Skip to main content
  • Research article
  • Open access
  • Published:

Prognostic value of 18F-FDG PET and PET/CT for assessment of treatment response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis

Abstract

Background

We performed a systematic review and meta-analysis to evaluate the prognostic significance of 18F-FDG PET and PET/CT for evaluation of responses to neoadjuvant chemotherapy (NAC) in breast cancer patients.

Methods

We searched PubMed, Embase, and the Cochrane Library databases until June 2020 to identify studies that assessed the prognostic value of 18F-FDG PET scans during or after NAC with regard to overall (OS) and disease-free survival (DFS). Hazard ratios (HRs) and their 95% confidence intervals (CIs) were pooled meta-analytically using a random-effects model.

Results

Twenty-one studies consisting of 1630 patients were included in the qualitative synthesis. Twelve studies investigated the use of PET scans for interim response evaluation (during NAC) and 10 studies assessed post-treatment PET evaluation (after NAC). The most widely evaluated parameter distinguishing metabolic responders from poor responders on interim or post-treatment PET scans was %ΔSUVmax, defined as the percent reduction of SUVmax compared to baseline PET, followed by SUVmax and complete metabolic response (CMR). For the 17 studies included in the meta-analysis, the pooled HR of metabolic responses on DFS was 0.21 (95% confidence interval [CI], 0.14–0.32) for interim PET scans and 0.31 (95% CI, 0.21–0.46) for post-treatment PET scans. Regarding the influence of metabolic responses on OS, the pooled HRs for interim and post-treatment PET scans were 0.20 (95% CI, 0.09–0.44) and 0.26 (95% CI, 0.14–0.51), respectively.

Conclusions

The currently available literature suggests that the use of 18F-FDG PET or PET/CT for evaluation of response to NAC provides significant predictive value for disease recurrence and survival in breast cancer patients and might allow risk stratification and guide rational management.

Introduction

Neoadjuvant chemotherapy (NAC) is the initial therapy for patients with inoperable or locally advanced breast cancer [1] and enables more patients with operable but large primary tumours to be treated with breast-conserving surgery [2]. Given a non-negligible proportion of patients treated with NAC cannot achieve an optimal response or have subsequent disease progression, accurate assessment of the therapeutic response is important to reduce toxicity from ineffective chemotherapy and guide selection of an alternative treatment option. Changes in tumour morphology and size on breast magnetic resonance imaging (MRI) after several cycles of NAC are widely used markers for assessment of the treatment response and prediction of patient outcome [3]. However, MRI studies have different predictive values across the various breast cancer subtypes [4, 5], and there is a limited evidence for their prognostic value. In addition, MRI techniques do not allow evaluation of newly developing distant metastasis during NAC.

Decreased glucose metabolism within breast cancer tissue on 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) is a useful indicator to assess the effectiveness of NAC [6]. Several meta-analyses have reported that 18F-FDG PET/CT scans performed during or after NAC could predict the final pathological response after completion of NAC [7,8,9,10]. A meta-analysis directly comparing PET/CT and breast MRI reported that PET/CT has a higher sensitivity and specificity for assessment of pathological response than conventional MRI when performed before 3 cycles of NAC [11]. More recently, accumulating evidence has suggested that assessment of the metabolic response using 18F-FDG PET or PET/CT has prognostic significance in breast cancer patients who underwent NAC [12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32].

The use of 18F-FDG PET or PET/CT for response evaluation is not yet established in clinical practice [1], in part due to lack of evidence supporting changes in treatment plans based on the results of PET scans performed to evaluate treatment responses and whether this strategy improves clinical outcomes [33]. However, prior to designing clinical trials evaluating the use of PET for response-adaptive treatment, a thorough review of the currently available data regarding the correlations between metabolic responses evaluated using PET or PET/CT scans and disease recurrence or survival, and of associated risk stratification of breast cancer patients during or after NAC, is warranted. In addition, differences in the timing of PET scans, response criteria and their threshold values across the available studies and their potential effects on survival also need to be assessed. Therefore, we performed a systematic review and meta-analysis of the currently available literature on the prognostic value of 18F-FDG PET or PET/CT for treatment response evaluation in breast cancer patients who underwent NAC.

Materials and methods

This meta-analysis adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [34]. The protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO) network (registration no.CRD42020188979).

Literature search and data extraction

The PubMed, Embase, and the Cochrane Library databases were searched from inception to June 4, 2020. Search queries included the related terms ‘breast cancer’, 18F-FDG PET’, ‘neoadjuvant therapy’, and ‘prognosis’, which are described in the Additional File 1. There was no language restriction for the electronic searches. The references of the extracted articles were also examined to identify additional relevant articles.

