Comparison 5: Pazopanib versus lapatinib, Outcome 2: Specific adverse events: gastrointestinal perforations or fistulae

Comparison 5: Pazopanib versus lapatinib, Outcome 3: Specific adverse events: haemorrhage

Figures and Tables -
Analysis 5.3

Comparison 5: Pazopanib versus lapatinib, Outcome 3: Specific adverse events: haemorrhage

Comparison 5: Pazopanib versus lapatinib, Outcome 4: Specific adverse events: thromboembolic events

Figures and Tables -
Analysis 5.4

Comparison 5: Pazopanib versus lapatinib, Outcome 4: Specific adverse events: thromboembolic events

Comparison 5: Pazopanib versus lapatinib, Outcome 5: Specific adverse events: hypertension

Figures and Tables -
Analysis 5.5

Comparison 5: Pazopanib versus lapatinib, Outcome 5: Specific adverse events: hypertension

Comparison 5: Pazopanib versus lapatinib, Outcome 6: Progression‐free survival

Figures and Tables -
Analysis 5.6

Comparison 5: Pazopanib versus lapatinib, Outcome 6: Progression‐free survival

Comparison 5: Pazopanib versus lapatinib, Outcome 7: Total adverse events

Figures and Tables -
Analysis 5.7

Comparison 5: Pazopanib versus lapatinib, Outcome 7: Total adverse events

Comparison 5: Pazopanib versus lapatinib, Outcome 8: Serious adverse events

Figures and Tables -
Analysis 5.8

Comparison 5: Pazopanib versus lapatinib, Outcome 8: Serious adverse events

Summary of findings 1. Bevacizumab plus chemotherapy compared to chemotherapy

Bevacizumab plus chemotherapy compared to chemotherapy for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Settings: hospital

Intervention: bevacizumab plus chemotherapy

Comparison: chemotherapy only

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with the chemotherapy only

Risk with bevacizumab plus chemotherapy

Overall survival

791 per 1000

701 per 1000
(621 to 774)

HR 0.77
(0.62 to 0.95)

452
(1 RCT)

⊕⊕⊝⊝
lowa,b

Maximum follow‐up was longer than 50 months. Median overall survival in the bevacizumab groups was 16.8 months (mortality rate 170/227 participants). Median overall survival in the control groups was 13.3 months (mortality rate 178/225 participants)

Specificadverse events: gastrointestinal perforations or fistulae

5 per 1,000

82 per 1,000
(11 to 608)

RR 18.00
(2.42 to 133.67)

440
(1 RCT)

⊕⊕⊕⊝
moderatea

At least 18 women (a maximum of 23) had fistulae events (N = 18) or perforation events (N = 5) in the bevacizumab group

Specific adverse events: serious haemorrhage

9 per 1,000

45 per 1,000
(10 to 205)

RR 5.00
(1.11 to 22.56)

440
(1 RCT)

⊕⊕⊝⊝
lowa,b

Specific adverse events: serious thromboembolic events

18 per 1,000

82 per 1,000
(28 to 238)

RR 4.50
(1.55 to 13.08)

440
(1 RCT)

⊕⊕⊕⊝
moderatea

Specific adverse events: hypertension

18 per 1,000

250 per 1,000
(92 to 678)

RR 13.75
(5.07 to 37.29)

440
(1 RCT)

⊕⊕⊕⊝
moderatea

Serious adverse events

361 per 1,000

519 per 1,000
(418 to 646)

RR 1.44
(1.16 to 1.79)

439
(1 RCT)

⊕⊕⊕⊝
moderatea

Economic evaluation

see comment

see comment

452
(1 RCT)

⊕⊕⊝⊝
lowa,c

The incremental cost‐effectiveness ratio (ICER) was USD 295,164 per quality‐adjusted life‐year (QALY), or USD 24,597 per quality‐adjusted life‐month

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious heterogeneity. Outcomes might be different in different subgroups in the only included study.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by one for serious risk of bias. This analysis was not planned in the study protocol.

Figures and Tables -
Summary of findings 1. Bevacizumab plus chemotherapy compared to chemotherapy
Summary of findings 2. Cediranib plus chemotherapy compared to placebo plus chemotherapy

Cediranib plus chemotherapy compared to placebo plus chemotherapy for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Settings: hospital

Intervention: cediranib plus chemotherapy

Comparison: placebo plus chemotherapy

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with placebo plus chemotherapy

Risk with cediranib plus chemotherapy

Overall survival

771 per 1000

750 per 1000
(543 to 912)

HR 0.94
(0.53 to 1.65)

69
(1 RCT)

⊕⊕⊝⊝
lowa,b

Median follow‐up was 24.2 months. Median overall survival in the cediranib group was 13.6 months (mortality rate 25/34 participants). Median overall survival in the placebo group was 14.8 months (mortality rate 27/35 participants)

