Next Article in Journal
Expanding Indications in Transplant Oncology
Previous Article in Journal
Cyclin-Dependent Kinase Inhibition in Prostate Cancer: Past, Present, and Future
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Modern Therapeutic Approaches in Anaplastic Thyroid Cancer: A Meta-Analytic Review of Randomised and Single Arm Studies on Efficacy and Survival

1
South West Wales Cancer Center, Swansea Bay University Health Board, Singleton Hospital, Swansea SA2 8QA, UK
2
Department of Medicine, Swansea Bay University Health Board, Swansea SA2 8QA, UK
3
School of Medicine, Swansea Bay University, Swansea SA2 8QA, UK
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(5), 777; https://doi.org/10.3390/cancers17050777
Submission received: 14 January 2025 / Revised: 9 February 2025 / Accepted: 15 February 2025 / Published: 24 February 2025
(This article belongs to the Section Cancer Therapy)

Simple Summary

The present meta-analysis aimed at the efficacy and safety of modern therapeutic approaches, including targeted therapies, in locally advanced, recurrent and metastatic anaplastic thyroid cancer (ATC). Analyzing 47 studies with 980 patients, the pooled overall response rate (ORR) was 29.7%, with a pooled median overall survival (OS) of 7.2 months and pooled median progression-free survival (PFS) of 5.4 months. Dabrafenib/trametinib (DT), with or without pembrolizumab and lenvatinib plus pembrolizumab (LP), demonstrated superior ORR, PFS and overall survival (OS). DT was identified as a promising first-line treatment for BRAFV600-mutated ATC, while LP showed potential in BRAFV600 wild-type and PDL1-overexpressing cases. Nearly half of the studies reported bio-marker analyses necessitating routine next-generation sequencing (NGS) testing (e.g., BRAFV600, PDL1, RAS). These findings underscore the role of biomarker-driven strategies in optimising ATC treatment.

Abstract

Background: Meta-analyses aimed to assess the effectiveness and safety of targeted and contemporary therapies utilised in locally advanced and metastatic anaplastic thyroid cancer (ATC). Methods: Employing PRISMA and MOOSE guidelines, PubMed, Scopus, Cochrane Library and Web of Science were explored from the inception of targeted therapy until December 2024. A meta-analysis was performed to evaluate the effectiveness, toxicity and survival outcomes of various mutationally directed agents, chemotherapy and radiotherapy in locally advanced/metastatic ATC cases. Results: A total of 47 studies (26 prospective phase II trials and 21 retrospective studies) involving 980 patients met the inclusion criteria. The pooled results showed an overall response rate (ORR) of 29.7% (95% CI: 25.4–34.2%; I2 = 42.4%; p < 0.0001). A total of 49.9% deaths were reported, although a significant number remained alive compared to baseline (mean difference [MD]: 2.07, 95% CI: 1.90–2.24; I2 = 88.6%; p < 0.0001). The pooled median progression-free survival (PFS) was 5.4 months (95% CI: 4.0–6.7 months; I2 = 97.9%; p < 0.0001). Dabrafenib/trametinib (DT) with and without pembrolizumab and lenvatinib plus pembrolizumab (LP) were associated with higher ORR rates and improved OS and PFS. About 51.% of studies mentioned bio-marker analysis (BRAFV600 [14.7%], PDL1 [9.2%], RAS [1.1%], PIK3CA [1.0%] and NTRK1/3 [0.7%]). Toxicity was reported in 94.7% of patients. Conclusions: This meta-analysis found that DT could be a promising first-line treatment option for BRAFV600-mutated ATC, with or without immunotherapy. Alternatively, LP shows potential in BRAFV600 wild-type and PDL1-overexpressing cases. Routine biomarker analysis remains critical for optimising ATC management strategies.

1. Introduction

Anaplastic thyroid cancer (ATC) is a highly aggressive and rare cancer, as defined by the European Union Rare Cancer Surveillance Program (RARECARE), with an incidence beneath six new cases per 100,000 person-years [1]. The majority of ATC patients are diagnosed at an advanced stage (IVB and IVC) associated with compressive symptoms, local invasion and distant metastasis, and only 10% are surgical candidates (IVA) [2,3]. Due to ATC’s ultimate nature, the median survival time is 3–4 months, and the 1-year survival rate is approximately 20% [4].
Due to its rarity, ATC has never been entrant in a large randomised remedial trial. Thus, available data have been primarily obtained from single-centre retrospective studies or phase II non-randomised trials [5]. Historically, doxorubicin monotherapy or a combination of two or more drugs, such as paclitaxel; cisplatin; or carboplatin, doxorubicin and docetaxel, has been considered the most effective treatment. Response rates range from 20% to 50% and significant toxicity [6]. The shift from conventional chemotherapy to precise, molecularly targeted therapies is a rapidly developing phenomenon. Tyrosine kinase inhibitors (TKIs) (lenvatinib, sorafenib, pazopanib) have been trialled since 2008. Lenvatinib has shown meaningful antitumor activity but limited clinical efficacy against ATC [7].
Genomic alterations are highly prevalent in ATC compared to differentiated thyroid cancer [8]. Studies indicate that 95.8% of ATC cases harbour at least one genetic alteration in the MAP kinase pathway or the PIK3/Akt pathway [8,9,10]. The most common mutation is BRAF V600E, observed in 8–87% of cases [8]. Personalised medicine, integrating genomic information, depends on next-generation sequencing (NGS) biomarker assays, which have significantly advanced in ATC. These assays facilitate the identification of targetable mutations and provide timely access to mutationally directed therapies (BRAF V600E mutations with dabrafenib plus trametinib [8]; TRK fusion alterations with entrectinib or larotrectinib [9] and RET fusion with selpercatinib [10]). Despite being based on low-powered studies, these findings have led to the approval of these treatments for ATC. In addition, several small studies of IO alone or combined with TKIs have yielded favourable response and progression-free survival (PFS) rates [11].
Despite current recommendations advocating using NGS and biomarker assays, global clinical practice for ATC management remains highly variable. These inconsistencies in implementation and access have highlighted the need for further evaluation, leading to our present meta-analysis. We aimed to clarify the efficacy and safety of mutational/anti-angiogenesis-directed therapies and contemporary therapies (chemotherapy, radiation therapy, chemoradiation), irrespective of their sequence, to provide some guidance on choosing the most effective regimen for locally advanced, recurrent and metastatic ATC.

2. Materials and Methods

We followed PRISMA (preferred reporting items for systemic reviews and meta-analyses) and MOOSE (meta-analyses of observational studies in epidemiology) recommendations to conduct and report present systematic review and meta-analysis [12,13]. The protocol has not been registered.

2.1. Intervention

This systematic review and meta-analysis was structured to evaluate the efficacy and survival outcomes of various mutational directed and conventional therapies in patients with unresectable, recurrent and metastatic anaplastic thyroid cancer, compared with tyrosine kinase inhibitors (TKI) [lenvatinib, sorafenib, pazopanib +/− RT, gefitinib, apatinib, sunitinib, imatinib], fosbretabulin (formerly combretastatin A4 phosphate), dabrafenib-trametinib (DT) (BRAF/MEK inhibitor), vemurafenib (BRAF inhibitor), immunotherapy (nivolumab, pembrolizumab, nivolumab–ipilimumab, spartalizumab) +/− lenvatinib +/− RT, everolimus (mTOR inhibitor), TRK inhibitors (larotrectinib, entrectinib), RET inhibitor (selpercatinib) and chemotherapy with paclitaxel or carboplatin +/− RT.

2.2. Inclusion and Exclusion Criteria

Inclusion criteria were (a) studies on patients with unresectable locally advanced, recurrent or metastatic ATC who had (b) received any of the treatment options as mentioned earlier in any sequence in (c) clinical trials, in either a controlled or single-arm setting, including prospective and retrospective observational studies available as complete publication, with (d) reported efficacy, toxicity and survival outcomes.
Exclusion criteria were (a) studies whose data were erratic for analysis, (b) review articles, case reports, or case series of less than five patients, (c) studies with abstracts only and (d) animal/cell studies.

2.3. Search Approach

We searched databases comprehensively (PubMed, Scopus, Cochrane Library, Web of Science and Clinicaltrial.gov) from the inception of targeted therapy until December 2024. The search keywords were: (“anaplastic thyroid cancer” [MeSH Terms] OR (“anaplastic” [All Fields] AND “thyroid cancer” [All Fields]) OR “thyroid cancer” [All Fields] AND (“tyrosine kinase inhibitors” [“Lenvatinib, Sorafenib, Pazopanib +/− RT, Gefitinib, Apatinib, Sunitinib, Imatinib”]) [All fields], (“Fosbretabulin”) [All fields], AND (“BRAF/MEK inhibitors” [“Vemurafenib, Dabrafenib/Trametinib, Encorafenib/binimetinib”]) [All fields], AND (“Immunotherapy” [“nivolumab, pembrolizumab, nivolumab–ipilimumab, atezolizumab, Spartalizumab”]) +/− “Lenvatinib” +/− “radiotherapy” [All fields], AND (“mTOR inhibitor” [“Everolimus, Sapanisertib”]) [All fields], AND (“TRK inhibitors” [“Larotrectinib, Entrectinib”]) [All fields], AND (“RET inhibitor” [“Selpercatinib”]) [All fields], and (“chemotherapy with Paclitaxel with Carboplatin or radiotherapy”) [All fields]. We also manually searched for any potential eligible study.
We used Endnote (Clarivate Analytics, Philadelphia, PA, USA) to remove duplicate data, and all retrieved studies were screened for titles/abstracts and full-text articles to assess relevance to the present meta-analysis. The publications reporting the highest number of patients were included in the studies derived from the same centre, with the possibility of data overlap.

2.4. Data Extraction

Two investigators independently reviewed the study selection. The investigators resolved any discrepancies through discussion. We recorded the information on the following attributes of included studies: previously given treatments, criteria for tumour response [complete response (CR), partial response (PR), stable disease (SD), overall response rate (ORR), disease control rate (DCR)], toxicity profile, biomarker analysis and reported progression-free survival (PFS) and overall survival (OS) rates.

2.5. Quality Assessment

A methodological index for non-randomised studies (MINORS) and a JBI critical appraisal checklist were applied to single-arm and retrospective studies [14,15]. The MINORS score consists of 8 items that are zero if not reported, 1 point when reported but inadequate and 2 points when adequately said. The maximum MINOR score is 16 points, and we categorised as poor a score <8, fair = 8 to 12 while excellent 13 to 16 scored points.

