Skip to main content

Discontinuation risk from adverse events: immunotherapy alone vs. combined with chemotherapy: a systematic review and network meta-analysis

Abstract

Background

While immunotherapy combined with chemotherapy (Chemo-IO) is generally recognized for providing superior outcomes compared to monotherapy (mono-IO), it is associated with a higher incidence of treatment-related adverse events (TRAEs), which may lead to treatment discontinuation. In this study, we compared the rates of treatment discontinuation between mono-IO and Chemo-IO as first-line treatments for various solid tumors.

Methods

We systematically reviewed clinical trials from databases (PubMed, Embase, Cochrane Library, and an additional source) published from January 1, 2018, to July 10, 2023. We included phase III randomized controlled trials (RCTs) that utilized immunotherapy agents in at least one arm as first-line treatments for a variety of solid tumors. Data extraction followed the Preferred Reporting Items for Systematic Reviews (PRISMA) extension statement for network meta-analysis. A random effects model was used for the network meta-analysis, with the risk of bias assessed using the Cochrane risk-of-bias tool II. The primary outcomes encompassed treatment discontinuation rates due to TRAEs among patients who underwent immunotherapy, either alone or combined with chemotherapy, for various solid tumors. Pooled relative risks (RRs) with 95% confidence intervals (CIs) were calculated to compare between treatment groups.

Results

From 29 RCTs, a total of 21,677 patients and 5 types of treatment were analyzed. Compared to mono-IO, Chemo-IO showed a significantly higher rate of discontinuation due to TRAEs (RR 2.68, 95% CI 1.98–3.63). Subgroup analysis for non-small cell lung cancer (NSCLC) patients also exhibited a greater risk of discontinuation due to TRAEs with Chemo-IO compared to mono-IO (RR 2.93, 95% CI 1.67–5.14). Additional analyses evaluating discontinuation rates due to either treatment emergent adverse events (TEAEs) or AEs regardless of causality (any AEs) consistently revealed an elevated risk associated with Chemo-IO.

Conclusions

Chemo-IO was associated with an elevated risk of treatment discontinuation not only due to TRAEs but also any AEs or TEAEs. Given that the treatment duration can impact clinical outcomes, a subset of patients might benefit more from mono-IO than combination therapy. Further research is imperative to identify and characterize this subset.

Peer Review reports

Background

Since the regulatory approval of ipilimumab, an anti-CTLA-4 agent, immunotherapies (IO) targeting other immune checkpoints such as programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have emerged [1]. These therapies have revolutionized cancer treatment and have become the standard-of-care for various cancer types [2]. While many predictive biomarkers have been scrutinized to select patients who might benefit from IO, their correlation with treatment responsiveness to IO remains elusive [3,4,5].

To further enhance the efficacy of IO, combinations with chemotherapy (Chemo-IO) have been introduced, leveraging their synergistic effects [6]. Correspondingly, numerous clinical trials exploring various combination regimens have been conducted. Some of these have already received approval as standard-of-care, while others are still in progress [7].

However, safety profiles significantly impact clinical outcomes, necessitating careful consideration of Chemo-IO’s safety alongside its efficacy. As might be intuitively expected, it is well-recognized that the addition of chemotherapy elevates the severity and frequency of treatment-related adverse events (TRAEs) when compared to IO alone [8, 9]. Nevertheless, the differences in discontinuation rates due to TRAEs between Chemo-IO and IO across various solid tumors have not been fully elucidated.

In this study, we evaluated the safety of Chemo-IO in comparison with IO monotherapy (mono-IO) as a first-line treatment, focusing on TRAEs leading to discontinuation in various types of solid tumors. Given the limited availability of randomized controlled trials (RCTs) directly comparing the safety between Chemo-IO and mono-IO, a network meta-analysis was conducted to establish an indirect comparison between these two groups.

Methods

The conduct of this meta-analysis adhered to the preferred reporting items for systematic review and meta-analysis (PRISMA) and the PRISMA extension statement for network meta-analysis (Supplementary Table 1) [10].

Types of adverse events

AEs of any cause (any AEs) are generally defined as any harmful changes in health or unanticipated side-effects experienced by an individual undergoing medical treatment or within a predetermined time period after the completion of treatment. Although any AEs are typically recorded after signing the informed consent form, treatment emergent adverse events (TEAEs) refer to any new events not that were not present prior to treatment initiation or any pre-existing conditions that escalate in severity or frequency post-exposure to the treatment. Therefore, TEAEs are collected after the initiation of the treatment. Moreover, both AEs or TEAEs can either be linked or unlinked to the treatment, while TRAEs are a subcategory of TEAEs deemed as being related to the treatment as determined by the investigator.

Data sources and search strategies

We systematically searched electronic databases (PubMed, Embase, Cochrane Library, and additional sources) for relevant clinical trials published in English between 01 January 2018 and 10 July 2023. The keywords for the literature search included: “clinical trial”, “phase III”, “first-line”, “immunotherapy”, “nivolumab”, “pembrolizumab”, “atezolizumab”, “PD-1 inhibitor”, “PD-L1 inhibitor”, “programmed cell death-1”, “programmed cell death-ligand 1”, “cemiplimab”, “sintilimab”, “tislelizumab”, “camrelizumab”, “durvalumab”, “avelumab”, “toripalimab”, “ipilimumab”, “sugemalimab”, “tremelimumab”, “serplulimab”, “adebrelimab”, “dostarlimab”, “cosibelimab”, “retifanlimab”, “CTLA-4 inhibitor”, and “cytotoxic T lymphocyte associated antigen-4”. More detailed search strategies are described in Supplementary Table 2.

