Skip to main content

SYSTEMATIC REVIEW article

Front. Oncol., 15 December 2022
Sec. Head and Neck Cancer

Survival prognostic factors for differentiated thyroid cancer patients with pulmonary metastases: A systematic review and meta-analysis

Hao ZhaoHao Zhao1Chun-Hao LiuChun-Hao Liu1Yue CaoYue Cao1Li-Yang ZhangLi-Yang Zhang1Ya ZhaoYa Zhao1Yue-Wu LiuYue-Wu Liu1Hong-Feng LiuHong-Feng Liu1Yan-Song Lin,Yan-Song Lin1,2Xiao-Yi Li*Xiao-Yi Li1*
  • 1Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
  • 2Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

Background: The prognostic factors for differentiated thyroid cancer (DTC) patients with pulmonary metastases (PM) remain scantly identified and analyzed. Therefore, this systematic review and meta-analysis were performed to identify and summarize the prognostic factors in adult DTC patients with PM to help distinguish patients with different prognoses and inform the rational treatment regimens.

Method: We performed a comprehensive search of the relevant studies published in the Cochrane Library, PubMed, Scopus, Embase, Wanfang database, VIP database, China National Knowledge Infrastructure, and Google Scholar from their inception until February 2021. The pooled hazard ratios (HR) for overall survival and/or progression-free survival (PFS) with 95% confidence intervals were applied to evaluate and identify the potential prognostic factors. Pooled OS at different time points were also calculated for the available data. A random-effects model was used in the meta-analysis.

Results: The review and meta-analysis included 21 studies comprising 2722 DTC patients with PM. The prognostic factors for poor OS were: age over 40 years (HR=7.21, 95% confidence interval [CI] 1.52-34.10, P=0.01, N=788), age over 45 years (HR=2.18, 95% CI 1.26-3.77, P<0.01, N=601), male gender (HR=1.01, 95% CI 1.01-1.19, P=0.03, N=1396), follicular subtype of thyroid cancer (HR=1.63, 95% CI 1.36-1.96, P<0.01, N=2110), iodine non-avidity (HR=3.10, 95% CI 1.79-5.37, P<0.01, N=646), and metastases to other organs (HR=3.18, 95% CI 2.43-4.16, P<0.01, N=1713). Factors associated with poor PFS included age over 45 years (HR=3.85, 95% CI 1.29-11.47, P<0.01, N=306), male gender (HR=1.36, 95% CI 1.06-1.75, P=0.02, N=546), iodine non-avidity (HR=2.93, 95% CI 2.18-3.95, P<0.01, N=395), pulmonary metastatic nodule size over 10mm (HR=2.56, 95% CI 2.02-3.24, P<0.01, N=513), and extra-thyroidal invasion (HR=2.05, 95% CI 1.15-3.67, P=0.02, N=271). The pooled 1, 3, 5, 10, 15, and 20-years OS were 95.24%, 88.46%, 78.36%, 64.86%, 56.57%, and 51.03%, respectively.

Conclusions: This review and meta-analysis identified the prognostic factors of DTC patients with PM. Notably, FTC, metastases to other organs, and iodine non-avidity were particularly associated with poor prognosis. The identified prognostic factors will help guide the clinical management of DTC patients with PM.

Systematic review registration: https://inplasy.com/inplasy-2022-2-0026/, identifier (INPLASY202220026).

1 Introduction

The past decades have witnessed a rapid increase in the global incidence of thyroid cancer. As the most common endocrine malignancy, thyroid cancer ranks ninth among all cancers in incidence (586,000 cases worldwide), with an increasing trend (1, 2). Over 90% of thyroid cancers are differentiated thyroid cancers (DTC), characterized by an excellent prognosis with a 10-year survival rate exceeding 90% (3, 4). Nevertheless, the survival rate of DTC drops significantly upon the occurrence of distant metastases (DM) (5, 6). The Surveillance, Epidemiology, and End Results (SEER) database reported a 76% and 64% 5-year overall survival (OS) rates for papillary thyroid cancer (PTC) and follicular thyroid cancer (FTC) with DM, respectively (7). DM is, thus, a significant prognostic factor for the survival of DTC patients.

Though DTCs with DM have an overall poor prognosis, there are still significant variations between subgroups. Studies postulate that the 10-year OS rate varies from 10% to 90% for different subgroups (8). For example, the 10-year OS rate for radioiodine avidity patients can reach 55% (911), while non-avidity patients have an OS of 10-18% (9, 12, 13). Meanwhile, treatment strategies for different subgroups, including the management options for localized lesions and optimal time to start systemic treatment like tyrosine kinase inhibitor (TKI) therapy, remain controversial (14). Therefore, identifying the prognostic factors of these patients could help with risk stratification and formulating treatment plans for different subgroups, thereby improving the long-term prognosis.

Distant metastases of DTCs commonly occur in the lungs (85%), followed by the bones (39.9%), while its occurrence in the brain (5.8%) and liver (3.6%) is relatively rare (15). Several studies have identified factors related to DM occurrence in DTCs, including old age, male gender, pathological subtypes, large primary tumor sizes, and extrathyroidal extension (ETE). However, only a few studies have focused on the prognostic factors of DTC patients with DM, especially of DTC patients with pulmonary metastases (PM). Previous studies suggest these factors, together with metastases size, multiorgan metastases, thyroglobulin (Tg) level, BRAFV600E mutation, radioiodine avidity, and TKI administration, among other factors, may affect the prognosis of DM (9, 10, 1530). Deciphering the role and impact of these factors on prognosis is thus crucial in aiding treatment decision-making.

This systematic review and meta-analysis aimed to identify and summarize the prognostic factors in adult DTC patients with PM to help distinguish patients with different prognoses and inform the rational design of treatment regimens.

2 Materials and methods

2.1 Guidelines and protocols

This systematic review and meta-analysis were conducted following the statement (31), and guidelines for systematic reviews and meta-analyses of prognostic factors (32). The protocol used for this systematic review and meta-analysis is registered in INPLASY (https://inplasy.com/) with ID INPLASY202220026.

2.2 Literature search

Two reviewers (Z.H. and L.C.H.) performed independent, comprehensive searches of relevant studies published in the Cochrane Library, PubMed, Scopus, Embase, Wanfang database, VIP database, China National Knowledge Infrastructure (CNKI) from their inception to February 2021 following the registered protocol. We also retrieved citations and references in Google Scholar to ensure no relevant literature was missed. There was no restriction on language and time of publication to limit publication bias. No search was done for the unpublished data and grey literature. Supplementary S1 outlines the detailed search strategy.

2.3 Study selection

We considered randomized clinical trials (RCTs), retrospective and prospective observation cohort studies for inclusion. Inclusion criteria were: studies enrolled mostly adult patients (≥18 years) with a confirmed diagnosis of DTC through histopathology and PM though chest CT, chest x-ray, or 131I whole-body scan (WBS); explored at least one prognostic factor for DTC patients with PM; reported OS and/or progression-free survival (PFS) as an outcome of interest; with an available English abstract and an accessible full text was required. Studies with fewer than 20 cases of DTC patients with PM, those impossible to extract or convert valid data, repeated published literature, case reports, reviews, conference reports, animal experiments, and in vitro cell experiments were excluded.

All the identified citations were imported into Rayyan (rayyan.qcri.org) to assess their eligibility after removing the duplicates. The two reviewers (Z.H. and L.C.H.) subsequently completed the screening process independently. A third reviewer (L.X.Y.) was invited to make the final decision if there was a disagreement between the two reviewers even after the discussion.

2.4 Data extraction and quality assessment

The researchers independently used standardized data extraction forms based on the CHARMS-PF checklist in Microsoft Excel to extract essential data (e.g., date, settings, study design, and prognostic factors) from eligible studies32. The data was subsequently reviewed, and all disputes were resolved by consensus based on discussion. Study authors were not contacted to obtain unpublished data.

The quality in prognosis studies (QUIPS) tool developed by Hayden et al. was used to assess the bias risk of included studies 33. This tool includes six domains. Studies with five or six low-risk domains were classified to have an overall low risk of bias, while those with two or more high-risk domains were classified as having an overall high risk of bias. The remaining studies were classified as having an overall moderate risk of bias. The assessment results of the risk of bias were summarized and presented graphically. An overall certainty assessment in pooled estimates was performed using the grading of recommendations, assessment, development, and evaluation (GRADE) approach adopted in prognostic studies.