2.6. Statistical Methods

Pooled PR, SD and ORR analysis was performed using R version 3.6.3. For binary endpoints (PFS and OS), we used arcsine log transformation along with inverse probability weighting (IPW). The random-effect model was utilised because it harboured a more significant standard error in the pooled estimate, making it suitable for conflicting or erratic estimates. The chi-square test (Cochrane Q test) calculated I-squared (I2) to measure heterogeneity among studies. A p value less than 0.1 was considered significant heterogeneity. I2 values ≥ 50% were suggestive of high heterogeneity. Publication bias was calculated using Begg’s test and Egger’s test. A p value < 0.05 was weighed up as statistically significant.

3. Results

The electronic search revealed 619 relevant citations. After screening, 263 full-text articles were retrieved for further assessment. Finally, 47 studies (26 prospective phase II trials met MINORS and 21 retrospective studies met JBI critical appraisal checklist criteria) were identified (Figure 1) (Appendix A, Table A1 and Table A2); the total population of these studies was 980 patients [8,9,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60].
The median age of the whole population was 62.9 years (range: 58–77). The majority of the population was previously heavily treated. Patient characteristics are given in Table 1 (Appendix A Table A3).
All studies examined were published from 2008 to 2024. Lenvatinib monotherapy was used in 11/47 studies (23.4%) and with pembrolizumab in five studies (10.6%). BRAF inhibitors, with DT, were used in ten studies (21.3%), Encorafenib plus binimetinib in one (2.1%) and vemurafenib in one study (2.1%). IO, +/− TKI, or DT radiation, was used in ten studies (21.3%).

3.1. Overall Response Rates (ORR) and Disease Control Rates (DCR)

The pooled ORR was seen in 291/980 patients (29.7% [95% CI, 25.4–34.2%; I2 = 42.4%; p = 0.0009]) (Figure 2).
In a subgroup analysis, studies using DT showed a significantly higher pooled ORR of 64.9% (95% CI, 46.3–83.4%; I2 = 77.5%; p < 0.001), while in studies using lenvatinib plus pemrolizumab (LP), the pooled ORR was 42% (95% CI, 28.6–55.3%; I2 = 55.8%; p = 0.04). Lenvatinib and other TKI monotherapy or IO alone did not result in meaningful ORR.
The pooled DCR was seen in 497 patients (51.0% [95% CI, 44.0–59%; I2 = 85.2%; p < 0.0001]). SD was obtained in 206 (21.0%). In the subgroup analysis, higher DCR rates were seen with DT (74.4% [95% CI, 57.0–81.0%]) and LP (64.2% [95% CI, 48.0–78.0%]).

3.2. Survival Outcomes

In total, 489/980 (49.9%) deaths were reported. A significant number of patients remained alive relative to baseline (mean difference [MD], 2.07 [95% CI: 1.90–2.24; I2 = 88.6%; p < 0.0001]) (Figure 3).
The pooled median OS was 7.2 months (95% CI: 5.6–8.8 months I2 = 87.8%; p < 0.0001); the cumulative OS rate at 6 months, 9 months, 12 months and 24 months was 58.2%, 44.3%, 38.5% and 21.2%, respectively (Appendix A Figure A1). In the subgroup analysis, improved OS rates were observed in DT with and without pembrolizumab (median OS 11.2 months, 95% CI: 5.3–17.1 months) and LP regimens (median OS 14.4 months, 95% CI: 7.6–21.1 months). Only two studies of lenvatinib monotherapy showed any OS benefit [16,17].
A total of 329/980 (33.6%) PFS outcomes were reported. Pooled PFS significantly improved relative to baseline (MD 1.50 [95% CI: 1.33–1.68, I2 = 90.9%; p < 0.0001]), while the median PFS was 5.4 months (95% CI, 4.0–6.7 months); I2 = 97.9%; p < 0.0001 (Figure 4), with the cumulative PFS rate at 6, 9, 12 and 24 months being 47.2%, 32.7%, 26.1% and 9.2%, respectively.
In the subgroup analysis, improved PFS rates were seen with DT with and without pembrolizumab (median PFS 12.1 months, 95% CI: 7.5–15.9 months) and LP (median PFS 10.0 months, 95% CI: 5.3–14.6 months). Only one study of lenvatinib monotherapy resulted in PFS improvement [16].
RT was administered to 76 out of 980 patients (7.2%). The RT regimen included a daily dose of 2 Gy per fraction over 33 days (totalling 66 Gy) combined with weekly paclitaxel, with or without pazopanib, in 71 patients ORR 31%) [30]. Alternatively, a bi-daily fractionation schedule was used in 5 patients [53], delivering 3.5 Gy per fraction at intervals of more than 6 h over 2 consecutive days (totalling 14 Gy) combined with pembrolizumab (ORR 40%).

3.3. Biomarker Analysis

Biomarker analysis was performed in 24/47 studies (51.1%) (Appendix A Table A3).
BRAF V600 mutations were reported in 18 out of 47 studies (38.3%), and 144 out of 980 patients (14.7%) were identified as BRAF mutated.
-
PIK3CA status was mentioned in 4 studies (8.5%) and was positive in 10 patients (1.0%).
-
RAS (KRAS and NRAS) status was reported in 4 studies (8.5%) and was positive in 11 patients (1.1%).
-
NTRK1/3 was mentioned in 1 study (2.1%) and was positive in 7 patients (0.7%).
-
P53 status was reported in 3 studies (6.3%) and was detected in 10 patients (1.0%).
-
Programmed death ligand-1 (PDL-1) status was reported in 10 studies (21.3%) and was positive in 90 patients (9.2%).

3.4. Toxicity Profile

Grade 3 and above toxicity was reported for 928/980 (94.7%) patients (Table 2).
In lenvatinib studies, the predominant grade 3 and above toxicities were hypertension (16.4%), diarrhoea (11.3%), loss of appetite (9.7%) and hand-foot syndrome (8.7%). Dose modifications, interruptions and withdrawal of lenvatinib were observed at rates of 30.2%, 22.6% and 8.7%, respectively.
For DT studies, notable grade 3 and above toxicities included anaemia (11.1%), fatigue (8.7%), hypertension (6.7%) and pneumonitis (6.77%). Dose adjustments, interruptions and withdrawal of DT occurred at 15%, 16.5% and 4.4%, respectively. Immunotherapy studies revealed lipase/amylase elevation (9.1%), adrenal insufficiency (3.0%), dermatitis (2.1%) and acute heart failure (2.1%) as common grade 3 and above toxicities.
The resultant funnel plot showed significant heterogeneity (Egger’s test p < 0.001) (Appendix A Figure A2).

4. Discussion

Locally advanced, recurrent and metastatic ATC remains one of the most challenging malignancies due to its highly aggressive nature and historically limited therapeutic options, which have primarily included systemic chemotherapy and neck irradiation. More recently, there has been a shift towards targeted therapies and IO, offering new avenues for treatment.
This meta-analysis represents the first comprehensive evaluation of various therapeutic approaches in ATC, encompassing mutationally targeted therapies, IO and novel chemotherapeutic agents to provide insights into therapeutic decision-making within this complex treatment landscape. In contrast, the prior meta-analysis by Huang D et al. focused solely on the efficacy of lenvatinib [7].
The observed ORR of 64.9% in patients receiving dual-targeted therapy (DT) underscores the potent efficacy of BRAF/MEK inhibition, consistent with preclinical and clinical data demonstrating the oncogenic reliance of ATC on the MAPK pathway [36,37]. Similarly, the combination of lenvatinib and pembrolizumab achieved an ORR of 42%, supporting the therapeutic potential of combining angiogenesis inhibition with immune checkpoint blockade in ATC. DCRs were also notable, with DT achieving 74.4% and lenvatinib plus pembrolizumab reaching 64.2%, suggesting that durable disease stabilisation is possible in patients harbouring actionable mutations. However, despite these encouraging response rates, the OS remains limited at 7.2 months, with the most extended median OS survival observed in DT-treated patients (11.2 months) and those receiving lenvatinib atinib plus pembrolizumab (14.4 months).

4.1. Next-Generation Sequencing (NGS) and Molecular-Driven Treatment

Current guidelines emphasise the routine use of biomarker analysis [61,62], given the high prevalence of genomic alterations in ATC. The most frequently observed mutations include BRAF V600E and MEK alterations (40.50%) [38], TP53 (63%) [55], RET and RAS mutations (22%) [56] and TERT promoter mutations (75%) [62]. Additionally, alterations in PIK3CA (18%) [53], EIF1AX (14%) and PTEN (14%) [63] have been documented, while the prevalence of NTRK fusions and other “non-actionable mutations” such as “NESTIN, CCND1, POU5F1, MCL1, MYBL2, IQGAP1, SOX2 and NANOG— remains unknown [64].
In the present meta-analysis, biomarker assessment was reported in only 51.1% of the included studies. Among the patient population, 14.7% were identified as BRAF V600E-mutated, while 9.2% were PD-L1 positive. NTRK1/3 fusions were detected in only 0.7% of cases (12 ATC cases among 83 total NTRK fusion-positive tumours), highlighting a rare but clinically significant subgroup that may benefit from TRK inhibitors. In this meta-analysis, entrectinib demonstrated an ORR of 20%, while larotrectinib achieved an ORR of 29%.
Additionally, the presence of RET fusions in some studies supports the potential role of selective RET inhibitors such as selpercatinib, as evidenced in the LIBRETTO-001 trial [65]. However, this trial was not included in the current meta-analysis, as it only enrolled two ATC patients among 166 individuals with RET-driven tumours. The detection of PIK3CA (1.0%) and RAS mutations (1.1%) suggests alternative pathways for targeted therapy. However, their clinical relevance remains uncertain due to the absence of approved targeted agents for these mutations in ATC. These findings highlight the necessity of incorporating comprehensive genomic profiling into routine clinical practice to optimise treatment selection and improve patient outcomes. The limited availability of molecular data in many studies underscores the need for standardised biomarker assessment in ATC to define treatment strategies better and identify patients most likely to benefit from mutational driven and IO approaches.

4.2. PD-L1 Expression in ATC

A total 21.3% of included studies reported PD-L1 expression, with 9.2% of ATC patients identified as PD-L1 positive. This is pretty low, as 22–29% of ATC tumour samples have been reported to express PD-L1 [66].
One of the primary challenges in utilising PD-L1 as a predictive biomarker in ATC is the absence of standardised cutoff values for clinical decision-making. Unlike non-small-cell lung cancer (NSCLC) or head and neck squamous cell carcinoma (HNSCC), where PD-L1 expression thresholds (1–50%) have been validated for pembrolizumab, no established ATC-specific cutoff values currently exist [67].
While our analysis demonstrates that lenvatinib combined with pembrolizumab achieves an ORR of 42% and a median OS of 14.4 months, these findings emphasise further investigation. Future clinical trials should prioritise standardising PD-L1 scoring thresholds and explore potential synergistic treatment strategies, including its combination with angiogenesis inhibitors or RT, to enhance IO efficacy in ATC.