Specific adverse events: gastrointestinal perforations or fistulae

0 per 1000

0 per 1000
(0 to 0)

RR 3.27
(0.14 to 77.57)

67
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: serious haemorrhage

0 per 1000

0 per 1000
(0 to 0)

RR 5.45
(0.27 to 109.49)

67
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: serious thromboembolic events

0 per 1000

0 per 1000
(0 to 0)

RR 3.41
(0.14 to 80.59)

60
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: serioushypertension

29 per 1000

10 per 1000
(1 to 246)

RR 0.36
(0.02 to 8.62)

67
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Serious adverse events

514 per 1000

591 per 1000
(386 to 915)

RR 1.15
(0.75 to 1.78)

67
(1 RCT)

⊕⊕⊝⊝
lowa,b

Economic evaluation ‐ not measured

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious heterogeneity. Outcomes might be different in different subgroups in the only included study.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by two for very serious imprecision. There were few events and the confidence interval included appreciable benefit and harm.

Figures and Tables -
Summary of findings 2. Cediranib plus chemotherapy compared to placebo plus chemotherapy
Summary of findings 3. Apatinib plus chemotherapy or chemotherapy/brachytherapy compared to chemotherapy or chemotherapy/brachytherapy

Apatinib plus chemotherapy or chemotherapy/brachytherapy compared with chemotherapy or chemotherapy/brachytherapy for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with recurrent or FIGO stage IVB cervical cancer

Settings: hospital

Intervention: apatinib plus chemotherapy or chemotherapy/brachytherapy

Comparison: chemotherapy or chemotherapy/brachytherapy

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with chemotherapy or chemotherapy/brachytherapy

Risk with apatinibplus chemotherapy or chemotherapy/brachytherapy

Overall survival

667 per 1000

628 per 1000
(161 to 429)

HR 0.90
(0.51 to 1.60)

52
(1 RCT)

⊕⊕⊝⊝
lowa,b

Median follow‐up was 14 months. Median overall survival in the apatinib group was 14.7 months (mortality rate 11/28 participants). Median overall survival in the control group was 12.8 months (mortality rate 16/24 participants)

Specific adverse events: gastrointestinal perforations or fistulae ‐ not reported

Specific adverse events: haemorrhage ‐ not reported

Specific adverse events: thromboembolic events ‐ not reported

Specific adverse events: hypertension

83 per 1000

428 per 1000
(107 to 1000)

RR 5.14
(1.28 to 20.73)

52
(1 RCT)

⊕⊕⊝⊝
lowa,c

Serious adverse events ‐ not reported

Economic evaluation ‐ not measured

*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; FIGO: International Federation of Obstetrics and Gynaecology; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious risk of bias. This single small study was funded by the drug manufacturer.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by one for serious risk of bias. Trial registration occurred later than trial completion.

Figures and Tables -
Summary of findings 3. Apatinib plus chemotherapy or chemotherapy/brachytherapy compared to chemotherapy or chemotherapy/brachytherapy
Summary of findings 4. Pazopanib plus lapatinib compared to lapatinib

Pazopanib plus lapatinib compared to lapatinib for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Setting: clinic

Intervention: pazopanib plus lapatinib

Comparison: lapatinib only

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with lapatinib only

Risk with pazopanib plus lapatinib

Overall survival

172 per 1000

401 per 1000
(197 to 697)

HR 2.71
(1.16 to 6.31)

117
(1 RCT)

⊕⊕⊝⊝
lowa,b

Maximum follow‐up was longer than 40 weeks. Median overall survival in the pazopanib plus lapatinib group was 25.7 weeks (mortality rate 14/59 participants). Median overall survival in the lapatinib group was 35.0 weeks (mortality rate 10/58 participants)

Specific adverse events: gastrointestinal perforations or fistulae

13 per 1000

26 per 1000
(3 to 284)

RR 2.00
(0.19 to 21.59)

152
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: haemorrhage

66 per 1000

132 per 1000
(47 to 367)

RR 2.00
(0.72 to 5.58)

152
(1 RCT)

⊕⊝⊝⊝
verylowa,c

At least 10 women (maximum of 29) with 29 bleeding events were included in the pazopanib plus lapatinib group; at least 5 women (maximum of 11) with 11 bleeding events were included in the lapatinib group

Specific adverse events: thromboembolic events

0 per 1000

0 per 1000
(0 to 0)

RR 3.00
(0.12 to 72.50)

152
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: hypertension

26 per 1000

316 per 1000
(77 to 1000)

RR 12.00
(2.94 to 49.01)

152
(1 RCT)

⊕⊕⊕⊝
moderatea

Serious adverse events

289 per 1000

420 per 1000
(272 to 654)

RR 1.45
(0.94 to 2.26)

152
(1 RCT)

⊕⊕⊝⊝
lowa,b

A total of 5 women In the pazopanib plus lapatinib group had fatal adverse events

Economic evaluation ‐ not measured

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious risk of bias. This single small study was funded by the drug manufacturer.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by two for very serious imprecision. There were few events and the confidence interval included appreciable benefit and harm.

