Scolaris Content Display Scolaris Content Display

Inmunoterapia (excluyendo los inhibidores del puesto de control) para el cáncer pulmonar no microcítico en estadio I a III tratado con cirugía o radioterapia con intención curativa

Contraer todo Desplegar todo

Antecedentes

El cáncer pulmonar no microcítico (CPNM) es el cáncer de pulmón más frecuente y representa aproximadamente del 80% al 85% de todos los casos. Se ha especulado que en las personas con CPNM localizado (estadios I a III), la inmunoterapia puede ser útil para reducir las tasas de recidiva posoperatoria o para mejorar los resultados clínicos del tratamiento actual de los tumores no resecables. Esta es una actualización de una revisión Cochrane publicada por primera vez en 2017 e incluye dos nuevos ensayos controlados aleatorizados (ECA).

Objetivos

Evaluar la efectividad y la seguridad de la inmunoterapia (excluyendo los inhibidores del puesto de control) en personas con CPNM localizado de estadios I a III que recibieron radioterapia o cirugía con intención curativa.

Métodos de búsqueda

Se realizaron búsquedas en las siguientes bases de datos desde su creación hasta el 19 de mayo de 2021: CENTRAL, MEDLINE, Embase, CINAHL y en cinco registros de ensayos. También se buscó en las actas de congresos y las listas de referencias de los ensayos incluidos.

Criterios de selección

Se incluyeron ECA realizados en adultos (≥ 18 años) con diagnóstico de CPNM en estadio I a III después de resección quirúrgica y adultos con CPNM en estadio III avanzado localmente no resecable que recibían radioterapia con intención curativa. Se incluyeron participantes que hubieran recibido tratamiento quirúrgico primario, radioterapia o quimiorradioterapia posoperatorias si se administraron la misma estrategia en los grupos de intervención y control.

Obtención y análisis de los datos

Dos autores de la revisión, de forma independiente, seleccionaron los ensayos elegibles, evaluaron el riesgo de sesgo y extrajeron los datos. Se utilizó el análisis de supervivencia para agrupar los datos de tiempo transcurrido hasta el evento mediante el cociente de riesgos instantáneos (CRI). Se utilizaron las razones de riesgos (RR) para los datos dicotómicos y las diferencias de medias (DM) para los datos continuos, con intervalos de confianza (IC) del 95%. Debido a la heterogeneidad clínica (agentes inmunoterapéuticos con diferentes mecanismos subyacentes), se combinaron datos mediante modelos de efectos aleatorios.

Resultados principales

Se incluyeron 11 ECA con 5128 participantes (que incluye dos nuevos ensayos con 188 participantes desde la última búsqueda el 20 de enero de 2017). Los participantes que habían recibido resección quirúrgica o radioterapia curativa se aleatorizaron a un grupo de inmunoterapia o uno de control. Las intervenciones inmunológicas fueron inmunoterapia activa Bacillus Calmette‐Guérin (BCG), transferencia adoptiva de células (es decir, factor de transferencia [FT]), linfocitos que infiltran tumores (LIT), linfocitos citolíticos inducidos por citocinas activados por las células dendrítricas (DC/CIK, por sus siglas en inglés), vacunas contra el cáncer basadas en antígenos específicos (antígeno 3 asociado con el melanoma [MAGE‐A3] y L‐BLP25) y linfocitos citolíticos naturales (células NK) selectivos. Siete ensayos tenían riesgo alto de sesgo para al menos un dominio de riesgo de sesgo. Tres ensayos tenían bajo riesgo de sesgo en todos los dominios y un pequeño ensayo tenía riesgo incierto de sesgo ya que no proporcionaba suficiente información. Se incluyeron datos de nueve de los 11 ensayos en los metanálisis (que implicaron a 4863 participantes).

No hubo evidencia de una diferencia entre los agentes inmunoterapéuticos y los controles en ninguno de los siguientes desenlaces: supervivencia global (CRI 0,94; IC del 95%: 0,84 a 1,05; p = 0,27; cuatro ensayos, 3848 participantes; evidencia de calidad alta), supervivencia sin progresión (CRI 0,94; IC del 95%: 0,86 a 1,03; p = 0,19; evidencia de calidad moderada), eventos adversos (RR 1,12; IC del 95%: 0,97 a 1,28; p = 0,11; cuatro ensayos, 4126 participantes evaluados; evidencia de calidad baja) y eventos adversos graves (RR 1,14; IC del 95%: 0,92 a 1,40; seis ensayos, 4546 participantes evaluados; evidencia de calidad baja).

Las tasas de supervivencia en diferentes puntos temporales no mostraron evidencia de una diferencia entre los agentes de inmunoterapia y los controles: tasa de supervivencia al año de seguimiento (RR 1,02; IC del 95%: 0,96 a 1,08; I2 = 57%; siete ensayos, 4420 participantes; evidencia de calidad baja), a los dos años de seguimiento (RR 1,02; IC del 95%: 0,93 a 1,12; siete ensayos, 4420 participantes; evidencia de calidad moderada), a los tres años de seguimiento (RR 0,99; IC del 95%: 0,90 a 1,09; siete ensayos, 4420 participantes; I2 = 22%; evidencia de calidad moderada) y a los 5 años de seguimiento (RR 0,98; IC del 95%: 0,86 a 1,12; I2 = 0%; siete ensayos, 4389 participantes; evidencia de calidad moderada).

Solo un ensayo proporcionó las tasas de respuesta general. Dos ensayos proporcionaron resultados de calidad de vida relacionada con la salud con resultados contradictorios.

Conclusiones de los autores

De acuerdo con esta revisión, la bibliografía actual no proporciona evidencia que indique un efecto beneficioso en la supervivencia al agregar inmunoterapia (excluyendo los inhibidores del puesto de control) a la cirugía o radioterapia curativas convencionales, en personas con CPNM localizado (estadios I a III). Varios ensayos en curso con inhibidores de los puestos de control inmunitarios (PD‐1/PD‐L1) podrían proporcionar nueva información sobre la función de la inmunoterapia en personas con CPNM estadios I a III.

PICO

Population
Intervention
Comparison
Outcome

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

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

Efecto de la inmunoterapia sobre el pronóstico del cáncer de pulmón de células no pequeñas en estadios I a III tratado con cirugía o radioterapia con intención curativa

Pregunta de la revisión

¿Los tratamientos que ayudan al sistema inmunitario del organismo a luchar contra las células cancerígenas (inmunoterapia) logran que los pacientes con cáncer pulmonar de células no pequeñas (CPCNP), que han recibido cirugía o radioterapia con el objetivo de una curación, vivan más tiempo?

Antecedentes

Muchos pacientes con CPCNP que han recibido cirugía o radioterapia para curar el cáncer con el tiempo mueren porque el cáncer reaparece, ya sea en el pecho o en otra parte del cuerpo. Con el transcurso de los años se han realizado varios ensayos que han analizado si la inmunoterapia ayuda a las personas a vivir más tiempo. Algunas parecieron mostrar efectos beneficiosos y otras no.

Características de los estudios

Se realizaron búsquedas en cuatro bases de datos informatizadas y cinco registros de ensayos hasta el 19 de mayo de 2021. Se buscaron todos los ensayos que asignaron al azar a los participantes a un tratamiento u otro (ensayos controlados aleatorizados [ECA]), y que incluyeron a adultos (18 años de edad o más) con CPCNP en estadios iniciales (estadios I a III), confirmado por pruebas de laboratorio de una muestra del tumor. Se encontraron 11 ECA que incluyeron a más de 5000 participantes que habían recibido cirugía o radioterapia curativa y los asignaron al azar a recibir inmunoterapia o ningún tratamiento adicional.

Resultados clave

Se encontró que la administración de inmunoterapia, principalmente en vacunas (con el objetivo de activar el sistema inmunitario del huésped para inducir una respuesta inmunitaria humana a antígenos específicos del tumor), después de la cirugía o la radioterapia no logró que los pacientes vivieran más tiempo. Las personas que recibieron la inmunoterapia en vacunas no parecieron experimentar más efectos secundarios que los demás. No se encontraron resultados que pudieran señalar si añadir la inmunoterapia mejoró la calidad de vida. Actualmente no existe evidencia para apoyar ni refutar la administración de inmunoterapia (principalmente en vacunas) a los pacientes con CPCNP localizado (estadios I a III). Hay ECA en curso que evalúan medicamentos nuevos más prometedores para la inmunoterapia (p.ej. inhibidores del puesto de control).

Calidad de la evidencia

Las evidencias que se encontraron acerca de la supervivencia general y la supervivencia sin progresión fue de calidad alta y moderada, respectivamente. Cuando se analizó, la evidencia de aproximadamente cuántos participantes seguían vivos al año y a los dos, tres o cinco años fue solamente de calidad moderada o baja, porque los ECA no se realizaron demasiado bien y los resultados no coincidieron entre sí. La evidencia para cualquier evento adverso y eventos adversos graves fue de calidad baja.

Authors' conclusions

Implications for practice

So far, based on this updated review, there is no evidence showing better survival outcomes associated with the addition of immunotherapy (excluding checkpoint inhibitors) for people with stages I to III NSCLC. However, the probability of experiencing adverse events may be greater for people receiving immunotherapy, especially the MAGE‐A3 vaccine. A major concern for the evidence on overall survival and progression‐free survival was the small proportion of included trials (36%; 45%) that contributed usable data for group comparisons. For yearly survival rate analyses, for which more data were available, we found no clear differences between the treatment groups for 1‐year, 2‐year, 3‐year, or 5‐year survival rates, with significant heterogeneity across the trials for the 1‐year rate, which could not be fully explained by either variation in study quality or the clinical differences in the trials.

Based on these results, we consider that at present, there is insufficient evidence to support or negate the use of adjuvant immunotherapy (excluding checkpoint inhibitors) for people with stages I to III NSCLC. Several planned or ongoing trials that aim to treat people with stages IB to IIIA NSCLC with adjuvant checkpoint inhibitors (PD‐1/PD‐L1) may provide new evidence for the use of immunotherapy.

Implications for research

We found no ongoing clinical trials on participants with stages I to III NSCLC that could provide additional evidence on the effectiveness of vaccine‐based immunotherapy from this updated review. We identified several planned or ongoing trials with a focus on adjuvant checkpoint inhibitors (PD‐1/PD‐L1). Future efforts should be put into the development of novel, effective immunotherapy, and the detection of more useful prognostic biomarkers to guide the use of these immunotherapeutic drugs.

Summary of findings

Open in table viewer
Summary of findings 1. Immunotherapy for surgically‐treated patients with non‐small cell lung cancer (NSCLC), with or without radiotherapy with curative intent

Immunotherapy for surgically‐treated NSCLC patients, with or without radiotherapy with curative intent

Patient or population: stages I to III NSCLC patients treated with surgery or radiotherapy with curative intent
Setting: hospital
Intervention: immunotherapy plus surgery (adjuvant chemotherapy or chemoradiotherapy was allowed, provided it was applied to both experimental and control groups)
Comparison: surgical treatment with placebo, best supportive care, no intervention

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(trials)

Quality of the evidence
(GRADE)

Comments

Assumed risk with surgical treatment only (control group)

Corresponding risk with immunotherapy plus surgery (experimental group)

Overall survival

(Duration of follow‐up varied between trials: median follow‐up time ranged from 37.7 months to 70 months

The median overall survival time ranged across control groups from 22.3 to 60.2 months

The median overall survival time ranged across experimental groups from 25.6 to 62.0 months

 

 

HR 0.94
(0.84 to 1.05)

3848

(4 RCTs)

⊕⊕⊕⊕
High

 

 

Progression‐free survival

The median progression‐free survival time ranged across control groups from 11.4 to 57.9 months

The median progression‐free survival time ranged across experimental groups from 14.2 to 60.5 months

HR 0.94
(0.86 to 1.03)

3861
(5 RCTs)

 

⊕⊕⊕⊝

Moderatea

 

 

Overall survival: 1‐year survival rate

 

 

Study population

RR 1.02
(0.96 to 1.08)

4420
(7 RCTs)

 

⊕⊕⊝⊝

Lowa,b

 

The presence of heterogeneity could be partly explained by the inclusion of data from trials with high risk of bias.

 

824 per 1000

 

841 per 1000
(792 to 891)

 

Overall survival: 2‐year survival rate

Study population

RR 1.02
(0.93 to 1.12)

4420
(7 RCTs)

⊕⊕⊕⊝

Moderatea

 

602 per 1000

605 per 1000 (551 to 664)

Overall survival: 3‐year survival rate

Study population

RR 0.99
(0.90 to 1.09)

4420
(7 RCTs)

⊕⊕⊕⊝

Moderatea

 

387 per 1000

367 per 1000
(333 to 404)

Overall survival: 5‐year survival rate

Study population

RR 0.98
(0.86 to 1.12)

 

4389
(7 RCTs)

 

⊕⊕⊕⊝
Moderatea

 

 

122 per 1000

81 per 1000
(71 to 92)

Adverse events: any

(Duration of follow‐up varied between trials: median follow‐up time ranged from 37.7 months to 70 months)

 

Study population

RR 1.12
(0.97 to 1.28)

4126
(4 RCTs)

⊕⊕⊝⊝
Lowa,b
 

 

The presence of heterogeneity could be explained by the different agents applied in different trials. By restricting to MAGE‐A3 trials, we observed statistically significant elevation in general adverse event risk (RR 1.23, 95% CI 1.18 to 1.29; I² = 0%).

805 per 1000

913 per 1000
(791 to 1044)

Adverse events: severe, grade > 2

(Duration of follow‐up varied between trials: median follow‐up time ranged from 37.7 months to 70 months)

 

Study population

RR 1.14
(0.92 to 1.40)

4546
(6 RCTs)

⊕⊕⊝⊝
Lowa,b

The presence of heterogeneity could be explained by the involvement of low‐quality trials.

237 per 1000

243 per 1000
(196 to 298)

*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; MAGE‐A3: melanoma‐associated antigen 3: NSCLC: non‐small cell lung cancer; 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 one level due to methodological limitations: inclusion of data from low‐quality trials.
bDowngraded one level due to inconsistency: significant heterogeneity was detected during analysis.

Background

Description of the condition

Lung cancer is the most common cancer in men, and the leading cause of cancer‐related death worldwide (GLOBOCAN 2020). In 2020, there were an estimated 2.2 million new lung cancer cases (accounting for 11.4% of all new cancer cases), making up to 18% of total cancer deaths worldwide (GLOBOCAN 2020). Clinically, there are two main types of lung cancer: non‐small cell lung cancer (NSCLC), the most common type, accounting for approximately 80% to 85% of all lung cancer cases, and small cell lung cancer (SCLC; Roy 2008). This classification is important for deciding treatment and predicting prognosis (Roy 2008). Another key issue for clinical management is lung cancer staging ‐ the assessment of the extent of spread of cancer from its original source (Detterbeck 2009). For NSCLC, the best outcomes are achieved with complete surgical resection of a stage IA tumour, with up to a 70% 5‐year survival rate (Mountain 1997). However, the corresponding rates for later stages drop sharply to less than 20% for stage IIIA; the worst survival is seen in stage IV patients, with 2% living longer than five years from diagnosis. 

Despite the recent advances in the treatment of NSCLC, there has been little improvement in overall survival (National Cancer Institute 2017). The current standard treatment is surgical resection (lobectomy and adequate mediastinal lymph node evaluation), with or without adjuvant chemotherapy, for those with early‐stage disease (Howington 2013). For people with unresectable, locally advanced tumours (some stage IIIA and all stage IIIB patients), curative radiotherapy combined with chemotherapy is usually offered (Ramnath 2013). Although there is a chance of being cured, overall outcomes for people with stages I to III are not very good. Even after curative treatment, many patients develop a local or distant recurrence (Lardinois 2005). We need more effective, and better tolerated therapeutic options, to prevent relapse and improve the rate of cure. Cancer immunotherapy is one possible approach.

Description of the intervention

'Cancer immunotherapy' generally refers to the use of a number of agents that activate or potentiate immune responses and increase anticancer immunity (Mellman 2011). Immunotherapy can be either active or passive. Active immunotherapy is treatment that stimulates the innate immune system to attack cancer cells (Hirschowitz 2006). Antigen‐specific cancer immunotherapeutics (ASCIs) use exogenous antigens, ideally as tumour‐specific as possible, to induce the immune system to produce an effective T‐cell response against cancer cells (Tyagi 2009). A strong adjuvant component to stimulate the immune response and a proper delivery system for promoting antigen presentation are needed for ASCIs to be effective (Mellman 2011). Passive immunotherapy provides immune effector molecules or effector cells made outside of the human body to modulate immunity. These include monoclonal antibodies and adoptive cell transfer of autologous T‐cells genetically engineered to attack tumour cells. 

Currently, immunotherapy has been used successfully in people with malignant melanoma and renal cell carcinoma, because of their high immunogenicity (Drake 2014). Although there are suggestions that lung cancer is a highly immunogenic tumour, initial attempts to administer immunotherapy to people with NSCLC have failed to show clinical benefit (Dasanu 2012). However, significant improvements may come from newer agents, by identifying more relevant, targeted antigens, and developing better adjuvants and delivery systems (Finn 2008Forde 2014). Trial reports have recorded significant objective response rates using novel agents that block immune checkpoint molecules (Topalian 2012). Also, with promising results from phase II trials, several new approaches have been tested in randomised phase III clinical trials, targeting different stages of NSCLC (Vansteenkiste 2013).

How the intervention might work

The idea of cancer immunotherapy originated with the improved understanding of immune surveillance, a process by which the immune system can recognise malignant cells as foreign, and then induce immune responses to eliminate them (Finn 2008). Physiologically, a normal cellular immune response starts with the uptake of tumour antigens by antigen‐presenting cells, such as dendritic cells or macrophages. Antigen‐presenting cells then process the antigens to T‐cells, by presenting them on their surfaces via major histocompatibility complex classes I and II. With assistance from co‐stimulatory signals, different downstream immune effectors (e.g. plasma cells, natural killer (NK) cells, and cytotoxic T‐cells) may be activated, consequently causing the apoptosis (death) of tumour cells (Figure 1). However, immune surveillance may be limited by other factors. If there is an immunosuppressive microenvironment, even malignant tumour cells that carry unusual antigens can escape an immune‐mediated attack (Drake 2006). One such resistance mechanism involves a series of immune checkpoint molecules presenting on the cell surface, including programmed death‐ligand 1 (PD‐L1) and other ligands to inhibitory T‐cell receptors, which can substantially suppress T‐cell proliferation and its killing capacity (Keir 2008). 


Schematic diagram of the immune components and events involved in cancer immunotherapyAPCs: antigen‐presenting cells; MHC: major histocompatibility complex; NK: natural killer

Schematic diagram of the immune components and events involved in cancer immunotherapy

APCs: antigen‐presenting cells; MHC: major histocompatibility complex; NK: natural killer

Therapeutic cancer vaccination and monoclonal antibodies that block immune checkpoints are the most widely used immunotherapies for NSCLC treatment, especially for stages I to III. Our main focus for the review was the effect of therapeutic cancer vaccines, which can be summarised by the type of antigens described below.

Cell‐based vaccines

Autologous cell vaccines, generated from lysate or whole cells from the tumours of individual patients, have the advantage of stimulating an immune response in a large variety of tumour‐specific antigens expressed by the patient's cancer cells. In contrast, allogeneic cell vaccines use mixtures of different cancer cell lines. For instance, the belagenpumatucel‐L vaccine is composed of several lung cancer cell lines (2 adenocarcinomas, 1 squamous cell carcinoma, and 1 large cell carcinoma), and two adjuvants, which form a major histocompatibility complex and antisense molecule, targeting transforming growth factor ß2 (TGF‐ß2; Giaccone 2013). Theoretically, expression of the TGF‐ß2 antisense molecule can undermine the TGF‐ß‐related immunosuppressive effect and potentiate dendritic cell activation, resulting in increased immunogenicity of gene‐modified cancer cells (Nemunaitis 2006). More recently, NK cells have been developed as another type of cell vaccine. Modulated NK cells using soluble α‐galactosylceramide (α‐GalCer) in vivo was found to be tolerated, however there was no promising clinical response due to the low levels of baseline NK cells (Hayes 2021). Furthermore, a clinical trial using ex vivo Hsp70‐derived peptide (TKDNNLLGRFELSG, TKD)/interleukin‐2 (IL‐2)‐activated  autologous NK cells show it was well‐tolerated, with positive clinical responses (Multhoff 2020). 

Compound‐directed vaccines

Peptide vaccines are based on amino acid sequences. However, since they can only target a few epitopes, their major shortcoming is poor immunogenicity (Kochenderfer 2007). This defect can be circumvented by incorporating an efficient delivery system or immunoadjuvants, such as the L‐BLP25 vaccine (known as stimuvax or tecemotide). The L‐BLP25 vaccine consists of a 25‐amino acid sequence from the glycoprotein mucin‐1 (MUC‐1) protein, along with an immunoadjuvant (monophosphoryl lipid A), and a liposomal delivery system (Sangha 2007). MUC‐1 is a highly glycosylated transmembrane protein found on the epithelial cell surface. In cancer cells, MUC‐1 was reported to be frequently overexpressed, with an abnormally glycosylated status (Bafna 2010). Cancer‐associated MUC‐1 can induce abnormal interactions between receptor tyrosine kinases and other cell surface receptors, which in turn lead to inappropriate activation of intracellular signalling pathways. These events then facilitate the growth, proliferation, and survival of cancer cells (Acres 2005Bafna 2010). In preclinical trials, the L‐BLP25 vaccine induced a cellular immune response, characterised by T‐cell proliferation in response to MUC‐1. 

Protein‐based vaccines can elicit an immune response targeting multiple epitopes, but efficient implementation requires that they are combined with immunoadjuvants. Melanoma‐associated antigen 3 (MAGE‐A3) is considered to be a highly exclusive tumour‐specific antigen, since normally it is only expressed in the testes and placenta, where it remains inaccessible to T‐cells because of the lack of major histocompatibility complex molecules to present the antigens (Simpson 2005). Therefore, the MAGE‐A3 vaccine is expected to be a well‐tolerated therapy with minimal side effects. In several types of cancer cells, MAGE‐A3 expression increases with tumour stage (Van den Eynde 1997). MAGE‐A3 is detected in about 35% to 50% of NSCLC tumours (Tyagi 2009).

Viral‐based vaccines

Finally, another way to produce cancer vaccines is to incorporate the target antigen into a viral backbone. One such vaccine, consisting of a modified Ankara virus, known as TG4010, has been developed to target MUC‐1 (Kochenderfer 2007).

Why it is important to do this review

Although immunotherapy for NSCLC showed disappointing results in earlier trials, more promising evidence of its efficacy has emerged in the last decade. For people with localised NSCLC (stages I to III), immunotherapy has been used to reduce the postoperative recurrence rate or negative clinical outcomes of current chemoradiotherapy for unresectable tumours. While several agents have now entered phase III clinical trials, there is a need for a systematic review to address the question of the effectiveness and safety of immunotherapy in such patients. It is also unclear what the most effective type of immunotherapeutic agents is, and which group of patients could benefit most from this treatment. Therefore, our subgroup analysis might offer supportive evidence to optimise its further development and application in the clinic. 

This review was first published in 2017, and updated in May 2021.

Objectives

To assess the effectiveness and safety of immunotherapy (excluding checkpoint inhibitors) among people with localised NSCLC of stages I to III who received curative intent of radiotherapy or surgery.

Methods

Criteria for considering studies for this review

Types of studies

We only included randomised controlled trials (RCTs).

Types of participants

We included all adults (18 years or older) with histologically‐confirmed early‐stage NSCLC (stages I to III) after surgical resection (with or without chemotherapy), and those with unresectable locally advanced stage III NSCLC who had received radiotherapy, or radiotherapy combined with chemotherapy with curative intent.

Types of interventions

  • Surgical treatment + immunotherapy agents versus surgical treatment with placebo, best supportive care, or no intervention.

  • Radical radiotherapy (with or without chemotherapy) + immunotherapy agents versus radical radiotherapy (with or without chemotherapy) with placebo, best supportive care, or no intervention.

Types of outcome measures

Primary outcomes

  • Overall survival: defined as the interval between the date of randomisation and the date of death from any cause.

  • Progression‐free survival: defined as the time from randomisation to either death or disease progression, whichever occurred first. Disease progression was defined according to response evaluation criteria in solid tumours (RECIST; Therasse 2000), as at least a 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum of the longest diameter recorded since the treatment starts, or the appearance of one or more new lesions.

Secondary outcomes

  • Overall survival rates: the percentage of participants in a study who were still alive for a certain period of time.

  • Adverse events or side effects: graded severity with the National Cancer Institute‐Common Terminology Criteria for Adverse Events (NCI‐CTCAE 2017), including the percentage of treatment‐related deaths.

  • Overall response: response assessed according to RECIST guidelines (Therasse 2000), or immune‐related response criteria (Wolchok 2009).

  • Health‐related quality of life: measured by a validated scale.

We included all primary outcomes, as well as parts of secondary outcomes in a summary of findings table (summary of findings Table 1).

Search methods for identification of studies

Electronic searches

We conducted a literature search to identify all published or unpublished RCTs. The literature search identified potential trials in all languages.

We searched the following electronic databases for potential trials.

  • Cochrane Central Register of Controlled Trials (CENTRAL; 2021, Issue 5) in the Cochrane Library, searched 19 May 2021 (Appendix 1).

  • MEDLINE PubMed (1966 to 19 May 2021; Appendix 2).

  • Embase (1988 to 19 May 2021; Appendix 3).

  • CINAHL (1982 to 19 May 2021).

The search string for MEDLINE was developed according to the Cochrane Highly Sensitive Search Strategy, sensitivity‐maximising version, as referenced in Chapter 6.4.11.1 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2021). We adapted the terms and the search strategies for CENTRAL, MEDLINE, and Embase to search CINAHL. 

Searching other resources

We searched Clinical Trial Registers (www.clinicaltrials.gov; www.controlled-trials.com), databases of the Food and Drug Administration (FDA), the European Medicines Agency (EMA), and the World Health Organization (WHO) International Clinical Trials Registry Platform (who.int/clinical-trials-registry), to identify information about ongoing trials. We searched all databases from their inception to 19 May 2021. 

We checked reference lists of all included trials and related reviews for additional references. We asked experts in the field and manufacturers of relevant drugs to provide details of outstanding clinical trials and any relevant unpublished material. We also contacted authors of identified trials and asked them to identify other published and unpublished trials.

We manually checked for potential trials in abstracts or reports from the following relevant conference proceedings (from 1990 to present): American Society of Clinical Oncology (ASCO), European Society of Medical Oncology (ESMO), European Cancer Conference Organisation (ECCO), and International Association for the Study of Lung Cancer (IASLC) World Lung Cancer Conference.

We searched for errata or retractions from eligible trials on www.ncbi.nlm.nih.gov/pubmed on 19 May 2021. 

Data collection and analysis

Selection of studies

Two review authors (YY and XW for this updated review, or OT and ET for the original review) independently screened titles and abstracts of all trials that we identified as a result of the search, and labelled them as 'retrieve' (eligible, potentially eligible, or unclear) or 'do not retrieve'. For the ones coded as 'retrieve', we then referred to their full‐text study reports or publication. Two review authors (HS, RL) independently screened the full‐text reports. The procedure of study identification for inclusion and exclusion was well documented by using standard screening forms. We resolved any disagreement through discussion, or consulted a third review author (CS).

We identified and excluded duplicates 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 (Liberati 2009), and 'Characteristics of excluded studies' table.

Data extraction and management

We used a data collection form for study characteristics and outcome data, which we tested on one study in the review. One review author (RL) extracted study characteristics from included trials. We extracted the following study characteristics.

  • Methods: study design (for example, parallel or cross‐over design), number of study centres and location (country), total duration (for example, date of study, follow‐up period, early stopping of trial), method of randomisation (including imbalanced randomisation ratio), methods of allocation concealment, blinding

  • Participants: N, age, gender, Eastern Co‐operative Oncology Group (ECOG) performance status, medical history, the severity of condition (stage), diagnostic criteria, inclusion criteria, exclusion criteria

  • Interventions: intervention, comparison, concomitant medications, excluded medications

  • Outcomes: primary and secondary outcomes specified and collected, time points reported

  • Notes: funding for trial, or any notable conflicts of interest of trial authors

Two review authors (YY and XW for this updated review, HS and RL for the original review) independently extracted outcome data from the included trials. We noted in the 'Characteristics of included studies' table if outcome data were reported in an unusable way. We resolved disagreements by consensus, or by involving a third review author (CS). One review author (RL) copied the data from the data collection form into the Review Manager file (Review Manager 2020). We double‐checked that the data were entered correctly by comparing the study reports with the data in the systematic review. A second review author (JZ) spot‐checked study characteristics for accuracy against the trial report.

Assessment of risk of bias in included studies

Two review authors (YY and XW for this updated review, HS and RL for the original review) independently assessed the risk of bias for each study, using RoB 1 (Higgins 2011). Any disagreements were resolved by discussion, or by involving a third assessor (JZ). We assessed the risk of bias according to the following domains.

  • Random sequence generation

  • Allocation concealment

  • Blinding of participants and personnel

  • Blinding of outcome assessment

  • Incomplete outcome data

  • Selective outcome reporting

  • Other potential bias

We graded each potential source of bias as high, low, or unclear, and provided a quote from the study report together with a justification for our judgement in the risk of bias table. We summarised the risk of bias judgements across different trials for each of the domains listed. For overall risk of bias, we considered trials that had adequate random sequence generation, adequate allocation concealment, adequate blinding, adequate handling of incomplete outcome data, no selective outcome reporting, and were without other bias risks, as being at overall low risk of bias. We considered trials that were assessed as being at high or unclear risk of bias in the majority of domains as being at overall high risk of bias; and the remaining trials to be at low risk of bias. We considered blinding separately for different key outcomes where necessary e.g. for unblinded outcome assessment, the risk of bias for all‐cause mortality may be very different than for a participant‐reported pain scale. Where information on the risk of bias related to unpublished data or correspondence with a trialist, we noted this in the risk of bias table.

When considering treatment effects, we took into account the risk of bias for the trials that contributed to that outcome.

Measures of treatment effect

We analysed the primary outcomes based on intention‐to‐treat (ITT) analyses, where available. We measured effect estimates by hazard ratios (HRs) for time‐to‐event variables, and risk ratios (RRs) for dichotomous variables. For continuous variables, we calculated mean differences (MDs) with their corresponding 95% confidence intervals (CIs) if trials used the same measurement, and standardised mean differences (SMDs) and 95% CIs when trials use different scales. We contacted the corresponding authors for missing information about standard deviations or standard errors. For reports without available data for pooling, we tried to measure rates from figures, and calculated effect estimates from P values, t statistics, ANOVA tables, or other statistics as appropriate.

Unit of analysis issues

We did not find any trials with non‐standard design (such as cluster‐randomised trials for potential 'unit of analysis error') for this updated review. But if such eligible trials emerge in future literature searches, we will carefully assess these trials (in terms of recruitment bias, baseline imbalance, loss of clusters, and comparability with individually‐randomised trials). Furthermore, we will apply proper statistical methods (such as multilevel models and generalised estimating equations) for analysis, according to the Handbook for Systematic Review of Interventions (Higgins 2021). The individual participant was the unit of analysis for this updated review.

If there had been data from a cross‐over trial with eligible intervention performances, we would have used the data from the first phase only, i.e. from randomisation to the point of cross‐over.

Had multiple trial arms been reported in a single trial, we would only have included the relevant arms. In future, if we enter two comparisons (e.g. drug A versus placebo and drug B versus placebo) from the same trial into the same meta‐analysis, we will halve the control group to avoid double counting.
 

Dealing with missing data

We contacted investigators or study sponsors to verify key study characteristics and obtain missing outcome data where applicable (e.g. when a study was identified as an abstract only). For full‐text reports with missing details relevant to our analysis, we also contacted the authors of the trials by email. In the case of non‐response after repeated attempts, we dropped these incomplete data from the analysis, stating this clearly in the Results section, and discussed it further under the Potential biases in the review process section of the Discussion.

Assessment of heterogeneity

We carried out tests for heterogeneity using the Chi² test, to assesses whether observed differences in results were compatible with chance alone. We used the I² statistic to quantify inconsistency across trials. The presence of heterogeneity was defined by P < 0.05 from the Chi² test, and I² > 50% (Higgins 2021). If we detected moderate or higher heterogeneity (50% to 100%), we applied a thorough exploration of possible sources of heterogeneity by means of subgroup and sensitivity analyses (as stated below). Given the limitations of the methods, the P value from the Chi² test and the value of I² were only referred to as a guide, and we exercised caution when interpreting the results.

Assessment of reporting biases

We attempted to contact study authors, asking them to provide missing outcome data. When this was not possible, and the missing data were thought to introduce serious bias, we explored the impact of including such trials in the overall assessment of results by conducting a sensitivity analysis.

We did not include sufficient trials for each outcome in the current updated review to create a funnel plot. But if we are able to pool more than 10 trials in future updates, we will do so to explore possible publication biases (intervention effect estimate versus standard error of intervention effect estimate). If we find funnel plot asymmetry, we will further investigate clinical diversity of trials as a possible explanation. If there are sufficient trials (> 10), we also will use the 'contour‐enhanced' funnel plot to differentiate asymmetry due to other factors (Peters 2008). If the supposed missing trials are in areas of higher statistical significance, the cause of the asymmetry is highly suggestive of being due to factors other than publication bias.

Data synthesis

We used Review Manager 5 for pooling data and for statistical analysis (Review Manager 2020). We used random‐effects models for primary analyses since the agents of interest had different mechanisms of action. In the future (since currently no subgroup analysis was successfully conducted), provided that the trials in some subgroups are found to be homogeneous (in terms of age, diagnostic subtype, intervention type, intervention duration), we will use both fixed‐effect and random‐effects models, and compare the results. In the absence of heterogeneity and significant reporting bias, these two models should yield the same results. In this case, we will report the results from the fixed‐effect model only. If the results are different, indicating significant heterogeneity, we will report the results from the random‐effects model only.

Subgroup analysis and investigation of heterogeneity

We had planned to perform the following exploratory subgroup analyses on the primary outcomes, using Review Manager 5 (Review Manager 2020).

  • Participants receiving immunotherapy who present with a different stage of NSCLC (stages I, II, or III).

  • Participants receiving different types of immunotherapy.

  • Participants with a specific biomarker: e.g. participants with a gene‐signature profile (MAGE‐A3‐positive).

In the current updated review, we did not include sufficient trials for each population (subgroup) of interest to conduct subgroup analyses. In future updates, we will perform a subgroup analysis for primary outcomes when we have at least three trials for a subgroup.

Considering the differences between subgroups, we will first examine them by visual inspection of their CIs; non‐overlapping CIs indicate a statistically significant difference in treatment effect between subgroups. Also, we will use the approach of Borenstein 2008 to formally investigate differences between two or more subgroups.

Sensitivity analysis

We performed sensitivity analyses, defined a priori, to assess the robustness of our conclusions. This was achieved by repeating the analyses to explore the influence of the following factors on effect size.

  • Exclusion of unpublished trials (in the current updated review, we did not include unpublished data, but will conduct this analysis if unpublished data are included in future updates).

  • Exclusion of lower quality trials (those at high or unclear risk of bias).

Summary of findings and assessment of the quality of the evidence

We created a summary of findings table presenting all our primary and secondary outcomes, except overall response and health‐related quality of life, using GRADEpro software (GRADEpro GDT). We used the five GRADE considerations (study limitations, consistency of effect, imprecision, indirectness, and publication bias) to assess the quality of the body of evidence as it related to the trials that contributed data to the meta‐analyses for the prespecified outcomes. We justified decisions to downgrade or upgrade the quality of the evidence using footnotes, and made comments to aid the reader's understanding of the review where necessary (Higgins 2021).

Summary of findings and assessment of the certainty of the evidence

We created a 'Summary of findings' table presenting all our primary and secondary outcomes except overall response and health‐related quality of life, using the GRADEpro software, version 3.2 (GRADEpro GDT). We used the five GRADE considerations (study limitations, consistency of effect, imprecision, indirectness, and publication bias) to assess the quality of the body of evidence as it related to the studies that contributed data to the meta‐analyses for the pre‐specified outcomes. We justified decisions to downgrade or upgrade the quality of the evidence using footnotes, and made comments to aid the reader's understanding of the review where necessary.

Results

Description of studies

Results of the search

Figure 2 shows details of the updated search results from 20/01/2017 to 19/05/2021. We identified 6218 citations in this updated version of the review (723 from CENTRAL, 3543 from MEDLINE, and 1952 from Embase). In addition, we found 3 ongoing trials in ClinicalTrials.gov (Merck 2015Saka 2017Wu 2011). We did not find any relevant abstract or trial from conference proceedings by handsearching.  

We retained 5439 references after excluding duplicates. By screening the titles and abstracts, we excluded 5429 citations, including the protocol published by Wu 2011 describing a potentially eligible trial which was terminated prematurely (16 September 2016 at ClinicalTrials.gov). We therefore had 10 new records considered to be highly relevant to our review. Among the 10 full‐text accessed, we excluded 8 trials with reasons (see Excluded studies). Two trials met our inclusion criteria (Katakami 2017Multhoff 2020). Combined with the nine full‐text reports from the original published version of the review (Butts 2014Fujisawa 1996Giovanni 1996Macchiarini 1991Matthay 1986Stanley 1986Vansteenkiste 2013Vansteenkiste 2016Zhao 2014), our update includes 11 trials.

Included studies

Overall, we included 11 trials after carefully evaluating potentially eligible articles, corresponding to 12 individual RCTs, with a total of 5128 participants (Butts 2014Giovanni 1996Vansteenkiste 2013Vansteenkiste 2016Stanley 1986Katakami 2017Macchiarini 1991Matthay 1986Multhoff 2020Fujisawa 1996Zhao 2014). Two trials after 20 January 2017, were newly included in this updated review. We summarised the characteristics of the included trials in the 'Characteristics of included studies' tables. 

Unfortunately, we did not manage to extract any relevant results from Matthay 1986, a small trial containing 48 Bacillus Calmette‐Guérin (BCG)‐treated and 40 control subjects. This trial reported survival time by tumour stage (stage I, stage II, and stage III), without providing any detailed data on overall survival, or adverse events, which were described in a very general way, where only fever and transient malaise were mentioned, without grading the severity. Also, because of the poor study quality of Zhao 2014, especially the conflicting data reported in the paper (survival rates in abstract, full text, and figures were different from each other), we decided not to include the results of this study in our analysis.

Study design

Four trials were double‐blind RCTs (Butts 2014Katakami 2017Vansteenkiste 2013Vansteenkiste 2016). With the exception of Stanley 1986, where the details of blinding were unclear, the other six had an open‐label design (Macchiarini 1991Matthay 1986Multhoff 2020Fujisawa 1996Giovanni 1996Zhao 2014). We included four multicentre international trials (Butts 2014Stanley 1986Vansteenkiste 2013Vansteenkiste 2016), enrolling participants from the USA, Europe, and Asia. Other trials recruited participants from China (Zhao 2014), Germany (Multhoff 2020), Italy (Giovanni 1996Macchiarini 1991), Japan (Fujisawa 1996Katakami 2017), or the USA (Matthay 1986).

Participants

All included trials enrolled participants with histologically‐confirmed NSCLC. Two trials enrolled participants with stages I to III NSCLC (Matthay 1986Vansteenkiste 2016). Three RCTs focused on stage I or II completely resected NSCLC (Fujisawa 1996Stanley 1986Vansteenkiste 2013); two trials were conducted on participants with stage II or III NSCLC (Giovanni 1996Macchiarini 1991); and the remaining four only included participants with locally advanced NSCLC (stage III; Butts 2014Katakami 2017Multhoff 2020Zhao 2014). Vansteenkiste 2013 and Vansteenkiste 2016 were separate phase II and phase III clinical trials on MAGE‐A3 immunotherapy, and they only included participants with MAGE‐A3‐positive NSCLC. Likewise, Multhoff 2020 was a phase II trial, which included participants with unresectable, membrane‐bound forms of Hsp70 (mHsp70)‐positive NSCLC. As stated above, 11 eligible trials enrolled a total of 5128 participants; after we excluded the two trials without usable information on outcomes of interest (Matthay 1986Zhao 2014), the total number of participants that contributed to the analyses was 4863. The mean age of analysed participants was 61 years, with a range of 19 to 89 years; 75.1% of them were men (Giovanni 1996 did not report the number of men and women).

Interventions

Three trials included participants with unresectable NSCLC, in two of which, participants were receiving chemoradiotherapy and randomly assigned to receive immunotherapy (L‐BLP25) or placebo (Butts 2014Katakami 2017). In another trial, participants were randomly assigned to natural killer (NK) cell‐based immunotherapy along with chemoradiotherapy, or chemoradiotherapy alone (Multhoff 2020). All other included RCTs included participants with surgically‐treated NSCLC. Four RCTs compared the immunotherapy group to a control group of either placebo, best supportive care, or no intervention (Fujisawa 1996Matthay 1986Stanley 1986Vansteenkiste 2013); the other four trials allowed adjuvant chemotherapy (Vansteenkiste 2016Zhao 2014), or chemoradiotherapy (Giovanni 1996Macchiarini 1991), for both experimental and control groups. It is noteworthy that the immunotherapy agents used in these trials differed over the years. Earlier trials mainly studied active immunotherapy, such as BCG injected into the intrapleural space (Macchiarini 1991Stanley 1986), or into the tumour (Matthay 1986). The focus of research then gradually moved to passive immunotherapy, with adoptive cell transfer (transfer factor (TF); Fujisawa 1996), tumour‐infiltrating lymphocytes (TIL; Giovanni 1996), dendritic cell/cytokine‐induced killer (DC/CIK; Zhao 2014), and natural killer (NK) cells (Multhoff 2020). Most recently, antigen‐specific cancer vaccines (MAGE‐A3; Vansteenkiste 2013Vansteenkiste 2016) and L‐BLP25 (Butts 2014Katakami 2017), were widely introduced and evaluated. 

Outcome measures

Apart from Multhoff 2020, the remaining trials reported overall survival time, although measured in different ways. Matthay 1986 reported survival time by tumour stage (stage I, stage II, and stage III); but they only provided survival probability curves for stage I and stage III. Therefore, we could not extract any survival outcome data from this study. Only the four recent trials measured the difference in survival between experimental and control groups by time‐to‐event analysis and hazard ratio (HR; (Butts 2014Katakami 2017Vansteenkiste 2013Vansteenkiste 2016). Fujisawa 1996 reported 5‐year and 10‐year survival rates and all other RCTs reported survival by a median or mean survival with ranges or standard deviation (SD). To enable the use of these data for meta‐analysis, we extracted data for 1‐year, 2‐year, 3‐year, and 5‐year survival rates from all included trials (where possible), from either text statements or survival curves, as secondary outcomes. Multhoff 2020 reported progression‐free survival as the primary outcome. We extracted progression‐free survival data, as HRs, from five RCTs (Butts 2014Katakami 2017Multhoff 2020Vansteenkiste 2013Vansteenkiste 2016). 

The overall response was reported in only one RCT (Multhoff 2020). Adverse events were mentioned in eight trials, but we only included data from six of them in the analysis of adverse events (Butts 2014Katakami 2017Multhoff 2020Stanley 1986Vansteenkiste 2013Vansteenkiste 2016), since Matthay 1986 and Giovanni 1996 reported adverse events in general, without grading the severity. Quality of life was reported in two RCTs (Multhoff 2020Vansteenkiste 2016), assessed by the European Organisation for Reasearch and Treatment of Cancer (EORTC) Quality of Life Questionnaire‐Core 30 (QLQ‐C30)  (Aaronson 1993) and EuroQoL‐5D (EQ‐5D)(EuroQol 1990) questionnaire, respectively.

Because of the inconsistencies in the duration of follow‐up in the different trials, these point estimates should be interpreted with caution. 

Excluded studies

Please see Characteristics of excluded studies.

Risk of bias in included studies

In Figure 3 and Figure 4, we summarised the risk of bias in the included trials. Overall, we considered three trials to be well‐designed and well‐conducted, and therefore we assessed these trials at low risk of bias (Butts 2014Vansteenkiste 2013Vansteenkiste 2016). However, for all of them, the involvement of the sponsors during study design, analysis, and results interpretation was mentioned in their reports. One trial had unclear risks of bias for two domains of assessment, owing to limited study information for risk assessment reported in the published paper, and we considered this trial to have a unclear risk of bias (Katakami 2017). The other seven trials were at high risk of bias, mainly because of non‐blinding design (Fujisawa 1996Giovanni 1996Macchiarini 1991Matthay 1986Multhoff 2020Stanley 1986Zhao 2014). 


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

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


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

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

Allocation

Four trials described how they carried out the randomisation procedure in detail (Butts 2014Matthay 1986Vansteenkiste 2013Vansteenkiste 2016). The other trials did not provide details on how they randomised the participants into different treatment arms, so we considered them to be at unclear risk of selection bias (Fujisawa 1996Giovanni 1996Macchiarini 1991Multhoff 2020Stanley 1986Zhao 2014Katakami 2017). For Butts 2014Katakami 2017Vansteenkiste 2013, and Vansteenkiste 2016, randomisation was done centrally, via the Internet, minimising the risk of lack of allocation concealment. Matthay 1986 randomised with a printed table of random numbers, which was concealed from investigators with a sealed envelope.

Blinding

We judged the three trials with a double‐blind, placebo‐controlled design to be at low risk of bias for blinding of participants and researchers, as well as blinding of outcome assessors (Butts 2014Vansteenkiste 2013Vansteenkiste 2016). Stanley 1986 had a placebo comparator, but did not provide a detailed explanation of blinding of participants, so we evaluated it to be at unclear risk. We considered six trials to be at high risk of performance bias because they had no placebo comparator (open‐label design; (Fujisawa 1996Giovanni 1996Macchiarini 1991Matthay 1986Multhoff 2020Zhao 2014)). 

To assess the influence of detection bias, we considered the concept of each outcome; we classified overall survival, yearly survival rates, and severe adverse events as objective effects of treatment, and so they are unlikely to be affected by the non‐blinding of outcome assessment. But progression‐free survival, response, quality of life, and less severe adverse events are subjective outcomes, and could be affected by the outcome assessors’ knowledge of treatment and the participant's awareness of the assignment status. We assessed all trials to be at low risk of detection bias for objective outcomes. For subjective outcomes, the risk of detection bias was low, when data were extracted with masking for assessors (progression‐free survival and any adverse event in Butts 2014Vansteenkiste 2013, and Vansteenkiste 2016, and quality life in Vansteenkiste 2016). And the risk of detection bias for subjective outcomes was high in the open‐label trial (progression‐free survival, overall response, and quality of life in Multhoff 2020), and unclear in the trial with blinding of the outcome assessor not specified (progression‐free survival, quality of life, and any adverse events in Katakami 2017). 

Incomplete outcome data

We considered all the included trials to be at low risk of bias, either because the number of participants missing from follow‐up was very low (dropout rates below 5%), or because the primary survival analysis was done on the intention‐to‐treat population of all participants randomly allocated to treatment.

Selective reporting

The protocols of five trials were available, where all their prespecified (primary and secondary) outcomes were described in detail (Butts 2014Katakami 2017Multhoff 2020Vansteenkiste 2013Vansteenkiste 2016). Since the pre‐published protocols were consistent with their reports, we judged these five trials to be at low risk of selective outcome reporting. We considered all other trials to be at high risk for selective reporting bias, because their study protocol was unavailable and they just reported part of the primary outcomes (Fujisawa 1996Giovanni 1996Macchiarini 1991Matthay 1986Stanley 1986Zhao 2014). Only Butts 2014Katakami 2017Vansteenkiste 2013, and Vansteenkiste 2016 reported overall survival, together with progression‐free survival. Others reported overall survival, but in different formats (Fujisawa 1996Giovanni 1996Macchiarini 1991Matthay 1986Stanley 1986Zhao 2014). Multhoff 2020 reported progression‐free survival as the primary outcome and did not report overall survival. 

Other potential sources of bias

Zhao 2014 reported conflicting data in their paper (survival rates in abstract, full text, and figures were different from each other), and so we considered this study to be at high risk of other potential bias. For the remaining trials, since there was no obvious potential source of bias, we classified them to be at low risk of other potential biases. 

However, four trials were funded by pharmaceutical companies (Butts 2014; Katakami 2017; Vansteenkiste 2013; Vansteenkiste 2016), and the sponsors played critical roles in study design, data collection, management and statistical analysis. Therefore, we carefully compared the final reports with their previously published protocols. The consistency between these two documents mitigated this concern, by indicating that in spite of the sponsors’ involvement, these trials were conducted as planned, and with full supervision by independent administrators. 

The other seven RCTs had no connection with these companies and confirmed their independence in the study implementation. 

Effects of interventions

See: Summary of findings 1 Immunotherapy for surgically‐treated patients with non‐small cell lung cancer (NSCLC), with or without radiotherapy with curative intent

Primary outcomes

Effect of immunotherapy on overall survival for participants with stages I to III NSCLC

To enable the maximum use of eligible data for the meta‐analysis of survival outcomes, we used two approaches to pool data, the first examined overall survival, and the second examined overall survival rates during different time slots. We have provided details of the second analyses under Secondary outcomes

We extracted hazard ratios (HRs) from four trials (Butts 2014Katakami 2017Vansteenkiste 2013Vansteenkiste 2016). In total, 4179 participants were involved in these trials, and 3848 of them were evaluated for overall survival (92% of all randomised participants). Using a random‐effects model, the pooled results illustrated that the study groups did not have a reduced risk of death compared to the control groups (HR 0.94, 95% confidence interval (CI) 0.84 to 1.05; high‐quality evidence; Analysis 1.1Figure 5); we did not detect heterogeneity across the trials (I² = 0%; P = 0.56). Since only three out of the four included trials were considered to be at low risk of bias, we performed sensitivity analysis after excluding Katakami 2017. Results were consistent with the main analysis (HR 0.94, 95% CI 0.84 to 1.05) and we did not observe heterogeneity (I² = 2%; P = 0.36). 


Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Overall survival.

Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Overall survival.

Effect of immunotherapy on progression‐free survival for participants with stages I to III NSCLC

Five trials reported progression‐free survival (Butts 2014Katakami 2017Multhoff 2020Vansteenkiste 2013Vansteenkiste 2016). Although different agents were used in these trials, we found no heterogeneity between the results of these five trials (I² = 0%; P = 0.49); using a random‐effects model, we found that immunotherapy plus surgery showed no advantage compared to surgery alone (HR 0.94, 95% CI 0.86 to 1.03; P = 0.19; moderate‐quality evidence; summary of findings Table 1Analysis 1.2Figure 6). We performed a further sensitivity analysis after we excluded two moderate‐/low‐quality trials (Katakami 2017Multhoff 2020). Similar to the main analysis, we did not observe heterogeneity (I² = 39%; P = 0.19), and we did not find a difference for progression‐free survival between the two arms (HR 0.93, 95% CI 0.81 to 1.07; moderate‐quality evidence).


Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Progression‐free survival.

Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Progression‐free survival.

Secondary outcomes

Effect of immunotherapy on overall survival rates for participants with stages I to III NSCLC

We added this outcome after assessing the data to capture as much information as we could on participant survival. We extracted 1‐year, 2‐year, and 3‐year survival rates from seven trials with 4420 participants (Butts 2014Giovanni 1996Katakami 2017Macchiarini 1991Stanley 1986Vansteenkiste 2013Vansteenkiste 2016). The results from a random‐effects model showed no clear difference in 1‐year survival probabilities for participants treated with immunotherapy compared to participants assigned to control groups (risk ratio (RR) 1.02, 95% CI 0.96 to 1.08; Analysis 1.3). The RRs were similar for 2‐year (1.02, 95% CI 0.93 to 1.12; Analysis 1.4) and 3‐year survival probability (0.99, 95% CI 0.90 to 1.09; Analysis 1.5). There was moderate heterogeneity for 1‐year (I² = 57%; P = 0.03) and 2‐year (I² = 51%; P = 0.06) survival rates; while 3‐year survival rates showed low between‐trial heterogeneity (I² = 22%; P = 0.26). Five‐year survival rates were available from seven trials that included 2834 participants from the experimental and 1555 participants from the control groups(Butts 2014; Fujisawa 1996; Katakami 2017Macchiarini 1991; Stanley 1986; Vansteenkiste 2013; Vansteenkiste 2016). Analysis showed there was no clear benefit for 5‐year survival by adding immunotherapy (RR 0.98, 95% CI 0.86 to 1.12; Analysis 1.6), and there was no heterogeneity (I² = 0%; P = 0.75). We concluded that the quality of evidence for the 1‐year survival rate was low, primarily due to the inconsistency of effect across the trials and the involvement of trials with unclear or high risks of bias; the quality of evidence for 2‐year, 3‐year and 5‐year survival rates was moderate, because of the involvement of trials with high or unclear risk of selection bias. 

Because of significant heterogeneity for the 1‐year survival rate, we repeated the analyses, restricting it to trials at low risk of bias. This sensitivity analysis suggested that the heterogeneity for 1‐year survival rates could be partly explained by the differences in study quality. And the inconsistency of effect size also was mitigated after the exclusion of low‐quality trials. 

Effect of immunotherapy on adverse events for participants with stages I to III NSCLC

Four trials, with a total of 4126 evaluated participants, provided data on the proportion of participants with any adverse event, graded for severity (Butts 2014Katakami 2017Vansteenkiste 2013Vansteenkiste 2016). Using a random‐effects model, our analysis showed that the addition of immunotherapy to surgery or curative radiotherapy lead to a similar risk of experiencing any adverse events among these participants (RR 1.12, 95% CI 0.97 to 1.28; P = 0.11; Analysis 1.7). The effect size varied substantially between trials (suggesting a drug‐specific effect on this outcome), which corresponded to a high level of heterogeneity between individual outcomes (I² = 95%; P < 0.00001). We also observed an increase in the general adverse event risk (RR 1.23, 95% CI 1.18 to 1.29; I² = 0%; P < 0.00001) by restricting the analysis to MAGE‐A3 trials (Vansteenkiste 2013Vansteenkiste 2016). 

Six trials (Butts 2014Katakami 2017Multhoff 2020Stanley 1986Vansteenkiste 2013Vansteenkiste 2016), with 2979 evaluated participants in the experimental and 1567 evaluated participants in the control groups, showed that participants receiving immunotherapy did not have different risk of severe adverse events (severity grade > 2) than their controls (RR 1.14, 95% CI 0.92 to 1.40; Analysis 1.8). We also found moderate heterogeneity among the results of these trials (I² = 56%; P = 0.04). Moreover, heterogeneity disappeared in the following sensitivity analysis where low‐quality trials were removed (RR 0.96, 95% CI 0.86 to 1.08; I² = 0%; P = 0.72), indicating that the inconsistency might have originated from the inclusion of two high risk of bias trials (Multhoff 2020Stanley 1986), and one unclear risk of bias trial (Katakami 2017). In summary, we considered there was low‐quality evidence for both any adverse event, and severe adverse events (due to the involvement of low‐quality trials and significant heterogeneity; summary of findings Table 1). 

Effect of immunotherapy on overall response rate for participants with stages I to III NSCLC

The overall response was assessed only in Multhoff 2020 according to RECIST guidelines (Therasse 2000). One participant in the experimental group showed complete response, and one participant showed partial response. One participant in the control arm had a partial response. The overall response rate was 33% (2/6) in the experimental group and 14% (1/7) in the control group. 

Effect of immunotherapy on health‐related quality of life for participants with stages I to III NSCLC

Vansteenkiste 2016 assessed the health‐related quality of life of their participants (1515 experimental and 757 control), using both EQ‐5D (EuroQol 1990) utility scores and visual analogue scores (VAS) (Aitken 1969). They did not find any evidence that the application of melanoma‐associated antigen 3 (MAGE‐A3) improved quality of life. Instead, the immunotherapy group reported significantly lower scores (indicating poorer life quality) on quality of life assessments on the day after the first and the third MAGE‐A3 administrations, compared to the control group. 

Multhoff 2020 assessed the quality of life of participants using the QLQ‐C30 (Aaronson 1993). Differences in means of sum scores of the QLQ‐C30 varied at multiple visits, and the experimental group, in general, had a better quality of life during the follow‐up period. However, there were no differences between both studied groups across all visits. 

Subgroup analysis

As described in our protocol, we had planned to perform subgroup analysis for primary outcomes according to participants at different tumour stages (I, II, or III), type of immunotherapy, and specific prognostic biomarkers that were used for participant selection. However, since we only found four trials with data for primary outcome analyses, we were not able to conduct any subgroup analyses at this stage. 

Discussion

Summary of main results

This updated review, which pooled survival data from trials that focused mainly on vaccine‐based immunotherapy, showed that there was no clear evidence of additional survival benefit from immunotherapy (excluding checkpoint inhibitors) for people with stages I to III NSCLC, who had undergone surgery or received radiotherapy with curative intent. For overall survival, pooled data from four (3848 participants analysed) of the 11 included trials suggested no difference in death (hazard ratio (HR) 0.94, 95% confidence interval (CI) 0.84 to 1.05) for those receiving immunotherapy, compared to their controls. Moreover, the pooled data from five (3861 participants analysed) of the 11 included trials also indicated a similar result in risk of progression (progression‐free survival HR 0.94, 95% CI 0.86 to 1.03) for those receiving immunotherapy. Similarly, survival rate analyses of data from seven trials (4420 participants) showed no improvement of 1‐year, 2‐year, 3‐year, or 5‐year survival associated with the addition of immunotherapy agents. Sensitivity analysis showed that the exclusion of low‐quality data could not explain the observed variation in study results. Due to the small number of trials, we could not perform subgroup analyses to detect the possible differences caused by tumour stage, type of immunotherapy, or the use of a specific prognostic biomarker.

Furthermore, we observed similar risk of experiencing any adverse event (12% for MAGE‐A3 and L‐BLP25 together (P = 0.11)) for participants who were assigned to immunotherapy, compared to the control groups. The risk of having severe adverse events also showed no clear increase (RR 1.14, 95% CI 0.92 to 1.40). 

Two of the 11 included trials reported controversial findings about quality of life after immunotherapy. The immunotherapy group in the MAGE‐A3 study reported significantly lower scores (indicating poorer life quality) on quality of life assessments on the day after the first and the third MAGE‐A3 administrations, however the experimental group of the NK cell study showed a better quality of life during the follow‐up period, compared to their controls. The immunotherapy group in one study reported significantly higher overall response rate, indicating better therapy response, compared with the control group.

Overall completeness and applicability of evidence

In order to reduce publication bias, our literature search sought unpublished or ongoing trials, without any limitation on publication language. However, the identified trials only partially addressed the objectives of the review, mainly because data about the effect of immunotherapy for participants with stages I to III NSCLC were insufficiently described. Survival outcomes were reported in various ways. Only five recent trials (45% of included trials) provided adequate data for overall survival and progression‐free survival (Butts 2014Katakami 2017Multhoff 2020Vansteenkiste 2013Vansteenkiste 2016). Due to the absence of a uniform approach for group comparison, data extraction and analyses from earlier trials was difficult. In addition, because of clinical (different agents used in each trial, and participants with different tumour stages) and statistical heterogeneity, the legitimacy of pooling results may be debatable. The limited number of included trials prevented us from detailed comparisons within more homogeneous subgroups. Also, some important outcomes, such as response rates and health‐related quality of life, could not be examined in this updated review, since response rates were only measured in one trial and quality of life from two trials was measured on different scales. 

At the time of our literature search, new immunotherapy agents, i.e. checkpoint inhibitors (PD‐1/PD‐L1), are mainly applied to people with either advanced stage or metastatic NSCLC. Several trials that aimed to assess the efficacy and safety of checkpoint inhibitor drugs are still ongoing (NCT02273375NCT02486718NCT02504372). Therefore, further reviews that focus on the effectiveness of checkpoint inhibitors for people with stage I to III NSCLC will be needed when results from several trials are available. 

Quality of the evidence

We identified 11 eligible RCTs. We could not extract useful data (for our outcomes of interest) from Matthay 1986, and we discarded results from Zhao 2014 because of poor study quality and contradictory reports of the primary outcomes, leaving only nine trials for further analysis. Data from trials performed before 2000 generally were of poor quality, because of the open‐label design and lack of information provided about randomisation methods (particularly allocation concealment). We considered the four newer double‐blinded RCTs to be of high or moderate quality (Butts 2014Katakami 2017Vansteenkiste 2013Vansteenkiste 2016).  

It should be noted that the available data for the meta‐analyses of primary outcomes was incomplete. All participants included in the final analysis were from either four or five trials, and evaluated three types of immunotherapies, although we did not downgrade the quality of the evidence because of this. Clinical and statistical heterogeneity leads to a problem of imprecision for most of our outcomes of interest. We found no suggestion of publication bias, and therefore, no serious limitation was assumed. In addition, we could not explain the inconsistency between individual trial results by our predefined subgroups.

Potential biases in the review process

For the primary analyses, we combined all intervention groups' data, regardless of the type of immunotherapeutic agents administered, and the specific biomarker used for participant selection. Consequently, our analyses were subject to high potential risk of between‐study heterogeneity, due to clinical diversity. Further, we observed significant statistical heterogeneity in the analysis of the 1‐year survival rate. But, because only a limited number of eligible trials was included in these analyses, we were not able to fully explore the reasons for the variation by subgroup or sensitivity analysis. The unexplained inconsistency between individual trial results may undermine the reliability of our effectiveness assessment.

Another issue relevant to the potential bias of our pooled results was the small proportion of trials included in the meta‐analyses of overall survival (36%) and progression‐free survival (45%), our primary outcomes (i.e. a large proportion of included trials were at high risk of selective reporting bias). Although these five trials contained more than 80% of all participants (4195/5128) from all eligible trials, such analyses were deemed to be incomplete, with over‐representation of the effects of most recent agents (Butts 2014Katakami 2017Multhoff 2020Vansteenkiste 2013Vansteenkiste 2016). This naturally limited our analysis to antigen‐specific vaccines (MAGE‐A3 and L‐BLP25) and cell‐based vaccines (NK cells). On the other hand, since other trials (with smaller numbers of participants) produced mixed results, we were unable to assess the efficacy of other types of immunotherapeutic agents in the current updated review. 

Two review authors independently carried out study selection, assessment of risks of bias, and data collection, without blinding. Regarding missing data and missing information related to study quality assessment, we tried to contact authors by email to obtain these details. However, even after repeated attempts, we did not receive any response from these authors (since these trials were conducted a long time ago, the non‐response rates were expected to be high). In the end, we were unable to access sufficient information to assess the eligibility of two trials (Macchiarini 1989; Schlieben 1984). For the other trials, we deemed that there were unclear risks of certain study bias (see Figure 4), and we extracted and used available data for the meta‐analysis for each outcome. Again, this could influence the accuracy and reliability of our results because of incomplete study assessment and data pooling. 

Agreements and disagreements with other studies or reviews

The survival benefit of immunotherapy for people with localised NSCLC has been discussed for decades. For early (stage I and II) or locally advanced (IIIA) NSCLC, the first attempts to use immunotherapy (mainly cellular immunity‐based therapeutic strategies) in such patients began in the 1980s, using active immunotherapy agents (e.g. Bacillus Calmette‐Guérin (BCG)) or adoptive cell transfer (e.g. transfer factor (TF)). Striking improvements in survival were observed in some clinical trials, including both RCTs (Fujisawa 1996Macchiarini 1991), and non‐randomised clinical trials; while null results were reported in Matthay 1986. These attempts were finally thought to fail since the largest RCT (with 441 participants) found no significant difference between experimental and control groups (Stanley 1986). New efforts were launched after 2000, mainly applying vaccine‐based immunotherapy agents. Promising outcomes from small trials inspired further explorations (Vansteenkiste 2013). However, disappointing results were reported from later phase III RCTs (Butts 2014Vansteenkiste 2016). Similar conclusions were drawn from other published reviews (Carrizosa 2015Reckamp 2015). Nevertheless, new trials in this area have been started, with a focus on novel agents, especially immune checkpoint inhibitors, for which superior progression‐free survival has been reported in the planned interim analysis of one phase III RCT trial (Antonia 2017).

Schematic diagram of the immune components and events involved in cancer immunotherapyAPCs: antigen‐presenting cells; MHC: major histocompatibility complex; NK: natural killer

Figuras y tablas -
Figure 1

Schematic diagram of the immune components and events involved in cancer immunotherapy

APCs: antigen‐presenting cells; MHC: major histocompatibility complex; NK: natural killer

original image

Figuras y tablas -
Figure 2

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

Figuras y tablas -
Figure 3

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

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

Figuras y tablas -
Figure 4

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

Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Overall survival.

Figuras y tablas -
Figure 5

Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Overall survival.

Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Progression‐free survival.

Figuras y tablas -
Figure 6

Forest plot of comparison: Effect of immunotherapy for surgically‐treated NSCLC patients: main analyses, outcome: Progression‐free survival.

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 1: Overall survival

Figuras y tablas -
Analysis 1.1

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 1: Overall survival

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 2: Progression‐free survival

Figuras y tablas -
Analysis 1.2

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 2: Progression‐free survival

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 3: Overall survival rate, 1‐year

Figuras y tablas -
Analysis 1.3

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 3: Overall survival rate, 1‐year

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 4: Overall survival rate, 2‐year

Figuras y tablas -
Analysis 1.4

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 4: Overall survival rate, 2‐year

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 5: Overall survival rate, 3‐year

Figuras y tablas -
Analysis 1.5

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 5: Overall survival rate, 3‐year

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 6: Overall survival rate, 5‐year

Figuras y tablas -
Analysis 1.6

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 6: Overall survival rate, 5‐year

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 7: Adverse events (any)

Figuras y tablas -
Analysis 1.7

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 7: Adverse events (any)

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 8: Adverse events (severe, grade > 2)

Figuras y tablas -
Analysis 1.8

Comparison 1: Effect of immunotherapy for surgically treated NSCLC patients: main analyses, Outcome 8: Adverse events (severe, grade > 2)

Summary of findings 1. Immunotherapy for surgically‐treated patients with non‐small cell lung cancer (NSCLC), with or without radiotherapy with curative intent

Immunotherapy for surgically‐treated NSCLC patients, with or without radiotherapy with curative intent

Patient or population: stages I to III NSCLC patients treated with surgery or radiotherapy with curative intent
Setting: hospital
Intervention: immunotherapy plus surgery (adjuvant chemotherapy or chemoradiotherapy was allowed, provided it was applied to both experimental and control groups)
Comparison: surgical treatment with placebo, best supportive care, no intervention

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(trials)

Quality of the evidence
(GRADE)

Comments

Assumed risk with surgical treatment only (control group)

Corresponding risk with immunotherapy plus surgery (experimental group)

Overall survival

(Duration of follow‐up varied between trials: median follow‐up time ranged from 37.7 months to 70 months

The median overall survival time ranged across control groups from 22.3 to 60.2 months

The median overall survival time ranged across experimental groups from 25.6 to 62.0 months

 

 

HR 0.94
(0.84 to 1.05)

3848

(4 RCTs)

⊕⊕⊕⊕
High

 

 

Progression‐free survival

The median progression‐free survival time ranged across control groups from 11.4 to 57.9 months

The median progression‐free survival time ranged across experimental groups from 14.2 to 60.5 months

HR 0.94
(0.86 to 1.03)

3861
(5 RCTs)

 

⊕⊕⊕⊝

Moderatea

 

 

Overall survival: 1‐year survival rate

 

 

Study population

RR 1.02
(0.96 to 1.08)

4420
(7 RCTs)

 

⊕⊕⊝⊝

Lowa,b

 

The presence of heterogeneity could be partly explained by the inclusion of data from trials with high risk of bias.

 

824 per 1000

 

841 per 1000
(792 to 891)

 

Overall survival: 2‐year survival rate

Study population

RR 1.02
(0.93 to 1.12)

4420
(7 RCTs)

⊕⊕⊕⊝

Moderatea

 

602 per 1000

605 per 1000 (551 to 664)

Overall survival: 3‐year survival rate

Study population

RR 0.99
(0.90 to 1.09)

4420
(7 RCTs)

⊕⊕⊕⊝

Moderatea

 

387 per 1000

367 per 1000
(333 to 404)

Overall survival: 5‐year survival rate

Study population

RR 0.98
(0.86 to 1.12)

 

4389
(7 RCTs)

 

⊕⊕⊕⊝
Moderatea

 

 

122 per 1000

81 per 1000
(71 to 92)

Adverse events: any

(Duration of follow‐up varied between trials: median follow‐up time ranged from 37.7 months to 70 months)

 

Study population

RR 1.12
(0.97 to 1.28)

4126
(4 RCTs)

⊕⊕⊝⊝
Lowa,b
 

 

The presence of heterogeneity could be explained by the different agents applied in different trials. By restricting to MAGE‐A3 trials, we observed statistically significant elevation in general adverse event risk (RR 1.23, 95% CI 1.18 to 1.29; I² = 0%).

805 per 1000

913 per 1000
(791 to 1044)

Adverse events: severe, grade > 2

(Duration of follow‐up varied between trials: median follow‐up time ranged from 37.7 months to 70 months)

 

Study population

RR 1.14
(0.92 to 1.40)

4546
(6 RCTs)

⊕⊕⊝⊝
Lowa,b

The presence of heterogeneity could be explained by the involvement of low‐quality trials.

237 per 1000

243 per 1000
(196 to 298)

*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; MAGE‐A3: melanoma‐associated antigen 3: NSCLC: non‐small cell lung cancer; 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 one level due to methodological limitations: inclusion of data from low‐quality trials.
bDowngraded one level due to inconsistency: significant heterogeneity was detected during analysis.

Figuras y tablas -
Summary of findings 1. Immunotherapy for surgically‐treated patients with non‐small cell lung cancer (NSCLC), with or without radiotherapy with curative intent
Comparison 1. Effect of immunotherapy for surgically treated NSCLC patients: main analyses

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Overall survival Show forest plot

4

Hazard Ratio (IV, Random, 95% CI)

0.94 [0.84, 1.05]

1.2 Progression‐free survival Show forest plot

5

Hazard Ratio (IV, Random, 95% CI)

0.94 [0.86, 1.03]

1.3 Overall survival rate, 1‐year Show forest plot

7

4420

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.96, 1.08]

1.4 Overall survival rate, 2‐year Show forest plot

7

4420

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.93, 1.12]

1.5 Overall survival rate, 3‐year Show forest plot

7

4420

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.90, 1.09]

1.6 Overall survival rate, 5‐year Show forest plot

7

4389

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.86, 1.12]

1.7 Adverse events (any) Show forest plot

4

4126

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.97, 1.28]

1.8 Adverse events (severe, grade > 2) Show forest plot

6

4546

Risk Ratio (M‐H, Random, 95% CI)

1.14 [0.92, 1.40]

Figuras y tablas -
Comparison 1. Effect of immunotherapy for surgically treated NSCLC patients: main analyses