4.3. Radiation Therapy in ATC

In this meta-analysis, RT was administered only to 7.2% of patients using two fractionation regimens. Most patients (n = 71) received conventional fractionation, consisting of a total dose of 66 Gy delivered in 33 fractions (2 Gy per fraction) over 33 days, combined with weekly paclitaxel, with or without pazopanib. In contrast, a smaller cohort (n = 5) underwent hypofractionated RT (HypoRT), receiving 14 Gy in 4 fractions (3.5 Gy per fraction) over two consecutive days in combination with pembrolizumab. HypoRT achieved a higher ORR (40%) than conventional RT (31%) [30,53].
This finding aligns with recent work by Oliinyk et al., [68] who evaluated the use of hypoRT in ATC patients treated with 3DRT or IMRT. In their cohort of 17 ATC patients, a cumulative radiation dose of <30 Gy was delivered, with four patients receiving concurrent chemotherapy (carboplatin with paclitaxel or doxorubicin weekly). The median OS was four months (range: 1–12 months), with survival rates of 82%, 55% and 36% at one, three and six months, respectively. Subsequent authors performed a systematic review of hypoRT in ATC, supporting its role as an integral component of multimodal treatment and demonstrating promising clinical outcomes. Given the increasing evidence favouring hypoRT, its incorporation into future clinical trials should be explored, particularly in combination with molecularly targeted agents and IO.
Toxicity profiles, which differed across various therapeutic approaches, were also examined. The studies involving Lenvatinib demonstrated the highest incidence of grade 3 and above adverse effects among all studies, like hypertension (16.4%) and diarrhoea (11.3%), leading to a higher frequency of dose modifications and interruptions. DT studies exhibited anaemia (11.1%) as a significant adverse effect. IO studies revealed lipase/amylase elevation (9.4%).
A recent meta-analysis by Cao and colleagues [69], based on six prospective trials of TKI monotherapy involving a pooled population of 140 ATC patients, reported a median OS of 4.8 months and a median PFS of 2.6 months. In contrast, our meta-analysis demonstrated a median OS of 7.2 months and a median PFS of 5.4 months, highlighting the enhanced efficacy of dual therapy (DT) with or without immunotherapy (IO) and combinations of TKI with IO. Combination therapy as a potential therapeutic direction for ATC patients is also supported by the recently published study by Zheng X et al. using TKI (erlotinib) with chemotherapy [70]. This study was not included in meta-analysis due to lack of full details.
The strengths of the present meta-analysis were as follows: (1) a comprehensive literature search, (2) rigorous inclusion and exclusion criteria, (3) a large sample size with inclusion of 47 studies involving 980 patients contributes to the robustness of the analysis, providing a substantial dataset.
Limitations: Although IPW and standard effect size were used to reduce bias and enable comparisons, the limited number of included randomised studies and variability in study designs among the included studies may have influenced the OS and PFS outcomes.

5. Conclusions

This meta-analysis comprehensively evaluated therapeutic strategies for locally advanced, recurrent and metastatic ATC, focusing on novel mutational-targeted therapy, IO, chemotherapy and RT. While dual-targeted therapy (BRAF/MEK inhibition) demonstrated the highest ORR at 64.9% and lenvatinib plus pembrolizumab achieved a 42% ORR, OS remains a challenge, with median OS reaching only 11.2 months for DT and 14.4 months for lenvatinib plus pembrolizumab. Despite these improvements in treatment outcomes, our meta-analysis showed underutilisation of biomarker-driven treatment, with genomic profiling reported in just over half of the included studies (51.1%), highlighting the critical need for standardised molecular testing. The limited data regarding PD-L1 expression (9.2% of patients) and the absence of validated cutoffs in ATC further complicate patient selection for IO. HypoRT showed promise with a higher ORR (40%) than conventional RT (31%), suggesting its potential within multimodal approaches. While combination therapy appeared superior compared to historical TKI monotherapy data, toxicity profiles varied across treatments, necessitating careful monitoring. These findings underscore the urgent need for well-designed prospective trials investigating optimal sequencing and combinations of DT, IO and RT, incorporating standardised molecular profiling (including NGS) to guide treatment decisions, validating PD-L1 cutoffs for IO and further evaluating the role of HypoRT in combination with systemic therapies. Such trials should also prioritise evaluating toxicity management strategies and exploring novel mutational-driven and targeted therapies to improve long-term outcomes for ATC patients.

Author Contributions

Conceptualisation, M.A.T. and D.H.; methodology, M.A.T., M.H.R. and Y.A.I.; software, W.M., M.A.T. and Y.A.I.; validation, D.H., B.E. and M.H.R.; formal analysis, M.A.T. and W.M.; investigation, M.A.T., D.H., B.E. and Y.A.I.; resources, M.A.T.; data curation, Y.A.I., K.K.N. and M.H.R.; writing—original draft preparation, M.A.T., K.K.N. and W.M.; writing—review and editing, M.A.T. and K.K.N.; visualisation, M.H.R. and B.E.; supervision, W.M.; project administration, M.A.T.; funding acquisition, none. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data supporting the results of this study are available within Appendix A and upon request from corresponding author. Raw data used to conduct the analysis is stored and available for validation purposes.

Acknowledgments

We acknowledge Muhammad Abdullah Tanveer/Katarina N for administrative and technical support.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ATCAnaplastic thyroid cancer
ORROverall response rate
DCRDisease control rate
MDMean difference
DTDabrafenib/trametinib
LPLenvatinib/pembrolizumab
OSOverall survival
PFSProgression-free survival
TKITyrosine kinase inhibitors
PD/PDL1Programmed death/ programmed death ligand 1
IOImmunotherapy
PRISMAPreferred reporting items for systemic reviews and meta-analyses
MOOSEMeta analyses for observational studies in epidemiology
CRComplete response
PRPartial response
SDStable disease
IPWInverse probability weighting
HypoRTHypofractionated radiotherapy
MINORSMythological index for non-randomised studies

Appendix A

Table A1. Checklist for critically appraising retrospective studies by JBI.
Table A1. Checklist for critically appraising retrospective studies by JBI.
Study/Year/NatureQ1Q2Q3Q4Q5Q6Q7Q8Q9Q10Overall
Yamazaki H, et al. [18] YesYesYesYesYes YesunclearYesYesYesInclude
Sparano C, et al. [19] YesYesYesYesYes YesYesYesYesYesInclude
Park J, et al. [20] YesYesYesYesYes YesYesYesYesYesInclude
Iwasaki H, et al. [21] YesYesYesYesYesYes unclearYesYesYesInclude
Ishihara S, et al. [22] YesYesYesYesYesYesYes YesYesYesInclude
Fukuda, N et al. [23] YesYesYesYesYes YesYesYesYesYesInclude
Kim SY, et al. [24] YesYesYesYesYes YesYesYesYesYesInclude
Iyer PC, et al. [26] YesYesYesYesYes YesYesYesYesYesInclude
Lorimer C, et al. [8]YesYesYesYesYes YesYesYesYesYesInclude
Bueno F, et al. [38] YesYesYesYesYes YesYesYesYesYesInclude
da Silva TN, et al. [39] YesYesYesYesYes UnclearYesYesYesYesInclude
Zhao X, et al. [40]YesYesYesYesYes YesYesYesYesYesInclude
Hatashima A, et al. [42] YesYesYesYesYes YesYesYesYesYesInclude
Dierks C, et al. [45]YesYesYesYesYes YesYesYesYesYesInclude
Iyer PC, et al. [47] YesYesYesYesYes YesYesYesYesYesInclude
Harris EJ, et al. [50]YesYesYesYesYes YesunclearYesYesYesInclude
Song Y, et al. [59] YesYes YesYesYesYesYesYesYesYesInclude
Soll D, et al. [56] YesYesYesYesYesUnclearYesYesYesYesInclude
Evans LK, et al. [55] YesYesYesYesYesYesunclearYesYesYesInclude
Tan JSH, et al. [53] YesYesYesYesYesYesYesYesYesYesInclude
Hamidi S, et al. [52] YesYesYesYesYesYesYesYesYesYesInclude
Table A2. A MINORS index for included non-randomised trials.
Table A2. A MINORS index for included non-randomised trials.
Study/Year/Nature12345678Total
Higashiyama T, et al. (HOPE) [16]2220221213
Wirth LJ, et al. [17]2120212212
Takahashi S, et al. [25] 2222022012
Iyer PC, et al. [47] 2222022012
Ito Y et al. [27] 2212021010
Savvides P, et al. [28] 2222222014
Bible KC, et al. [29]2222022012
Pennell NA, et al. [31] 2212021010
Zhao Q, et al. [32] 2222022216
Doebele, et al. [51] 2222022216
Ha HT, et al. [33]2222022012
Mooney CJ, et al. [34] 2222022216
Sosa JA, et al. [35]2212021010
Subbiah V, et al. [36] 2222022216
ROAR Study [37]2222022216
Hyman DM, et al. [41] 2120212212
Hatashima A, et al. [42]2120212212
Canabillas ME, et al. [57]2120212212
Capdevila J, et al. [43]2222022216
Lorch JH, et al. [44]2222022216
ATLEP trial [46]2222022216
Lim SM, et al. [48] 2120212212
Sehgal L, et al. [58] 2112112212
Tahara M, et al. [54]2222222216
Waguespack SG, et al. [9]2222222216
Hanna GJ, et al. [49]2120212212
Sehgal K, et al. [60] 2222022216
Table A3. A demographic data on mutations analysed and event rates.
Table A3. A demographic data on mutations analysed and event rates.
Study/Year/NatureFemale/MaleMutation Analysed (n) Median Follow Up (Months)Progression (Events)Deaths (Events)
Higashiyama T, et al. (HOPE) [16]
Prospective 2022
NRNR121137
Wirth LJ, et al. [17]
Prospective 2021
13/21NR142623
Yamazaki H, et al. [18]
Retrospective 2021
9/11NR241518
Sparano C, et al. [19]
Retrospective 2021
9/6NR128NR
Park J, et al. [20]
Retrospective 2021
NRNR648
Iwasaki H, et al. [21]
Retrospective 2021
14/18NR601225
Ishihara S, et al. [22]
Retrospective 2021
3/7NR14411
Fukuda, N et al. [23]
Retrospective 2020
4/9NR1446
Kim M, et al. [24]
Retrospective 2020
5/9NR1416
Takahashi S, et al. [25]
Prospective 2019
6/11NR121NR
Iyer PC, et al. [26]
Retrospective 2018
NRNR1448
Ito Y et al. [27]
Prospective 2017
5/6NR1249
Savvides P, et al. [28]
Prospective 2013
7/13NR24NR19
Bible KC, et al. [29]
Prospective 2012
10/5NR121513
NRG/RTOG 0912 [30] Prospective 2023 37/34NR1215 Pazopanib plus XRT 11 in XRT33 Pazopanib plus XRT 30 in XRT
Pennell NA, et al. [31] Prospective 2008NRNR17.555
Zhao Q, et al. [32] Prospective 2022NRNR10.72NR
Ha HT, et al. [33]
Prospective 2010
5/6NR616
Mooney CJ, et al. [34]
Prospective 2009
10/16NR81921
Sosa JA, et al. [35]
Prospective 2014
NRNR12NR20 CP/fosbretabulin 8 CP
Subbiah V, et al. [36]
Prospective 2018
10/6BRAF V600 = 15
No mutation = 1
11.752NR
ROAR Study [37]
Prospective 2022
20/16BRAF V600 = 3311.12224
Lorimer C, et al. [8]
Retrospective 2023
NRBRAF V600 = 17123NR
Bueno F, et al. [38]
Retrospective 2023
2/3BRAF V600 = 56002
da Silva TN, et al. [39]
retrospective 2023
NRBRAF V600 = 2712NR18
Zhao X, et al. [40]
Retrospective 2023
27/30BRAF V600 = 57241149
Hyman DM, et al. [41]
Prospective 2015
3/4BRAF V600 = 7124NR
Hatashima A, et al. [42]
Retrospective 2022
6/7BRAF V600 = 7
PDL1 = 10
13.5118
Capdevila J, et al. [43]
Prospective 2020
15/23BRAF V600 = 12261022
Lorch JH, et al. [44]
Prospective 2020
NRPDL1 = 1024NRNR
Dierks C, et al. [45]
Retrospective 2021
3/3PDL1 = 64016
ATLEP trial [46]
Prospective 2022
NRPDL1 = 2920322
Iyer PC, et al. [47]
Retrospective 2018
4/8PDL1 = 1040.5117
Lim SM, et al. [48]
Prospective 2013
NR NR2067
Hanna GJ, et al. [49]
Prospective 2018
NRPIK3CA = 7255NR
Harris EJ, et al. [50]
Retrospective 2019
1/4BRAF V600 = 1
PIK3CA = 1
16.935
Waguespack SG, et al. [9]
Prospective 2022
5/2NTRK1 = 3
NTRK3 = 4
2455
Doebele RC, et al. [51]
Prospective 2020
NRNR12.955
Hamidi S, et al. [52]
Retrospective 2024
NRBRAF V600 = 71110NRNR
Tan JSH, et al. [53]
Retrospective 2024
4/1BRAF V600 = 1
NRAS p.Q61K = 2
KRAS p.L19F = 1
TP53 p.M169 = 2
PIK3CA p.H1047R = 1
PDL1 = 3
32.633
Tahara M, et al. [54]
Prospective 2024
NRBRAF V600 = 511.544
Evans LK, et al. [55]
Retrospective 2025
22/19BRAF V600 = 15
KRAS p.G12R = 1
TP53 = 9
NRAS = 2
P63 = 2
PDL1 = 5
65NR33
Soll D, et al. [56]
Retrospective 2024
2/3BRAF V600 = 2
KRAS p.G12R = 1
TP53 p.P153fs = 1
PIK3CA p.E545K = 1
PDL1 = 3
12.714
Cabanillas ME, et al. [57]
Prospective 2024
22/20BRAF V600 = 18
MEK = 21
VEGF = 3
42.13129
Sehgal K, et al. [58]
Prospective 2024
NRNRNR18NR
Song Y, et al. [59]
Retrospective 2024
11/7BRAF V600 = 9
PDL1= 9
14NR10
Sehgal K, et al. [60]
Prospective 2024
NR BRAF V600 = 3
NRAS codon 61 = 4
PDL1 = 5
24 84
Figure A1. Cumulative overall survival.
Figure A1. Cumulative overall survival.
Cancers 17 00777 g0a1
Figure A2. A Funnel plot of publication bias.
Figure A2. A Funnel plot of publication bias.
Cancers 17 00777 g0a2

