Prognostic value of 18F-FDG PET/CT tumor metabolic parameters and Ki-67 in pre-treatment diffuse large B-cell lymphoma
Original Article

Prognostic value of 18F-FDG PET/CT tumor metabolic parameters and Ki-67 in pre-treatment diffuse large B-cell lymphoma

Huixia Geng1,2, Ke Lian2,3, Wanchun Zhang1,2

1Department of Nuclear Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China; 2Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; 3Department of Lymphoma, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China

Contributions: (I) Conception and design: H Geng, W Zhang; (II) Administrative support: W Zhang; (III) Provision of study materials or patients: H Geng, K Lian; (IV) Collection and assembly of data: H Geng, K Lian; (V) Data analysis and interpretation: H Geng, W Zhang; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Wanchun Zhang, MD. Department of Nuclear Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Longcheng Street No. 99, Taiyuan 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. Email: zhang_wanchun@126.com.

Background: Diffuse large B-cell lymphoma (DLBCL) is a highly aggressive lymphoma. Rituximab-based conventional chemotherapy still leads to drug resistance or relapse in 30–40% of patients. Therefore, early identification of high-risk patients and accurate assessment of prognosis are very important for clinical decision-making. The aim of this study is to investigate the value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) of tumor metabolic, clinical and biological parameters in the prognostic risk stratification of DLBCL before treatment.

Methods: We retrospectively collected clinical data on 63 patients with newly diagnosed DLBCL admitted to Shanxi Bethune Hospital during the period from November 2016 to April 2020 who underwent 18F-FDG PET/CT prior to treatment in a cohort study. Metabolic, clinical, and biological parameters were analyzed by Cox regression. Kaplan-Meier curves of patient survival were compared by the log-rank test.

Results: The median follow-up was 21 months. The 2-year progression-free survival (PFS) was 47.62%, and the overall survival (OS) was 53.97%. The subtype, double expression, Ann Abor stage, NCCN-IPI score, Ki-67, maximum standardized uptake value (SUVmax), bulk volume glycolysis (BVG), total lesion glycolysis (TLG), total metabolic tumor volume (TMTV) were the influencing factors for PFS and OS (P<0.050) in univariate analysis. BVG (PFS: HR =6.62, P<0.001; OS: HR =3.53, P=0.029), TLG (PFS: HR =8.56, P<0.001; OS: HR =5.20, P=0.004), TMTV (PFS: HR =12.02, P=0.001; OS: HR =5.05, P=0.033) and Ki-67 were found to be independent prognostic risk stratification parameters affecting PFS and OS by multivariate regression analysis. The 2-year PFS and OS rates for patients with high BVG (≥288.00 cm3), TLG (≥1,854.00 cm3), TMTV (≥103.00 cm3), and Ki-67 (≥85%) were 20% and 28.57%, 9.68% and 22.58%, 20.51%, and 30.77%, and 25% and 33.33%, respectively; and the 2-year PFS and OS rates for patients with low BVG (<288.00 cm3), TLG (<1,854.00 cm3), TMTV (<103.00 cm3), and Ki-67 (<85%) patients were 82.14% and 85.71%, 84.37% and 84.37%, 91.67% and 91.67%, and 61.54% and 66.67%, respectively. Patients with high BVG, TLG, TMTV, and Ki-67 had a worse 2-year PFS as well as OS rate (Ki-67: P=0.0018/P=0.0025; P<0.0001 for the rest of the groups).

Conclusions: Our findings suggest that BVG, TLG, TMTV, and Ki-67 are independent prognostic indicators for survival in patients with pre-treatment DLBCL, especially BVG, which is a novel prognostic indicator that has to be validated in future research.

Keywords: Diffuse large B-cell lymphoma (DLBCL); 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT); prognosis


Submitted May 19, 2023. Accepted for publication Oct 19, 2023. Published online Nov 30, 2023.

doi: 10.21037/qims-23-702


Introduction

Diffuse large B-cell lymphoma (DLBCL) is a prevalent lymphoma, contributing to about one-third of non-Hodgkin’s lymphomas (NHL) (1). Although the combination of rituximab (R) and cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) in the chemotherapy regimen of diffuse large B patients have increased patient curability (2), drug resistance or relapse still occurs in about 30–40% of patients (3). Therefore, the prognosis should be accurately evaluated before conventional treatment, and the high-risk patients should be identified early. It is of great value in determining clinical treatment options.

The international prognostic index (IPI) (4), the subsequent revision of IPI (5), and the emergence of National Comprehensive Cancer Network IPI (NCCN-IPI) (6) in recent years have provided risk stratification for NHL. However, adverse outcomes have yet to be found in a sufficient number of patients (7). Other prognostic factors, such as the germinal center B-cell (GCB) and non-GCB (8) or dual expression (DE) of MYC and BCL2 (9,10) and Ki-67 (11), have been recently identified, but their value in guiding treatment decisions is still debatable. Therefore, new strategies are urgently needed to better evaluate the prognosis of DLBCL.