Selection criteria

The literature incorporated in this study comprises prospective phase III RCT data pertaining to first-line treatments that utilize either IO agents in at least one arm for patients with solid cancer. To simplify the comparison in this study, we focused on IO agents targeting CTLA-4, PD-1, and PD-L1. Studies providing data on the number of discontinuations of any components of treatment due to TRAEs were included in our analysis. For clinical trials with multiple updated results, we referenced the initial peer-reviewed publication reporting discontinuation rates. This decision was based on the observation that the majority of immune-related adverse events (irAEs) or AEs typically occur within the first 15 weeks of treatment [11, 12].

Our exclusion criteria encompassed: (1) abstracts, posters, conference presentations and unpublished results, (2) inability to obtain full-text or repeated publications, (3) studies with incomplete or ambiguous data, or lack of original extractable data, (4) absence of evaluation indicators, (5) studies combined with other drugs or treatment (e.g., tyrosine kinase inhibitors and radiation therapy), and (6) studies in the adjuvant, neoadjuvant or maintenance treatment setting.

Screening was conducted on titles and abstracts prior to the assessment of full-texts to ascertain eligibility. An online spreadsheet was used to double-check all the included trials to ensure adherence to the inclusion criteria.

Data extraction

Two investigators, Sangwon Shin and Jimin Moon, independently conducted the database search, and scrutinized the titles, abstracts and full-texts to assess eligibility of studies and extract relevant data. Any disagreements encountered during this process were resolved via discussions with a third investigator, Seunghwan Shin. The extracted data includes elements such as clinical trial names, first author and publication year, indication, treatment group, IO agent name, number of participants included in safety analysis, reported outcomes (types of AEs leading to treatment discontinuation) and median follow-up duration. This information was then collated in an online spreadsheet.

Risk of bias (RoB) and quality assessment

The quality of methodologies of the included trials was evaluated using the Cochrane Collaboration’s tool (2.0) for assessing the RoB in RCTs [13]. The five bias domains examined were those arising from the randomization process, deviations from intended interventions, missing outcome data, measurement of the outcome, and selection of the reported result. Each of these was evaluated for low risk, high risk or having “some concerns” of bias. Subsequently, an overall RoB for each individual RCT was evaluated.

Additional analysis of the rate of treatment discontinuation due to either TEAEs or any AEs

In this network meta-analysis, the primary results were the rates of treatment discontinuation due to TRAEs. However, in instances where studies did not report discontinuation rates due to TRAEs, we also gathered data on the rates of discontinuation resulting from either any AEs or TEAEs using the same approach for additional analysis. Given that the incidence of AEs between the time of informed consent signing and the initiation of treatment is expected to be minimal, we conducted an aggregated analysis, combining the discontinuation rates due to either TEAEs or any AEs.

Statistical analysis

We conducted a network meta-analysis based on the frequentist approach to estimate the relative risks (RRs) for the treatment discontinuation rate due to TRAEs, along with 95% CIs compared to each control group in RCTs. RRs greater than others represented to have more risk of treatment discontinuation. Every treatment arm in each RCT was reclassified into one of five categories: mono-IO (defined as the use of a single IO drug), dual-IO (defined as the use of a combination of IO drugs), Chemo-only (defined as the use of a chemotherapy alone), Chemo-IO (defined as the use of a combination of only one IO drug and other chemotherapy drugs), and Chemo-dual-IO (defined as the use of a combination of two IO drugs and other chemotherapy drugs). In cases where multiple reclassified arms existed in a single RCT, we selected a control arm with an identical chemotherapy regimen to better delineate the impact of IO agents.

The heterogeneity within study designs and inconsistency between designs were assessed using Cochran’s Q statistics and its associated P-value. A random effects model was considered when substantial heterogeneity existed. The publication bias of studies was evaluated by Egger’s test with a funnel plot. Analyses were conducted separately for the risks of discontinuation due to TRAEs and those of discontinuation due to either any AEs or TEAEs. As the majority of the first-line trials involving IO were published in NSCLC, we conducted a subgroup analysis specific to NSCLC. A two-sided P-value < 0.05 was considered statistically significant. All statistical analyses were performed using R version 4.2.3 with meta and netmeta libraries. In addition, the geometry of the network was demonstrated by using the netgraph function in the netmeta library.

Results

Characteristics of the literature

We initially identified a total of 1,620 records from major databases and 18 supplemented publications from NEJM evidence (https://evidence.nejm.org/). After eliminating duplicates and non-relevant literatures through title and abstract screening, we found 151 literatures suitable for full-text review. Of these, 29 RCTs met our inclusion criteria (Fig. 1). Detailed evaluation was conducted on these 29 trials with results of discontinuation rate due to TRAEs (Tables 1 and Fig. 2A). A total of 21,677 patients were included in the safety analysis across various cancer types: NSCLC (11 trials, n = 8,442), esophageal squamous cell carcinoma (ESCC) (5 trials, n = 3,385), gastrointestinal cancer (GI cancer) (3 trials, n = 3,014), biliary tract cancer (BTC) (2 trials, n = 1,743), small cell lung cancer (SCLC) (2 trials, n = 1,047) and other cancers (6 trials, n = 4,046). For the Chemo-IO groups, the following IO agents were used: camrelizumab in 4 arms (n = 830), nivolumab in 3 arms (n = 1,451), pembrolizumab in 2 arms (n = 779), serplulimab in 2 arms (n = 771), durvalumab in 2 arms (n = 672) and other IO agents across 6 arms (n = 1,842). For the mono-IO groups, the following IO agents were used: pembrolizumab in 3 arms (n = 1,044), durvalumab in 3 arms (n = 1,304), and nivolumab in an arm (n = 391).