References

  1. Casali, P.G.; Trama, A. Rationale of the rare cancer list: A consensus paper from the Joint Action on Rare Cancers (JARC) of the European Union (EU). ESMO Open 2020, 5, e000666. [Google Scholar] [CrossRef]
  2. Molinaro, E.; Romei, C.; Biagini, A.; Sabini, E.; Agate, L.; Mazzeo, S.; Materazzi, G.; Sellari-Franceschini, S.; Ribechini, A.; Torregrossa, L.; et al. Anaplastic thyroid carcinoma: From clinicopathology to genetics and advanced therapies. Nat. Rev. Endocrinol. 2017, 13, 644–660. [Google Scholar] [CrossRef]
  3. Nagaiah, G.; Hossain, A.; Mooney, C.J.; Parmentier, J.; Remick, S.C. Anaplastic thyroid cancer: A review of epidemiology, pathogenesis, and treatment. J. Oncol. 2011, 2011, 542358. [Google Scholar] [CrossRef] [PubMed]
  4. Smallridge, R.C.; Ain, K.B.; Asa, S.L.; Bible, K.C.; Brierley, J.D.; Burman, K.D.; Kebebew, E.; Lee, N.Y.; Nikiforov, Y.E.; Rosenthal, M.S.; et al. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid 2012, 22, 1104–1139. [Google Scholar] [CrossRef] [PubMed]
  5. Maniakas, A.; Dadu, R.; Busaidy, N.L.; Wang, J.R.; Ferrarotto, R.; Lu, C.; Williams, M.D.; Gunn, G.B.; Hofmann, M.C.; Cote, G.; et al. Evaluation of Overall Survival in Patients With Anaplastic Thyroid Carcinoma, 2000–2019. JAMA Oncol. 2020, 6, 1397–1404. [Google Scholar] [CrossRef]
  6. Wu, S.S.; Lamarre, E.D.; Yalamanchali, A.; Brauer, P.R.; Hong, H.; Reddy, C.A.; Yilmaz, E.; Woody, N.; Ku, J.A.; Prendes, B.; et al. Association of Treatment Strategies and Tumor Characteristics With Overall Survival Among Patients With Anaplastic Thyroid Cancer: A Single-Institution 21-Year Experience. JAMA Otolaryngol. Head. Neck Surg. 2023, 149, 300–309. [Google Scholar] [CrossRef] [PubMed]
  7. Huang, D.; Zhang, J.; Zheng, X.; Gao, M. Efficacy and Safety of Lenvatinib in Anaplastic Thyroid Carcinoma: A Meta-Analysis. Front. Endocrinol. 2022, 13, 920857. [Google Scholar] [CrossRef]
  8. Lorimer, C.; Cheng, L.; Chandler, R.; Garcez, K.; Gill, V.; Graham, K.; Grant, W.; Sardo Infirri, S.; Wadsley, J.; Wall, L.; et al. Dabrafenib and Trametinib Therapy for Advanced Anaplastic Thyroid Cancer—Real-World Outcomes From UK Centres. Clin. Oncol. (R Coll. Radiol.) 2023, 35, e60–e66. [Google Scholar] [CrossRef] [PubMed]
  9. Waguespack, S.G.; Drilon, A.; Lin, J.J.; Brose, M.S.; McDermott, R.; Almubarak, M.; Bauman, J.; Casanova, M.; Krishnamurthy, A.; Kummar, S.; et al. Efficacy and safety of larotrectinib in patients with TRK fusion-positive thyroid carcinoma. Eur. J. Endocrinol. 2022, 186, 631–643. [Google Scholar] [CrossRef] [PubMed]
  10. Vodopivec, D.M.; Hu, M.I. RET kinase inhibitors for RET-altered thyroid cancers. Ther. Adv. Med. Oncol. 2022, 14, 17588359221101691. [Google Scholar] [CrossRef] [PubMed]
  11. Ma, M.; Lin, B.; Wang, M.; Liang, X.; Su, L.; Okose, O.; Li, J. Immunotherapy in anaplastic thyroid cancer. Am. J. Transl. Res. 2020, 12, 974–988. [Google Scholar]
  12. Page, M.J.; Moher, D.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. PRISMA 2020 explanation and elaboration: Updated guidance and exemplars for reporting systematic reviews. BMJ 2021, 372, n160. [Google Scholar] [CrossRef] [PubMed]
  13. Faber, T.; Ravaud, P.; Riveros, C.; Perrodeau, E.; Dechartres, A. Meta-analyses including non-randomized studies of therapeutic interventions: A methodological review. BMC Med. Res. Methodol. 2016, 16, 35. [Google Scholar] [CrossRef]
  14. Slim, K.; Nini, E.; Forestier, D.; Kwiatkowski, F.; Panis, Y.; Chipponi, J. Methodological index for non-randomized studies (minors): Development and validation of a new instrument. ANZ J. Surg. 2003, 73, 712–716. [Google Scholar] [CrossRef] [PubMed]
  15. Zeng, X.; Zhang, Y.; Kwong, J.S.; Zhang, C.; Li, S.; Sun, F.; Niu, Y.; Du, L. The methodological quality assessment tools for preclinical and clinical studies, systematic review and meta-analysis, and clinical practice guideline: A systematic review. J. Evid. Based Med. 2015, 8, 2–10. [Google Scholar] [CrossRef] [PubMed]
  16. Higashiyama, T.; Sugino, K.; Hara, H.; Ito, K.I.; Nakashima, N.; Onoda, N.; Tori, M.; Katoh, H.; Kiyota, N.; Ota, I.; et al. Phase II study of the efficacy and safety of lenvatinib for anaplastic thyroid cancer (HOPE). Eur. J. Cancer 2022, 173, 210–218. [Google Scholar] [CrossRef] [PubMed]
  17. Wirth, L.J.; Brose, M.S.; Sherman, E.J.; Licitra, L.; Schlumberger, M.; Sherman, S.I.; Bible, K.C.; Robinson, B.; Rodien, P.; Godbert, Y.; et al. Open-Label, Single-Arm, Multicenter, Phase II Trial of Lenvatinib for the Treatment of Patients With Anaplastic Thyroid Cancer. J. Clin. Oncol. 2021, 39, 2359–2366. [Google Scholar] [CrossRef] [PubMed]
  18. Yamazaki, H.; Iwasaki, H.; Suganuma, N.; Toda, S.; Masudo, K.; Nakayama, H.; Rino, Y.; Masuda, M. Inflammatory biomarkers and dynamics of neutrophil-to-lymphocyte ratio in lenvatinib treatment for anaplastic thyroid carcinoma. Gland Surg. 2021, 10, 852–860. [Google Scholar] [CrossRef] [PubMed]
  19. Sparano, C.; Godbert, Y.; Attard, M.; Do Cao, C.; Zerdoud, S.; Roudaut, N.; Joly, C.; Berdelou, A.; Hadoux, J.; Lamartina, L.; et al. Limited efficacy of lenvatinib in heavily pretreated anaplastic thyroid cancer: A French overview. Endocr. Relat. Cancer 2021, 28, 15–26. [Google Scholar] [CrossRef] [PubMed]
  20. Park, J.; Jung, H.A.; Shim, J.H.; Park, W.Y.; Kim, T.H.; Lee, S.H.; Kim, S.W.; Ahn, M.J.; Park, K.; Chung, J.H. Multimodal treatments and outcomes for anaplastic thyroid cancer before and after tyrosine kinase inhibitor therapy: A real-world experience. Eur. J. Endocrinol. 2021, 184, 837–845. [Google Scholar] [CrossRef] [PubMed]
  21. Iwasaki, H.; Toda, S.; Murayama, D.; Kato, S.; Matsui, A. Relationship between adverse events associated with lenvatinib treatment for thyroid cancer and patient prognosis. Mol. Clin. Oncol. 2021, 14, 28. [Google Scholar] [CrossRef] [PubMed]
  22. Ishihara, S.; Onoda, N.; Noda, S.; Tauchi, Y.; Morisaki, T.; Asano, Y.; Kashiwagi, S.; Takashima, T.; Ohira, M. Treatment of anaplastic thyroid cancer with tyrosine kinase inhibitors targeted on the tumor vasculature: Initial experience in clinical practice. Endocr. J. 2021, 68, 63–68. [Google Scholar] [CrossRef]
  23. Fukuda, N.; Toda, K.; Fujiwara, Y.U.; Wang, X.; Ohmoto, A.; Urasaki, T.; Hayashi, N.; Sato, Y.; Nakano, K.; Yunokawa, M.; et al. Neutrophil-to-Lymphocyte Ratio as a Prognostic Marker for Anaplastic Thyroid Cancer Treated With Lenvatinib. In Vivo 2020, 34, 2859–2864. [Google Scholar] [CrossRef]
  24. Kim, M.; Ahn, J.; Song, D.E.; Yoon, J.H.; Kang, H.C.; Lim, D.J.; Kim, W.G.; Kim, T.Y.; Kim, W.B.; Shong, Y.K.; et al. Real-world experience of lenvatinib in patients with advanced anaplastic thyroid cancer. Endocrine 2021, 71, 427–433. [Google Scholar] [CrossRef] [PubMed]
  25. Takahashi, S.; Kiyota, N.; Yamazaki, T.; Chayahara, N.; Nakano, K.; Inagaki, L.; Toda, K.; Enokida, T.; Minami, H.; Imamura, Y.; et al. A Phase II study of the safety and efficacy of lenvatinib in patients with advanced thyroid cancer. Future Oncol. 2019, 15, 717–726. [Google Scholar] [CrossRef]
  26. Iyer, P.C.; Dadu, R.; Ferrarotto, R.; Busaidy, N.L.; Habra, M.A.; Zafereo, M.; Gross, N.; Hess, K.R.; Gule-Monroe, M.; Williams, M.D.; et al. Real-World Experience with Targeted Therapy for the Treatment of Anaplastic Thyroid Carcinoma. Thyroid 2018, 28, 79–87. [Google Scholar] [CrossRef] [PubMed]