18F-deoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) has become a routine tool for staging, restaging, and monitoring the development of lymphomas (12,13). In recent years, the value of PET/CT metabolic indicators in predicting the risk of lymphoma has attracted a growing amount of attention due to advancements in software programs and image-processing techniques. The metabolic parameters of maximum standardized uptake value (SUVmax), total metabolic tumor volume (TMTV) and total lesion glycolysis (TLG) are increasingly being used in tumor prognostic studies (14,15). However, metabolic parameters as pre-treatment predictors in DLBCL are less well studied, and their results are inconsistent (16-20). Consequently, additional studies are required to assess the predictive value of PET/CT metabolic parameters for risk stratification of pre-treatment DLBCL.

Lymphomatous lesions greater than 6 to 10 cm have been regarded as worse prognostic factors (21,22), but their value in DLBCL remains uncertain. It has been reported that 3D measurement of the metabolic bulk volume (MBV) in 18F-FDG PET/CT may be associated with poor prognosis (23). However, bulk volume glycolysis (BVG), which combines the volume and metabolic parameters of the largest lesion on PET/CT images is more representative of or close to the actual tumor burden. Yet, it has not been researched to the best of our knowledge.

The purpose of this study is to investigate the prognostic value of 18F-FDG PET/CT of tumor metabolic, clinical, and biological parameters in pre-treatment DLBCL. We present this article in accordance with the STROBE reporting checklist (available at https://qims.amegroups.com/article/view/10.21037/qims-23-702/rc).


Methods

Patients

We retrospectively analyzed 63 patients with DLBCL admitted to Shanxi Bethune Hospital during the period from November 2016 to April 2020 in a cohort study. Inclusion criteria were: (I) newly diagnosed adult DLBCL; (II) PET/CT examination prior to the therapy; and (III) R-CHOP and similar therapeutic regimens; exclusion criteria were: patients with central nerve lymphoma or secondary tumors, history of surgery, chemotherapy, radiotherapy, or incomplete follow-up or therapy (Figure 1). The following indicators were collected from medical records: age, gender, NCCN-IPI level, Ann Abor stage, DLBCL subtypes (GCB and non-GCB) (24), DE of BCL2 and MYC protein, and Ki-67. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Ethics Committee of Shanxi Bethune Hospital (No. YXLL-2022-146) and informed consent was taken from all the patients.

Figure 1 Flow chart of patient selection.

Image acquisition and analysis

All images were acquired using a PET/CT scanner [Discovery VCT 64 (GE Healthcare, Milwaukee, Wisconsin, USA)]. Patients were required to fast for at least 6 h and have a blood glucose level of less than 200 mg/dL prior to drug injection. After intravenous 18F-FDG (4.44 MBq/kg), patients were instructed to rest for 50–60 min before the examination. PET reconstructed images based on CT attenuation correction and ordered subset expectation maximum (OSEM) algorithm.

On an advanced workstation (GE ADW 4.6), CT and PET images were displayed independently and in infusion mode in axial, coronal, and sagittal planes. Abnormal lesions were determined by consensus between two experienced nuclear medicine physicians who were blinded to patient outcomes. PET/CT tumor metabolic parameters were calculated using PETVCAR software (GE ADW 4.6). The boundaries of the tumor were automatically generated using the 41% threshold recommended by the European Association of Nuclear Medicine (25). SUVmax and MTV were calculated using the software. BVG was defined as the metabolic value of the lesion with the largest volume on 18F-FDG PET/CT (MTV × SUVmean of the largest lesion). TLG was the sum of MTV × SUVmean of all lesions.

Statistical analysis

All statistical analyses were performed using Statistical Package for the Social Sciences Version 26.0 software (SPSS Inc., Chicago, IL, USA) and R 4.0.5 software. The definition of progression-free survival (PFS) was the time between the date of diagnosis and the first relapse, progression, mortality from any cause, or the date of the last follow-up. Overall survival (OS) was defined as the duration between the date of diagnosis and the date of mortality from any cause or last follow-up. Optimal cut off values for SUVmax, BVG, TMTV, TLG, and Ki-67 were sought and evaluated by receiver operating characteristic (ROC) curves. PET metabolic, clinical, and biological parameters that significantly affected prognosis in the univariate regression statistical analysis were subsequently included in the multivariate statistical analysis. The Kaplan-Meier survival curve evaluated the survival status of patients, and the comparison between groups was performed by log-rank test. P<0.05 was considered statistically significant.


Results

Patient characteristics

The clinical, biological, and metabolic parameters of the 63 enrolled participants (24 women and 39 men) are summarized in Table 1. The median age of the participants was 66 years (range, 28–87 years). At a median follow-up of 21 months (range, 3–87 months), 33 patients experienced disease progression, and 29 died. The 2-year PFS and OS were 47.62% and 53.97%, respectively.

Table 1

Patient characteristics

Characteristic Value (n=63)
Age (years)
   Mean (SD) 62.86 (13.75)
   Median (P25, P75) 66.00 (55.00, 71.00)
   >60, n (%) 40 (63.49)
   ≤60, n (%) 23 (36.51)
Sex, n (%)
   Male 39 (61.90)
   Female 24 (38.10)
Subtype, n (%)
   GCB 20 (31.75)
   Non-GCB 43 (68.25)
Double expression, n (%)
   Yes 25 (39.68)
   No 38 (58.73)
Ann-Abor stage, n (%)
   I–II 22 (34.92)
   III–IV 41 (65.08)
NCCN-IPI score, n (%)
   0–3 24 (38.10)
   ≥4 39 (61.90)
SUVmax (g/mL)
   Mean (SD) 23.51 (11.32)
   Median (P25, P75) 23.25 (14.28, 32.40)
BVG (cm3)
   Mean (SD) 1,683.57 (2,611.56)
   Median (P25, P75) 392.00 (64.00, 1,718.00)
TLG (cm3)
   Mean (SD) 3,888.76 (4,753.77)
   Median (P25, P75) 1,749.00 (168.00, 7,658.00)
TMTV (cm3)
   Mean (SD) 376.48 (577.17)
   Median (P25, P75) 152.00 (17.00, 470.00)
Ki-67 (%)
   Mean (SD) 74.68 (15.73)
   Median (P25, P75) 80.00 (60.00, 90.00)

GCB, germinal center B-cell; SD, standard deviation; NCCN-IPI, National Comprehensive Cancer Network International Prognostic Index; SUVmax, standardized maximum uptake value; BVG, bulk volume glycolysis; TLG, total lesion glycolysis; TMTV, total tumor metabolic volume.

ROC analysis

The best cut-off values of SUVmax, BVG, TMTV, TLG, and Ki-67 for PFS were 19.41 g/mL, 288.00 cm3, 103.00 cm3, 1,854.00 cm3, and 85%, respectively, based on the analysis of the ROC curves. For clinicians, the OS of a patient is more important, and hence the OS cut-off values were selected for subsequent analysis (Table S1).

Univariate and multivariate analyses of prognostic risk factors

Univariate analysis showed that double expression subtype, NCCN-IPI score, Ann-Abor stage, Ki-67, SUVmax, BVG, TLG, and TMTV were predictive factors for PFS and OS (P<0.05) (Table 2). Statistical analysis of Spearman’s rank correlation showed that BVG, TLG, and TMTV were correlated with each other (P<0.001) (Table S2). Therefore, the univariate significant SUVmax, BVG, TLG, and TMTV were analyzed separately using multivariate Cox regression analysis (Table 3). The results revealed that BVG (PFS: HR =6.62, 95% CI: 2.50–17.56, P<0.001; OS: HR =3.53, 95% CI: 1.13–11.01, P=0.029), TLG (PFS: HR =8.56, 95% CI: 3.18–23.03, P<0.001; OS: HR =5.20, 95% CI: 1.71–15.76, P=0.004), TMTV (PFS: HR =12.02, 95% CI: 2.72–53.13, P=0.001; OS: HR =5.05, 95% CI: 1.14–22.40, P=0.033) and Ki-67 were found to be independent prognostic risk stratification parameters affecting PFS and OS by multivariate regression analysis.

Table 2

Univariate Cox proportional hazard regression analysis

Characteristic N PFS OS
HR (95% CI) P value HR (95% CI) P value
Age (≤60/>60 years) 23/40 2.64 (1.14–6.08) 0.023 2.12 (0.90–4.97) 0.085
Sex (male/female) 39/24 1.04 (0.51–2.11) 0.924 1.23 (0.59–2.58) 0.582
Subtype (non-GCB/GCB) 43/20 0.21 (0.08–0.61) 0.004 0.25 (0.09-0.73) 0.011
Double expression (no/yes) 38/25 2.70 (1.35–5.38) 0.005 2.85 (1.36–5.96) 0.005
Ann Abor stage (I–II/III–IV) 22/41 8.72 (2.65–28.68) <0.001 10.20 (2.42–42.98) 0.002
NCCN-IPI score (0–3/≥4) 24/39 9.68 (2.94–31.90) <0.001 29.18 (3.94–216.24) 0.001
Ki-67 (<85%/≥85%) 39/24 2.83 (1.42–5.64) 0.003 2.96 (1.42–6.18) 0.004
SUVmax (<19.41/≥19.41 g/mL) 23/40 2.43 (1.09–5.39) 0.030 3.32 (1.34–8.22) 0.009
BVG (<288.00/≥288.00 cm3) 28/35 8.03 (3.07–21.04) <0.001 8.96 (3.06–26.22) <0.001
TLG (<1,854.00/≥1,854.00 cm3) 32/31 13.96 (5.26–37.00) <0.001 10.32 (3.79–28.14) <0.001
TMTV (<103.00/≥103.00 cm3) 24/39 18.23 (4.33–76.83) <0.001 14.49 (3.40–61.69) <0.001

PFS, progression-free survival; OS, overall survival; HR, hazard ratio; CI, confidence interval; GCB, germinal centre B cells; Double expression, dual expression of MYC and BCL2; NCCN-IPI, National Comprehensive Cancer Network International Prognostic Index; SUVmax, maximum standardised uptake value; BVG, metabolic volume; TLG, total lesion glycation; TMTV, total metabolic tumor volume.

Table 3

Multivariate Cox proportional hazard regression analysis

Characteristic N PFS OS
HR (95% CI) P value HR (95% CI) P value
Model 1
   SUVmax (<19.41/≥19.41 g/mL) 23/40 0.82 (0.30–2.22) 0.693 1.58 (0.56–4.46) 0.392
   NCCN-IPI score (0–3/≥4) 24/39 4.77 (1.36–16.65) 0.014 18.97 (2.48–145.40) 0.005
   Ki-67 (<85%/≥85%) 39/24 2.81 (1.39–5.66) 0.004 3.71 (1.72–8.03) 0.001
   Ann-Abor stage (I–II/III–IV) 22/41 3.84 (1.01–13.35) 0.035
Model 2
   BVG (<288.00/≥288.00 cm3) 28/35 6.62 (2.50–17.56) <0.001 3.53 (1.13–11.01) 0.029
   NCCN-IPI score (0–3/≥4) 24/39 9.25 (1.10–77.74) 0.040
   Ki-67 (<85%/≥85%) 39/24 2.57 (1.27–5.20) 0.008 3.45 (1.56–7.63) 0.0002
   Ann-Abor stage (I–II/III–IV) 22/41 6.50 (1.96–21.53) 0.002
   Subtype (non-GCB/GCB) 43/20 3.27 (1.01–10.45) 0.046
Model 3
   TLG (<1,854.00/≥1,854.00 cm3) 32/31 8.56 (3.18–23.03) <0.001 5.20 (1.71–15.76) 0.004
   NCCN-IPI score (0–3/≥4) 24/39 12.53 (1.63–96.39) 0.015
   Ki-67 (<85%/≥85%) 39/24 2.13 (1.05–4.33) 0.036 3.39 (1.56–7.39) 0.002
   Ann-Abor stage (I–II/III–IV) 22/41 4.80 (1.42–16.21) 0.012 4.38 (1.02–18.86) 0.047
Model 4
   TMTV (<103.00/≥103.00 cm3) 24/39 12.02 (2.72–53.13) 0.001 5.05 (1.14–22.40) 0.033
   NCCN-IPI score (0–3/≥4) 24/39 12.16 (1.57–94.01) 0.017
   Ki-67 (<85%/≥85%) 39/24 3.11 (1.53–6.32) 0.002 4.18 (1.89–9.27) <0.001
   Subtype (non-GCB/GCB) 43/20 3.77 (1.15–12.36) 0.029

Model 1 included age, subtype, double expression, Ann Abor stage, NCCN-IPI score, Ki-67 and SUVmax; Model 2 included age, subtype, double expression, Ann Abor stage, NCCN-IPI score, Ki-67 and BVG; Model 3 included age, subtype, double expression, Ann Abor stage, NCCN-IPI score, Ki-67 and TLG; Model 4 included age, subtype, double expression, Ann Abor stage, NCCN-IPI score, Ki-67 and TMTV. – indicates that the indicator did not enter the model. PFS, progression-free survival; OS, overall survival; HR, hazard ratio; CI, confidence interval; SUVmax, standardized maximum uptake value; NCCN-IPI, National Comprehensive Cancer Network international prognostic index; BVG, bulk volume glycolysis; GCB, germinal center B-cell; TLG, total lesion glycolysis, TMTV, total metabolic tumor volume.

Survival status analysis

The optimal cut-off values of BVG, TLG, TMTV, and Ki-67 were analyzed by Kaplan-Meier curves and compared using log-rank tests. The 2-year PFS and OS rates for patients with high BVG (≥288.00 cm3, n=35) were 20% and 28.57%, respectively, compared with low BVG (<288.00 cm3, n=28) 82.14% and 85.71% (c2=25.3/c2=22.7, both P<0.0001). The 2-year PFS and OS rates for patients with high TLG (≥1,854.00 cm3, n=31) were 9.68% and 22.58%, respectively, compared with 84.37% and 84.37% for low TLG (<1,854.00 cm3, n=32) (c2=45.2/c2=30.0, both P<0.0001). The 2-year PFS and OS rates for patients with high TMTV (≥103.00 cm3, n=39) were 20.51% and 30.77%, respectively, compared with low TMTV (<103.00 cm3, n=24), 91.67% and 91.67% (c2=29.6/c2=25.1, both P<0.0001). The 2-year PFS and OS rates for patients with high Ki-67 (≥85%, n=24) were 25% and 33.33%, respectively, compared with patients with low Ki-67 (<85%, n=39) were 61.54% and 66.67% (c2=9.7/c2=9.2, P=0.0018, P=0.0025, Figure 2).

Figure 2 Kaplan-Meier curves for PFS and OS based on BVG, TLG, TMTV and Ki-67 in all patients. (A) PFS in relation to BVG; (B) OS in relation to BVG; (C) PFS in relation to TLG; (D) OS in relation to TLG; (E) PFS in relation to TMTV; (F) OS in relation to TMTV; (G) PFS in relation to Ki-67; (H) OS in relation to Ki-67. PFS, progression-free survival; OS, overall survival; BVG, bulk volume glycolysis; TLG, total lesion glycolysis; TMTV, total metabolic tumor volume.

Discussion

In this study, we focused on the prognostic value of PET/CT-derived tumor metabolic, clinical, and biological parameters in risk stratification of DLBCL before treatment. We found that BVG, TLG, TMTV, and Ki-67 were found to be independent prognostic risk stratification parameters affecting PFS and OS. To the best of our knowledge, the predictive value of BVG has not been investigated, and these indicators can help clinicians identify high-risk patients early, which is very important for clinical treatment decision-making.

The maximum transverse diameter (MTD) of the largest lesion is considered to be a factor of poor prognosis (21,22,26). However, MTD is a single-dimensional measure, and the MTD cutoff range of 6 to 10 cm in previous studies does not completely represent the tumor burden of the largest lesion, so the results are debatable (16,23,27). In this study, we attempted to incorporate the BVG of the largest lesion, which combines the three-dimensional measured volume of the largest lesion with metabolic values and more accurately reflects the tumor load of large lesions. The present study showed that BVG were found to be independent prognostic risk stratification parameters affecting PFS and OS. Elevated levels of BVG (≥288.00 cm3) correlated with a poorer prognosis and were more difficult to treat compared with low BVG (<288.00 cm3). The relationship between chemotherapy outcomes and pretreatment tumor volume in patients with DLBCL was investigated by Tout et al. (28). They found that as tumor volume increased, it increased the difficulty in chemotherapeutic drug penetration, which may have contributed to the poor prognosis observed in this study.

SUVmax is a widely used metabolic index to reflect tumor invasiveness. Wu et al. discovered that SUVmax could be used to guide DLBCL risk stratification (29), whereas Xie et al. rejected this claim (30). In our study, TLG and TMTV but not SUVmax were found to be independent prognostic risk stratification parameters affecting PFS and OS; patients with high TLG (≥1,854.00 cm3) and high TMTV (≥103.00 cm3) had a poorer prognosis than those with low TLG and low TMTV. This result is consistent with the findings of prior research (31,32). Jiang et al. discovered that TLG ≥1,852 cm3 was associated with an adverse prognosis (8,33), and our study had a similar cut-off value for TLG (≥1,854.00 cm3). However, previous studies only included one of these, and did not consider the correlation between TLG and TMTV (8,33). The advantage of this study is that multivariate regression model was developed separately for each of the associated indicators, and the indicator correction was more comprehensive at the level of available data. TMTV and TLG reflect the total body tumor load from a three-dimensional perspective, whereas SUVmax only reflects the highest metabolic pixel value in the tumor, therefore, TMTV and TLG can be more effective indicators for prognostic assessment in DLBCL.

Ki-67 is a pathologic indicator of cell proliferation, and studies on its prognostic stratification value have been divergent in recent years (34); Huber et al. retrospectively analyzed 58 cases of DLBCL, and they found that OS was significantly worse in those with high Ki-67 (>70%) than low Ki-67 (≤70%), but PFS was not significantly different (11). Gaudio et al. found that Ki-67 expression was more than 80% correlated with OS and PFS (35). In accordance with the findings of Gaudio et al. (35) our findings indicated that Ki-67 was an independent prognostic risk stratification indicator affecting PFS and OS. However, the cut-off values were marginally different, and we found that patients in the Ki-67 (≥85%) group had worse 2-year PFS as well as OS than those in Ki-67 (<85%) group.

Some studies have reported separately that other clinical and biological parameters such as IPI score ≥4, non-GCB cell origin subtype, and dual MYC and BCL2 protein expression are likely correlated with worse survival outcomes (11,36); but the results of this study found that they were associated with PFS and OS in a univariate regression analysis, and a multifactorial regression analysis did not demonstrate independent prognostic value; thus, further studies with larger groups are warranted.

The limitations of this study stem from its retrospective nature, the need to expand the sample size, and the lack of standardization of thresholds for describing tumors, which may have contributed to inconsistent results. We used the criteria recommended by the European Association of Nuclear Medicine as there was better inter-observer agreement (25).


Conclusions

Our study demonstrates that the PET/CT tumor metabolic parameters BVG, TLG, TMTV, and Ki-67 are independent prognostic risk stratification indicators affecting PFS and OS. These indicators can help clinicians identify high-risk patients early and guide intensive treatment and clinical trials. In particular, BVG, a novel prognostic indicator, requires confirmation through prospective studies.


Acknowledgments

We would like to acknowledge the hard and dedicated work of all the staff that implemented the intervention and evaluation components of the study.

Funding: This work was supported by the Shanxi Provincial External Expert Studio.


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://qims.amegroups.com/article/view/10.21037/qims-23-702/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://qims.amegroups.com/article/view/10.21037/qims-23-702/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Ethics Committee of Shanxi Bethune Hospital (No. YXLL-2022-146) and informed consent was taken from all the patients.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Jiang J, Lyu W, Chen N. A bibliometric analysis of diffuse large B-cell lymphoma research from 2001 to 2020. Comput Biol Med 2022;146:105565. [Crossref] [PubMed]
  2. Wang J, Zhang R, Ding X, Jin Y, Qin R, Xia B, Liao Q, Hu H, Song W, Wang Z, Zhang X, Xu J. Pathologically complete remission to combination of invariant NK T cells and anti-CD20 antibody in a refractory HIV+ diffuse large B-cell lymphoma patient. Immunotherapy 2022;14:599-607. [Crossref] [PubMed]
  3. He MY, Kridel R. Treatment resistance in diffuse large B-cell lymphoma. Leukemia 2021;35:2151-65. [Crossref] [PubMed]
  4. A predictive model for aggressive non-Hodgkin's lymphoma. N Engl J Med 1993;329:987-94. [Crossref] [PubMed]
  5. Sehn LH, Berry B, Chhanabhai M, Fitzgerald C, Gill K, Hoskins P, Klasa R, Savage KJ, Shenkier T, Sutherland J, Gascoyne RD, Connors JM. The revised International Prognostic Index (R-IPI) is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 2007;109:1857-61. [Crossref] [PubMed]
  6. Zhou Z, Sehn LH, Rademaker AW, Gordon LI, Lacasce AS, Crosby-Thompson A, Vanderplas A, Zelenetz AD, Abel GA, Rodriguez MA, Nademanee A, Kaminski MS, Czuczman MS, Millenson M, Niland J, Gascoyne RD, Connors JM, Friedberg JW, Winter JN. An enhanced International Prognostic Index (NCCN-IPI) for patients with diffuse large B-cell lymphoma treated in the rituximab era. Blood 2014;123:837-42. [Crossref] [PubMed]
  7. Ruppert AS, Dixon JG, Salles G, Wall A, Cunningham D, Poeschel V, Haioun C, Tilly H, Ghesquieres H, Ziepert M, Flament J, Flowers C, Shi Q, Schmitz N. International prognostic indices in diffuse large B-cell lymphoma: a comparison of IPI, R-IPI, and NCCN-IPI. Blood 2020;135:2041-8. [Crossref] [PubMed]
  8. Jiang C, Teng Y, Zheng Z, Zhou Z, Xu J. Value of total lesion glycolysis and cell-of-origin subtypes for prognostic stratification of diffuse large B-cell lymphoma patients. Quant Imaging Med Surg 2021;11:2509-20. [Crossref] [PubMed]
  9. Augustyn A, Medeiros LJ, Ludmir EB, Gunther J, Fang P, Li S, et al. The impact of cell-of-origin, MYC/Bcl-2 dual expression and MYC rearrangement on disease relapse among early stage diffuse large B-cell lymphoma patients treated with combined modality therapy. Leuk Lymphoma 2021;62:1361-9. [Crossref] [PubMed]
  10. Xu-Monette ZY, Wei L, Fang X, Au Q, Nunns H, Nagy M, et al. Genetic Subtyping and Phenotypic Characterization of the Immune Microenvironment and MYC/BCL2 Double Expression Reveal Heterogeneity in Diffuse Large B-cell Lymphoma. Clin Cancer Res 2022;28:972-83. [Crossref] [PubMed]
  11. Huber F, Zwickl-Traxler E, Pecherstorfer M, Singer J. Evaluation of Ki-67 as a Prognostic Marker in Diffuse Large B-Cell Lymphoma-A Single-Center Retrospective Cohort Study. Curr Oncol 2021;28:4521-9. [Crossref] [PubMed]
  12. Juweid ME, Mueller M, Alhouri A. A-Risheq MZ, Mottaghy FM. Positron emission tomography/computed tomography in the management of Hodgkin and B-cell non-Hodgkin lymphoma: An update. Cancer 2021;127:3727-41. [Crossref] [PubMed]
  13. Zanoni L, Bezzi D, Nanni C, Paccagnella A, Farina A, Broccoli A, Casadei B, Zinzani PL, Fanti S. PET/CT in Non-Hodgkin Lymphoma: An Update. Semin Nucl Med 2023;53:320-51. [Crossref] [PubMed]
  14. Feng X, Wen X, Li L, Sun Z, Li X, Zhang L, Wu J, Fu X, Wang X, Yu H, Ma X, Zhang X, Xie X, Han X, Zhang M. Baseline Total Metabolic Tumor Volume and Total Lesion Glycolysis Measured on 18F-FDG PET-CT Predict Outcomes in T-Cell Lymphoblastic Lymphoma. Cancer Res Treat 2021;53:837-46. [Crossref] [PubMed]
  15. Wan X, Guo W, Wang X, Li J, Zhao Y, Feng X, Young KH, Bai O. Improving the prognostic ability of PET/CT SUVmax to identify follicular lymphoma with early treatment failure. Am J Cancer Res 2022;12:3857-69.
  16. Sasanelli M, Meignan M, Haioun C, Berriolo-Riedinger A, Casasnovas RO, Biggi A, Gallamini A, Siegel BA, Cashen AF, Véra P, Tilly H, Versari A, Itti E. Pretherapy metabolic tumour volume is an independent predictor of outcome in patients with diffuse large B-cell lymphoma. Eur J Nucl Med Mol Imaging 2014;41:2017-22. [Crossref] [PubMed]
  17. Kim TM, Paeng JC, Chun IK, Keam B, Jeon YK, Lee SH, Kim DW, Lee DS, Kim CW, Chung JK, Kim IH, Heo DS. Total lesion glycolysis in positron emission tomography is a better predictor of outcome than the International Prognostic Index for patients with diffuse large B cell lymphoma. Cancer 2013;119:1195-202. [Crossref] [PubMed]
  18. Esfahani SA, Heidari P, Halpern EF, Hochberg EP, Palmer EL, Mahmood U. Baseline total lesion glycolysis measured with (18)F-FDG PET/CT as a predictor of progression-free survival in diffuse large B-cell lymphoma: a pilot study. Am J Nucl Med Mol Imaging 2013;3:272-81.
  19. Islam P, Goldstein J, Flowers CR. PET-derived tumor metrics predict DLBCL response and progression-free survival. Leuk Lymphoma 2019;60:1965-71. [Crossref] [PubMed]
  20. Gallicchio R, Mansueto G, Simeon V, Nardelli A, Guariglia R, Capacchione D, Soscia E, Pedicini P, Gattozzi D, Musto P, Storto G. F-18 FDG PET/CT quantization parameters as predictors of outcome in patients with diffuse large B-cell lymphoma. Eur J Haematol 2014;92:382-9. [Crossref] [PubMed]
  21. Song MK, Chung JS, Sung-Yong O, Lee GW, Kim SG, Seol YM, Shin HJ, Choi YJ, Cho GJ, Shin DH, Yun EY. Clinical impact of bulky mass in the patient with primary extranodal diffuse large B cell lymphoma treated with R-CHOP therapy. Ann Hematol 2010;89:985-91. [Crossref] [PubMed]
  22. Cheson BD, Fisher RI, Barrington SF, Cavalli F, Schwartz LH, Zucca E, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol 2014;32:3059-68. [Crossref] [PubMed]
  23. Delaby G, Hubaut MA, Morschhauser F, Besson A, Huglo D, Herbaux C, Baillet C. Prognostic value of the metabolic bulk volume in patients with diffuse large B-cell lymphoma on baseline (18)F-FDG PET-CT. Leuk Lymphoma 2020;61:1584-91. [Crossref] [PubMed]
  24. Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, Müller-Hermelink HK, Campo E, Braziel RM, Jaffe ES, Pan Z, Farinha P, Smith LM, Falini B, Banham AH, Rosenwald A, Staudt LM, Connors JM, Armitage JO, Chan WC. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 2004;103:275-82. [Crossref] [PubMed]
  25. Boellaard R, Delgado-Bolton R, Oyen WJ, Giammarile F, Tatsch K, Eschner W, et al. FDG PET/CT: EANM procedure guidelines for tumour imaging: version 2.0. Eur J Nucl Med Mol Imaging 2015;42:328-54. [Crossref] [PubMed]
  26. Pfreundschuh M, Ho AD, Cavallin-Stahl E, Wolf M, Pettengell R, Vasova I, Belch A, Walewski J, Zinzani PL, Mingrone W, Kvaloy S, Shpilberg O, Jaeger U, Hansen M, Corrado C, Scheliga A, Loeffler M, Kuhnt E. Prognostic significance of maximum tumour (bulk) diameter in young patients with good-prognosis diffuse large-B-cell lymphoma treated with CHOP-like chemotherapy with or without rituximab: an exploratory analysis of the MabThera International Trial Group (MInT) study. Lancet Oncol 2008;9:435-44. [Crossref] [PubMed]
  27. Mikhaeel NG, Smith D, Dunn JT, Phillips M, Møller H, Fields PA, Wrench D, Barrington SF. Combination of baseline metabolic tumour volume and early response on PET/CT improves progression-free survival prediction in DLBCL. Eur J Nucl Med Mol Imaging 2016;43:1209-19. [Crossref] [PubMed]
  28. Tout M, Casasnovas O, Meignan M, Lamy T, Morschhauser F, Salles G, Gyan E, Haioun C, Mercier M, Feugier P, Boussetta S, Paintaud G, Ternant D, Cartron G. Rituximab exposure is influenced by baseline metabolic tumor volume and predicts outcome of DLBCL patients: a Lymphoma Study Association report. Blood 2017;129:2616-23. [Crossref] [PubMed]
  29. Wu X, Pertovaara H, Korkola P, Vornanen M, Järvenpää R, Dastidar P, Eskola H, Kellokumpu-Lehtinen PL. Early interim PET/CT predicts post-treatment response in diffuse large B-cell lymphoma. Acta Oncol 2014;53:1093-9. [Crossref] [PubMed]
  30. Xie M, Zhai W, Cheng S, Zhang H, Xie Y, He W. Predictive value of F-18 FDG PET/CT quantization parameters for progression-free survival in patients with diffuse large B-cell lymphoma. Hematology 2016;21:99-105. [Crossref] [PubMed]
  31. Zhou M, Chen Y, Huang H, Zhou X, Liu J, Huang G. Prognostic value of total lesion glycolysis of baseline 18F-fluorodeoxyglucose positron emission tomography/computed tomography in diffuse large B-cell lymphoma. Oncotarget 2016;7:83544-53. [Crossref] [PubMed]
  32. Vercellino L, Cottereau AS, Casasnovas O, Tilly H, Feugier P, Chartier L, et al. High total metabolic tumor volume at baseline predicts survival independent of response to therapy. Blood 2020;135:1396-405. [Crossref] [PubMed]
  33. Shagera QA, Cheon GJ, Koh Y, Yoo MY, Kang KW, Lee DS, Kim EE, Yoon SS, Chung JK. Prognostic value of metabolic tumour volume on baseline (18)F-FDG PET/CT in addition to NCCN-IPI in patients with diffuse large B-cell lymphoma: further stratification of the group with a high-risk NCCN-IPI. Eur J Nucl Med Mol Imaging 2019;46:1417-27. [Crossref] [PubMed]
  34. He X, Chen Z, Fu T, Jin X, Yu T, Liang Y, Zhao X, Huang L. Ki-67 is a valuable prognostic predictor of lymphoma but its utility varies in lymphoma subtypes: evidence from a systematic meta-analysis. BMC Cancer 2014;14:153. [Crossref] [PubMed]
  35. Gaudio F, Giordano A, Perrone T, Pastore D, Curci P, Delia M, Napoli A. de' Risi C, Spina A, Ricco R, Liso V, Specchia G. High Ki67 index and bulky disease remain significant adverse prognostic factors in patients with diffuse large B cell lymphoma before and after the introduction of rituximab. Acta Haematol 2011;126:44-51. [Crossref] [PubMed]
  36. Savage KJ, Slack GW, Mottok A, Sehn LH, Villa D, Kansara R, Kridel R, Steidl C, Ennishi D, Tan KL, Ben-Neriah S, Johnson NA, Connors JM, Farinha P, Scott DW, Gascoyne RD. Impact of dual expression of MYC and BCL2 by immunohistochemistry on the risk of CNS relapse in DLBCL. Blood 2016;127:2182-8. [Crossref] [PubMed]
Cite this article as: Geng H, Lian K, Zhang W. Prognostic value of 18F-FDG PET/CT tumor metabolic parameters and Ki-67 in pre-treatment diffuse large B-cell lymphoma. Quant Imaging Med Surg 2024;14(1):325-334. doi: 10.21037/qims-23-702

Download Citation