Figures and Tables -
Summary of findings 4. Pazopanib plus lapatinib compared to lapatinib
Summary of findings 5. Pazopanib compared to lapatinib

Pazopanib compared to lapatinib for persistent, recurrent, or metastatic cervical cancer

Patient or population: women with persistent, recurrent, or metastatic cervical cancer

Setting: clinic

Intervention: pazopanib only

Comparison: lapatinib only

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of evidence
(GRADE)

Comments

Risk with lapatinib only

Risk with pazopanib only

Overall survival

795 per 1000

781 per 1000
(654 to 888)

HR 0.96
(0.67 to 1.38)

152
(1 RCT)

⊕⊕⊝⊝
lowa,b

Maximum follow‐up was longer than 150 weeks. Median overall survival in the pazopanib group was 49.7 weeks (mortality rate 56/74 participants). Median overall survival in the lapatinib group was 44.1 weeks (mortality rate 62/78 participants)

Specific adverse events: gastrointestinal perforations or fistulae

13 per 1000

14 per 1000
(1 to 212)

RR 1.03
(0.07 to 16.12)

150
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: haemorrhage

66 per 1000

68 per 1000
(20 to 224)

RR 1.03
(0.31 to 3.40)

150
(1 RCT)

⊕⊝⊝⊝
verylowa,c

At least 5 women (maximum of 13) in the pazopanib group had 13 bleeding events; at least 5 women (maximum of 11) in the lapatinib group had 11 bleeding events

Specific adverse events: thromboembolic events

0 per 1000

0 per 1000
(0 to 0)

RR 3.08
(0.13 to 74.42)

150
(1 RCT)

⊕⊝⊝⊝
verylowa,c

Specific adverse events: hypertension

26 per 1000

311 per 1000
(76 to 1,000)

RR 11.81
(2.89 to 48.33)

150
(1 RCT)

⊕⊕⊕⊝
moderatea

Serious adverse events

289 per 1000

379 per 1000
(240 to 599)

RR 1.31
(0.83 to 2.07)

150
(1 RCT)

⊕⊕⊝⊝
lowa,b

Economic evaluation ‐ not measured

*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; HR: hazard ratio; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

aDowngraded by one for serious risk of bias. This single small study was funded by the drug manufacturer.

bDowngraded by one for serious imprecision. Optimal information size was not met.

cDowngraded by two for very serious imprecision. There were few events and the confidence interval included appreciable benefit and harm.

Figures and Tables -
Summary of findings 5. Pazopanib compared to lapatinib
Comparison 1. Bevacizumab plus chemotherapy versus chemotherapy only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Overall survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

1.2 Specific adverse events: gastrointestinal perforations or fistulae Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.3 Specific adverse events: serious haemorrhage Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.4 Specific adverse events: serious thromboembolic events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.5 Specific adverse events: hypertension Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.6 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

1.7 Quality of life Show forest plot

1

Other data

No numeric data

1.8 Total adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.9 Serious adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 1. Bevacizumab plus chemotherapy versus chemotherapy only
Comparison 2. Cediranib plus chemotherapy versus chemotherapy only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Overall survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

2.2 Specific adverse events: gastrointestinal perforations or fistulae Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.3 Specific adverse events: serious haemorrhage Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.4 Specific adverse events: serious thromboembolic events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.5 Specific adverse events: serious hypertension Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.6 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

2.7 Quality of life Show forest plot

1

Other data

No numeric data

2.8 Total adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.9 Serious adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 2. Cediranib plus chemotherapy versus chemotherapy only
Comparison 3. Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 Overall survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

3.2 Specific adverse events: hypertension Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3.3 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 3. Apatinib plus chemotherapy or chemotherapy/brachytherapy versus chemotherapy or chemotherapy/brachytherapy only
Comparison 4. Pazopanib plus lapatinib versus lapatinib only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Overall survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

4.2 Specific adverse events: gastrointestinal perforations or fistulae Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.3 Specific adverse events: haemorrhage Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.4 Specific adverse events: thromboembolic events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.5 Specific adverse events: hypertension Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.6 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

4.7 Total adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.8 Serious adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 4. Pazopanib plus lapatinib versus lapatinib only
Comparison 5. Pazopanib versus lapatinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Overall survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

5.2 Specific adverse events: gastrointestinal perforations or fistulae Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.3 Specific adverse events: haemorrhage Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.4 Specific adverse events: thromboembolic events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.5 Specific adverse events: hypertension Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.6 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

Totals not selected

5.7 Total adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.8 Serious adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 5. Pazopanib versus lapatinib