  27. Ito, Y.; Onoda, N.; Ito, K.I.; Sugitani, I.; Takahashi, S.; Yamaguchi, I.; Kabu, K.; Tsukada, K. Sorafenib in Japanese Patients with Locally Advanced or Metastatic Medullary Thyroid Carcinoma and Anaplastic Thyroid Carcinoma. Thyroid 2017, 27, 1142–1148. [Google Scholar] [CrossRef] [PubMed]
  28. Savvides, P.; Nagaiah, G.; Lavertu, P.; Fu, P.; Wright, J.J.; Chapman, R.; Wasman, J.; Dowlati, A.; Remick, S.C. Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid 2013, 23, 600–604. [Google Scholar] [CrossRef]
  29. Bible, K.C.; Suman, V.J.; Menefee, M.E.; Smallridge, R.C.; Molina, J.R.; Maples, W.J.; Karlin, N.J.; Traynor, A.M.; Kumar, P.; Goh, B.C.; et al. A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. J. Clin. Endocrinol. Metab. 2012, 97, 3179–3184. [Google Scholar] [CrossRef] [PubMed]
  30. Sherman, E.J.; Harris, J.; Bible, K.C.; Xia, P.; Ghossein, R.A.; Chung, C.H.; Riaz, N.; Gunn, G.B.; Foote, R.L.; Yom, S.S.; et al. Radiotherapy and paclitaxel plus pazopanib or placebo in anaplastic thyroid cancer (NRG/RTOG 0912): A randomised, double-blind, placebo-controlled, multicentre, phase 2 trial. Lancet Oncol. 2023, 24, 175–186. [Google Scholar] [CrossRef]
  31. Pennell, N.A.; Daniels, G.H.; Haddad, R.I.; Ross, D.S.; Evans, T.; Wirth, L.J.; Fidias, P.H.; Temel, J.S.; Gurubhagavatula, S.; Heist, R.S.; et al. A phase II study of gefitinib in patients with advanced thyroid cancer. Thyroid 2008, 18, 317–323. [Google Scholar] [CrossRef] [PubMed]
  32. Zhao, Q.; Feng, H.; Yang, Z.; Liang, J.; Jin, Z.; Chen, L.; Zhan, L.; Xuan, M.; Yan, J.; Kuang, J.; et al. The central role of a two-way positive feedback pathway in molecular targeted therapies-mediated pyroptosis in anaplastic thyroid cancer. Clin. Transl. Med. 2022, 12, e727. [Google Scholar] [CrossRef] [PubMed]
  33. Ha, H.T.; Lee, J.S.; Urba, S.; Koenig, R.J.; Sisson, J.; Giordano, T.; Worden, F.P. A phase II study of imatinib in patients with advanced anaplastic thyroid cancer. Thyroid 2010, 20, 975–980. [Google Scholar] [CrossRef] [PubMed]
  34. Mooney, C.J.; Nagaiah, G.; Fu, P.; Wasman, J.K.; Cooney, M.M.; Savvides, P.S.; Bokar, J.A.; Dowlati, A.; Wang, D.; Agarwala, S.S.; et al. A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid 2009, 19, 233–240. [Google Scholar] [CrossRef]
  35. Sosa, J.A.; Elisei, R.; Jarzab, B.; Balkissoon, J.; Lu, S.P.; Bal, C.; Marur, S.; Gramza, A.; Yosef, R.B.; Gitlitz, B.; et al. Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid 2014, 24, 232–240. [Google Scholar] [CrossRef] [PubMed]
  36. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.; Cabanillas, M.E.; Urbanowitz, G.; et al. Dabrafenib and Trametinib Treatment in Patients With Locally Advanced or Metastatic BRAF V600-Mutant Anaplastic Thyroid Cancer. J. Clin. Oncol. 2018, 36, 7–13. [Google Scholar] [CrossRef] [PubMed]
  37. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.C.; Cabanillas, M.E.; Boran, A.; et al. Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: Updated analysis from the phase II ROAR basket study. Ann. Oncol. 2022, 33, 406–415. [Google Scholar] [CrossRef]
  38. Bueno, F.; Smulever, A.; Califano, I.; Guerra, J.; Del Grecco, A.; Carrera, J.M.; Giglio, R.; Rizzo, M.; Lingua, A.; Voogd, A.; et al. Dabrafenib plus trametinib treatment in patients with anaplastic thyroid carcinoma: An Argentinian experience. Endocrine 2023, 80, 134–141. [Google Scholar] [CrossRef] [PubMed]
  39. da Silva, T.N.; Rodrigues, R.; Saramago, A.; Pires, C.; Rito, M.; Horta, M.; Martins, C.; Leite, V.; Cavaco, B.M. Target therapy for BRAF mutated anaplastic thyroid cancer: A clinical and molecular study. Eur. J. Endocrinol. 2023, 188, lvac011. [Google Scholar] [CrossRef] [PubMed]
  40. Zhao, X.; Wang, J.R.; Dadu, R.; Busaidy, N.L.; Xu, L.; Learned, K.O.; Chasen, N.N.; Vu, T.; Maniakas, A.; Eguia, A.A.; et al. Surgery After BRAF-Directed Therapy Is Associated with Improved Survival in BRAFV600EMutant Anaplastic Thyroid Cancer: A Single-Center Retrospective Cohort Study. Thyroid 2023, 33, 484–491. [Google Scholar] [CrossRef]
  41. Hyman, D.M.; Puzanov, I.; Subbiah, V.; Faris, J.E.; Chau, I.; Blay, J.Y.; Wolf, J.; Raje, N.S.; Diamond, E.L.; Hollebecque, A.; et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N. Engl. J. Med. 2015, 373, 726–736. [Google Scholar] [CrossRef] [PubMed]
  42. Hatashima, A.; Archambeau, B.; Armbruster, H.; Xu, M.; Shah, M.; Konda, B.; Lott Limbach, A.; Sukrithan, V. An Evaluation of Clinical Efficacy of Immune Checkpoint Inhibitors for Patients with Anaplastic Thyroid Carcinoma. Thyroid 2022, 32, 926–936. [Google Scholar] [CrossRef] [PubMed]
  43. Capdevila, J.; Wirth, L.J.; Ernst, T.; Ponce Aix, S.; Lin, C.C.; Ramlau, R.; Butler, M.O.; Delord, J.P.; Gelderblom, H.; Ascierto, P.A.; et al. PD-1 Blockade in Anaplastic Thyroid Carcinoma. J. Clin. Oncol. 2020, 38, 2620–2627. [Google Scholar] [CrossRef] [PubMed]
  44. Lorch, J.H.; Barletta, J.A.; Nehs, M.; Uppaluri, R.; Alexander, E.K.; Haddad, R.I. A phase II study of nivolumab (N) plus ipilimumab (I) in radioidine refractory differentiated thyroid cancer (RAIR DTC) with exploratory cohorts in anaplastic (ATC) and medullary thyroid cancer (MTC). J. Clin. Oncol. 2020, 38, 6513. [Google Scholar] [CrossRef]
  45. Dierks, C.; Seufert, J.; Aumann, K.; Ruf, J.; Klein, C.; Kiefer, S.; Rassner, M.; Boerries, M.; Zielke, A.; la Rosee, P.; et al. Combination of Lenvatinib and Pembrolizumab Is an Effective Treatment Option for Anaplastic and Poorly Differentiated Thyroid Carcinoma. Thyroid 2021, 31, 1076–1085. [Google Scholar] [CrossRef]
  46. Dierks, C.; Seufert, J.; Aumann, K.; Ruf, J.; Klein, C.; Kiefer, S.; Rassner, M.; Boerries, M.; Zielke, A.; la Rosee, P.; et al. Phase II ATLEP trial: Final results for lenvatinib/pembrolizumab in metastasized anaplastic and poorly differentiated thyroid carcinoma. Ann. Oncol. 2022, 33 (Suppl. S7), S750–S757. [Google Scholar] [CrossRef]
  47. Iyer, P.C.; Dadu, R.; Gule-Monroe, M.; Busaidy, N.L.; Ferrarotto, R.; Habra, M.A.; Zafereo, M.; Williams, M.D.; Gunn, G.B.; Grosu, H.; et al. Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J. Immunother. Cancer 2018, 6, 68. [Google Scholar] [CrossRef] [PubMed]
  48. Lim, S.M.; Chang, H.; Yoon, M.J.; Hong, Y.K.; Kim, H.; Chung, W.Y.; Park, C.S.; Nam, K.H.; Kang, S.W.; Kim, M.K.; et al. A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes. Ann. Oncol. 2013, 24, 3089–3094. [Google Scholar] [CrossRef] [PubMed]
  49. Hanna, G.J.; Busaidy, N.L.; Chau, N.G.; Wirth, L.J.; Barletta, J.A.; Calles, A.; Haddad, R.I.; Kraft, S.; Cabanillas, M.E.; Rabinowits, G.; et al. Genomic Correlates of Response to Everolimus in Aggressive Radioiodine-refractory Thyroid Cancer: A Phase II Study. Clin. Cancer Res. 2018, 24, 1546–1553. [Google Scholar] [CrossRef]
  50. Harris, E.J.; Hanna, G.J.; Chau, N.; Rabinowits, G.; Haddad, R.; Margalit, D.N.; Schoenfeld, J.; Tishler, R.B.; Barletta, J.A.; Nehs, M.; et al. Everolimus in Anaplastic Thyroid Cancer: A Case Series. Front. Oncol. 2019, 9, 106. [Google Scholar] [CrossRef]
  51. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. trial investigators. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef] [PubMed]
  52. Hamidi, S.; Iyer, P.C.; Dadu, R.; Gule-Monroe, M.K.; Maniakas, A.; Zafereo, M.E.; Wang, J.R.; Busaidy, N.L.; Cabanillas, M.E. Checkpoint Inhibition in Addition to Dabrafenib/Trametinib for BRAFV600E-Mutated Anaplastic Thyroid Carcinoma. Thyroid 2024, 34, 336–346. [Google Scholar] [CrossRef] [PubMed]