2.5 Data synthesis

In this prognostic meta-analysis, the pooled HR of OS and/or PFS with 95% confidence intervals (CI) was calculated for potential prognostic factors to investigate the effects of the factors. If possible, an indirect extraction of the HR values was done when unreported using the method described by Perneger et al. 34. A meta-analysis of OS estimates was conducted in cases with sufficient data at different time points (at 1, 3,5, 10, 15, and 20 years as reported). The interstudy heterogeneity was accessed using I2 value. I2 ≤ 50% indicated low heterogeneity and the fixed-effects model was adopted, while I2>50% indicated significant heterogeneity and the random-effects model was adopted. If necessary, meta regression was performed to explore the potential source of heterogeneity. Evidence of publication bias was accessed by funnel plot analysis and Egger’s test if at least ten studies were included (present in the Supplementary). All analyses were performed using the “metafor” package in R version 4.0.2 (R Core Team, R Foundation for Statistical Computing, Vienna, Austria). The significance threshold was set at P<0.05 or a 95% CI excluding 1.00.

3 Results

3.1 Search results

3.1.1 Identification of the relevant studies

Our initial database search retrieved 6542 eligible studies. Exclusion of 1930 duplications, 4393 irrelevant studies based on the title and abstract, and 219 studies after full-text reading left 17 suitable studies. An additional four papers were identified from reference lists and citation tracking (Figure 1), totaling 21 studies comprising 2722 DTC patients with PM, which were included in the analysis.

FIGURE 1
www.frontiersin.org

Figure 1 Flowchart of summarising reference search and study selection.

3.1.2 Characteristics of the included studies

Supplementary Table 1 summarises the characteristics of the included studies. Of the 21 studies, 12 were from Asia, five were from Europe, three were from North America, and one was from South America. Most studies were published between 2010 and 2020. All studies were retrospective, while one was a multi-centered study.

3.1.3 Quality of the included studies

The bias risk assessment of the included studies using the QUIPS tool was shown in Supplementary Figure 1. Most of the studies were low-intermediate risk (n=17, 80.95%), with only four studies classified as high risk. The four studies were judged to have a risk of bias in study attrition and confounding, statistical analysis, and reporting.

3.2 Synthesis results

Table 1 outlines pooled hazard ratios, 95% CI, and GRADE level of certainty for potential prognostic factors. The results are based on different clinical endpoints.

TABLE 1
www.frontiersin.org

Table 1 Prognostic factors and associated hazard ratio of survival.

3.2.1 Age

Figure 2A shows the calculated pooled hazard ratios upon assessing the impact of age at four reported age thresholds. Patients under 45 years had a better OS than those over 45 years (HR=2.18 [CI 1.26-3.77], P<0.01, N=601, Figure 2A) (5, 15, 23, 27, 3335). Similarly, patients under 40 years had a better OS than those over 40 years (HR=7.21 [CI 1.52 - 34.10], P=0.01, N=788, Supplementary Figure 3) (22, 25, 36). A single study reported that patients above 25 years had a poor OS (HR=2.36 [CI 1.67-3.04], P<0.01, N=42) (37). In the same line, the PFS was significantly better in the under-45 group than in the over-45 group (HR=3.85 [CI 1.29-11.47], P=0.02, N=306, Supplementary Figure 4) (20, 26). However, there was no significant difference in PFS between the under and over 55 groups (HR=1.30 [CI 0.06-30.17], P=0.87, N=136, Supplementary Figure 5) (24, 38).

FIGURE 2
www.frontiersin.org

Figure 2 Forest plot for the association of: (A) age with OS (>45 years vs. ≤45 years), (B) gender with OS (male vs. female), (C) gender with PFS (male vs. female), (D) histology subtype with OS (FTC vs. PTC), and (E) metastases to other organs with OS (with vs. without).

3.2.2 Gender

Eleven and four studies were included in OS and PFS analysis based on gender, respectively (10, 15, 19, 20, 22, 2426, 3339). Females had a significantly better OS than males (HR=1.01 [CI 1.01-1.19], P=0.03, N=1396, Fig 2B). Similarly, they had a significantly better PFS than males (HR=1.36 [CI 1.06-1.75], P=0.02, N=546, Figure 2C).

3.2.3 Histology subtype

Twelve studies comprising 1738 DTC patients with PM were included for histology subtype analysis (5, 9, 10, 19, 22, 23, 25, 27, 33, 34, 36, 37). The FTC group had a significantly poor OS than the PTC group (HR=1.63 [CI 1.36-1.96], P<0.01, N=2110, Figure 2D), consistent with the trend of the pooled OS (Figure 3B). However, a single study reported an insignificant difference in PFS between the two groups (HR=1.32 [CI 0.54-3.21], P=0.541, N=107, Supplementary Table 2) (20).

3.2.4 Metastases to other organs

Ten studies comprising 1713 patients with DM were included in this meta-analysis (5, 9, 15, 19, 22, 25, 27, 34, 36, 37). DTC patients with metastases to other organs had a poorer OS than patients with PM alone (HR=3.18 [CI 2.43-4.16], P<0.01, N=1713, Figure 2E). This trend was similar to that of pooled OS (Figure 3C). Of note, one study also reported a poorer PFS in patients with metastases to other organs than those with PM only (HR=2.38 [CI 1.1-5.15], P=0.028, N=107) (20).

3.2.5 Iodine avidity

Seven studies were included in the meta-analysis of iodine avidity: four studies for OS and three studies for PFS separately (15, 19, 20, 2326). The iodine non-avidity group had a poorer prognosis than the iodine avidity group for the OS (HR=3.10 [CI 1.79-5.37], P<0.01, N=646) and PFS (HR=2.93 [CI 2.18-3.95], P<0.01, N=395) (Figures 4A, B). A similar trend was observed for the pooled OS (Figure 3D).

FIGURE 3
www.frontiersin.org

Figure 3 Pooled OS rate point estimates: (A) all included patients; (B) PTC versus FTC; (C) pulmonary metastases only versus with metastases to other organs; (D) iodine avidity versus iodine non-avidity.

FIGURE 4
www.frontiersin.org

Figure 4 Forest plot for the association of: (A) iodine avidity with OS (non-avidity vs. avidity), (B) iodine avidity with PFS (non-avidity vs. avidity), (C) metastatic nodule size with PFS (≥ 10mm vs. <10mm), (D) extra thyroid invasion with PFS (with vs. without), (E) LNM with OS (with vs. without), (F) LNM with PFS (with vs. without).

3.2.6 Metastatic nodule size

Four studies comprising 513 patients were included in the analysis of the metastatic nodule size threshold of 10mm for PFS (15, 20, 24, 26). Notably, patients with a metastatic nodule size >10mm had a significantly poor PFS than those with metastatic nodule size ≤10mm (HR=2.56 [CI 2.02-3.24], P <0.01, N=513, Figure 4C). One study yielded a similar result in OS (HR=2.58 [CI 1.34-4.98], P<0.001, N=113) (27). Furthermore, another study reported that patients with a negative chest CT had a better OS than those with metastatic nodule size less (HR=1.9 [CI 1.1-3.4], P<0.01) or greater than 10mm (HR=3.5 [CI 2.1-5.8], P<0.01) in 372 patients (25).

3.2.7 ETE

Three studies comprising 272 patients were included in the meta-analysis of ETE for PFS (15, 20, 38). Patients with ETE had significantly poorer PFS than those without ETE (HR=2.05 [CI 1.15-3.67], P=0.02, N=271, Figure 4D). In particular, one study reported a significantly poor OS in patients with ETE than those without (HR=2.4 [CI 1.10-5.15], P=0.03, N=152) (15).

3.2.8 Lymph node metastases

Six studies were included in the meta-analysis for LNM; three studies each for OS and three studies for PFS (15, 20, 24, 26, 33, 37). There were no significant differences in OS (HR=0.68 [CI 0.41–1.12], P=0.13, N=501) and PFS (HR=0.94 [CI 0.47-1.90], P=0.86, N=526) between patients with or without LNM (Figures 4E, F).