The inclusion criteria were based upon the Patient/Intervention/Comparator/Outcome/Study design (PICOS) criteria as follows [34]: (1) female ‘patients’ with breast cancer; (2) 18F-FDG PET, PET/CT, or PET/MRI during or after NAC as ‘intervention’; (3) no ‘comparator’ for the study; (4) overall (OS) and disease-free survival (DFS) as ‘outcome’; and (5) prospective or retrospective studies published as original articles as ‘study design’. The exclusion criteria were as follows: (1) small sample size (< 10 patients); (2) other publication types including conference abstracts, review articles, editorials, and letters; (3) papers irrelevant to the research question; (4) insufficient information regarding survival analysis provided for the study; and (5) overlapping study populations. When the study populations may have overlapped, we selected the publication with the largest population.

Data extraction and quality assessment

The outcomes, study, and patient characteristics of the included studies were extracted using a standardised form. The methodological quality was appraised using the Quality in Prognostic Studies (QUIPS) tool based on key questions of prompting items and considerations for six bias domains which consist of study participation, study attrition, prognostic factor measurement, outcome measurement, study confounding, statistical analysis, and reporting [35]. Study selection, data extraction, and quality assessment were performed by two independent reviewers (S.H. and J.Y.C) with any discrepancy resolved through discussion.

Statistical analyses

Results from the survival analyses within individual articles, including survival rates, univariate and multivariate hazard ratios (HRs), and p values from log-rank tests were extracted. When the Kaplan-Meier curves were provided without corresponding HRs, survival probability at each time point was extracted by means of Engauge Digitizer software (version 10.4, http://markummitchell.github.io/engauge-digitizer/) and individual patient data were reconstructed using the methodology proposed by Guyot et al. [36]. Then, Cox regression analyses were performed to derive HRs and their 95% confidence intervals (CIs); if no events were observed in one arm, Cox regression with Firth’s penalised likelihood was used.

The HRs and their 95% CIs from the univariate Cox regression analyses comparing metabolic responders and poor responders on PET scans were pooled meta-analytically using the DerSimonian-Liard method for calculating weights. If multiple HRs for a single PET parameter were provided in an individual study due to different cut-offs or regions of interest, we selected the HR with the best prognostic value for the meta-analyses. Of note, the terms ‘interim PET’ and ‘post-treatment PET’ were defined as PET studies conducted during (i.e., after one, two, or three cycles) and after NAC, respectively. Higgins I2 statistics were used to assess heterogeneity [37]. Funnel plots with Egger’s test were drawn to identify the presence of publication bias [38]. The ‘survHE’, ‘coxphf’, ‘meta’, and ‘metafor’ packages in R (R Foundation for Statistical Computing, version 3.6.0) were used for the statistical analyses.

Results

Study characteristics

The PRISMA study selection process is described in Fig. 1. The initial literature search yielded 1682 articles. After removing 437 duplicates, screening of the remaining 1245 titles and abstracts yielded 37 potentially eligible papers. We excluded 16 of the 37 articles for the following reasons: palliative chemotherapy (n = 2), neoadjuvant endocrine therapy only (n = 1), no survival analysis (n = 3), overlapping patient populations (n = 8), PET for baseline assessment (n = 1), and only kinetic analyses of dynamic PET scans (n = 1). Thus, 21 studies with 1630 patients were included in the qualitative synthesis [12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32]. Eleven studies were prospectively conducted, where ten were retrospective studies. For the quantitative synthesis, we included only studies where HRs for metabolic responses assessed by PET scans either during or after NAC were available. A total of 17 studies (1279 patients) were included in the quantitative synthesis [12, 16,17,18,19,20,21, 23,24,25,26,27,28,29,30,31,32]. Of note, there was one study in which Kaplan-Meier curves were separately plotted according to the therapeutic regimen [19]; these patients were incorporated into the meta-analysis as separate cohorts. Tables 1 and 2 summarise the patient and study characteristics.

Fig. 1
figure 1

PRISMA flow chart showing the study selection process

Table 1 Study characteristics of the included studies
Table 2 Patient characteristics in the included studies

Quality assessment

The quality assessment performed using the QUIPS tool is illustrated in Fig. 2. The specific number of studies at risk of bias and reasons were as follows: Five studies presented a moderate risk in selection of participants because of the retrospective study designs, lack of clarity regarding whether the patients were enrolled in a consecutive manner, and/or the unclear inclusion and exclusion criteria [12, 23, 25, 26, 32]. All studies were rated as having a low risk of attrition bias. For prognostic factor measurement, nine studies had a high risk of bias due to the use of data-dependent cut-off values [12,13,14, 18, 20, 24, 27, 31, 32]. Regarding outcome measurements, fifteen studies had a moderate risk of bias because the definition or methods for measuring disease recurrence were unclear [12,13,14,15,16, 22,23,24, 26,27,28,29,30,31,32]. Six studies presented a moderate risk of confounding bias as no adjustment for potential confounders was performed [12, 18, 20, 21, 24, 28]. With regard to statistical analysis domains, five studies had a moderate risk of bias as it was unclear which variables were incorporated into the multivariate analyses, or too many variables were included in the multivariate analyses considering the number of patients in the study population [13, 15, 17, 19, 30]. One study had a high risk of bias in the statistical analyses due to possible selective reporting, as the results of survival analysis were provided for only a subset of the study population, and not the whole population [12].

Fig. 2
figure 2

Quality assessment using the QUIPS tool

Qualitative synthesis

The outcomes of the included articles are summarised in Table 3. PET scanning for evaluation of the patients’ response to NAC was performed during (interim PET) or after treatment (post-treatment PET) in 12 and 10 studies, respectively. The most widely evaluated PET parameter was the percent reduction of maximum standardised uptake value (SUVmax) compared to baseline SUVmax (%ΔSUVmax), followed by SUVmax and complete metabolic response (CMR) on interim or post-treatment PET scans. Of note, %ΔSUVmax is defined as (SUVmax at baseline PET − SUVmax at interim or post-treatment PET) / SUVmax at baseline PET × 100%. The CMR in the included studies was defined as negative FDG uptake [21], or according to the European Organisation for Research and Treatment of Cancer (EORTC) or Positron Emission Tomography Response Criteria in Solid Tumours (PERCIST) criteria [17, 26, 30]. Regarding the timing of interim assessment, PET or PET/CT were performed after two cycles of NAC in five studies [17,18,19, 30, 32], one cycle in four [20, 21, 28, 31], and three cycles in two studies [13, 29]. Specifically, higher %ΔSUVmax at interim evaluation was significantly associated with longer DFS in seven of ten studies [13, 17,18,19,20,21, 28, 29, 31, 32], and longer OS in two of six studies [13, 17, 20, 21, 28, 29]. In addition, CMR and lower SUVmax on interim PET scan was significantly associated with longer DFS in one of two studies [17, 30], and one study [18], respectively. Regarding post-treatment PET scans, %ΔSUVmax was a significant prognostic factor for DFS in four of six studies [12, 13, 17, 22, 25, 27]. Lower SUVmax was significantly associated with longer DFS in all five studies [12, 17, 22, 23, 25]. CMR was correlated with better DFS in three studies [16, 17, 26]; however, CMR was no longer statistically significant after completion of multivariate analyses of the data from two of them [17, 26]. Two studies reported that metabolic tumour volume and total lesion glycolysis and their reduction rates on post-treatment PET scans were significant predictors for better DFS [22, 25].

Table 3 Summary of outcomes in the included studies

The five studies exclusively included specific hormonal subtype of either TN or HR+/HER2−. Of two studies for HR+/HER2− subtype [18, 20], higher %ΔSUVmax on interim PET assessment was significantly associated with better DFS and OS. Likewise in three studies for TN subtype [19, 21, 27], higher %ΔSUVmax on interim PET scans or post-treatment PET scans was a significant predictor for longer DFS.

Quantitative synthesis

Meta-analytical pooling of HRs for interim and post-treatment PET analyses on DFS and OS was performed. With regard to the influence of metabolic responses on DFS, the pooled HR for interim PET scans was 0.21 (95% CI, 0.14–0.32; Fig. 3a) with no heterogeneity (I2 = 0%). No publication bias was shown in the funnel plot and Egger’s test (P = 0.8654; Fig. 4a). PET response analyses performed after one, two, and three cycles of NAC showed comparable prognostic values for DFS with pooled HRs of 0.18 (95% CI, 0.09–0.35), 0.25 (95% CI, 0.14–0.45), and 0.22 (95% CI, 0.09–0.51), respectively (P for subgroup difference = 0.7661). The pooled HR for metabolic responses on post-treatment PET/CT was 0.31 (95% CI, 0.21–0.46; Fig. 3b). No heterogeneity was found (I2 = 0%), and no publication bias was present (P = 0.3199; Fig. 4b). No statistical difference was found between the pooled HRs of interim and post-treatment PET response analyses (P = 0.1942). For studies using combined PET/CT, the pooled HRs for interim and post-treatment PET/CT were 0.23 (95% CI, 0.15–0.37) and 0.30 (95% CI, 0.20–0.43), respectively. The results of subgroup analyses according to PET response parameters are provided in Table 4.

Fig. 3
figure 3

Forest plots showing the pooled HRs of the PET response on interim (a) and post-treatment (b) 18F-FDG PET scans for disease-free survival

Fig. 4
figure 4

Funnel plots of studies assessing the PET response on interim (a) and post-treatment (b) 18F-FDG PET scans for disease-free survival

Table 4 Subgroup analysis according to PET timing and parameters

Among nine studies assessing the prognostic value of %ΔSUVmax for DFS on interim PET scans, six studies included patients with initial clinical stage II–III cancers; %ΔSUVmax was a significant predictor of DFS in Stage II–III breast cancer with a pooled HR of 0.21 (95% CI, 0.13–0.34; Additional file 2: Fig. S1). Of these, there were four studies which included either ER+/HER2− or triple-negative breast cancer population; %ΔSUVmax was also a significant prognostic factor in stage II–III ER+/HER2− or triple-negative breast cancer (pooled HRs = 0.20 [95% CI, 0.08–0.47] and 0.26 [95% CI, 0.11–0.61], respectively). Meta-regression analyses were performed according to clinical variables (including age, stage, histologic type and grade, receptors, subtypes, and pathological complete response) for these nine studies, and no variable was found to significantly influence the HRs (Additional file 3: Table S1).

With regard to the influence of metabolic response on OS, the pooled HR for the interim PET scans was 0.20 (95% CI, 0.09–0.44; Fig. 5a), and no heterogeneity was present (I2 = 0%). The pooled HRs for the metabolic response on post-treatment PET scans was 0.26 (95% CI, 0.14–0.51; Fig. 5b). No heterogeneity was found (I2 = 0%). No statistical difference was found between the pooled HRs of interim and post-treatment PET response analyses (P = 0.6137). Publication bias could not be evaluated because of the limited number of included studies. No statistical difference was found upon subgroup analyses in accordance with PET parameters (Table 4), although the paucity of studies limited the statistical power. For studies using combined PET/CT, the pooled HRs for interim and post-treatment PET/CT were 0.33 (95% CI, 0.08–1.37) and 0.24 (95% CI, 0.11–0.51), respectively.

Fig. 5
figure 5

Forest plots showing the pooled HRs of the PET response on interim (a) and post-treatment (b) 18F-FDG PET scans for overall survival

Discussion

In this meta-analysis, we assessed the prognostic value of 18F-FDG PET or PET/CT for evaluation of the response to treatment in breast cancer patients. Our major findings were as follows; (1) PET metabolic response during and after NAC is a significant predictor of disease recurrence and death; (2) more evidence is available regarding PET scans for prediction of DFS than OS (18 and 7 studies included in meta-analyses for DFS and OS, respectively); (3) %ΔSUVmax was the most frequently evaluated response parameter in both interim and post-treatment PET scans; (4) no differences were found between the prognostic values of interim and post-treatment PET scans in term of both DFS and OS; (5) regarding the timing of the interim assessment, PET is mostly commonly performed after 2 cycles (followed by PET after 1 cycle) and yielded significant prognostic values; and (6) in more than half of the included studies, the cut-off values for each PET parameter were data-dependent and therefore differed greatly across studies.

Notably, a metabolic response identified on PET scans is significantly associated with the pathological response in surgical specimens after NAC [7,8,9], a well-established predictor of patient outcomes [39]. In addition, in clinical practice, it is plausible that 18F-FDG PET or PET/CT is highly likely to have an incremental prognostic value on pathological response of breast cancer given (1) PET scans enable early assessment of patient responses to NAC, which may support decisions to cease ineffective treatment and select alternative treatment options, whereas pathological response can only be assessed after completion of surgical resection; (2) twelve of 15 included studies in which either multivariate Cox regression analyses or subgroup analyses were performed reported the metabolic response as having independent prognostic significance to pathological response [13,14,15,16,17, 19, 22,23,24,25,26,27,28,29, 31]. We could not pool HRs from multivariate Cox regression in the meta-analysis because variables included in the models differed widely across the studies that would directly affect the values of HR; (3) the results of our meta-regression indicated the pathological complete response did not influence the pooled HR, which may indirectly support the independent prognostic role of the metabolic response.

Upon thorough inspection of the clinical characteristics of the included studies, our study population mainly consisted of Stage II–III breast cancer patients, which was consistent with the types of patients who typically receive NAC [33]. We also found higher proportions of patients with HER2+ and triple-negative subtypes in the included studies (HER2+ regardless of hormonal receptor status: 26 [402/1558]; and TN: 28 [374/1357]) compared to general breast cancer population referred to cancer statistics published by the US National Cancer Institute: the prevalence of the HER2+ and triple-negative subtypes was 14% and 10%, respectively [40]. HER2+ or triple-negative subtypes are aggressive subtypes, with patients typically presenting with higher FDG uptake at baseline. Therefore, these subtypes of breast cancer are promising targets for evaluation of the metabolic response using 18F-FDG PET or PET/CT [33]. In addition, our analyses indicated that a metabolic response on interim PET also has prognostic significance in the ER+/HER2− group [18, 20], a subtype in which MRI is of limited utility for evaluation of patient responses to NAC [4, 5].

The studies included in our qualitative synthesis varied widely in terms of PET timing, and PET criteria for defining a metabolic response. %ΔSUVmax, the percent reduction of SUVmax between the baseline and interim or post-treatment PET scans, is the most frequently evaluated parameter and is associated with disease recurrence and survival. This ‘ratio’ has the advantage that it may offset the potential effect of noise, reconstruction, image sampling, and smoothing on SUVmax, as long as the PET scans at baseline and during or after NAC are performed using the same machine and protocols; otherwise, it may limit the applicability of results across PET facilities [41].

There were a comparable number of studies and prognostic significance regarding interim vs. post-treatment PET. As it can allow early response evaluation and subsequent modification in treatment, interim PET scans may have better clinical values than post-treatment PET. Regarding specific timing of interim PET, there were no apparent differences between the prognostic values of interim PET assessments performed at different times during NAC; however, the number of studies was insufficient to assess statistical significance. We found that in the majority of studies addressing interim PET scans the response was assessed after 1–2 cycles of NAC, and evidence of their prognostic value was found. Moreover, the better predictive values for pathological response when performing PET scans after 1–2 cycles of NAC (compared to after 3 cycles of NAC) were reported in previous meta-analyses [7, 10]. Given early assessment of response to NAC is important for timely modification of the therapeutic strategy, it might be advisable for interim PET to be performed after 1–2 cycles.

There were several limitations of our study. First, a substantial portion of the included studies were performed retrospectively. Second, there was considerable heterogeneity of hormonal subtype of tumour, PET scan timing, and response parameters among the studies. Therefore, caution is required when considering the applicability of our pooled estimates. Third, approximately half of the included studies used data-dependent cut-off values for the assessment of PET parameters (i.e., optimal cut-off on receiver operating characteristics analysis for predicting pathological response) which may overestimate the prognostic values of 18F-FDG PET or PET/CT. Fourth, the number of studies included for meta-analysis of OS was small, though the pooled HR was statistically significant. However, DFS was regarded as a valid surrogate for OS which requires long-term follow-up for the assessment of efficacy [42].

Conclusions

A metabolic response to NAC as detected by 18F-FDG PET or PET/CT is a significant prognostic factor in terms of DFS and OS. Meta-analytically pooled HRs for DFS nor OS were not significantly different for interim or post-treatment PET scans. %ΔSUVmax, defined as the percent reduction of SUVmax compared with that obtained from the baseline PET, is the most widely evaluated PET response parameter. For the interim assessment of patient responses to NAC, PET scans were commonly performed after 1–2 cycles of NAC and provided significant prognostic values. Evaluation of the metabolic response to NAC may be helpful not only in HER2+ or triple-negative subtypes which are known to be FDG-avid, but also in hormone receptor-positive tumours. These results suggest that 18F-FDG PET or PET/CT may provide accurate risk stratification of breast cancer patients and support risk-adapted therapeutic management based on metabolic response in clinical practice or trials.

Availability of data and materials

Not applicable (The current study was performed based on published literature and no datasets were generated.)

References

  1. Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, Blair SL, Burstein HJ. Breast cancer, version 4.2020, NCCN clinical practice guidelines in oncology; 2020.

    Google Scholar 

  2. Fisher B, Bryant J, Wolmark N, Mamounas E, Brown A, Fisher ER, Wickerham DL, Begovic M, DeCillis A, Robidoux A, et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol. 1998;16(8):2672–85.

    Article  CAS  PubMed  Google Scholar 

  3. Hylton NM, Gatsonis CA, Rosen MA, Lehman CD, Newitt DC, Partridge SC, Bernreuter WK, Pisano ED, Morris EA, Weatherall PT, et al. Neoadjuvant chemotherapy for breast cancer: functional tumor volume by MR imaging predicts recurrence-free survival-results from the ACRIN 6657/CALGB 150007 I-SPY 1 TRIAL. Radiology. 2016;279(1):44–55.

    Article  PubMed  Google Scholar 

  4. McGuire KP, Toro-Burguete J, Dang H, Young J, Soran A, Zuley M, Bhargava R, Bonaventura M, Johnson R, Ahrendt G. MRI staging after neoadjuvant chemotherapy for breast cancer: does tumor biology affect accuracy? Ann Surg Oncol. 2011;18(11):3149–54.

    Article  PubMed  Google Scholar 

  5. Loo CE, Straver ME, Rodenhuis S, Muller SH, Wesseling J, Vrancken Peeters MJ, Gilhuijs KG. Magnetic resonance imaging response monitoring of breast cancer during neoadjuvant chemotherapy: relevance of breast cancer subtype. J Clin Oncol. 2011;29(6):660–6.

    Article  PubMed  Google Scholar 

  6. Wahl RL, Zasadny K, Helvie M, Hutchins GD, Weber B, Cody R: Metabolic monitoring of breast cancer chemohormonotherapy using positron emission tomography: initial evaluation 1993, 11(11):2101–2111.

  7. Tian F, Shen G, Deng Y, Diao W, Jia Z. The accuracy of (18)F-FDG PET/CT in predicting the pathological response to neoadjuvant chemotherapy in patients with breast cancer: a meta-analysis and systematic review. Eur Radiol. 2017;27(11):4786–96.

    Article  PubMed  Google Scholar 

  8. Mghanga FP, Lan X, Bakari KH, Li C, Zhang Y. Fluorine-18 fluorodeoxyglucose positron emission tomography-computed tomography in monitoring the response of breast cancer to neoadjuvant chemotherapy: a meta-analysis. Clin Breast Cancer. 2013;13(4):271–9.

    Article  PubMed  Google Scholar 

  9. Liu Q, Wang C, Li P, Liu J, Huang G, Song S. The role of (18)F-FDG PET/CT and MRI in assessing pathological complete response to neoadjuvant chemotherapy in patients with breast cancer: a systematic review and meta-analysis. Biomed Res Int. 2016;2016:3746232.

    PubMed  PubMed Central  Google Scholar 

  10. Wang Y, Zhang C, Liu J, Huang G. Is 18F-FDG PET accurate to predict neoadjuvant therapy response in breast cancer? A meta-analysis. Breast Cancer Res Treat. 2012;131(2):357–69.

    Article  CAS  PubMed  Google Scholar 

  11. Chen L, Yang Q, Bao J, Liu D, Huang X, Wang J. Direct comparison of PET/CT and MRI to predict the pathological response to neoadjuvant chemotherapy in breast cancer: a meta-analysis. Sci Rep. 2017;7(1):8479.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Akimoto E, Kadoya T, Kajitani K, Emi A, Shigematsu H, Ohara M, Masumoto N, Okada M. Role of (18)F-PET/CT in predicting prognosis of patients with breast cancer after neoadjuvant chemotherapy. Clin Breast Cancer. 2018;18(1):45–52.

    Article  PubMed  Google Scholar 

  13. Champion L, Lerebours F, Alberini JL, Fourme E, Gontier E, Bertrand F, Wartski M. 18F-FDG PET/CT to predict response to neoadjuvant chemotherapy and prognosis in inflammatory breast cancer. J Nucl Med. 2015;56(9):1315–21.

    Article  CAS  PubMed  Google Scholar 

  14. Chen S, Ibrahim NK, Yan Y, Wong ST, Wang H, Wong FC. Complete metabolic response on interim (18)F-fluorodeoxyglucose positron emission tomography/computed tomography to predict long-term survival in patients with breast cancer undergoing neoadjuvant chemotherapy. Oncologist. 2017;22(5):526–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dunnwald LK, Doot RK, Specht JM, Gralow JR, Ellis GK, Livingston RB, Linden HM, Gadi VK, Kurland BF, Schubert EK, et al. PET tumor metabolism in locally advanced breast cancer patients undergoing neoadjuvant chemotherapy: value of static versus kinetic measures of fluorodeoxyglucose uptake. Clin Cancer Res. 2011;17(8):2400–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Emmering J, Krak NC, Van der Hoeven JJ, Spreeuwenberg MD, Twisk JW, Meijer S, Pinedo HM, Hoekstra OS. Preoperative [18F] FDG-PET after chemotherapy in locally advanced breast cancer: prognostic value as compared with histopathology. Ann Oncol. 2008;19(9):1573–7.

    Article  CAS  PubMed  Google Scholar 

  17. Garcia Vicente AM, Amo-Salas M, Relea Calatayud F, Munoz Sanchez Mdel M, Pena Pardo FJ, Jimenez Londono GA, Alvarez Cabellos R, Espinosa Aunion R, Soriano Castrejon A. Prognostic role of early and end-of-neoadjuvant treatment 18F-FDG PET/CT in patients with breast cancer. Clin Nucl Med. 2016;41(7):e313–22.

    Article  PubMed  Google Scholar 

  18. Groheux D, Sanna A, Majdoub M, de Cremoux P, Giacchetti S, Teixeira L, Espie M, Merlet P, de Roquancourt A, Visvikis D, et al. Baseline tumor 18F-FDG uptake and modifications after 2 cycles of neoadjuvant chemotherapy are prognostic of outcome in ER+/HER2- breast cancer. J Nucl Med. 2015;56(6):824–31.

    Article  CAS  PubMed  Google Scholar 

  19. Groheux D, Biard L, Giacchetti S, Teixeira L, Hindie E, Cuvier C, Vercellino L, Merlet P, de Roquancourt A, de Cremoux P et al: (1)(8)F-FDG PET/CT for the early evaluation of response to neoadjuvant treatment in triple-negative breast cancer: influence of the chemotherapy regimen. J Nucl Med 2016, 57(4):536–543.

  20. Humbert O, Berriolo-Riedinger A, Cochet A, Gauthier M, Charon-Barra C, Guiu S, Desmoulins I, Toubeau M, Dygai-Cochet I, Coutant C, et al. Prognostic relevance at 5 years of the early monitoring of neoadjuvant chemotherapy using (18)F-FDG PET in luminal HER2-negative breast cancer. Eur J Nucl Med Mol Imaging. 2014;41(3):416–27.

    Article  CAS  PubMed  Google Scholar 

  21. Humbert O, Riedinger JM, Vrigneaud JM, Kanoun S, Dygai-Cochet I, Berriolo-Riedinger A, Toubeau M, Depardon E, Lassere M, Tisserand S, et al. 18F-FDG PET-derived tumor blood flow changes after 1 cycle of neoadjuvant chemotherapy predicts outcome in triple-negative breast cancer. J Nucl Med. 2016;57(11):1707–12.

    Article  CAS  PubMed  Google Scholar 

  22. Hyun SH, Ahn HK, Park YH, Im YH, Kil WH, Lee JE, Nam SJ, Cho EY, Choi JY. Volume-based metabolic tumor response to neoadjuvant chemotherapy is associated with an increased risk of recurrence in breast cancer. Radiology. 2015;275(1):235–44.

    Article  PubMed  Google Scholar 

  23. Ishiba T, Nakagawa T, Sato T, Nagahara M, Oda G, Sugimoto H, Kasahara M, Hosoya T, Kubota K, Fujioka T, et al. Efficiency of fluorodeoxyglucose positron emission tomography/computed tomography to predict prognosis in breast cancer patients received neoadjuvant chemotherapy. Springerplus. 2015;4:817.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Jung SY, Kim SK, Nam BH, Min SY, Lee SJ, Park C, Kwon Y, Kim EA, Ko KL, Park IH, et al. Prognostic impact of [18F] FDG-PET in operable breast cancer treated with neoadjuvant chemotherapy. Ann Surg Oncol. 2010;17(1):247–53.

    Article  PubMed  Google Scholar 

  25. Kim TH, Yoon JK, Kang DK, Kang SY, Jung YS, Han S, Kim JY, Yim H, An YS. Value of volume-based metabolic parameters for predicting survival in breast cancer patients treated with neoadjuvant chemotherapy. Medicine (Baltimore). 2016;95(41):e4605.

    Article  CAS  Google Scholar 

  26. Kitajima K, Nakatani K, Yamaguchi K, Nakajo M, Tani A, Ishibashi M, Hosoya K, Morita T, Kinoshita T, Kaida H, et al. Response to neoadjuvant chemotherapy for breast cancer judged by PERCIST - multicenter study in Japan. Eur J Nucl Med Mol Imaging. 2018;45(10):1661–71.

    Article  PubMed  Google Scholar 

  27. Kiyoto S, Sugawara Y, Hosokawa K, Nishimura R, Yamashita N, Ohsumi S, Mochizuki T. Predictive ability of (18)F-fluorodeoxyglucose positron emission tomography/computed tomography for pathological complete response and prognosis after neoadjuvant chemotherapy in triple-negative breast cancer patients. Asia Oceania J Nuclear Med Biology. 2016;4(1):3–11.

    Google Scholar 

  28. Kolesnikov-Gauthier H, Vanlemmens L, Baranzelli MC, Vennin P, Servent V, Fournier C, Carpentier P, Bonneterre J. Predictive value of neoadjuvant chemotherapy failure in breast cancer using FDG-PET after the first course. Breast Cancer Res Treat. 2012;131(2):517–25.

    Article  CAS  PubMed  Google Scholar 

  29. Lee HW, Lee HM, Choi SE, Yoo H, Ahn SG, Lee MK, Jeong J, Jung WH. The prognostic impact of early change in 18F-FDG PET SUV after neoadjuvant chemotherapy in patients with locally advanced breast cancer. J Nucl Med. 2016;57(8):1183–8.

    Article  CAS  PubMed  Google Scholar 

  30. Lian W, Liu C, Gu B, Zhang J, Lu L, Pan H, Yao Z, Wang M, Song S, Zhang Y, et al. The early prediction of pathological response to neoadjuvant chemotherapy and prognosis: comparison of PET response criteria in solid tumors and European Organization for Research and Treatment of Cancer criteria in breast cancer. Nucl Med Commun. 2020;41(3):280–7.

    Article  PubMed  Google Scholar 

  31. Lim I, Noh WC, Park J, Park JA, Kim HA, Kim EK, Park KW, Lee SS, You EY, Kim KM, et al. The combination of FDG PET and dynamic contrast-enhanced MRI improves the prediction of disease-free survival in patients with advanced breast cancer after the first cycle of neoadjuvant chemotherapy. Eur J Nucl Med Mol Imaging. 2014;41(10):1852–60.

    Article  CAS  PubMed  Google Scholar 

  32. Zucchini G, Quercia S, Zamagni C, Santini D, Taffurelli M, Fanti S, Martoni AA. Potential utility of early metabolic response by 18F-2-fluoro-2-deoxy-D-glucose-positron emission tomography/computed tomography in a selected group of breast cancer patients receiving preoperative chemotherapy. Eur J Cancer. 2013;49(7):1539–45.

    Article  CAS  PubMed  Google Scholar 

  33. Groheux D, Mankoff D, Espié M, Hindié E. 18F-FDG PET/CT in the early prediction of pathological response in aggressive subtypes of breast cancer: review of the literature and recommendations for use in clinical trials. Eur J Nucl Med Mol Imaging. 2016;43(5):983–93.

    Article  CAS  PubMed  Google Scholar 

  34. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Hayden JA, van der Windt DA, Cartwright JL, Cote P, Bombardier C. Assessing bias in studies of prognostic factors. Ann Intern Med. 2013;158(4):280–6.

    Article  PubMed  Google Scholar 

  36. Guyot P, Ades AE, Ouwens MJ, Welton NJ. Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan-Meier survival curves. BMC Med Res Methodol. 2012;12:9.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ (Clinical research ed). 2003;327(7414):557–60.

    Article  Google Scholar 

  38. Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ (Clinical research ed). 1997;315(7109):629–34.

    Article  CAS  Google Scholar 

  39. von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, Eiermann W, Hilfrich J, Huober J, et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol. 2012;30(15):1796–804.

    Article  Google Scholar 

  40. SEER Cancer Stat Facts: Female breast cancer subtypes. National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/statfacts/html/breast-subtypes.html.

  41. Soret M, Bacharach SL, Buvat I. Partial-volume effect in PET tumor imaging. J Nucl Med. 2007;48(6):932–45.

    Article  PubMed  Google Scholar 

  42. Hirai T, Nemoto A, Ito Y, Matsuura M. Meta-analyses on progression-free survival as a surrogate endpoint for overall survival in triple-negative breast cancer. Breast Cancer Res Treat. 2020;181(1):189–98.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (Ministry of Science and ICT) (No. NRF-2020M2D9A1094072), which had no role in study design, data collection, analysis, interpretation, or writing of the report. No conflicts of interest for all authors.

Author information

Authors and Affiliations

Authors

Contributions

SH and JYC have contributed to study concept and design, data acquisition, data analysis and interpretation, drafting and critical revision of the manuscript. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Joon Young Choi.

Ethics declarations

Ethics approval and consent to participate

This article does not contain any studies with human participants or animals performed by any of the authors.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Supplementary information

Additional file 1.

The queries and results of electronic searches of the PubMed, Embase, and Cochrane Library databases. We provided queries and results of electronic searches of the PubMed, Embase, and Cochrane Library databases as tables.

Additional file 2: Figure S1.

Forest plots of studies assessing the HRs of %ΔSUVmax at the interim evaluation for disease-free survival in Stage II–III breast cancer. We provided forest plots supporting the pooled HRs for the influence of %ΔSUVmax on disease-free survival in stage II–III breast cancer patients at the interim evaluation. (PPTX 78 kb)

Additional file 3: Table S1.

Meta-regression analyses of nine studies where hazard ratios for the influence of %ΔSUVmax on disease-free survival at interim PET scan were available. We provided detailed results of meta-regression analyses including regression coefficients and P values.

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

Han, S., Choi, J.Y. Prognostic value of 18F-FDG PET and PET/CT for assessment of treatment response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis. Breast Cancer Res 22, 119 (2020). https://0-doi-org.brum.beds.ac.uk/10.1186/s13058-020-01350-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13058-020-01350-2

Keywords