  53. Tan, J.S.H.; Tay, T.K.Y.; Ong, E.H.W.; Fehlings, M.; Tan, D.S.; Sukma, N.B.; Chen, E.X.; Sng, J.H.; Yip, C.S.P.; Lim, K.H.; et al. Combinatorial Hypofractionated Radiotherapy and Pembrolizumab in Anaplastic Thyroid Cancer. Eur. Thyroid J. 2024, 13, e230144. [Google Scholar] [CrossRef] [PubMed]
  54. Tahara, M.; Kiyota, N.; Imai, H.; Takahashi, S.; Nishiyama, A.; Tamura, S.; Shimizu, Y.; Kadowaki, S.; Ito, K.I.; Toyoshima, M.; et al. A Phase 2 Study of Encorafenib in Combination with Binimetinib in Patients with Metastatic BRAF-Mutated Thyroid Cancer in Japan. Thyroid 2024, 34, 467–476. [Google Scholar] [CrossRef]
  55. Evans, L.K.; Chen, H.; Taki Labib, M.; Cronkite, D.A.; Yu, A.C.; Ashendouek, M.; Elashoff, D.; Chai-Ho, W.; Wong, D.J.; St John, M. Improved Survival of Advanced-Stage Anaplastic Thyroid Cancer With Systemic Therapy. Laryngoscope 2025, 135, 478–484. [Google Scholar] [CrossRef] [PubMed]
  56. Soll, D.; Bischoff, P.; Frisch, A.; Jensen, M.; Karadeniz, Z.; Mogl, M.T.; Horst, D.; Penzkofer, T.; Spranger, J.; Keilholz, U.; et al. First effectiveness data of lenvatinib and pembrolizumab as first-line therapy in advanced anaplastic thyroid cancer: A retrospective cohort study. BMC Endocr. Disord. 2024, 24, 25. [Google Scholar] [CrossRef]
  57. Cabanillas, M.E.; Dadu, R.; Ferrarotto, R.; Gule-Monroe, M.; Liu, S.; Fellman, B.; Williams, M.D.; Zafereo, M.; Wang, J.R.; Lu, C.; et al. Rare Tumor Initiative Team. Anti-Programmed Death Ligand 1 Plus Targeted Therapy in Anaplastic Thyroid Carcinoma: A Nonrandomized Clinical Trial. JAMA Oncol. 2024, 10, 1672–1680. [Google Scholar] [CrossRef] [PubMed]
  58. Sehgal, K.; Serritella, A.; Liu, M.; ONeill, A.; Nangia, C.; Pappa, T.; Demeure, M.J.; Worden, F.P.; Haddad, R.; Lorch, J. A phase I/II trial of sapanisertib in advanced anaplastic and radioiodine refractory differentiated thyroid carcinoma. J. Clin. Endocrinol. Metab. 2024, dgae443. [Google Scholar] [CrossRef]
  59. Song, Y.; Zhang, Y.; Bai, Y.; Wang, T.; Xu, G.; Ma, X.; Fei, K.; Zhang, B. Combination kinase inhibitors and immunotherapy for unresectable anaplastic thyroid carcinoma: A retrospective single-center study. Oral Oncol. 2024, 159, 107067. [Google Scholar] [CrossRef]
  60. Sehgal, K.; Pappa, T.; Shin, K.Y.; Schiantarelli, J.; Liu, M.; Ricker, C.; Besson, N.R.; Jones, S.M.; Welsh, E.L.; Pfaff, K.L.; et al. Dual Immune Checkpoint Inhibition in Patients With Aggressive Thyroid Carcinoma: A Phase 2 Nonrandomized Clinical Trial. JAMA Oncol. 2024, 10, 1663–1671. [Google Scholar] [CrossRef]
  61. Silver Karcioglu, A.; Iwata, A.J.; Pusztaszeri, M.; Abdelhamid Ahmed, A.H.; Randolph, G.W. The American Thyroid Association (ATA) integrates molecular testing into its framework for managing patients with anaplastic thyroid carcinoma (ATC): Update on the 2021 ATA ATC guidelines. Cancer Cytopathol. 2022, 130, 174–180. [Google Scholar] [CrossRef] [PubMed]
  62. Hamidi, S.; Dadu, R.; Zafereo, M.E.; Ferrarotto, R.; Wang, J.R.; Maniakas, A.; Gunn, G.B.; Lee, A.; Spiotto, M.T.; Iyer, P.C.; et al. Initial Management of BRAF V600E-Variant Anaplastic Thyroid Cancer: The FAST Multidisciplinary Group Consensus Statement. JAMA Oncol. 2024, 10, 1264–1271. [Google Scholar] [CrossRef] [PubMed]
  63. Jannin, A.; Escande, A.; Al Ghuzlan, A.; Blanchard, P.; Hartl, D.; Chevalier, B.; Deschamps, F.; Lamartina, L.; Lacroix, L.; Dupuy, C.; et al. Anaplastic Thyroid Carcinoma: An Update. Cancers 2022, 14, 1061. [Google Scholar] [CrossRef]
  64. Mahdiannasser, M.; Khazaei, S.; Akhavan Rahnama, M.; Soufi-Zomorrod, M.; Soutodeh, F.; Parichehreh-Dizaji, S.; Rakhsh-Khorshid, H.; Samimi, H.; Haghpanah, V. Illuminating the role of lncRNAs ROR and MALAT1 in cancer stemness state of anaplastic thyroid cancer: An exploratory study. Noncoding RNA Res. 2023, 8, 451–458. [Google Scholar] [CrossRef] [PubMed]
  65. Wirth, L.J.; Sherman, E.; Robinson, B.; Solomon, B.; Kang, H.; Lorch, J.; Worden, F.; Brose, M.; Patel, J.; Leboulleux, S.; et al. Efficacy of Selpercatinib in RET-Altered Thyroid Cancers. N. Engl. J. Med. 2020, 383, 825–835. [Google Scholar] [CrossRef] [PubMed]
  66. Ahn, S.; Kim, T.H.; Kim, S.W.; Ki, C.S.; Jang, H.W.; Kim, J.S.; Kim, J.H.; Choe, J.H.; Shin, J.H.; Hahn, S.Y.; et al. Comprehensive screening for PD-L1 expression in thyroid cancer. Endocr. Relat. Cancer 2017, 24, 97–106. [Google Scholar] [CrossRef] [PubMed]
  67. Crosta, S.; Boldorini, R.; Bono, F.; Brambilla, V.; Dainese, E.; Fusco, N.; Gianatti, A.; L’Imperio, V.; Morbini, P.; Pagni, F. PD-L1 Testing and Squamous Cell Carcinoma of the Head and Neck: A Multicenter Study on the Diagnostic Reproducibility of Different Protocols. Cancers 2021, 13, 292. [Google Scholar] [CrossRef]
  68. Oliinyk, D.; Augustin, T.; Koehler, V.F.; Rauch, J.; Belka, C.; Spitzweg, C.; Käsmann, L. Hypofractionated Radiotherapy for Anaplastic Thyroid Cancer: Systematic Review and Pooled Analysis. Cancers 2020, 12, 2506. [Google Scholar] [CrossRef] [PubMed]
  69. Cao, R.B.; Ge, Y.; Zhang, W.X.; Lin, G.H.; Kuang, B.H.; Wang, B.C. The efficacy and safety of antiangiogenesis tyrosine kinase inhibitors in patients with advanced anaplastic thyroid cancer: A meta-analysis of prospective studies. Medicine 2024, 103, e38679. [Google Scholar] [CrossRef] [PubMed]
  70. Zheng, X.; Wang, J.; Ye, T.; Tang, W.; Pan, X.; Wang, S.; Liu, J. Efficacy and safety of anlotinib-based chemotherapy for locally advanced or metastatic anaplastic thyroid carcinoma. Endocrine 2023, 81, 540–546. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of the studies’ selection strategy.
Figure 1. Flow diagram of the studies’ selection strategy.
Cancers 17 00777 g001
Figure 2. Forest plot of overall response rates in included studies.
Figure 2. Forest plot of overall response rates in included studies.
Cancers 17 00777 g002
Figure 3. Forest plot of overall survival in included studies.
Figure 3. Forest plot of overall survival in included studies.
Cancers 17 00777 g003
Figure 4. Forest plot of progression survival rate in included studies.
Figure 4. Forest plot of progression survival rate in included studies.
Cancers 17 00777 g004
Table 1. Characteristics of included studies.
Table 1. Characteristics of included studies.
Study/Year/
Nature
CountryN
(Median Age)
TNM
(n)
Previous Treatment (%)InterventionEndpointsORR (%)Median PFS (Months)Median OS
(Months)
Higashiyama T, et al. (HOPE) [16]
Prospective
2022
Japan42
(73y)
T4 = 37
N1a = 3
N1b = 23
M0 = 17
M1 = 21
CTH = 40
XRT = 21.4
Sx = 54.7
Lenvatinib Primary: OS
Secondary:
PFS, ORR, DCR, safety
5 (11.9)16.518.5
Wirth LJ, et al. [17]
Prospective
2021
US, UK, France, Italy, Australia,34
(65y)
M1 = 34CTH = 62
TKI = 9
IO = 9
XRT = 65
Sx = 71
LenvatinibPrimary: ORR, safety
Secondary:
PFS, OS
1 (2.9)2.6 (1.4–2.8)10.6 (3.8–19.8)
Yamazaki H, et al. [18]
Retrospective
2021
Japan20
(73.6y)
T4 = 14Sx = 6Lenvatinib-2 (10.0)4.1 (1.1–12.2)-
Sparano C, et al. [19]
Retrospective
2021
France, Italy15
(63y)
Recurrent/
M1 = 15
XRT = 71
CTH = 73.2
TKI = 26.5
Lenvatinib-5 (33.3)2.7 (1.9–3.5)3.1 (0.6–5.5)
Park J, et al. [20]
Retrospective
2021
South Korea11
(66.4y)
T4/
M1 = 11
Sx = 64.2
XRT = 53.3
CTH = 24.2
TKI = 15.8
Lenvatinib-3 (27.3)2.7 (0.1–12.7)12.4 (1.7–47.7)
Iwasaki H, et al. [21]
Retrospective
2021
Japan32
(77y)
T4/
M1 = 32