3.2.9 Other factors

Several studies reported the primary tumor size and number of foci amongst the patients included in those studies (Supplementary Table 2). There were no significant differences in OS (HR=1.28 [CI 0.58 -2.80], P=0.54, N=96, Supplementary Figure 3) and PFS (HR=1.41 [CI 0.79-2.52], P=0.24, N=196, Supplementary Figure 4) between patients with primary tumor sizes over and under 40mm. Similarly, there was no significant difference in PFS between the unifocal group and the multifocal group (HR=1.81 [CI 0.77-4.25], P=0.18, N=154, Supplementary Figure 5).

3.2.10 Survival at different time points

We also evaluated and conducted a meta-analysis on the OS of DTC patients with PM at different time points (Supplementary Table 3). The pooled 1-year OS was 95.67% (95% CI: 92.93%-98.42%). However, the OS rate declined by approximately 15% every 5 years (Figure 3A), with the pooled OS at 20 years declining to 51.03% (95% CI: 36.52%-65.54%). Only two studies reported the PFS of patients at different time points (26, 38). The pooled 1-year PFS was 93.20% (95% CI: 62.55%-100%), while the pooled 10-year PFS was only 38.93% (95% CI: 23.53%-56.90%).

Subgroup analyses were only conducted on the histology subtype, metastases to other organs, and iodine avidity because of the lack of relevant data. On all three factors, there were significant differences in pooled OS between groups at different time points (P<0.001, Supplementary Table 3). PTC patients had a better pooled OS than FTC patients. The pooled 10-year and 20-year OS of the two groups were 61.66% vs. 31.96% and 46.65% vs. 18.27%, respectively (Figure 3B). Notably, patients with metastases to other organs had a poorer OS than patients with lung metastases alone (Figure 3C). The pooled 10-year and 20-year OS of the two groups were 65.21% vs. 31.21% and 54.68% vs. 7.25%, respectively. Similarly, non-avidity patients had a worse pooled OS than iodine avidity patients (Figure 3D). The pooled 10-year and 20-year OS were of the two groups were 50.22% vs. 15.95% and 32.30% vs. 1.0%, respectively.

3.2.11 Interstudy heterogeneity and publication bias

Variables including year of publication, sample size, region, and study quality were analyzed by meta-regression. However, no significant source of heterogeneity was identified in the meta-regression for OS of histology subtypes and metastases to other organs (P>0.05 for each, Supplementary Table 4). In addition, the funnel plots (Supplementary Figure 9-11) and Egger’s test showed no evidence of publication bias for the OS of gender (t=-0.45, P=0.66), histology subtypes (t=-0.14, P=0.89), and metastases to other organs (t=-0.96, P=0.36).

4 Discussion

A review of the relevant prognostic factors for DTC patients with PM was conducted to confirm their roles. To the best of the authors’ knowledge, this is the first systematic review and meta-analysis to evaluate the prognostic factors for DTCs with PM. The prognostic factors include:

4.1 Age

Age is an important prognostic factor and a consideration in staging for DTC patients (40). Though the age cutoff point changed to 55 years in the 8th TNM (41), 45 years was still the most commonly used cutoff point for the included studies in this review. DTC patients over 45 years and with PM had a poorer OS and PFS, consistent with previous studies (40, 42, 43). Studies suggest that 55 years may be a better cutoff point to predict the recurrence and survival of DTC patients (44). However, there were no differences in PFS between the two groups (≤55 years vs. >55 years) according to this meta-analysis. Of note, the number of included studies in the analysis was small, causing a high heterogeneity (I2 = 68%, P=0.08). The predictive value for 55 years as a cutoff point on DTC patients with PM thus remains to be explored. Studies suggest that the elevation of the cutoff point underestimates the prognostic risk of younger patients with risk features. For instance, Adam et al. showed that LNMs were associated with a 32% increase in mortality risk for stage I patients under 45 years, indicating that the current age cutoff point might be under staging young patients, translating to their undertreatment (42). In this meta-analysis, the maximum value of HRs was observed when the cutoff point was set at 40 years (Supplementary Figure 2). This cutoff point may predict the prognosis better and avoid underestimating the risk for younger patients considering the coexistence of PM.

4.2 Gender

Though the incidence of thyroid cancer is three-fold higher in females than in males, most studies showed higher mortality (HR 1.47-2.53) and shorter PFS in males than in females (4547). Similarly, male DTC patients with PM had a poorer OS (HR=1.36) and PFS (HR=1.10). Male DTC patients with PM should thus be subjected to more aggressive therapy than females.

4.3 Histological subtype

PTC and FTC account for 80%-85% and 10%-15% of DTC, respectively (48). Most studies suggest a poorer prognosis of FTC than PTC (4951), which was also observed in patients in this review. The pooled 20-year OS of FTC was less than 50% that for PTC patients.

FTC is characterized by early vascular infiltration, making it more likely to cause distant metastases through blood flow to the bones and lungs than PTC (24, 52, 53). The proportion of DM in FTC was much higher than in PTC (4.1%-14.1% vs. 0.5%-3.6%) (5456), an important reason why FTC has a poorer prognosis than PTC. However, the reasons for the poor survival prognosis of FTC than PTC when PM exists remains unclear. Lungs are the most common site of metastases in DTC, followed by bones, liver, and brain. Several studies revealed that the proportion of bone metastases in FTC is twice that in PTC (7-28% vs. 1.4-7%) (5759), with incidences of liver and brain metastases in FTC also being higher than in PTC (60). Generally, these studies suggest that FTC patients have a higher probability of combining with metastases to other organs than PTC patients when lung metastases exist. For instance, Wang et al., reported that FTC patients were more likely to progress from single organ metastases (SODM) to multiple organ metastases in 5 years than PTC patients (37.5% vs. 23.7%) (61). MODM could be the reason FTC patients with PM have a poorer prognosis than PTC patients, as observed in this review. FTC combined with PM is thus a histological subtype that requires high priority.

4.4 Metastases to other organs

PM patients with metastases to other organs (a condition of MODM) had a significant increase in mortality risk, which was thrice higher than that of patients with PM alone (HR=3.18, P<0.01, N=1713). The pooled 20-year OS was only 7.25% for MODM patients but remained 54.68% for patients with PM alone. Several studies suggest that DTC patients with MODM have a poor prognosis than patients with SODM. In a retrospective study including 111 PTC patients with DM, Haq et al. reported a poor survival for MODM patients than patients with PM alone (CSS: HR=2.70 [CI 1.38-5.26], P=0.02) or other single sites (13). Toraih et al. analyzed 1819 DTC patients with DM from the SEER database and reported a five- to six-fold increase in mortality risk for MODM patients than SODM patients (62). A more active treatment strategy may thus be necessary for PM patients with metastases to other organs.

4.5 Metastatic nodule size

Radioiodine therapy is the first line of choice for DTC patients with DM (63). However, the average penetration distance of β-irradiation from 131I inside the tissue is only 1 mm (4), resulting in larger lesions requiring higher doses of 131I, poorer outcomes, and more side effects (64). The pulmonary metastatic nodules are divided into micro-nodular and macro-nodular by 10mm. In this review, the micro-nodular group had a significantly better PFS than the macro-nodular group. Yang et al. also reported a significant difference in OS between the two groups (HR=2.58, P<0.001, N=113). Furthermore, it has been reported that PM patients with negative chest CT can achieve a better prognosis (27). Song et al. reported a 15-year OS of 0% vs. 50.1% vs. 75.8% (P<0.001, N=372) when comparing the macro-nodular group and micro-nodular group with patients who had negative chest CT, respectively (25). Qiu et al. tried 5mm as the cutoff point of the PM nodule size but reported no significant differences in PFS between groups (38). The optimal cutoff point of the metastatic nodule size should thus be explored further.

4.6 ETE

ETE is a recognized prognostic factor for DTC and is divided into minimal extrathyroid extension (mETE) and extensive extrathyroid extension (eETE) (65, 66). A large retrospective study by Youngwirth et al. that included 241,118 cases reported that patients with ETE, especially eETE (HR=1.74, P<0.01), had a poorer survival prognosis than those without (67). In this meta-analysis, ETE remains significantly affected thyroid cancer recurrence in PM patients (HR=2.05, P=0.02, N=271). Hirsch et al. also reported that PM patients with ETE have a higher mortality risk (HR=2.4 [CI 1.1-5.4], P=0.032, N=101) (15). Of note, the studies included in this meta-analysis did not categorize ETE into mETE and eETE. The prognostic effect of mETE on DTC patients with PM remains controversial (68, 69) and should thus be further explored.

4.7 Primary tumor size, LNM, and multifocality

The primary tumor size is a determinant for outcome in DTC (70). Previous studies postulate that the prognosis is worse for patients with larger tumor size (71, 72). However, the results of this meta-analysis revealed no significant differences in survival and recurrence between groups with 40mm as the cutoff point. In the same line, Qiu et al. reported insignificant differences in PFS among three subgroups of PM patients divided based on the primary tumor size (<20mm vs. 20-40mm and <20mm vs. ≥40mm, P>0.05, N=47) (38). Previous studies suggest that the risk of LNM and DM increases with increasing tumor size, leading to a worse prognosis for DTC patients (73, 74). However, the prognostic impact of distant metastases overrides the impact of tumor size upon PM occurrence, explaining why the impact of tumor size is not shown in PM patients. Similarly, the meta-analysis results did not show the impact of LNM on the OS (HR=0.68, P=0.13, N=510) and PFS (HR=0.97, P=0.94, N=526) of patients with PM. Although the presence of LNM leads to an increased risk of DM and a worse prognosis, its prognostic role is masked by the impact of DM when combined with PM (75). Multifocality does not affect patient prognosis in patients with DTC and PM because of similar mechanisms (76, 77). Meta-analysis results revealed that multifocality did not affect the PFS of PM patients (HR=1.81, P=0.18, N=154). Similarly, Leite et al. also reported that multifocality did not affect the PFS of PM patients (HR=1.804, P=0.917, N=54) (34).

4.8 RAI therapy, targeted therapy, and immunotherapy

Radioiodine therapy is the fundamental therapy for DM patients. The uptake status of radioactive iodine plays a crucial role in the prognosis of DM patients, depending on the size of the lesion and the iodine uptake capacity (78). The loss of ability to take up radioactive iodine can be congenital or acquired during RAI therapy (79). The prognosis for the patients is poor when iodine resistance is present regardless of the cause for losing iodine uptake ability (80). The same effect applies to DTC patients with PM (4). In this meta-analysis, iodine non-avidity patients with PM had a three-fold higher risk of mortality and recurrence than avidity patients. The pooled 20-year OS of the non-avidity patients was merely 0%, while that of avidity patients remained 34.3%. This result suggested that the sensitivity to radioiodine therapy is crucial to the overall outcome for PM patients, and identifying RAI-Refractory DTC (RAIRD) has important implications for treatment decisions (81).

Radioiodine therapy in PM patients is not ideal. Studies report that one-third to one-half of DTC patients with DM have RAI refractory (82, 83). The pooled overall efficacy rate of RAI therapy is 58% in PM patients (84). It is foreseeable that quite a significant portion of PM patients have a poor prognosis if they receive only standard treatment, including surgery, RAI therapy, and TSH suppression therapy. Promisingly, the emergence of TKIs offers new treatment options for thyroid cancer and has been employed as the first-line treatment for RAIRD (85, 86). Several randomized controlled clinical trials have reported encouraging results for TKIs, such as lenvatinib and sorafenib in RAIRD (8790). For instance, the SELECT trial showed that RAIRD patients with PM had a duration of overall response (DOR) of 29.9 months (95% CI 17.5-37.8) after treatment with lenvatinib (88). Moreover, a post hoc analysis of the SELECT trial showed that the lenvatinib group had a longer median OS (44.7 months vs. 33.1 months, P<0.01, N=392) than the placebo group in patients with PM lesions ≥10mm (30). Previous studies postulate that combining TKIs and immune checkpoint inhibitors (ICIs) could improve RAIRD treatment (91). A recent study reported better clinical benefit on PFS when lenvatinib was combined with pembrolizumab for RAIRD than using lenvatinib alone (92). However, further studies are needed to ascertain the effectiveness of these combinations in DTC patients with PM. The use of TKIs and ICIs in DTCs is still at an early stage and is thus important to identify the right target and the optimal treatment time (4, 86). Current ATA guidelines recommend using TKIs in patients with metastases, rapid progression, and symptomatic and life-threatening diseases. However, some researchers argue that patients potentially miss the optimal treatment time before meeting these requirements (86). This meta-analysis suggested that DTC patients with PM have a poor prognosis, especially those with FTC, multi-organ metastases, or iodine non-avidity (Figure 3). It is thus worth considering and exploring whether these patients should be treated with TKIs more aggressively before significant tumor progression.

4.9 Other prognostic factors

Serum Tg is the main biochemical tumor marker used to detect postoperative recurrence in patients with DTC (93). Previous studies postulate that serum Tg levels ≥50μg/L are associated with poorer PFS and OS in patients with DTC (94). However, the prognostic role of serum Tg in DTC patients with PM remains to be further investigated. Chopra et al. reported a poorer DFS in the ≥50μg/L group than the <50μg/L group (10 years DFS: 70% vs. 35%, HR=4.59 [CI 1.02-20.62], P=0.047, N=42) (21). In contrast, Chen et al. reported no significant differences in PFS between the two groups (HR=4.37 [CI 0.59-32.47], P=0.15, N=103) (20). Tg doubling time (TgDT) is a more predictable factor in PM patients than serum Tg levels (95). Miyauchi et al. reported that the 10-year cancer-specific survival (CSS) in PTC patients was only 50% when TgDT was <1 year, which was significantly worse than in patients with TgDT of 1-3 years (CSS: 95%) or >3 years (CSS: 100%) (96). However, no studies have reported the role of TgDT in PM patients.

Serum anti-thyroglobulin antibody (TgAb) level has been reported to predict persistent or recurrent diseases in DTC patients (97, 98). However, there are no sufficient studies to ascertain whether TgAb can be used as a prognosis indicator in PM patients. Qiu et al. evaluated the association between TgAb levels and the prognosis of DTC patients with PM and reported insignificant differences in PFS at 5 years (P=0.725) and 10 years (P=0.739) between different TgAb level groups in 47 patients (38).

BRAFV600E mutations, RAS mutations, and TERT promoter mutations are enriched in primary tumors of DTC with DM (99). Numerous studies confirm their association with aggressive biological behavior and worse prognosis (100104). Xing et al. reported a significantly higher mortality rate in patients with BRAFV600E mutation in primary tumor than unmutated DTC patients with DM (51.5% vs. 18.2%, P<0.001, N=1849) (105), attributed to the RAI resistance caused by BRAFV600E mutation (106). Song et al. reported that the recurrence risks for DTC patients with TERT promoter mutations in the primary tumor were 5.79 times and 3.6 times higher than those without mutations among ATA high-risk patients and stage III/IV patients, respectively (107). To date, no studies have evaluated the prognostic impact of the gene mutations in the primary tumor on DTC patients with PM and their impact on metastatic lesions.

4.10 Strengths and limitations of the study

This study is characterized by several strengths. It follows the recent guidelines on the meta-analysis of prognostic factor studies, which recommends an evaluation of the overall quality and reliability of the pooled results for clinical guidance using the GRADE approach. The study fully used the data from the included studies and systematically reviewed the prognostic factors for DTC patients with DM for the first time. The systematic review and meta-analysis revealed important prognostic factors for DTC patients with PM, such as histological subtype, extrapulmonary metastases, and iodine avidity.

Nevertheless, this study was limited by several factors. First, all the included studies were retrospective cohort studies, and thus most of them had confounding factors, including missing baseline and prognostic survival data because of the lack of a pre-established study protocol. These factors could have potentially affected the identification and assessment of prognostic survival factors (108). Second, high statistical heterogeneity was observed in most meta-analysis, but the source of heterogeneity was not well recognized. Since the included studies fewer than ten, meta-regression was not performed on most prognostic factors. The large time span (1985–2019) that induces confounding factors may explain part of the heterogeneity, however, unexplained heterogeneity was inevitable because the study was a meta-analysis and review of prognostic survival factors (32). Additionally, due to insufficient data, most individual meta-analyses included a small number of studies, and some important prognostic factors could not be performed. Finally, because of the lack of literature available, the prognostic impact of molecular biomarkers was not thoroughly discussed here. Hopefully, future research in this field will fill some of these gaps.

5 Conclusions

In this systematic review and meta-analysis, we evaluated the prognostic factors for DTC patients with PM based on the clinicopathological, therapeutic, and biochemical aspects. We found that these patients had a poor overall prognosis, especially those with FTC, metastases to other organs, or iodine non-avidity. In additional, emerging therapies have yielded encouraging results in these patients but still requires further investigation. These findings will help in better prognostic risk stratification and clinical management in DTC patients with PM.

Data availability statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding author.

Author contributions

Protocol and design of study: HZ, C-HL,X-YL,YC. Acquisition and/or management of data: HZ, C-HL, YZ. Analysis and/or interpretation of data: HZ, C-HL, Y-YL. Drafting and revising the manuscript: HZ, X-YL, L-YZ, Y-WL, H-FL, and Y-SL. All authors contributed to the article and approved the submitted version.

Acknowledgments

The authors would like to thank Yanlong Li, Ph.D., for statistical consultation during data collection and analysis, and MogoEdit (https://www.mogoedit.com) for its English editing during the preparation of this manuscript.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2022.990154/full#supplementary-material

References

1. Vigneri R, Malandrino P, Russo M. Is thyroid cancer increasing in incidence and aggressiveness? J Clin Endocrinol Metab (2020) 105(7):dgaa223. doi: 10.1210/clinem/dgaa223

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin (2021) 71(3):209–49. doi: 10.3322/caac.21660

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Cabanillas ME, McFadden DG, Durante C. Thyroid cancer. Lancet (2016) 388(10061):2783–95. doi: 10.1016/S0140-6736(16)30172-6

PubMed Abstract | CrossRef Full Text | Google Scholar

4. American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer, Cooper DS, Doherty GM, Haugen BR, Hauger BR, Kloos RT, et al. Revised American thyroid association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid (2009) 19(11):1167–214. doi: 10.1089/thy.2009.0110

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Cho SW, Choi HS, Yeom GJ, Lim JA, Moon JH, Park DJ, et al. Long-term prognosis of differentiated thyroid cancer with lung metastasis in Korea and its prognostic factors. Thyroid (2014) 24(2):277–86. doi: 10.1089/thy.2012.0654

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Ito Y, Higashiyama T, Takamura Y, Kobayashi K, Miya A, Miyauchi A. Clinical outcomes of patients with papillary thyroid carcinoma after the detection of distant recurrence. World J Surg (2010) 34(10):2333–7. doi: 10.1007/s00268-010-0712-0

PubMed Abstract | CrossRef Full Text | Google Scholar

7. SEER*Explorer:An interactive website for SEER cancer statistics. In: Surveillance research program. National Cancer Institute.

Google Scholar

8. O’Neill CJ, Oucharek J, Learoyd D, Sidhu SB. Standard and emerging therapies for metastatic differentiated thyroid cancer. Oncologist (2010) 15(2):146–56. doi: 10.1634/theoncologist.2009-0190

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Schlumberger M, Challeton C, De Vathaire F, Travagli JP, Gardet P, Lumbroso JD, et al. Radioactive iodine treatment and external radiotherapy for lung and bone metastases from thyroid carcinoma. J Nucl Med (1996) 37(4):598–605.

PubMed Abstract | Google Scholar

10. Ronga G, Filesi M, Montesano T, Di Nicola AD, Pace C, Travascio L, et al. Lung metastases from differentiated thyroid carcinoma. a 40 years’ experience. Q J Nucl Med Mol Imaging (2004) 48(1):12–9.

PubMed Abstract | Google Scholar

11. Bernier MO, Leenhardt L, Hoang C, Aurengo A, Mary JY, Menegaux F, et al. Survival and therapeutic modalities in patients with bone metastases of differentiated thyroid carcinomas. J Clin Endocrinol Metab (2001) 86(4):1568–73. doi: 10.1210/jcem.86.4.7390

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Mihailovic J, Stefanovic L, Malesevic M, Markoski B. The importance of age over radioiodine avidity as a prognostic factor in differentiated thyroid carcinoma with distant metastases. Thyroid (2009) 19(3):227–32. doi: 10.1089/thy.2008.0186

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Haq M, Harmer C. Differentiated thyroid carcinoma with distant metastases at presentation: prognostic factors and outcome. Clin Endocrinol (Oxf) (2005) 63(1):87–93. doi: 10.1111/j.1365-2265.2005.02304.x

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Liu JW, Chen C, Loh EW, Chu CC, Wang MY, Ouyang HJ, et al. Tyrosine kinase inhibitors for advanced or metastatic thyroid cancer: a meta-analysis of randomized controlled trials. Curr Med Res Opin (2018) 34(5):795–803. doi: 10.1080/03007995.2017.1368466

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Hirsch D, Levy S, Tsvetov G, Gorshtein A, Slutzky-Shraga I, Akirov A, et al. Long-term outcomes and prognostic factors in patients with differentiated thyroid cancer and distant metastases. Endocr Pract (2017) 23(10):1193–200. doi: 10.4158/EP171924.OR

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Sabra MM, Sherman EJ, Tuttle RM. Tumor volume doubling time of pulmonary metastases predicts overall survival and can guide the initiation of multikinase inhibitor therapy in patients with metastatic, follicular cell-derived thyroid carcinoma. Cancer (2017) 123(15):2955–64. doi: 10.1002/cncr.30690

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Ruegemer JJ, Hay ID, Bergstralh EJ, Ryan JJ, Offord KP, Gorman CA. Distant metastases in differentiated thyroid carcinoma: a multivariate analysis of prognostic variables. J Clin Endocrinol Metab (1988) 67(3):501–8. doi: 10.1210/jcem-67-3-501

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Showalter TN, Siegel BA, Moley JF, Baranski TJ, Grigsby PW. Prognostic factors in patients with well-differentiated thyroid cancer presenting with pulmonary metastasis. Cancer Biother Radiopharm (2008) 23(5):655–9. doi: 10.1089/cbr.2008.0501

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Casara D, Rubello D, Saladini G, Masarotto G, Favero A, Girelli ME, et al. Different features of pulmonary metastases in differentiated thyroid cancer: natural history and multivariate statistical analysis of prognostic variables. J Nucl Med (1993) 34(10):1626–31.

PubMed Abstract | Google Scholar

20. Chen P, Feng HJ, Ouyang W, Wu JQ, Wang J, Sun YG, et al. Risk factors for nonremission and progression-free survival after I-131 therapy in patients with lung metastasis from differentiated thyroid cancer: a single-institute, retrospective analysis in southern China. Endocr Pract (2016) 22(9):1048–56. doi: 10.4158/EP151139.OR

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Chopra S, Garg A, Ballal S, Bal CS. Lung metastases from differentiated thyroid carcinoma: prognostic factors related to remission and disease-free survival. Clin Endocrinol (Oxf) (2015) 82(3):445–52. doi: 10.1111/cen.12558

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Durante C, Haddy N, Baudin E, Leboulleux S, Hartl D, Travagli JP, et al. Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: benefits and limits of radioiodine therapy. J Clin Endocrinol Metab (2006) 91(8):2892–9. doi: 10.1210/jc.2005-2838

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Sampson E, Brierley JD, Le LW, Rotstein L, Tsang RW. Clinical management and outcome of papillary and follicular (differentiated) thyroid cancer presenting with distant metastasis at diagnosis. Cancer (2007) 110(7):1451–6. doi: 10.1002/cncr.22956

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Sohn SY, Kim HI, Kim YN, Kim TH, Kim SW, Chung JH. Prognostic indicators of outcomes in patients with lung metastases from differentiated thyroid carcinoma during long-term follow-up. Clin Endocrinol (Oxf) (2018) 88(2):318–26. doi: 10.1111/cen.13489

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Song HJ, Qiu ZL, Shen CT, Wei WJ, Luo QY. Pulmonary metastases in differentiated thyroid cancer: efficacy of radioiodine therapy and prognostic factors. Eur J Endocrinol (2015) 173(3):399–408. doi: 10.1530/EJE-15-0296

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Sabra MM, Ghossein R, Tuttle RM. Time course and predictors of structural disease progression in pulmonary metastases arising from follicular cell-derived thyroid cancer. Thyroid (2016) 26(4):518–24. doi: 10.1089/thy.2015.0395

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Yang J, Liang M, Jia Y, Wang L, Lin L, Geng J, et al. Therapeutic response and long-term outcome of differentiated thyroid cancer with pulmonary metastases treated by radioiodine therapy. Oncotarget (2017) 8(54):92715–26. doi: 10.18632/oncotarget.21570

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Qutbi M, Shafeie B, Amoui M, Tabeie F, Azizmohammadi Z, Mahmoud-Pashazadeh A, et al. Evaluation of prognostic factors associated with differentiated thyroid carcinoma with pulmonary metastasis. Clin Nucl Med (2016) 41(12):917–21. doi: 10.1097/RLU.0000000000001426

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Lin YS, Yang H, Ding Y, Cheng YZ, Shi F, Tan J, et al. Donafenib in progressive locally advanced or metastatic radioactive iodine-refractory differentiated thyroid cancer: Results of a randomized, multicenter phase II trial. Thyroid (2021) 31(4):607–15. doi: 10.1089/thy.2020.0235

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Tahara M, Kiyota N, Hoff AO, Badiu C, Owonikoko TK, Dutcus CE, et al. Impact of lung metastases on overall survival in the phase 3 SELECT study of lenvatinib in patients with radioiodine-refractory differentiated thyroid cancer. Eur J Cancer (2021) 147:51–7. doi: 10.1016/j.ejca.2020.12.032

PubMed Abstract | CrossRef Full Text | Google Scholar

31. 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. doi: 10.1136/bmj.n71

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Riley RD, Moons KGM, Snell KIE, Ensor J, Hooft L, Altman DG, et al. A guide to systematic review and meta-analysis of prognostic factor studies. BMJ (2019) 364:k4597. doi: 10.1136/bmj.k4597

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Qiu ZL, Shen CT, Luo QY. Clinical management and outcomes in patients with hyperfunctioning distant metastases from differentiated thyroid cancer after total thyroidectomy and radioactive iodine therapy. Thyroid (2015) 25(2):229–37. doi: 10.1089/thy.2014.0233

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Leite AKN, Kulcsar MAV, de Godoi Cavalheiro B, de Mello ES, Alves VAF, Cernea CR, et al. Death related to pulmonary metastasis in patients with differentiated thyroid cancer. Endocr Pract (2017) 23(1):72–8. doi: 10.4158/EP161431.OR

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Machens A, Dralle H. Correlation between the number of lymph node metastases and lung metastasis in papillary thyroid cancer. J Clin Endocrinol Metab (2012) 97(12):4375–82. doi: 10.1210/jc.2012-1257

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Samaan NA, Schultz PN, Haynie TP, Ordonez NG. Pulmonary metastasis of differentiated thyroid carcinoma: treatment results in 101 patients. J Clin Endocrinol Metab (1985) 60(2):376–80. doi: 10.1210/jcem-60-2-376

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Ilgan S, Karacalioglu AO, Pabuscu Y, Atac GK, Arslan N, Ozturk E, et al. Iodine-131 treatment and high-resolution CT: results in patients with lung metastases from differentiated thyroid carcinoma. Eur J Nucl Med Mol Imaging (2004) 31(6):825–30. doi: 10.1007/s00259-004-1460-x

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Qiu ZL, Shen CT, Sun ZK, Song HJ, Zhang GQ, Luo QY. Lung metastases from papillary thyroid cancer with persistently negative thyroglobulin and elevated thyroglobulin antibody levels during radioactive iodine treatment and follow-up: Long-term outcomes and prognostic indicators. Front Endocrinol (Lausanne) (2019) 10:903. doi: 10.3389/fendo.2019.00903

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Lin JD, Chao TC, Hsueh C. Follicular thyroid carcinomas with lung metastases: a 23-year retrospective study. Endocr J (2004) 51(2):219–25. doi: 10.1507/endocrj.51.219

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Franssila KO. Prognosis in thyroid carcinoma. Cancer (1975) 36(3):1138–46. doi: 10.1002/1097-0142(197509)36:3<1138::AID-CNCR2820360346>3.0.CO;2-8

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Amin MB, . Meyer LR. AJCC cancer staging manual (2018). 8th. New York: Springer.

Google Scholar

42. Adam MA, Thomas S, Hyslop T, Scheri RP, Roman SA, Sosa JA. Exploring the relationship between patient age and cancer-specific survival in papillary thyroid cancer: Rethinking current staging systems. J Clin Oncol (2016) 34(36):4415–20. doi: 10.1200/JCO.2016.68.9372

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Eichhorn W, Tabler H, Lippold R, Lochmann M, Schreckenberger M, Bartenstein P. Prognostic factors determining long-term survival in well-differentiated thyroid cancer: an analysis of four hundred eighty-four patients undergoing therapy and aftercare at the same institution. Thyroid (2003) 13(10):949–58. doi: 10.1089/105072503322511355

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Nam SH, Bae MR, Roh JL, Gong G, Cho KJ, Choi SH, et al. A comparison of the 7th and 8th editions of the AJCC staging system in terms of predicting recurrence and survival in patients with papillary thyroid carcinoma. Oral Oncol (2018) 87:158–64. doi: 10.1016/j.oraloncology.2018.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Rahbari R, Zhang L, Kebebew E. Thyroid cancer gender disparity. Future Oncol (2010) 6(11):1771–9. doi: 10.2217/fon.10.127

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Sipos JA, Mazzaferri EL. Thyroid cancer epidemiology and prognostic variables. Clin Oncol (R Coll Radiol) (2010) 22(6):395–404. doi: 10.1016/j.clon.2010.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Liu C, Chen T, Zeng W, Wang S, Xiong Y, Liu Z, et al. Reevaluating the prognostic significance of male gender for papillary thyroid carcinoma and microcarcinoma: a SEER database analysis. Sci Rep (2017) 7(1):11412. doi: 10.1038/s41598-017-11788-8

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Yoo SK, Song YS, Park YJ, Seo JS. Recent improvements in genomic and transcriptomic understanding of anaplastic and poorly differentiated thyroid cancers. Endocrinol Metab (Seoul) (2020) 35(1):44–54. doi: 10.3803/EnM.2020.35.1.44

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Liu Z, Zeng W, Huang L, Wang Z, Wang M, Zhou L, et al. Prognosis of FTC compared to PTC and FVPTC: findings based on SEER database using propensity score matching analysis. Am J Cancer Res (2018) 8(8):1440–8.

PubMed Abstract | Google Scholar

50. Aboelnaga EM, Ahmed RA. Difference between papillary and follicular thyroid carcinoma outcomes: an experience from Egyptian institution. Cancer Biol Med (2015) 12(1):53–9. doi: 10.7497/j.issn.2095-3941.2015.0005

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Sugino K, Ito K, Nagahama M, Kitagawa W, Shibuya H, Ohkuwa K, et al. Prognosis and prognostic factors for distant metastases and tumor mortality in follicular thyroid carcinoma. Thyroid (2011) 21(7):751–7. doi: 10.1089/thy.2010.0353

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Lin JD, Huang MJ, Juang JH, Chao TC, Huang BY, Chen KW, et al. Factors related to the survival of papillary and follicular thyroid carcinoma patients with distant metastases. Thyroid (1999) 9(12):1227–35. doi: 10.1089/thy.1999.9.1227

PubMed Abstract | CrossRef Full Text | Google Scholar

53. DeGroot LJ, Kaplan EL, Shukla MS, Salti G, Straus FH. Morbidity and mortality in follicular thyroid cancer. J Clin Endocrinol Metab (1995) 80(10):2946–53. doi: 10.1210/jcem.80.10.7559879

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Yu L, Hong H, Han J, Leng SX, Zhang H, Yan X. Comparison of survival and risk factors of differentiated thyroid cancer in the geriatric population. Front Oncol (2020) 10:42. doi: 10.3389/fonc.2020.00042

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Kuo EJ, Roman SA, Sosa JA. Patients with follicular and hurthle cell microcarcinomas have compromised survival: a population level study of 22,738 patients. Surgery (2013) 154(6):1246–53. doi: 10.1016/j.surg.2013.04.033

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Yu XM, Schneider DF, Leverson G, Chen H, Sippel RS. Follicular variant of papillary thyroid carcinoma is a unique clinical entity: A population-based study of 10,740 cases. Thyroid (2013) 23(10):1263–8. doi: 10.1089/thy.2012.0453

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Choksi P, Papaleontiou M, Guo C, Worden F, Banerjee M, Haymart M. Skeletal complications and mortality in thyroid cancer: A population-based study. J Clin Endocrinol Metab (2017) 102(4):1254–60. doi: 10.1210/jc.2016-3906

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Muresan MM, Olivier P, Leclère J, Sirveaux F, Brunaud L, Klein M, et al. Bone metastases from differentiated thyroid carcinoma. Endocr Relat Cancer (2008) 15(1):37–49. doi: 10.1677/ERC-07-0229

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Kato S, Demura S, Shinmura K, Yokogawa N, Shimizu T, Tsuchiya H. Current management of bone metastases from differentiated thyroid cancer. Cancers (2021) 13(17):4429. doi: 10.3390/cancers13174429

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Hugen N, Sloot YJE, Netea-Maier RT, van de Water C, Smit JWA, Nagtegaal ID, et al. Divergent metastatic patterns between subtypes of thyroid carcinoma results from the nationwide Dutch pathology registry. J Clin Endocrinol Metab (2020) 105(3):e299. doi: 10.1210/clinem/dgz078

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Wang LY, Palmer FL, Nixon IJ, Thomas D, Patel SG, Shaha AR, et al. Multi-organ distant metastases confer worse disease-specific survival in differentiated thyroid cancer. Thyroid (2014) 24(11):1594–9. doi: 10.1089/thy.2014.0173

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Toraih EA, Hussein MH, Zerfaoui M, Attia AS, Marzouk Ellythy A, Mostafa A, et al. Site-specific metastasis and survival in papillary thyroid cancer: The importance of brain and multi-organ disease. Cancers (2021) 13(7):1625. doi: 10.3390/cancers13071625

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Choi JH, Byun BH, Lim I, Moon H, Park J, Chang KJ, et al. The predictive values of lesion size, f-18 FDG avidity and I-131 avidity for the clinical outcome of I-131 treatment in patients with metastatic differentiated thyroid carcinoma only in the lung. Nucl Med Mol Imaging (2018) 52(2):135–43. doi: 10.1007/s13139-017-0502-9

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Xu Z, Li C, Fang F, Zhang L, Wang H, Fu H, et al. The value of 18F-FDG PET/CT in avoiding overtreatment of 131l avidity pulmonary metastasis of differentiated thyroid cancer (2021). Available at: https://www.researchsquare.com/article/rs-34450/v1.

Google Scholar

65. Ito Y, Miyauchi A, Kihara M, Kobayashi K, Miya A. Prognostic values of clinical lymph node metastasis and macroscopic extrathyroid extension in papillary thyroid carcinoma. Endocr J (2014) 61(8):745–50. doi: 10.1507/endocrj.EJ14-0138

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Xu B, Ghossein R. Crucial parameters in thyroid carcinoma reporting – challenges, controversies, and clinical implications. Histopathology (2018) 72(1):32–9. doi: 10.1111/his.13335

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Youngwirth LM, Adam MA, Scheri RP, Roman SA, Sosa JA. Extrathyroidal extension is associated with compromised survival in patients with thyroid cancer. Thyroid (2017) 27(5):626–31. doi: 10.1089/thy.2016.0132

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Hay ID, Johnson TR, Thompson GB, Sebo TJ, Reinalda MS. Minimal extrathyroid extension in papillary thyroid carcinoma does not result in increased rates of either cause-specific mortality or postoperative tumor recurrence. Surgery (2016) 159(1):11–9. doi: 10.1016/j.surg.2015.05.046

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Woo CG, Sung CO, Choi YM, Kim WG, Kim TY, Shong YK, et al. Clinicopathological significance of minimal extrathyroid extension in solitary papillary thyroid carcinomas. Ann Surg Oncol (2015) 22 Suppl 3:S728–733. doi: 10.1245/s10434-015-4659-0

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Edge SB, Compton CC. The American joint committee on cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol (2010) 17(6):1471–4. doi: 10.1245/s10434-010-0985-4

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Ito Y, Kudo T, Kihara M, Takamura Y, Kobayashi K, Miya A, et al. Prognosis of low-risk papillary thyroid carcinoma patients: Its relationship with the size of primary tumors. Endocr J (2012) 59(2):119–25. doi: 10.1507/endocrj.EJ11-0288

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Zhang J, Cheng X, Su B, Wang X, Wang L, Jayachandran M, et al. The increased risk of thyroid cancer-specific mortality with tumor size in stage IVB patients. Front Oncol (2020) 10:1946. doi: 10.3389/fonc.2020.560203

CrossRef Full Text | Google Scholar

73. Nguyen XV, Roy Choudhury K, Tessler FN, Hoang JK. Effect of tumor size on risk of metastatic disease and survival for thyroid cancer: Implications for biopsy guidelines. Thyroid (2018) 28(3):295–300. doi: 10.1089/thy.2017.0526

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Sun QH, Zhang L, Yang JB, Liu YW, Liang F, Shi H, et al. [Related factors analysis for lymph node metastasis in papillary thyroid carcinoma: a series of 2 073 patients]. Zhonghua Wai Ke Za Zhi (2017) 55(8):592–8. doi: 10.3760/cma.j.issn.0529-5815.2017.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Machens A, Holzhausen HJ, Lautenschläger C, Thanh PN, Dralle H. Enhancement of lymph node metastasis and distant metastasis of thyroid carcinoma. Cancer (2003) 98(4):712–9. doi: 10.1002/cncr.11581

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Qu N, Zhang L, Ji QH, Zhu YX, Wang ZY, Shen Q, et al. Number of tumor foci predicts prognosis in papillary thyroid cancer. BMC Cancer (2014) 14:914. doi: 10.1186/1471-2407-14-914

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Kim H, Kwon H, Moon BI. Association of multifocality with prognosis of papillary thyroid carcinoma: A systematic review and meta-analysis. JAMA Otolaryngol Head Neck Surg (2021) 147(10):847–54. doi: 10.1001/jamaoto.2021.1976

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Kreissl MC, Janssen MJR, Nagarajah J. Current treatment strategies in metastasized differentiated thyroid cancer. J Nucl Med (2019) 60(1):9–15. doi: 10.2967/jnumed.117.190819

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Pacini F, Ito Y, Luster M, Pitoia F, Robinson B, Wirth L. Radioactive iodine-refractory differentiated thyroid cancer: unmet needs and future directions. Expert Rev Endocrinol Metab (2012) 7(5):541–54. doi: 10.1586/eem.12.36

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Gosain R, Alexander JS, Gill A, Perez C. Radioactive iodine-refractory differentiated thyroid cancer in the elderly. Curr Oncol Rep (2018) 20(10):82. doi: 10.1007/s11912-018-0736-4

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Mu ZZ, Zhang X, Lin YS. Identification of radioactive iodine refractory differentiated thyroid cancer. Chonnam Med J (2019) 55(3):127–35. doi: 10.4068/cmj.2019.55.3.127

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Aashiq M, Silverman DA, Na’ara S, Takahashi H, Amit M. Radioiodine-refractory thyroid cancer: Molecular basis of redifferentiation therapies, management, and novel therapies. Cancers (2019) 11(9):1382. doi: 10.3390/cancers11091382

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Hong CM, Ahn BC. Redifferentiation of radioiodine refractory differentiated thyroid cancer for reapplication of I-131 therapy. Front Endocrinol (2017) 8:260. doi: 10.3389/fendo.2017.00260

CrossRef Full Text | Google Scholar

84. Zhang X, DS L, Luan ZS, Zhang F, Liu XH, Zhou W, et al. Efficacy of radioiodine therapy for treating 20 patients with pulmonary metastases from differentiated thyroid cancer and a meta-analysis of the current literature. Clin Transl Oncol (2018) 20(7):928–35. doi: 10.1007/s12094-017-1792-1

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Gild ML, Bullock M, Robinson BG, Clifton-Bligh R. Multikinase inhibitors: a new option for the treatment of thyroid cancer. Nat Rev Endocrinol (2011) 7(10):617–24. doi: 10.1038/nrendo.2011.141

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Gild ML, Tsang VHM, Clifton-Bligh RJ, Robinson BG. Multikinase inhibitors in thyroid cancer: timing of targeted therapy. Nat Rev Endocrinol (2021) 17(4):225–34. doi: 10.1038/s41574-020-00465-y

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Gianoukakis AG, Dutcus CE, Batty N, Guo M, Baig M. Prolonged duration of response in lenvatinib responders with thyroid cancer. Endocr Relat Cancer (2018) 25(6):699–704. doi: 10.1530/ERC-18-0049

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet (2014) 384(9940):319–28. doi: 10.1016/S0140-6736(14)60421-9

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. New Engl J Med (2015) 372(7):621–30. doi: 10.1056/NEJMoa1406470

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Lin Y, Qin S, Li Z, Yang H, Fu W, Li S, et al. Apatinib vs placebo in patients with locally advanced or metastatic, radioactive iodine–refractory differentiated thyroid cancer: The REALITY randomized clinical trial. JAMA Oncol (2021) 8(2):242–50. doi: 10.1001/jamaoncol.2021.6268

CrossRef Full Text | Google Scholar

91. D’Andréa G, Lassalle S, Guevara N, Mograbi B, Hofman P. From biomarkers to therapeutic targets: the promise of PD-L1 in thyroid autoimmunity and cancer. Theranostics (2021) 11(3):1310–25. doi: 10.7150/thno.50333

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Haugen B, French J, Worden FP, Konda B, Sherman EJ, Dadu R, et al. Lenvatinib plus pembrolizumab combination therapy in patients with radioiodine-refractory (RAIR), progressive differentiated thyroid cancer (DTC): Results of a multicenter phase II international thyroid oncology group trial. JCO (2020) 38(15_suppl):6512–2. doi: 10.1200/JCO.2020.38.15_suppl.6512

CrossRef Full Text | Google Scholar

93. Spencer C, Petrovic I, Fatemi S, LoPresti J. Serum thyroglobulin (Tg) monitoring of patients with differentiated thyroid cancer using sensitive (Second-generation) immunometric assays can be disrupted by false-negative and false-positive serum thyroglobulin autoantibody misclassifications. J Clin Endocrinol Metab (2014) 99(12):4589–99. doi: 10.1210/jc.2014-1203

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Piccardo A, Arecco F, Puntoni M, Foppiani L, Cabria M, Corvisieri S, et al. Focus on high-risk DTC patients: High postoperative serum thyroglobulin level is a strong predictor of disease persistence and is associated to progression-free survival and overall survival. Clin Nucl Med (2013) 38(1):18–24. doi: 10.1097/RLU.0b013e318266d4d8

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Rössing RM, Jentzen W, Nagarajah J, Bockisch A, Görges R. Serum thyroglobulin doubling time in progressive thyroid cancer. Thyroid (2016) 26(12):1712–8. doi: 10.1089/thy.2016.0031

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Miyauchi A, Kudo T, Miya A, Kobayashi K, Ito Y, Takamura Y, et al. Prognostic impact of serum thyroglobulin doubling-time under thyrotropin suppression in patients with papillary thyroid carcinoma who underwent total thyroidectomy. Thyroid (2011) 21(7):707–16. doi: 10.1089/thy.2010.0355

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Hsieh CJ, Wang PW. Sequential changes of serum antithyroglobulin antibody levels are a good predictor of disease activity in thyroglobulin-negative patients with papillary thyroid carcinoma. Thyroid (2014) 24(3):488–93. doi: 10.1089/thy.2012.0611

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Kim WG, Yoon JH, Kim WB, Kim TY, Kim EY, Kim JM, et al. Change of serum antithyroglobulin antibody levels is useful for prediction of clinical recurrence in thyroglobulin-negative patients with differentiated thyroid carcinoma. J Clin Endocrinol Metab (2008) 93(12):4683–9. doi: 10.1210/jc.2008-0962

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Song E, Song DE, Ahn J, Kim TY, Kim WB, Shong YK, et al. Genetic profile of advanced thyroid cancers in relation to distant metastasis. Endocr Relat Cancer (2020) 27(5):285–93. doi: 10.1530/ERC-19-0452

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Li M, Jia H, Qian Q, Wen P, Chen C, Hua Y, et al. Genomic characterization of high-recurrence risk papillary thyroid carcinoma in a southern Chinese population. Diagn Pathol (2020) 15(1):49. doi: 10.1186/s13000-020-00962-8

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Mady LJ, Grimes MC, Khan NI, Rao RH, Chiosea SI, Yip L, et al. Molecular profile of locally aggressive well differentiated thyroid cancers. Sci Rep (2020) 10(1):8031. doi: 10.1038/s41598-020-64635-8

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Melo M, da Rocha AG, Vinagre J, Batista R, Peixoto J, Tavares C, et al. TERT promoter mutations are a major indicator of poor outcome in differentiated thyroid carcinomas. J Clin Endocrinol Metab (2014) 99(5):E754–765. doi: 10.1210/jc.2013-3734

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Shen X, Zhu G, Liu R, Viola D, Elisei R, Puxeddu E, et al. Patient age-associated mortality risk is differentiated by BRAF V600E status in papillary thyroid cancer. J Clin Oncol (2018) 36(5):438–45. doi: 10.1200/JCO.2017.74.5497

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Vuong HG, Duong UNP, Altibi AMA, Ngo HTT, Pham TQ, Tran HM, et al. A meta-analysis of prognostic roles of molecular markers in papillary thyroid carcinoma. Endocr Connect (2017) 6(3):R8–17. doi: 10.1530/EC-17-0010

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Xing M, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, et al. Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. JAMA (2013) 309(14):1493–501. doi: 10.1001/jama.2013.3190

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Durante C, Puxeddu E, Ferretti E, Morisi R, Moretti S, Bruno R, et al. BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism. J Clin Endocrinol Metab (2007) 92(7):2840–3. doi: 10.1210/jc.2006-2707

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Song YS, Lim JA, Choi H, Won JK, Moon JH, Cho SW, et al. Prognostic effects of TERT promoter mutations are enhanced by coexistence with BRAF or RAS mutations and strengthen the risk prediction by the ATA or TNM staging system in differentiated thyroid cancer patients. Cancer (2016) 122(9):1370–9. doi: 10.1002/cncr.29934

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Noordzij M, Dekker FW, Zoccali C, Jager KJ. Study designs in clinical research. Nephron Clin Pract (2009) 113(3):c218–221. doi: 10.1159/000235610

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: differentiated thyroid cancer, pulmonary metastases, prognosis, meta-analysis, systematic review

Citation: Zhao H, Liu C-H, Cao Y, Zhang L-Y, Zhao Y, Liu Y-W, Liu H-F, Lin Y-S and Li X-Y (2022) Survival prognostic factors for differentiated thyroid cancer patients with pulmonary metastases: A systematic review and meta-analysis. Front. Oncol. 12:990154. doi: 10.3389/fonc.2022.990154

Received: 09 July 2022; Accepted: 30 November 2022;
Published: 15 December 2022.

Edited by:

Guy Andry, Université libre de Bruxelles, Belgium

Reviewed by:

Jiandi Zhang, Quanticision Diagnostics, Inc., United States
Chentian Shen, Shanghai Sixth People’s Hospital, China

Copyright © 2022 Zhao, Liu, Cao, Zhang, Zhao, Liu, Liu, Lin and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xiao-Yi Li, li.xiaoyi@263.net

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.