Fig. 1
figure 1

Flowchart of study selection and design aAdditional records were identified from NEJM evidence (https://evidence.nejm.org/). AE indicates adverse event; ICI, immune checkpoint inhibitor; IO, immunotherapy; NSCLC, non-small cell lung cancer; RCT, randomized controlled trial; TEAE, treatment emergent adverse event; TRAE, treatment-related adverse event

Table 1 Baseline characteristics of studies included in the network meta-analysis
Fig. 2
figure 2

Network plot illustrating comparisons of the risk of discontinuation due to TRAEs among different treatments

(A) For various cancer types, (B) For the NSCLC subgroup

The width of lines is proportional to the number of trials that compare directly in the same trial. Chemo-dual-IO indicates dual-immunotherapy combined with chemotherapy; Chemo-IO, mono-immunotherapy combined with chemotherapy; Chemo-only, chemotherapy alone; dual-IO, dual-immunotherapy; mono-IO, mono-immunotherapy; NSCLC, non-small cell lung cancer; TRAE, treatment-related adverse event.

Assessment of RoB and quality assessment

The summary of the RoB assessment for the included studies is presented in Supplementary Fig. 1. Given that this network meta-analysis focuses on treatment discontinuation, deviations from intended interventions inevitably raise some concerns or result in a high risk. Furthermore, due to the variation in median follow-up durations across the RCTs, the selection of the reported result domain was unavoidably rated as high risk. As a result, all RCTs were found to have a high overall RoB. Nevertheless, based on the funnel plot and Egger’s test, there is no definite evidence of publication bias concerning the discontinuation rate due to TRAEs (see Supplementary Fig. 2).

Comparison of the rate of treatment discontinuation due to TRAEs across all solid cancers

Substantial heterogeneity within study designs was observed among the results of each study (Q = 50.69, P < 0.01), leading to the application of a random effects model. Additionally, the Q statistic for testing inconsistency between designs, when using a full design-by-treatment interaction random effect model, was no longer significant (changing from Q = 31.93, P < 0.01 to Q = 13.69, P = 0.13).

In direct comparisons with Chemo-only, Chemo-IO exhibited a substantially higher rate of treatment discontinuation due to TRAEs (RR 1.60, 95% CI 1.36–1.88), whereas mono-IO showed a reduced rate of discontinuation due to TRAEs (RR 0.67, 95% CI 0.50–0.90). Additionally, a significant, albeit small, difference was observed between dual-IO and Chemo-only (RR 1.35, 95% CI 1.04–1.74). Dual-IO exhibited nearly twice the rate of discontinuation compared to mono-IO (RR 2.09, 95% CI 1.51–2.91). Outcomes from direct comparisons with only a limited number of trials, such as between Chemo-IO and mono-IO, were considered less reliable.

In the overall comparison, which combines the outcomes of direct comparison with indirect comparisons from the network meta-analysis, Chemo-IO exhibited a notably higher rate of discontinuation due to TRAEs compared to mono-IO (RR 2.68, 95% CI 1.98–3.63). Furthermore, when compared to Chemo-only, Chemo-IO presented an increased rate of discontinuation (RR 1.68, 95% CI 1.44–1.98), while mono-IO displayed a decreased rate (RR 0.63, 95% CI 0.48–0.82). A comprehensive summary of all RRs for discontinuation due to TRAEs across five treatment types can be found in Fig. 3.

Fig. 3
figure 3

Relative risks (RRs) of discontinuation due to TRAEs across various solid tumors

(A) Forest plot of RR of treatment discontinuation due to TRAEs. (B) Direct and overall comparisons of discontinuation due to TRAEs. Each cell contains pooled RR with 95% CI. An RR greater than 1 means that upper-row treatment has a greater risk of treatment discontinuation. The overall comparison outcomes, which combine direct and indirect comparison results from the network meta-analysis are displayed in the left lowerhalf, while results from pairwise meta-analysis are shown in the right upper half, if available. Significant results are in bold. Chemo-dual-IO indicates dual-immunotherapy combined with chemotherapy; Chemo-IO, mono-immunotherapy combined with chemotherapy; Chemo-only, chemotherapy alone; CI, confidence interval; dual-IO, dual-immunotherapy; mono-IO, mono-immunotherapy; NA, not available; RR, relative risk; TRAE, treatment-related adverse event.

Subgroup analysis of the rate of treatment discontinuation due to TRAEs in NSCLC

Out of 29 RCTs reporting on the rate of treatment discontinuation due to TRAEs, 11 RCTs focused on NSCLC (Fig. 2B). In line with the analysis conducted across various cancer types, a random effects model was applied due to substantial heterogeneity with designs (Q = 17.54, P < 0.01). Additionally, after applying a random effects model, the significance indicating inconsistency between designs was no longer positive (changing from Q = 8.56, P = 0.07 to Q = 2.82, P = 0.59).

When directly compared to Chemo-only, Chemo-IO was associated with a significantly higher rate of discontinuation due to TRAEs (RR 2.45, 95% CI 1.60–3.76), On the other hand, mono-IO presented a decreased rate of discontinuation due to TRAEs, although this was not statistically significant (RR 0.87, 95% CI 0.59–1.29). Through indirect comparison, we observed that Chemo-IO had a considerably higher rate of discontinuation owing to TRAEs when compared to mono-IO (RR 2.93, 95% CI 1.67–5.14). All RRs for discontinuation due to TRAEs are summarized in Fig. 4.

Fig. 4
figure 4

Relative risks (RRs) of discontinuation due to TRAEs in NSCLC.

(A) Forest plot of RR of treatment discontinuation due to TRAEs. (B) Direct and overall comparisons of discontinuation due to TRAEs. Each cell contains pooled RR with 95% CI. An RR greater than 1 means that upper-row treatment has a greater risk of treatment discontinuation. The overall comparison outcomes, which combine direct and indirect comparison results from the network meta-analysis are displayed in the left lower half, while results from pairwise meta-analysis are shown in the right upper half, if available. Significant results are in bold. Chemo-dual-IO indicates dual-immunotherapy combined with chemotherapy; Chemo-IO, mono-immunotherapy combined with chemotherapy; Chemo-only, chemotherapy alone; CI, confidence interval; dual-IO, dual-immunotherapy; mono-IO, mono-immunotherapy; NA, not available; NSCLC, non-small cell lung cancer; RR, relative risk; TRAE, treatment-related adverse event

Comparison of the rate of treatment discontinuation due to any AEs or TEAEs

For the studies not reporting discontinuation rates due to TRAEs, we collected the discontinuation rates associated with either any AEs or TEAEs. These data were sourced from 33 RCTs, encompassing a safety analysis of 18,482 patients (Fig. 1 and Supplementary Table 3). Using these data, we conducted an aggregate analysis integrating the discontinuation rates stemming from either any AEs or TEAEs (Supplementary Fig. 3).

Given that significant within-design heterogeneity was evident (Q = 96.47, P < 0.01), a random effects model was adopted. Under this model, the inconsistency between designs continued to be non-significant, as indicated by the change in Q value (from Q = 8.66, P = 0.12 to Q = 2.70, P = 0.75). Overall, when combining direct and indirect comparison results, Chemo-IO demonstrated a significantly higher rate of discontinuation due to any AEs/TEAEs compared to mono-IO (RR 2.48, 95% CI 1.93–3.17) (Supplementary Fig. 4). Similarly, for NSCLC patients, Chemo-IO showed a markedly elevated rate of discontinuation due to any AEs/TEAEs in comparison to mono-IO, as deduced from the indirect comparisons of Chemo-IO or mono-IO against Chemo-only (RR 2.09, 95% CI 1.29–3.39) (Supplementary Fig. 5).

Discussion

In our network meta-analysis evaluating first-line treatments for diverse solid tumors, we found that patients treated with Chemo-IO experienced a higher rate of treatment discontinuation due to TRAEs than those receiving mono-IO.

The advent of immunotherapy has ushered in a new era in cancer treatment. Starting with mono-IO, the field has since evolved to incorporate more complex regimens, such as IO-IO combinations and Chemo-IO, which have shown improved survival outcomes over Chemo-only or mono-IO [2, 6, 7, 14]. These advancements have led to approvals from the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for various cancer treatment settings [7, 8, 15, 16].

However, due to the additive nature of adverse effects, Chemo-IO is generally associated with a higher incidence of TRAEs compared to mono-IO [8, 9]. Consistent with these findings, our study observed a significantly increased rate of TRAE-induced discontinuation in the Chemo-IO group compared to the mono-IO group across various cancer types. Similarly, among NSCLC patients, Chemo-IO also demonstrated an elevated rate of discontinuation due to TRAEs compared with mono-IO.

The impact of treatment discontinuation due to AEs on clinical outcomes remains a subject of debate. Some studies have observed significantly poorer outcomes in the discontinuation group compared to the non-discontinuation group [17,18,19], while some studies have found no significant difference in clinical outcomes between groups that discontinued treatment due to AEs and those that did not [20,21,22], Although treatment discontinuation may occur because of AEs, several factors—including the timing of AEs, the patient’s response prior to discontinuation, the severity of the AEs or other variables—could be associated with clinical outcomes after discontinuation [17, 19, 23]. For instance, one study found that early AEs, occurring before 12 weeks and often leading to discontinuation, were associated with worse survival outcomes than later AEs [17]. Another study demonstrated that patients achieving complete response (CR) or partial response (PR) before the occurrence of the first irAE showed no difference in clinical outcomes between those who discontinued treatment and those who did not or those who resumed treatment. Conversely, patients who did not achieve CR or PR before the first irAE experienced significantly poorer outcomes in the discontinuation group compared to the non-discontinuation or re-treatment group [19]. Furthermore, mild irAEs might be indicative of a favorable response to immunotherapy, while severe irAEs (grade ≥ 3), often life-threatening or leading to treatment discontinuation, may not be indicative of favorable clinical outcomes [23]. TRAEs that occur during IO or Chemo-IO treatment could be linked to life-threatening conditions or mortality, leading physicians to generally consider them as potentially detrimental. In this study, we primarily suggested that IO-chemotherapy combination treatments are associated with a high rate of discontinuation. And there is a need for additional studies to investigate the association between AE- or TRAE-induced discontinuation and clinical outcomes.

Furthermore, the optimal duration for IO treatment continues to be a topic of ongoing debate, several trials, including KEYNOTE-189 [24], KEYNOTE-010 [25], and CheckMate-153 [26], suggest the possibility of poor survival outcomes or disease progression after discontinuing IO treatment following one or two years of administration [27]. Limited research has managed to follow patients who discontinue treatment due to AEs. However, one retrospective study that examined the clinical outcomes of patients who discontinued IO treatment due to immune-related AEs found that 20% of patients experienced disease progression within six months of discontinuation and 10% of patients died [28]. This evidence further emphasizes the potential impact of treatment discontinuation on patient outcomes.

In susceptible populations, such as elderly patients or those with a poor Eastern Cooperative Oncology Group performance status (ECOG PS), the administration of Chemo-IO could potentially be associated with a poorer prognosis due to an increased incidence of severe TRAEs and subsequent treatment discontinuation owing to these AEs [29]. Moreover, even with Chemo-only regimens, certain patient subgroups such as those of advanced age, poor performance status, individuals with anemia, impaired renal function, hearing impairment, or history of falls, are known to have an increased risk of toxicity [30]. This heightened toxicity could potentially lead to treatment discontinuation in these subgroups. However, enrolling these specific subgroups in oncological trials poses challenges, leaving unresolved questions and concerns about the extent of harm caused by treatment discontinuation in these populations.

For high-risk patients predicted to cease treatment due to TRAEs, a treatment strategy focusing on mono-IO might offer a lower discontinuation rate, thus improving treatment continuity. Furthermore, considering that several studies have reported comparable effects of IO to Chemo-IO in certain subgroups, such as patients aged 75 or over, or those who have received prior treatments [16, 31], there is a pressing need for further research into potential biomarkers or clinical factors that can aid in identifying patients who would benefit sufficiently from mono-IO instead of Chemo-IO, especially since these populations may experience higher rates of treatment discontinuation with Chemo-IO.

Several predictive biomarkers, including PD-L1, tumor mutation burden (TMB), and tumor-infiltrating lymphocytes (TILs), have been proposed to predict the efficacy of IO, but their roles remain inconclusive [3, 4, 8, 32]. Consequently, novel approaches are being explored to improve prediction accuracy, including the application of artificial intelligence (AI). Some studies have even reported that AI-assisted methods, such as the evaluation of pretreatment contrast-enhanced CT images, PD-L1 expression, or the spatial analysis of TIL, can yield better predictions of survival outcomes in NSCLC patients undergoing IO treatment [33,34,35]. Advancing this line of research might not only refine predictions of IO efficacy but also help in anticipating and managing treatment discontinuation, thus facilitating more personalized treatment strategies and potentially improving patient outcomes.

Limitations

While this meta-analysis provides valuable insights, it does have certain limitations. First, the potential for bias due to confounding effects could vary based on the types of immunotherapies used across the different regimens and the diversity in types of solid tumors. However, due to the lack of sufficient data from the RCTs, we did not differentiate among these agents in our analysis. Second, our study is primarily based on published literature and clinical trial results, which could potentially lead to publication bias, as studies with negative results are less likely to be published. Third, the duration of follow-up in each RCT differed, which could have potentially influenced the uncertainty of our pooled results. However, it is important to note that the majority of irAEs or AEs typically occur within the first 15 weeks of treatment. Therefore, the impact of varying follow-up durations on our findings is likely to be minimal. Last, we did not have access to individual patient data, which restricted us from performing subgroup analysis based on various clinical factors, including age, gender, as well as types and grades of TRAEs.

Conclusions

While the efficacy of Chemo-IO is generally acknowledged as superior to that of mono-IO, our study has supported that Chemo-IO is linked with a significantly higher rate of treatment discontinuation due to TRAEs. This finding underscores the importance of personalized treatment approaches. For certain patients, particularly those at a heightened risk of TRAEs, mono-IO might be the preferable option due to its lower discontinuation rate. Conversely, for other patients, the potential superior efficacy of Chemo-IO could outweigh the higher risk of discontinuation. Future research should focus on the identification of novel biomarkers and patient characteristics, which could guide the selection of the most appropriate treatment modality, taking into account both efficacy and the likelihood of treatment discontinuation. By customizing treatment strategies to align with individual patient profiles, we aim to enhance therapeutic efficacy while minimizing the risk of premature treatment discontinuation. This balanced approach emphasizes the need for a more nuanced understanding of individual patient responses to different cancer therapies, guiding the evolution of oncology towards a more adaptable and patient-focused discipline.

Data availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request. Supplementary information is also accessible in the supplementary materials.

Abbreviations

AI:

Artificial intelligence

AE:

Adverse event

any AE:

AEs regardless of causality

BTC:

Biliary tract cancer

Chemo-dual-IO:

The use of a combination of two IO drugs and other chemotherapy drugs

Chemo-IO:

The use of a combination of only one IO drug and other chemotherapy drugs

Chemo-only:

The use of a chemotherapy alone

CI:

Confidence interval

dual-IO:

The use of a combination of IO drugs

ECOG PS:

Eastern Cooperative Oncology Group performance status

EMA:

European Medicines Agency

ESCC:

Esophageal squamous cell carcinoma

FDA:

Food and Drug Administration

GI cancer:

Gastrointestinal cancer

HCC:

Hepatocellular carcinoma

HNSCC:

Head and neck squamous cell carcinoma

IO:

Immunotherapy

irAE:

Immune-related adverse event

MIBC:

Muscle invasive bladder cancer

mono-IO:

The use of a single IO drug

NA:

Not applicable

NPC:

Nasopharyngeal carcinoma

NSCLC:

Non-small cell lung cancer

PD-1:

Programmed cell death protein 1

PD-L1:

Programmed cell death ligand 1

PRISMA:

Preferred reporting items for systematic review and meta-analysis

RCT:

Randomized controlled trial

RoB:

Risk of bias

RR:

Relative risk

SCLC:

Small cell lung cancer

TEAE:

Treatment emergent adverse event

TIL:

Tumor-infiltrating lymphocyte

TMB:

Tumor mutation burden

TRAE:

Treatment related adverse event

References

  1. Hoos A. Development of immuno-oncology drugs - from CTLA4 to PD1 to the next generations. Nat Rev Drug Discov. 2016;15:235–47.

    Article  CAS  PubMed  Google Scholar 

  2. Sharma P, Goswami S, Raychaudhuri D, Siddiqui BA, Singh P, Nagarajan A, et al. Immune checkpoint therapy-current perspectives and future directions. Cell. 2023;186:1652–69.

    Article  CAS  PubMed  Google Scholar 

  3. Davis AA, Patel VG. The role of PD-L1 expression as a predictive biomarker: an analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J Immunother Cancer. 2019;7:278.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Samstein RM, Lee C-H, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ryan E, Sheahan K, Creavin B, Mohan HM, Winter DC. The current value of determining the mismatch repair status of colorectal cancer: a rationale for routine testing. Crit Rev Oncol Hematol. 2017;116:38–57.

    Article  CAS  PubMed  Google Scholar 

  6. Salas-Benito D, Pérez-Gracia JL, Ponz-Sarvisé M, Rodriguez-Ruiz ME, Martínez-Forero I, Castañón E, et al. Paradigms on Immunotherapy combinations with Chemotherapy. Cancer Discov. 2021;11:1353–67.

    Article  CAS  PubMed  Google Scholar 

  7. Larroquette M, Domblides C, Lefort F, Lasserre M, Quivy A, Sionneau B, et al. Combining immune checkpoint inhibitors with chemotherapy in advanced solid tumours: a review. Eur J Cancer. 2021;158:47–62.

    Article  CAS  PubMed  Google Scholar 

  8. Saxena P, Singh PK, Malik PS, Singh N. Immunotherapy alone or in combination with Chemotherapy as First-Line treatment of Non-small Cell Lung Cancer. Curr Treat Options Oncol. 2020;21:69.

    Article  PubMed  Google Scholar 

  9. Xu C, Chen Y-P, Du X-J, Liu J-Q, Huang C-L, Chen L, et al. Comparative safety of immune checkpoint inhibitors in cancer: systematic review and network meta-analysis. BMJ. 2018;363:k4226.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Martins F, Sofiya L, Sykiotis GP, Lamine F, Maillard M, Fraga M, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16:563–80.

    Article  CAS  PubMed  Google Scholar 

  12. Tang S-Q, Tang L-L, Mao Y-P, Li W-F, Chen L, Zhang Y, et al. The pattern of time to Onset and Resolution of Immune-related adverse events caused by Immune checkpoint inhibitors in Cancer: a pooled analysis of 23 clinical trials and 8,436 patients. Cancer Res Treat. 2021;53:339–54.

    Article  CAS  PubMed  Google Scholar 

  13. Sterne JAC, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:l4898.

    Article  PubMed  Google Scholar 

  14. Dafni U, Tsourti Z, Vervita K, Peters S. Immune checkpoint inhibitors, alone or in combination with chemotherapy, as first-line treatment for advanced non-small cell lung cancer. A systematic review and network meta-analysis. Lung Cancer. 2019;134:127–40.

    Article  PubMed  Google Scholar 

  15. Wang C, Qiao W, Jiang Y, Zhu M, Shao J, Wang T, et al. The landscape of immune checkpoint inhibitor plus chemotherapy versus immunotherapy for advanced non-small‐cell lung cancer: a systematic review and meta‐analysis. J Cell Physiol. 2020;235:4913–27.

    Article  CAS  PubMed  Google Scholar 

  16. Akinboro O, Vallejo JJ, Mishra-Kalyani PS, Larkins EA, Drezner NL, Tang S, et al. Outcomes of anti-PD-(L1) therapy in combination with chemotherapy versus immunotherapy (IO) alone for first-line (1L) treatment of advanced non-small cell lung cancer (NSCLC) with PD-L1 score 1–49%: FDA pooled analysis. JCO. 2021;39 15suppl:9001–1.

    Article  Google Scholar 

  17. Naqash AR, Ricciuti B, Owen DH, Florou V, Toi Y, Cherry C, et al. Outcomes associated with immune-related adverse events in metastatic non-small cell lung cancer treated with nivolumab: a pooled exploratory analysis from a global cohort. Cancer Immunol Immunother. 2020;69:1177–87.

    Article  CAS  PubMed  Google Scholar 

  18. Ksienski D, Wai ES, Croteau N, Fiorino L, Brooks E, Poonja Z, et al. Efficacy of Nivolumab and Pembrolizumab in patients with Advanced non–small-cell Lung Cancer needing treatment interruption because of adverse events: a Retrospective Multicenter Analysis. Clin Lung Cancer. 2019;20:e97–106.

    Article  CAS  PubMed  Google Scholar 

  19. Santini FC, Rizvi H, Plodkowski AJ, Ni A, Lacouture ME, Gambarin-Gelwan M, et al. Safety and Efficacy of re-treating with Immunotherapy after Immune-related adverse events in patients with NSCLC. Cancer Immunol Res. 2018;6:1093–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schadendorf D, Wolchok JD, Hodi FS, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Efficacy and Safety Outcomes in Patients With Advanced Melanoma Who Discontinued Treatment With Nivolumab and Ipilimumab Because of Adverse Events: A Pooled Analysis of Randomized Phase II and, Trials III et al. JCO. 2017;35:3807–14.

  21. Sun L, Bleiberg B, Hwang W-T, Marmarelis ME, Langer CJ, Singh A et al. Association between Duration of Immunotherapy and overall survival in Advanced Non-small Cell Lung Cancer. JAMA Oncol. 2023;:e231891.

  22. Okauchi S, Miyazaki K, Shiozawa T, Yamada H, Satoh H, Hizawa N. Discontinuation of Immune checkpoint inhibitor due to irAEs in NSCLC patients with EGFR Mutation. Cancer Diagn Progn. 2023;3:244–50.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Zhou X, Yao Z, Yang H, Liang N, Zhang X, Zhang F. Are immune-related adverse events associated with the efficacy of immune checkpoint inhibitors in patients with cancer? A systematic review and meta-analysis. BMC Med. 2020;18:87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rodríguez-Abreu D, Powell SF, Hochmair MJ, Gadgeel S, Esteban E, Felip E, et al. Pemetrexed plus platinum with or without pembrolizumab in patients with previously untreated metastatic nonsquamous NSCLC: protocol-specified final analysis from KEYNOTE-189. Ann Oncol. 2021;32:881–95.

    Article  PubMed  Google Scholar 

  25. Herbst RS, Garon EB, Kim D-W, Cho BC, Perez-Gracia JL, Han J-Y, et al. Long-term outcomes and retreatment among patients with previously treated, programmed death-ligand 1–Positive, Advanced non–small-cell Lung Cancer in the KEYNOTE-010 study. J Clin Oncol. 2020;38:1580–90.

    Article  CAS  PubMed  Google Scholar 

  26. Waterhouse DM, Garon EB, Chandler J, McCleod M, Hussein M, Jotte R, et al. Continuous versus 1-Year fixed-duration nivolumab in previously treated Advanced Non-small-cell Lung Cancer: CheckMate 153. J Clin Oncol. 2020;38:3863–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yin J, Song Y, Tang J, Zhang B. What is the optimal duration of immune checkpoint inhibitors in malignant tumors? Front Immunol. 2022;13:983581.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lievense LA, Heukels P, van Walree NC. Clinical outcomes of patients with metastatic NSCLC after discontinuation of Immunotherapy because of Immune-related adverse effects. JTO Clin Res Rep. 2023;4:100441.

    PubMed  Google Scholar 

  29. Fujimoto D, Miura S, Yoshimura K, Wakuda K, Oya Y, Haratani K, et al. A real-world study on the effectiveness and safety of Pembrolizumab Plus Chemotherapy for Nonsquamous NSCLC. JTO Clin Res Rep. 2022;3:100265.

    PubMed  Google Scholar 

  30. Hurria A, Togawa K, Mohile SG, Owusu C, Klepin HD, Gross CP, et al. Predicting chemotherapy toxicity in older adults with cancer: a prospective multicenter study. J Clin Oncol. 2011;29:3457–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang Y, Han H, Zhang F, Lv T, Zhan P, Ye M, et al. Immune checkpoint inhibitors alone vs immune checkpoint inhibitors—combined chemotherapy for NSCLC patients with high PD-L1 expression: a network meta-analysis. Br J Cancer. 2022;127:948–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18:842–59.

    Article  CAS  PubMed  Google Scholar 

  33. Trebeschi S, Drago SG, Birkbak NJ, Kurilova I, Cǎlin AM, Delli Pizzi A, et al. Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Ann Oncol. 2019;30:998–1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Choi S, Cho SI, Ma M, Park S, Pereira S, Aum BJ, et al. Artificial intelligence-powered programmed death ligand 1 analyser reduces interobserver variation in tumour proportion score for non-small cell lung cancer with better prediction of immunotherapy response. Eur J Cancer. 2022;170:17–26.

    Article  CAS  PubMed  Google Scholar 

  35. Park S, Ock C-Y, Kim H, Pereira S, Park S, Ma M, et al. Artificial Intelligence-powered spatial analysis of Tumor-infiltrating lymphocytes as complementary biomarker for Immune Checkpoint Inhibition in Non-small-cell Lung Cancer. J Clin Oncol. 2022;40:1916–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cheng Y, Han L, Wu L, Chen J, Sun H, Wen G, et al. Effect of First-Line Serplulimab vs Placebo added to Chemotherapy on Survival in patients with extensive-stage small cell Lung Cancer: the ASTRUM-005 Randomized Clinical Trial. JAMA. 2022;328:1223–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Song Y, Zhang B, Xin D, Kou X, Tan Z, Zhang S, et al. First-line serplulimab or placebo plus chemotherapy in PD-L1-positive esophageal squamous cell carcinoma: a randomized, double-blind phase 3 trial. Nat Med. 2023;29:473–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kang Y-K, Chen L-T, Ryu M-H, Oh D-Y, Oh SC, Chung HC, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022;23:234–47.

    Article  CAS  PubMed  Google Scholar 

  39. Zhou C, Chen G, Huang Y, Zhou J, Lin L, Feng J, et al. Camrelizumab plus carboplatin and pemetrexed versus chemotherapy alone in chemotherapy-naive patients with advanced non-squamous non-small-cell lung cancer (CameL): a randomised, open-label, multicentre, phase 3 trial. Lancet Respir Med. 2021;9:305–14.

    Article  CAS  PubMed  Google Scholar 

  40. Ren S, Chen J, Xu X, Jiang T, Cheng Y, Chen G, et al. Camrelizumab Plus Carboplatin and Paclitaxel as First-Line treatment for advanced squamous NSCLC (CameL-Sq): a phase 3 trial. J Thorac Oncol. 2022;17:544–57.

    Article  CAS  PubMed  Google Scholar 

  41. Wang J, Zhou C, Yao W, Wang Q, Min X, Chen G, et al. Adebrelimab or placebo plus carboplatin and etoposide as first-line treatment for extensive-stage small-cell lung cancer (CAPSTONE-1): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022;23:739–47.

    Article  CAS  PubMed  Google Scholar 

  42. Yang Y, Qu S, Li J, Hu C, Xu M, Li W, et al. Camrelizumab versus placebo in combination with gemcitabine and cisplatin as first-line treatment for recurrent or metastatic nasopharyngeal carcinoma (CAPTAIN-1st): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2021;22:1162–74.

    Article  CAS  PubMed  Google Scholar 

  43. Hellmann MD, Ciuleanu T-E, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, et al. Nivolumab plus Ipilimumab in Lung Cancer with a high Tumor Mutational Burden. N Engl J Med. 2018;378:2093–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Doki Y, Ajani JA, Kato K, Xu J, Wyrwicz L, Motoyama S, et al. Nivolumab Combination Therapy in Advanced Esophageal squamous-cell carcinoma. N Engl J Med. 2022;386:449–62.

    Article  CAS  PubMed  Google Scholar 

  45. Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet. 2021;398:27–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Baas P, Scherpereel A, Nowak AK, Fujimoto N, Peters S, Tsao AS, et al. First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): a multicentre, randomised, open-label, phase 3 trial. Lancet. 2021;397:375–86.

    Article  CAS  PubMed  Google Scholar 

  47. Paz-Ares L, Ciuleanu T-E, Cobo M, Schenker M, Zurawski B, Menezes J, et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): an international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021;22:198–211.

    Article  CAS  PubMed  Google Scholar 

  48. Powles T, van der Heijden MS, Castellano D, Galsky MD, Loriot Y, Petrylak DP, et al. Durvalumab alone and durvalumab plus tremelimumab versus chemotherapy in previously untreated patients with unresectable, locally advanced or metastatic urothelial carcinoma (DANUBE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol. 2020;21:1574–88.

    Article  CAS  PubMed  Google Scholar 

  49. Gogishvili M, Melkadze T, Makharadze T, Giorgadze D, Dvorkin M, Penkov K, et al. Cemiplimab plus chemotherapy versus chemotherapy alone in non-small cell lung cancer: a randomized, controlled, double-blind phase 3 trial. Nat Med. 2022;28:2374–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Luo H, Lu J, Bai Y, Mao T, Wang J, Fan Q, et al. Effect of Camrelizumab vs Placebo added to Chemotherapy on Survival and Progression-Free Survival in patients with Advanced or metastatic esophageal squamous cell carcinoma: the ESCORT-1st Randomized Clinical Trial. JAMA. 2021;326:916–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhou C, Wang Z, Sun Y, Cao L, Ma Z, Wu R, et al. Sugemalimab versus placebo, in combination with platinum-based chemotherapy, as first-line treatment of metastatic non-small-cell lung cancer (GEMSTONE-302): interim and final analyses of a double-blind, randomised, phase 3 clinical trial. Lancet Oncol. 2022;23:220–33.

    Article  CAS  PubMed  Google Scholar 

  52. Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022;1:EVIDoa2100070.

    Article  Google Scholar 

  53. Monk BJ, Colombo N, Oza AM, Fujiwara K, Birrer MJ, Randall L, et al. Chemotherapy with or without avelumab followed by avelumab maintenance versus chemotherapy alone in patients with previously untreated epithelial ovarian cancer (JAVELIN ovarian 100): an open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22:1275–89.

    Article  CAS  PubMed  Google Scholar 

  54. Psyrri A, Fayette J, Harrington K, Gillison M, Ahn M-J, Takahashi S, et al. Durvalumab with or without tremelimumab versus the EXTREME regimen as first-line treatment for recurrent or metastatic squamous cell carcinoma of the head and neck: KESTREL, a randomized, open-label, phase III study. Ann Oncol. 2023;34:262–74.

    Article  CAS  PubMed  Google Scholar 

  55. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive non–small-cell Lung Cancer. N Engl J Med. 2016;375:1823–33.

    Article  CAS  PubMed  Google Scholar 

  56. Mok TSK, Wu Y-L, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019;393:1819–30.

    Article  CAS  PubMed  Google Scholar 

  57. Shitara K, Van Cutsem E, Bang Y-J, Fuchs C, Wyrwicz L, Lee K-W, et al. Efficacy and safety of Pembrolizumab or Pembrolizumab Plus Chemotherapy vs Chemotherapy alone for patients with First-line, Advanced Gastric Cancer: the KEYNOTE-062 phase 3 Randomized Clinical Trial. JAMA Oncol. 2020;6:1571–80.

    Article  PubMed  Google Scholar 

  58. Kelley RK, Ueno M, Yoo C, Finn RS, Furuse J, Ren Z, et al. Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2023;401:1853–65.

    Article  PubMed  Google Scholar 

  59. Rizvi NA, Cho BC, Reinmuth N, Lee KH, Luft A, Ahn M-J, et al. Durvalumab with or without Tremelimumab vs Standard Chemotherapy in First-line treatment of metastatic non-small cell Lung Cancer: the MYSTIC Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020;6:661–74.

    Article  PubMed  PubMed Central  Google Scholar 

  60. de Castro G, Rizvi NA, Schmid P, Syrigos K, Martin C, Yamamoto N, et al. NEPTUNE: phase 3 study of First-Line Durvalumab Plus Tremelimumab in patients with metastatic NSCLC. J Thorac Oncol. 2023;18:106–19.

    Article  PubMed  Google Scholar 

  61. Lu Z, Wang J, Shu Y, Liu L, Kong L, Yang L, et al. Sintilimab versus placebo in combination with chemotherapy as first line treatment for locally advanced or metastatic oesophageal squamous cell carcinoma (ORIENT-15): multicentre, randomised, double blind, phase 3 trial. BMJ. 2022;377:e068714.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Johnson ML, Cho BC, Luft A, Alatorre-Alexander J, Geater SL, Laktionov K, et al. Durvalumab with or without Tremelimumab in Combination with Chemotherapy as First-Line Therapy for Metastatic non–small-cell Lung Cancer: the Phase III POSEIDON Study. JCO. 2023;41:1213–27.

    Article  CAS  Google Scholar 

  63. Xu J, Kato K, Raymond E, Hubner RA, Shu Y, Pan Y, et al. Tislelizumab plus chemotherapy versus placebo plus chemotherapy as first-line treatment for advanced or metastatic oesophageal squamous cell carcinoma (RATIONALE-306): a global, randomised, placebo-controlled, phase 3 study. Lancet Oncol. 2023;24:483–95.

    Article  CAS  PubMed  Google Scholar 

  64. Oh D-Y, Ruth He A, Qin S, Chen L-T, Okusaka T, Vogel A, et al. Durvalumab plus Gemcitabine and Cisplatin in Advanced biliary tract Cancer. NEJM Evid. 2022;1:EVIDoa2200015.

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by Lunit.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

Substantially contributed to conception or design of the work: YL, CYO, SS‡. Contributed to acquisition, analysis, or interpretation of data: SS, JM, CO, SS‡. Drafted the manuscript for important content: SS, JM, SK. Critically revised the manuscript: SIC, SS‡. Read and approved the final manuscript: All authors.‡Corresponding author.

Corresponding author

Correspondence to Seunghwan Shin.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Sangwon Shin, Jimin Moon, Chiyoon Oum, Seulki Kim, Soo Ick Cho, Yoojoo Lim, Chan-Young Ock, and Seunghwan Shin are employed by Lunit and/or have stock/stock options in Lunit.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The past version of this study, a conventional meta-analysis, was presented as a poster in part at the Society for Immunotherapy of Cancer 2022 (November 8–12, 2022), Boston, MA.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shin, S., Moon, J., Oum, C. et al. Discontinuation risk from adverse events: immunotherapy alone vs. combined with chemotherapy: a systematic review and network meta-analysis. BMC Cancer 24, 152 (2024). https://doi.org/10.1186/s12885-024-11897-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-11897-4

Keywords