-
Lenvatinib-6 (18.8)-3.2 (0.5–28.9)
Ishihara S, et al. [22]
Retrospective
2021
Japan11
(74y)
Recurrent = 5
T4 = 4
T3b = 2
CTH = 63.6
XRT = 63.6
TKI = 27.3
Lenvatinib-3 (30.0)-4.7 (1.9–13.1)
Fukuda, N et al. [23]
Retrospective
2020
Japan13
(68y)
T4 = 10
N1 = 7
M1 = 11
Sx = 77
XRT = 46
CTH = 31
Lenvatinib-3 (23.0)3.8 (1.8–6.4)10.2 (3.7–17.6)
Kim M, et al. [24]
Retrospective
2020
South Korea14
(64.9y)
T4 = 14Sx = 33
CTH = 100
XRT = 100
Lenvatinib-4 (28.6)5.7 (2.2–8.3)6.7 (3.0–8.4)
Takahashi S, et al. [25]
Prospective
2019
Japan, US17
(65y)
T4 = 17Sx = 82
CTH = 41
XRT = 53
LenvatinibPrimary:
Safety
Secondary:
PFS, ORR, DCR, OS
4 (23.5)7.4 (1.7–12.9)10.6 (3.8–19.8)
Iyer PC, et al. [26]
Retrospective
2018
US10
(67y)
T3-4/N1 = 4
M1 = 6
Sx = 50
CTH = 60
XRT = 44
Lenvatinib-3 (30.0)2.6 (2.8-NR)3.9 (2.5-NR)
Ito Y et al. [27]
Prospective
2017
Japan10
(72y)
M1 = 8
T4 = 2
Sx = 70
CTH = 60
XRT = 70
SorafenibPrimary:
Safety
Secondary:
PFS, ORR, DCR, OS
3 (40.0)2.8 (0.7–5.6)5 (0.7–5.7)
Savvides P, et al. [28]
Prospective
2013
US20
(59y)
M1 = 20CTH = 100
XRT = 90
Sx = 90
SorafenibPrimary:
ORR, DCR
Secondary:
PFS, OS, safety
2 (10.0)1.9 (1.3–3.6)3.9 (2.2–7.1)
Bible KC, et al. [29]
Prospective
2012
US15
(63)
T4 = 2
N1 = 1
M1 = 12
CTH = 100
XRT = 90
Sx = 90
PazopanibPrimary:
ORR, DCR
Secondary:
PFS, OS, safety
5 (33.3)2.1 (NR)3.7 (0.5–35)
NRG/RTOG 0912 [30] Prospective 2023 US71
(63y)
T4 = 71
N1 = 52
M1 = 26
Sx = 57.5a. Pazopanib + weekly Paclitaxel +IMRT 66 Gy/33 fractions (36pts) b. Weekly Paclitaxel +IMRT 66 Gy/33 fractionsPrimary:
OS
Secondary:
PFS, safety
11 (30.5)
11 (31.4)
-5.7 (4.0–12.8)
7·3 (4.3–10.6)
Pennell NA, et al. [31] Prospective 2008US5/27
(65y)
T4/M1 = 5CTH = 22
Sx = 94
XRT = 85
GefitinibPrimary:
ORR, DCR
Secondary:
PFS, OS, safety
2 (40.0)3.717.5
Zhao Q, et al. [32] Prospective 2022China17
(61y)
T4 = 3
N1 = 7
M1 = 6
-ApatinibPrimary:
ORR, DCR
7 (41.1)--
Ha HT, et al. [33]
Prospective
2010
US11
(65y)
Recurrent = 7
M1 = 7
Sx = 64
CTH = 55
ImatinibPrimary:
ORR
2 (18.2)5.8 (2–29)3.2 (2–37)
Mooney CJ, et al. [34]
Prospective
2009
US26
(59y)
Recurrent = 24
M1 = 7
CTH = 50 Sx = 61 XRT = 61FosbretabulinPrimary:
Safety
Secondary:
8 (30.7)-4.7 (2.5–6.4)
Sosa JA, et al. [35]
Prospective
2014
US80
(63y)
Recurrent = 20 Sx = 25 XRT = 13.8 CTH =5Fosbretabulin +CP
Or
CP alone
Primary:
OS
13 (32.5)
13 (32.5)
3.3 (2.3–5.6)
3.1 (2.7–5.4)
5.2 (3.1–9)
4.0 (2.8–6.2)
Subbiah V, et al. [36]
Prospective
2018
US16
(72y)
Recurrent = 16Sx = 88
XRT = 81
CTH = 38
DTPrimary:
ORR, DCR
Secondary:
PFS, OS, safety
11 (68.7)NANA
ROAR Study [37]
Prospective
2022
US, France, south Korea36
(71y)
T4 = 1
M1 = 35
Sx = 83
XRT = 83
CTH = 42
RAI = 31
TKI = 19
IO = 11
DTPrimary:
ORR
Secondary:
PFS, OS, DOR
18 (50)6.7 (4.7–13.8)14.5 (6.8–23.2)
Lorimer C, et al. [8]
Retrospective
2023
UK17
(68y)
T4/M1 = 17Sx = 59
CTH = 12
RAI = 12
XRT = 18
DT-14 (82.2)4.7 (1.4–7.8)6.9 (2.46-NR)
Bueno F, et al. [38]
Retrospective
2023
Argentina5
(70y)
M1 = 5Sx = 60
XRT = 20
DT-4 (80.0)-20 (18-NR)
da Silva TN, et al. [39]
retrospective
2023
Portugal27
(77y)
Recurrent/
M1 = 27
Sx = 40.7
XRT = 40.7
CTH = 25.9
TKI = 7.4
DT-8 (29.6)9.0 (4.9–13.0)17.9 (15.9–19.8)
Zhao X, et al. [40]
Retrospective
2023
US57
(67.2y)
T4 = 20
M1 = 37
IO = 75.4
XRT = 47.4
DT-18 (31.5)34.2 (15.8–NA)39.2 (NR)
Hyman DM, et al. [41]
Prospective
2015
US, UK, Germany, France, Spain7/122
(65y)
Recurrent/ M1 = 7Any = 78 XRT = 67VemurafenibPrimary:
ORR
Secondary:
PFS, OS
2 (29.0)--
Hatashima A, et al. [42]
Retrospective
2022
US13
(70y)
T4 = 2
M1 = 11
-Pembrolizumab (12 patients) Nivolumab (1 patient)-2 (16.0)1.94.4 (4.0–29)
Capdevila J, et al. [43]
Prospective
2020
US, Canada, Germany, Italy, Switzerland, France, Poland42
(62.5y)
Recurrent/M1 = 42XRT = 71.4
Sx = 66.7
SpartalizumabPrimary:
ORR
Secondary:
Safety
7 (16.6)1.7 (1.2–1.9)5.9 (2.4-NR)
Lorch JH, et al. [44]
Prospective
2020
US10/49
(65y)
T4/M1 = 10-Ipilimumab + NivolumabPrimary:
ORR
3 (30.0)--
Dierks C, et al. [45]
Retrospective 2021
Germany6
(63.5y)
Recurrent/
M1 = 6
Sx = 100
XRT = 87.5
CTH = 75
RAI = 25
LP-4 (66.0)17.718.5
ATLEP trial [46]
Prospective
2022
Germany29
(63y)
N1/M1 = 29Sx = 90
XRT = 90
CTH = 90
LP 12 (41.2)9.510.25 (NR)
Iyer PC, et al. [47]
Retrospective
2018
US11
(67y)
T4/M1 = 11 Sx = 50
XRT = 44
CTH = 69
LP-5 (42.0)2.9 (2.2–3.7)6.93 (1.7–12.1)
Lim SM, et al. [48]
Prospective
2013
South Korea6/40
(61y)
T4/M1 = 6Sx = 68 CTH = 22 XRT = 15 TKI = 2EverolimusPrimary:
ORR
Secondary:
PFS, OS
1 (16.0)2.3 (1.1–3.7)-
Hanna GJ, et al. [49]
Prospective
2018
US7/50
(62y)
Recurrent/ M1 = 7Sx = 71 XRT = 57 RAI = 28 CTH = 28 TKI = 14EverolimusPrimary:
ORR
Secondary:
Safety
1 (14.2)-4.6 (<1–29.9)
Harris EJ, et al. [50]
Retrospective
2019
US5
(75y)
Recurrent/ M1 = 5XRT = 60 CTH = 60 RAI = 40Everolimus-1 (20.0)-7.4 (<1–40)
Waguespack SG, et al. [9]
Prospective
2022
US, Canada, Ireland, Italy, Germany, S. Korea7/29
(60y)
Recurrent/ T4 = 7Sx =100 XRT = 71 CTH = 43 LarotrectinibPrimary:
ORR
Secondary:
PFS, OS, Safety
2 (29.0)2.2 (<1–6)2.5 (<1–6)
Doebele RC, et al. [51]
Prospective
2020
US, UK, Australia, Italy, Hong Kong, Spain, S. Korea, Poland, Netherlands5/54
(58y)
--EntrectinibPrimary:
ORR
1 (20.0)--
Hamidi S, et al. [52]
Retrospective
2024
US71T4 = 23
M1 = 48
Sx/XRT23 = DT
48 = DTP
Primary: OS
Secondary:
PFS
7 (30.4)
15
(31.3)
DT = 4
DTP =11
DT = 9
DTP = 17
Tan JSH, et al. [53]
Retrospective
2024
Singapore 5
(63.6y)
T4 = 3
M1 = 2
-Quad-XRT/LP = 2. Quad-XRT/P = 3Primary: OS
Secondary:
PFS, ORR
2 (40.0)7.66.2
Tahara M, et al. [54]
Prospective
2024
Japan 5/22T4/M1 = 5-Encorafenib plus binimetinibPrimary: OS
Secondary:
PFS, ORR
4 (80.0)11.511.5
Evans LK, et al. [55]
Retrospective
2025
US41
(67.4y)
Recurrent/T4/M1 = 41-LP = 18
DT = 9
Primary: OS6 (33.3)
3 (33.3)
-7.6
Soll D, et al. [56]
Retrospective
2024
Germany 5
(65y)
M1 = 5-LPPrimary: OS
Secondary:
PFS
1 (20.0)4.76.4
Cabanillas ME, et al. [57]
Prospective
2024
US39T4 = 12 M1 = 30XRT = 12 CTH = 38.1VCA = 18 CA = 21 Primary: OS
Secondary:
PFS
2 (11.1)
1 (5.7)
VCA = 13.9
CA = 8.7
BA = 6.2
VCA = 43
CA = 13.9
BA = 8.7
Sehgal K, et al. [58]
Prospective
2024
US20/26Recurrent /M1 = 20TKI = 20SapanisertibPrimary: ORR, PFS6 (30.0)1.6-
Song Y, et al. [59]
Retrospective
2024
China18T4/M1-DTP, LP, anlotinib sintilimab, or camrelizumabPrimary: ORR, PFS, OS11 (61.1)-14
Sehgal K, et al. [60]
Prospective
2024
US10/49 (65y)Recurrent/metastatic/T4 = 10XRT =80 TKI = 70Ipilimumab/ nivolumab Primary:
ORR
Secondary:
PFS, OS, Safety
3 (30.0)--
N = number of patients, TNM = tumour, node, stage = CR = complete response, PR = partial response, ORR, overall response rate, DCR = disease control rate, PFS = progression-free survival, OS = overall survival, CTH = chemotherapy, XRT = radiation therapy, Sx = surgery, RAI = radioactive iodine, TKI = tyrosine kinase inhibitors, IMRT = intensity modulated radiation therapy. CP = carboplatin/paclitaxel, D/D = docetaxel/doxorubicin, DT = dabrafenib/trametinib, DTP = dabrafenib/trametinib/pembrolizumab, VCA = vemurafenib/cobimetinib plus atezolizumab, CA = cobimetinib plus atezolizumab.
Table 2. Toxicity Profile of Included Studies.
Table 2. Toxicity Profile of Included Studies.
G3/>ToxicityLenvatinib
(n = 266)
Sorafenib
(n = 30)
Pazopanib
(n = 51)
Gefitinib
(n = 5)
Apatinib
(n = 17)
Imatinib
(n = 11)
Chemotherapy
(n = 81)
mTORi
(n = 18)
BRAFi
(n = 206)
IO
(n = 232)
NTRKi
(n = 11)
Loss of appetite29 (11.0%)1 (3.0%)2 (5.8%)2 (40.0%)-1 (9.0%)2 (2.3%)5 (27.7%)10 (4.8%)4 (1.7%)1 (9.0%)
Weight loss23 (8.6%)--1 (20.0%)-1 (9.0%)1 (1.2%)4 (22.2%)8 (3.8%)4 (1.7%)1 (9.0%)
Fatigue21 (7.9%)-7 (13.7%)1 (20.0%)--1 (1.2%)1 (5.5%)18 (8.7%)4 (1.7%)3 (27.3%)
Hypertension37 (14.0%)-1 (1.9%)-6 (35.3%)-2 (2.3%)-14 (6.7%)--
HFS20 (7.5%)3 (9.0%)------ 4 (1.7%)-
Nausea12 (4.5%)2 (6.0%)4 (7.8%)1 (20.0%)--11 (13.6%)5 (27.7%)8 (3.8%)-1 (9.0%)
Diarrhoea24 (9.0%)3 (9.0%)-3 (60.0%)--12 (14.8%)-8 (3.8%)-1 (9.0%)
Anaemia3 (1.1%)-2 (3.9%)--2 (18.0%)8 (9.8%)-23 (11.1%)2 (0.8%)1 (9.0%)
Thrombocytopenia8 (3.0%)-1 (1.9%)-1 (5.9%)5 (45.5%)14 (17.3%)----
Lymphopenia4 (1.5%)-5 (9.8%)-1 (5.9%)-8 (9.8%)2 (11.0%)---
Neutropenia2 (1.0%)-7 (13.7%)-1 (5.9%)1 (9.0%)2 (2.3%)5 (27.7%)1 (0.5%)--
Hypophosphatemia-1 (3.0%)1 (1.9%)--- 2 (11.0%)-2 (0.8%)-
Hyperglycaemia- ----1 (1.2%)2 (11.0%)8 (3.8%)2 (0.8%)-
Hyponatremia-2 (6.0%)------11 (5.3%)3 (1.3%)-
Voice alteration3 (1.1%) 3 (5.8%)-5 (29.4%)----1 (0.4%)-
Proteinuria9 (3.4%)1 (3.0%)------1 (0.5%)--
Eczema/dermatitis3 (1.1%)3 (9.0%)13 (25.5%)-----2 (0.9%)5 (2.1%)-
Mucositis oral3 (1.1%)3 (9.0%)5 (9.8%)---1 (1.2%)7 (38.8%)---
Arthralgia2 (1.0%)1 (3.0%)---2 (18.0%)----1 (9.0%)
Non-tumour bleeding4 (1.5%)--------1 (0.4%)
Vomiting11 (4.1%)3 (9.0%)1 (1.9%)--1 (9.0%)1 (1.2%)1 (5.5%)6 (2.9%)1 (0.4%)-
Skin ulceration5 (1.9%)--- -- ---
Aspartate aminotransferase increased8 (3.0%)1 (3.0%)12 (23.5%)-1 (5.9%)--2 (11.0%)-11 (4.7%)1 (9.0%)
Alkaline phosphatase increased--2 (3.9%)-------1 (9.0%)
Amylase/lipase elevated---------21 (9.1%)-
Hypothyroidism7 (2.6%)--------3 (1.3%)-
Hypophysitis---------2 (0.8%)-
Dyspnoea7 (2.6%)--------4 (1.7%)-
Tracheal fistula4 (1.5%)--------2 (0.8%)-
Fistula2 (0.8%)1 (3.0%)1 (1.9%)------3 (1.3%)
Aspiration pneumonia/
pneumonitis
6 (2.2%)1 (3.0%)3 (5.8%)----1 (5.5%)14 (6.7%)1 (0.4%)--
Pneumothorax1 (0.4%)--------3 (1.3%)-
Pancreatitis1 (0.4%)----------
ECG QT prolonged3 (1.1%)-----6 (7.4%)----
Acute heart failure1 (0.4%)----1 (9.0%)1 (1.2%)--5 (2.1%)-
Adrenal insufficiency---------7 (3.0%)-
Dose modifications59 (22.2%)7 (23.0%)16 (31.4%)---24 (29.6%)7 (38.8%)31 (15.0%)-4 (36.4%)
Interruptions44 (16.5%)6 (20.0%)9 (17.6%)---25 (30.8%)6 (33.3%)34 (16.5%)--
Withdrawals17 (6.3%)1 (3.0%)12 (23.5%)---12 (14.8%)1 (5.5%)9 (4.4%)5 (2.1%)-
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tunio, M.A.; Hinder, D.; Emery, B.; Riaz, M.H.; Ibraheem, Y.A.; Nayak, K.K.; Mohamed, W. Modern Therapeutic Approaches in Anaplastic Thyroid Cancer: A Meta-Analytic Review of Randomised and Single Arm Studies on Efficacy and Survival. Cancers 2025, 17, 777. https://doi.org/10.3390/cancers17050777

AMA Style

Tunio MA, Hinder D, Emery B, Riaz MH, Ibraheem YA, Nayak KK, Mohamed W. Modern Therapeutic Approaches in Anaplastic Thyroid Cancer: A Meta-Analytic Review of Randomised and Single Arm Studies on Efficacy and Survival. Cancers. 2025; 17(5):777. https://doi.org/10.3390/cancers17050777

Chicago/Turabian Style

Tunio, Mutahar A., Donna Hinder, Blaise Emery, Muhammad H. Riaz, Yusef A. Ibraheem, Krishnendu Kumar Nayak, and Wael Mohamed. 2025. "Modern Therapeutic Approaches in Anaplastic Thyroid Cancer: A Meta-Analytic Review of Randomised and Single Arm Studies on Efficacy and Survival" Cancers 17, no. 5: 777. https://doi.org/10.3390/cancers17050777

APA Style

Tunio, M. A., Hinder, D., Emery, B., Riaz, M. H., Ibraheem, Y. A., Nayak, K. K., & Mohamed, W. (2025). Modern Therapeutic Approaches in Anaplastic Thyroid Cancer: A Meta-Analytic Review of Randomised and Single Arm Studies on Efficacy and Survival. Cancers, 17(5), 777. https://doi.org/10.3390/cancers17050777

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop