Next Article in Journal
Analysis of the Use of Sample Size and Effect Size Calculations in a Temporomandibular Disorders Randomised Controlled Trial—Short Narrative Review
Previous Article in Journal
The Role of Oral Supplementation for the Management of Age-Related Macular Degeneration: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

The Long Journey towards Personalized Targeted Therapy in Poorly Differentiated Thyroid Carcinoma (PDTC): A Case Report and Systematic Review

by
Odysseas Violetis
1,†,
Panagiota Konstantakou
1,†,
Ariadni Spyroglou
1,†,
Antonios Xydakis
1,
Panagiotis B. Kekis
2,
Sofia Tseleni
3,
Denise Kolomodi
4,5,
Manousos Konstadoulakis
1,
George Mastorakos
1,
Maria Theochari
6,
Javier Aller
7 and
Krystallenia I. Alexandraki
1,*
1
2nd Department of Surgery, Aretaieio Athens Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Endocrine Surgery Department, Athens Medical Center, 15125 Athens, Greece
3
Department of Pathology, Medical School, University of Athens, 11527 Athens, Greece
4
European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Ekpa-Laiko Center, 11527 Athens, Greece
5
IATROPOLIS Private Medical Center, 11521 Athens, Greece
6
Department of Oncology, Ippokrateio Athens General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
7
Endocrinology Department, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Pers. Med. 2024, 14(6), 654; https://doi.org/10.3390/jpm14060654
Submission received: 20 May 2024 / Revised: 1 June 2024 / Accepted: 10 June 2024 / Published: 18 June 2024
(This article belongs to the Section Molecular Targeted Therapy)

Abstract

:
Background: Poorly differentiated thyroid carcinoma (PDTC) has an intermediate prognosis between indolent well-differentiated thyroid carcinoma (TC) and anaplastic carcinoma. Herein, we present a case report with a PDTC component, along with a systematic review of the literature. Case Report: We report a case of a 45-year-old man diagnosed with a PDTC component, along with hobnail and tall-cell variant features positive for BRAFV600E mutation, after a total thyroidectomy and neck dissection. Radioactive iodine (RAI)-131 therapy was applied, but an early recurrence led to complementary surgeries. The anti-Tg rise, the presence of new lymph nodes, and the negative whole-bodyradioiodine scan were suggestive of a radioiodine-resistant tumor. Lenvatinib, sorafenib, dabrafenib/trametinib, cabozantinib and radiotherapy were all administered, controlling the tumor for a period of time before the patient ultimately died post-COVID infection. Systematic Review: We searched PubMed, Scopus, and WebofScience to identify studies reporting clinicopathological characteristics, molecular marker expression, and management of non-anaplastic TC with any proportion of PDTC in adult patients. Of the 2007 records retrieved, 82were included in our review (PROSPERO-ID545847). Conclusions: Our case, together with the systematic review, imply that a combination of molecular-targetedtreatments may be safe and effective in patients with RAI-resistantBRAF-mutated advanced PDTC when surgery has failed to control tumor progression.

1. Introduction

Nowadays, thyroid cancer (TC) is estimated to cause approximately 44,000new invasive cancer cases, with a higher prevalence in females [1]. Until recently, it was deemed one of the fastest cancer entities [2,3,4,5], largely as a consequence of oversurveillance and overdiagnosis of small, differentiated thyroid carcinoma (DTC) [3], attributed to significant developments in diagnostic imaging techniques [4,6,7]. However, the adoption of more restrictive criteria for the diagnosis of TC led to a drop in incidence by 2% since 2014. Five-year survival has remained generally stable over the years, being more than 92% in the United States [1]. DTC, including papillary thyroid cancer (PTC) and follicular thyroid cancer (FTC), accounts for more than 90% of TC [8]. De-differentiated TC is rare, classified as poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC), with reported incidence of 2–15% and 1.7%, respectively [9,10]. A PDTC prognosis is intermediate between DTC and ATC [9], while ATC is associated with the highest mortality risk of any thyroid-arising tumor [11,12].
Most DTC patients have a good prognosis, with a 5-year survival reaching 99% [1,13,14,15,16]. However, at the other end of the spectrum, some PTC variants are associated with aggressive behaviors, such as the recently described hobnail variant, the tall-cell variant, the columnar variant, the solid-trabecular variant [17,18,19,20,21,22,23], the diffuse sclerosing variant [24], the cribriform-morular variant (associated with FAP and APC gene alterations) [25,26], and the oncocytic variant [27]. Approximately 10–15% of all TCs exhibit aggressive behavior and high disease-specific mortality [28]. Among the array of genetic mutations identified in different histologic subtypes of TC, BRAFV600E has been extensively studied because it appears as the most common driver mutation in thyroid PTC, while PTC without BRAF mutations mostly harbors RAS mutations. The presence of BRAF or RAS mutations dictates significant assets of TC. Sequential acquisition of key genetic alterations promotes progressive dedifferentiation to more advanced TC, namely high-grade differentiated thyroid carcinoma (HGGTC), PDTC, and ATC [29]. Monotherapy with a BRAF inhibitor (BRAF-I) may result in temporary clinical remission, but acquired resistance is frequently observed, leading to significant relapse in the majority of cases [30]. The combination of BRAF-I and MEK inhibitors (MEK-I) in conducted clinical trials has shown delay in the development of resistance mechanisms and was recently approved by the Food and Drug Administration (FDA) as a therapy for BRAFV600E-mutant ATC [31].
We describe herein a patient with multiconstituent TC with poorly differentiated, hobnail, and tall-cell components, ab initio. The patient demonstrated refractoriness to radioactive iodine (RAI) treatment, was treated with tyrosine kinase inhibitors (TKI) and BRAF-I/MEK-I combination, and rechallenged with TKI with established disease control. Because of sparse clinical data and a lack of explicit management guidelines, we also conducted a systematic review of the literature regarding PDTC cases to compare the different approaches followed for this rare yet aggressive entity and to correlate them with our case. Herein, histopathological and clinical features, along with treatment and genetic profiling of TC with any component of PDTC, are addressed.

2. Materials and Methods

Aiming at presenting demographics and histopathological and molecular features of PDTC, we conducted a systematic review of the literature scrutinizing three databases, namely PubMed, Scopus, and Web of Science. This systematic review was performed following recommendations of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) [32]. Without limitations based on the type of study (case reports, case series cohort studies, case–control), adult patients with non-ATC with any proportion of PDTC from the year 2017 to April 2024 were included in the present study, whereas studies in pediatric populations or those lacking relevant demographic, histopathological, or management data or data derived from animal models were excluded. Cases presenting with any component of ATC were also excluded. Review articles, conference abstracts, and opinion articles were not included in the present study either. Since there are differences in the diagnostic criteria of PDTC communicated in the bibliography, we did not use specific criteria for the diagnosis and relied on the PDTC characterization of each study. Although PDTC has been recognized as a distinct pathologic entity based on the WHO classification since 2004, the 4th WHO classification only adopted specific unanimous diagnostic criteria (Turin criteria) in 2017, and they indicated that any poorly differentiated component should be mentioned in the pathology report. Thus, we decided to narrow our search to the period of 2017–2024. The clinicopathological characteristics included in our review consisted of age at diagnosis, gender, histological subtypes, nodal status, distant metastases, extrathyroidal extension, molecular subtypes, surgical performance, the administration of radiotherapy and/or chemotherapy, and outcomes. We used the following terms for our search in the abstract or title areas: poorly differentiated, thyroid cancer/neoplasm/carcinoma, PDTC. Boolean operators (AND, OR) were also used to narrow down the search.
Two independent reviewers (O.V., A.S.) screened the titles and abstracts of the articles and selected potentially relevant studies. Full-text articles were then studied, and the final articles were selected based on the inclusion and exclusion criteria. Disagreements in the review process were resolved through discussion and judgment by a third reviewer (K.I.A.). All studies were carefully compared to avoid the inclusion of duplicate or overlapping samples. Any duplicates identified were removed using EndNote 21. The systematic review is registered in the international prospective register of international reviews, PROSPERO (ID 545847). This study was approved by the local institutional ethics committee with reference number 363/13-10-2021.

3. Case Presentation

A 45-year-old man was referred to our department after undergoing a total thyroidectomy with central and lateral lymph node dissection in October 2012 followed by 80mCi of RAI treatment four weeks postoperatively in November 2012 with post-iodine uptake only in the thyroid bed. The pathology report described a PTC of the left lobe with a tall-cell variant, a maximum diameter 5 cm with extrathyroidal extension (ETE), and metastases in three out of 28 resected lymph nodes (isthmus area and transition area between isthmus and left lobe). The TNM status was Τ4Ν1aΜ0 (Stage ΙΙI) [33], which along with the tall-cell variant, placed the patient at intermediate risk. Thus, he was referred to our clinic, and the levothyroxine dose was titrated to achieve a suppressed thyroid-stimulating hormone (TSH). His past medical history was remarkable for hypercalciuria on hydrochlorothiazide, nephrolithiasis, chronic sinusitis, and positive thyroid-stimulating immunoglobulin (TSI) without documented thyroid dysfunction prior to the thyroidectomy. The patient reported no family history of thyroid disease, malignancy, or personal history of radiation exposure.
A documented increase in anti-Tg levels and a suspicious left neck lymph node enlargement, revealed by an ultrasound scan, were suggestive of disease recurrence. In March 2013, the patient underwent a complementary left lateral lymph node dissection, with 1/11 lymph nodes positive for the aforementioned carcinoma. In June 2014, diagnostic WBS after recombinant thyrotropin alfa stimulation was performed, and one lymph node 3.7 cm in the left compartment was visualized, along with a further rise in anti-Tg title (1081 IU/mL; Tg < 0.04 ng/mL; TSH 130.7 μIU/mL) under maximal stimulation. The stimulation with thyrotropin alfa was repeated during 18F-FDG-PET/CT, and no further uptake was evident other than the described lymph node. An ultrasound (US) scan and neck magnetic resonance imaging (MRI) scan pinpointed two additional heterogeneous 5–10 mm lymph nodes, as well as a 8 × 5 mm left submandibular lymph node with central vascularization. At this stage, the patient agreed to review the first histology, where a component of PDTC was seen (Figure 1), along with hobnail and tall-cell variant features, as previously reported [34]. Importantly, the molecular and immunohistochemical analysis of previous biopsies showed a positive BRAFV600E mutation (exon 15, c.1799 T > A [p.V600E,COSM 476]). The metastatic focus pathologic review showed a histological pattern reminiscent of the differentiated component. Subsequently, the patient was submitted to a third surgical intervention and underwent a left lateral lymph node dissection in July 2014 with resection of one large metastatic lymph node and 14 lymph nodes free of disease, while the pathology report reconfirmed the origin from the differentiated neoplastic component. An additional RAI treatment dose of 150 mCi was administered in October 2014. A decline in anti-Tg levels was temporarily observed, but later, anti-Tg levels progressively increased, while a suspicious lymph node became apparent in the US in September 2015. The WBS did not show a clear uptake, suggesting RAI refractory (RAIR) disease (anti-Tg 222.6 IU/mL; Tg 0.079 ng/mL; TSH 120.9 μIU/mL), and the patient was submitted to a fourth surgery in November 2015 with deep central compartment dissection of the suspicious lymph node (1/1). Histology once again was consistent with the original differentiated component, with hobnail features staining positive for thyroglobulin and galectin. The follow-up US in May 2016 revealed another infiltrated lymph node internally from the upper part of the left carotid artery and jugular vein 17 × 11 × 19 mm, which increased to 23 × 30 × 30 mm in the subsequent US in September 2016. A fifth surgery took place in October 2016, with complementary left lateral dissection and removal of the large lymph node and two smaller lymph nodes in the bulb of the common carotid artery, all infiltrated by the neoplasm with the tall-cell variant features. However, 3 months later, another suspicious lymph node was seen along the left jugular vein, measuring 7 × 4mm. The next US in June 2017 showed an increased number of infiltrated lymph nodes in the left side levels IIα, ΙΙβ, ΙIΙ, and IV, and a fine needle aspiration confirmed lymph node metastases from the hobnail variant.
At that point, in July 2017, therapy with full-dose lenvatinib was initiated. As early as within the first month of lenvatinib treatment, our patient presented adverse effects (AEs, according the CTCAE Version 5.0) in the form of palmar–plantar erythrodysesthesia syndrome and abdominal distension. At the completion of 12 months of treatment, he also reported diarrhea (grade 1), fatigue, severe headaches lasting from one to two days per week (all grade 2), and arterial hypertension (grade 2) that prompted the initiation of antihypertensive treatment in April 2018. Because of the headaches fluctuating in severity between grade 2 and 3 and the stable disease course, he was switched to full-dose sorafenib. However, as he soon manifested additional AEs, one week later, he restarted treatment with lenvatinib for a total period of 22 months. In the meantime, the infiltrated lymph nodes increased, and he received radiotherapy in the form of tomotherapy (Helical Tomo therapy along with daily image-guided radiation therapy) from January to March 2019 (33 sessions cyber knife: total dose 66 Gy; daily dose 2 Gy), resulting in a progressive size reduction of the infiltrated lymph nodes. In January 2020, because of a persistent headache, a brain CT was performed. Two hypervascular lesions were seen in the parietal and occipital lobe without edema, suspicious for metastatic foci, along with an increase in the size of the sphenoid sinus due to a heterogeneous material (suspicious for malignant invasion in the sphenoid sinus). These suspicious findings prompted earlier restaging. The 18F-FDG-PET/CT was extensively positive, revealing increased uptake in the thyroid bed (SUV:11.4), uptake in the lymph nodes in the levels of IIβ (SUV max 7.7), Vβ (SUV max 3.2) of the left neck, in the liver (SUV max 8), the left sphenoid sinus with extension to the right (SUV max 18.7), cervical vertebra C5 (SUV max 7.8), and laterally to the thyroid cartilage (SUV max 13.4) (Figure 2). All the suspicious areas were confirmed using a brain MRI, an abdominal CT, and bone scanning. Thus, the patient received cyber knife therapy for the brain metastases and conventional radiotherapy for the cervical vertebra metastatic focus. For the later finding, he received 120 mg of denosumab monthly for 3 months.
According to recent clinical data [35,36], and taking into account the intolerance and the progressive resistance to multi-kinase inhibitor treatment (lenvatinib, sorafenib) in our case, the patient received a therapeutic trial with a combination of BRAF-I/MEK-I in April 2020 with sorafenib as a bridging therapy until the approval reporting fatigue (grade 3). Conventional doses were well tolerated by our patient (dabrafenib 150 mg PO BID, trametinib 2 mg PO QD). Repeated brain and neck MRIs in July 2020 showed radiological improvement with shrinkage of the brain lesions, the left sphenoid sinus, and the area surrounding the thyroid cartilage. Upon subsequent imaging in January 2021, the abdominal MRI was unremarkable, while significant improvement was noted in the brain MRI, with a decreased size of both the occipital (from 6 mm to 5 mm) and parietal (from 20 mm to 8 mm) lesions, a decreased size of the sphenoid sinus, and no new foci. The follow-up neck CT in September 2021 showed a decreased size of the lymph node in the left compartment at the posterior border of the sternocleidomastoid muscle (from 1.3 × 1.7 cm to 1.6 × 2 cm) and a stable size of a lymph node 1 cm in the right submandibular area. Quite a few lymph nodes in the submental area, the right submandibular area, and along the right internal jugular vein remained stable in size as well. Initially, the dabrafenib/trametinib combination treatment was well tolerated, but the patient discontinued his treatment due to fatigue later on. A new brain CT in April 2022 showed edema in the lower part of the parieto-occipital area and in the right frontal area. The neck MRI showed a mass invading the left larynx and the median line with extension to the hypopharynx. In parallel, an increase in the number and size of the lymph nodes of the left neck was seen, together with the presence of a new lymph node in the supraclavicular area. An attempt to obtain a biopsy from this mass failed. A frozen biopsy from the latest operation in 2016 was sent for molecular analysis, but no alteration was detected in genes NTRK1, NTRK2, NTRK3, or RET. In May 2022, and without being on any therapeutic scheme, the patient was admitted to the hospital with acute dyspnea, and a laryngeal edema was documented, which was controlled by a tracheotomy. Subsequently, treatment with 60 mg of cabozantinib was decided on the basis of the COSMIC-311 trial [37], and an objective improvement was already evident one-month post-therapy (Figure 3). The patient complained of persistent pain and discomfort at the site of the tracheotomy, which was removed according to his request. However, the patient was then diagnosed with COVID-19, and he was admitted to the hospital in the COVID unit. During his admission, he refused to have an angiography to further clarify a small hemorrhagic area in his neck identified by the neck surgeons, probably due to perforation (grade 4) caused by the treatment. He signed to be discharged against medical recommendations and died one week later at home due to hemorrhage. The treatment timeline of the patient is also depicted in Figure 4.

4. Systematic Review Results

The electronic search from the databases yielded 2007 candidate studies, out of which 1872 were excluded (duplicates, reviews, title or abstract irrelevant to the inclusion criteria), leaving 135 studies for full-text review. After the full manuscript consideration, we finally included 82 studies fulfilling the inclusion criteria (Figure 5). The characteristics of the included studies are shown in Table 1. The excluded studies, along with the reasons for their exclusion, are shown in Figure 5.

5. Discussion

The present case shows that the natural history of a non-classic multi-component PTC may be dictated by the more aggressive histological phenotype. The early conversion into a RAIR tumor may also be predictive of shorter survival. The stepwise treatment approach has to be individualized based on molecular and clinical features owing to the lack of large series to guide the best treatment sequence. In the present systematic review, several characteristics of PDTC, such as clinicopathological traits and treatment options, are covered and discussed based on a total of 82case reports and cohort studies found in addition to our case.

5.1. Age and Sex

PDTC represents a rare but aggressive cancer subtype originating from thyroid follicular cells and making up approximately 2–7% of all TC [120]. Our systematic review has confirmed the limited data on PDTC cases, as only 82articles were included in our review, and so far, few cohort studies have been conducted. The age range registered herein is 12–93 years, with the majority of the patients being older than 50 years. We observed similar incidences between male and female patients, with a slight female predominance in a few cohort studies. Generally, PDTC is more common in adulthood, with a mean patient age of 60 years; though, cases of young patients have also been described [121]. The disease shows a slight predominance in females, which is concordance with DTC incidence in women [122].

5.2. Surgery and RAI Treatment

Since cytological diagnosis of PDTC based on FNAC is challenging, our review has shown that some patients underwent a hemithyroidectomy or subtotal thyroidectomy with a completion thyroidectomy after histological diagnosis [38,44,52,54,59,65,69,85,103,104,107,108,115,117], but the majority of them were submitted to a total thyroidectomy as the initial operation [40,41,42,43,46,48,49,50,51,53,54,55,56,58,60,61,62,64,67,68,69,72,73,75,79,80,81,82,85,86,88,89,90,91,92,93,95,96,100,101,102,103,106,107,108,109,110,112,113,117]. Lymph node dissection was applied in most patients [40,41,42,46,47,49,50,58,61,62,67,71,73,74,75,77,78,79,80,81,82,94,95,96,99,100,101,102,103,106,107,109,110,112,114,117], but the general philosophy was to carry out central or lateral neck dissection, provided that clinical or radiological enlargement of the nodes was evident. Accordingly, in several of the revised cases, sequential local resections were applied, depending on the respective recurrence.
While surgical resection remains the mainstay of treatment, the role of RAI remains controversial. PDTC as an intermediate between DTC and ATC may retain its ability to express thyroglobulin and uptake radioiodine, reflecting its partially well-differentiated features. Compared to DTC, PDTC does not usually respond to therapy, despite partial iodine avidity. Given the higher rate of ETE, R1 surgery, lymph node positivity, and distant metastases, adjuvant treatment should be considered. Sanders et al. [9] recommended considering adjuvant RAI in all PDTC patients, thereby gaining a potential benefit without risking significant morbidity. However, despite the RAI avidity in a high percentage of PDTC cases, no significant impact on survival has been reported after RAI treatment. Unlike in DTC [123,124], RAI appears to be relatively ineffective in the control of distant metastases in PDTC; thus, it is reasonable to consider the use of RAI for metastatic/advanced disease that demonstrates definite iodine uptake [125,126]. Likewise, most cases in our review received at least one cycle of RAI treatment, even without detectable metastatic foci [38,39,40,41,42,43,46,48,49,50,51,53,54,55,58,59,60,61,62,63,64,65,66,67,68,69,73,74,75,80,81,82,85,86,87,88,90,91,97,99,101,102,104,105,106,107,108,109,110,111,112,113,115,117]. In our case report, RAIR disease was evident after two RAI treatments. External beam radiation therapy (EBRT) is a viable option for controlling local disease in patients with PDTC and was utilized in the subset of patients with unresected, locally advanced disease or with metastatic foci such as bone lesions [42,49,50,58,60,61,62,63,64,67,68,69,70,74,75,80,81,85,90,91,97,98,102,106,107,108,109,112,113,116].

5.3. Histology

Our patient’s tumor histology was confirmatory of concurrent PDTC, hobnail, and tall-cell variant, all supportive of its aggressive nature, while all lymph node metastases from the first half of the long journey of this disease originated from the differentiated component of the tumor. The aggressive concomitant histological subtypes present in our case, hobnail PTC is a particularly rare variant (approximately 1% of PTC) [19], with frequent BRAF, p53, and hTERT mutations [127], usually adopting an aggressive clinical course with distant metastases, RAI refractoriness, and mortality in a significant subset of patients has been reviewed elsewhere [34]. The identification of malignancy patterns of growth (solid, trabecular, or insular) is usually the first hint of the diagnosis of PDTC. Our systematic review showed that an insular pattern is more common than the other two, although they can coexist. According to the 5th edition of the WHO classification of tumors of endocrine organs, when a mixture of differentiated and poorly differentiated areas are incorporated in the same tumor, the least differentiated tumor component, even if non-predominant, should be recorded. However, it remains debatable whether the cut-off value for the poorly differentiated area should be reported in PDTC tumors. Based on our review, several cases had a multiconstituent histology of well-differentiated (mostly PTC and FTC) with poorly differentiated areas without elaborating on their extent [49,50,55,56,58,69,71,72,73,91,100,103,107,109,112,113]. Interestingly, the Japanese society of thyroid surgery emphasizes that the presence of a poorly differentiated component should be acknowledged as a distinct entity from DTC, independent of its histological extent within the tumor [128]. Studies have utilized the poorly differentiated proportion to distinguish PDTC and DTC with poorly differentiated areas, advocating its difference in terms of natural history and prognosis [91]. In the majority of studies in our review, the extent of the poorly differentiated area was not reported, and a PDTC definition was decided to address this component. A large number of patients with PDTC reviewed herein presented with locally advanced disease (T3 or T4, extrathyroidal extension, and nodal involvement) [40,44,46,47,49,50,51,54,55,57,58,60,61,62,64,67,68,69,70,71,73,74,75,76,77,78,80,81,82,83,84,85,86,87,88,90,91,92,94,97,98,99,100,101,102,107,108,109,111,112,113,114,115,116,117,119] and distant metastases at presentation or during later stages, mostly in the lungs and bones [40,41,42,43,44,45,46,48,49,50,51,54,55,57,58,60,61,62,63,66,67,68,69,70,71,72,73,74,75,76,80,81,82,83,84,85,87,88,89,90,91,92,93,94,95,97,98,101,102,103,106,107,108,109,110,111,112,114,116,117,119]. These findings imply that even in our case and despite the fact of the failure to have a biopsy during the progression of the disease the metastases originated from the poorly differentiated component.

5.4. Molecular Events in PDTC

The currently accepted model of TC oncogenesis is the multistep model. The fact that DTC can harbor foci of PDTC and ATC strengthens this hypothesis [29,129,130]. Progression from follicular cells to DTC is marked by activating mutations of the MAPK (mitogen-activated protein kinase) or/and PI3K/AKT (phosphatidylinositol 3-kinase/protein kinase B) pathways. Further progression to PDTC and ATC is characterized by additional mutations and genetic and epigenetic events that favor genetic instability and oncogenesis [29]. The development of PDTC is hypothesized to be secondary to this “multi-hit” process, with TERT promoter mutations being the most common molecular findings in PDTC (40%), with stepwise increases from PTC (9%) [131] to PDTC and ATC (65–73%) [132,133]. Concomitant mutations in RET/PTC, RAS, and BRAF promoters have been observed in advanced PTC and confer an unfavorable prognosis [134,135,136,137,138]. BRAFV600E is reported as the most common mutation, and its presence is associated with higher mortality [139], higher risk of recurrence [140], and loss of NIS (sodium/iodide symporter) expression [141]. However, this can also be observed in low-risk PTCs [142,143]. In our case report the presence of the BRAFV600E mutation correlated with a highly recurrent malignancy, in line with previous findings regarding the increased aggressiveness of BRAF-positive tumors. Interestingly, throughout our systematic review, the most commonly detected genetic events in PDTC were BRAF, RAS, and the TERT promoter. Additionally, for some of the cases, PTEN and/or TP53 alterations were identified (Table 1).

5.5. Survival

The modern criteria for PDTC were established in the 2007 Turin consensus proposal and have been reaffirmed by the 5th edition of the WHO classification of endocrine and neuroendocrine tumors [144]. Being morphologically and biologically intermediate between DTC and ATC, PDTCs usually exhibit more aggressive behaviors than DTCs, namely ETE and distant metastasis, coinciding with poorer outcomes [145]. Despite its rarity, PDTC accounts for most fatalities from non-anaplastic follicular cell-derived TC, holding an intermediate position between DTC and ATC in terms of five-year overall survival (OS), disease-specific survival (DSS), and disease-free survival. [146]. This is also depicted in our review, where few patients went disease-free [36,39,45,52,53,54,56,58,59,64,65,68,71,72,87,96,99,103,104,106,108,117,118]. Distant metastases represent the most significant cause of death in PDTC, with the majority of patients succumbing to the disease.

5.6. Chemotherapy

At present, there is no effective targeted chemotherapeutic regime for PDTC. Cisplatin/doxorubicin and carboplatin/paclitaxel are similar combinations that are used to modulate disease progression, and likewise, only a few of the revised studies used these schemes for select patients [42,57,60,78,97,114,119].

5.7. Tyrosine Kinase Inhibitors

The role of tyrosine kinase inhibitors is evolving as a promising approach for treating PDTC in the near future. Sorafenib and lenvatinib have been approved by the U.S. Food and Drug Administration (FDA) for progressive, recurrent, or metastatic follicular cell-derived RAIR TCs. This was based on two seminal phase 3 trials (DECISION and SELECT, respectively), which included a limited number of PDTC cases, with the respective subgroup analyses available in the appendix of the studies [147,148]. In fact, in the cases discussed herein, TKIs, especially lenvatinib and sorafenib, were widely used in metastatic disease [42,44,50,55,60,67,68,73,75,80,81,84,89,90,94,98,109,110,111]. The actual benefit in terms of patient survival remains to be seen, since, in some patients, control was obtained [42,44,64,67,73,80,81,84,97,110], while others succumbed to the disease [55,60,86,89,90,94,97,98,109,111]. In the sole cohort study including 8 PDTC patients who had all received lenvatinib, this TKI treatment achieved a median progression-free survival (PFS) of 12 months [94]. In our case, where lenvatinib was administered alternatively with sorafenib, due to their AEs, the combined PFS had reached 30 months.
While Sorafenib [147] and Lenvatinib [148] are still considered first-line treatments in advanced RAIR TCs, patients with the BRAFV600E mutation may benefit from treatment with BRAF-I once they have progressed on standard-of-care treatments. BRAF-Is (dabrafenib, vemurafenib) have been implemented for the management of TC [30,149]. BRAF-Is have not yet been FDA approved for the treatment of BRAF-mutant RAIR-DTC. A dabrafenib/tramatenib combination was recently FDA approved for the treatment of BRAF-mutated ATC [150], while a phase II randomized clinical trial studying the use of a BRAF-I (dabrafenib) and BRAF-I/MEK-I (dabrafenib/tramatenib) combination in BRAF-mutated DTC demonstrated high objective response rates and efficacy of both the single agent and the combination [35]. An ongoing global, multicenter, randomized, double-blind, placebo-controlled phase III study aims to evaluate the efficacy and safety of dabrafenib/trametinib in adult patients with locally advanced or metastatic BRAFV600E mutation-positive RAIR-DTC that have progressed following prior VEGFR targeted therapy. Patients are randomized to either dabrafenib/trametinib or a placebo and stratified based on the number of prior VEGFR targeted therapies (1 versus 2) and prior lenvatinib treatment (NCT04940052) [151]. In our BRAF V600E-positive case, due to drug intolerance and disease progression, a change from sorafenib/lenvatinib to the BRAF/MEK-I combination resulted in remarkable shrinkage of all lesions, leading to a further PFS of 24 months. A similar combination of dabrafenib/trametinib, as administered in our patient, was used to treat two PDTC patients, one of them presenting disease remission in one of the revised studies [92]. These data suggest potential benefits and a better tolerability profile of the dabrafenib/trametinib combination in PDTC patients, which should urge for further studies investigating appropriate sequelae of treatments. We could speculate that an earlier administration may increase the PFS in these patients. Another newly employed treatment approach in PDTC cases is the co-administration of lenvatinib and pembrolizumab. The combination of both TKI and classic immunotherapy achieved a median PFS of 17.7 months in two PDTC patients [60].
The critical limitations of targeted therapies are their major AEs profile, as in our case, that occasionally cannot be counteracted by dosede-escalation and/or adjunct agents and, most importantly, the development of escape mechanisms developed by the tumor. Although targeted therapies exhibited significant efficacy in our case, the related AEs were also notable. The balance between drug efficacy, management of AEs, and drug resistance is truly challenging. The fact that the occurrence of certain AEs has been proposed as a predictive response marker (hypertension correlated with improved outcomes on lenvatinib treatment) [152] calls for the timely implementation of preventive strategies to avoid toxicity and prolong therapeutic use.
There are certain limitations of our systematic review that should be addressed. Although we narrowed down our search to the time period after 2017, we did not evaluate the histopathological reviews of the presented studies to inspect their unanimity. We relied on the diagnosis of PDTC presented in each study, whether a detailed pathology review was provided or not. Furthermore, since the majority of the studies did not report the exact percentage of the PDTC component in DTC, we decided to include any relevant study with any component of poorly differentiated area in the pathology review. Additionally, despite the exclusion of pediatric populations, we decided to include a few cohort studies that also included, among others, minor patients [54,58,74,91,112,113], since they could shed light on the characteristics of PDTC in a wider population.

6. Conclusions

PDTC is a histological entitiy that isclassified as intermediate in the spectrum of TC in terms of biological behavior and prognosis. In terms of therapeutics, it poses a challenge, as itis frequently not RAI avid, limiting current therapeutic options. However, with insight into molecular mechanisms rapidly increasing, TC therapy should be individualized on a case-by-case basis, employing molecular diagnostics in everyday practice. Molecular testing is now warranted to plan the treatment strategy for both anaplastic and PDTC. The detection of BRAF mutations, RET fusions, or NTRK rearrangements is of utmost importance for diagnosis and treatment, considering the reported promising results of novel targeted therapies. Herein, we report a case of metastatic mixed PDTC and present the efficacy of the sequential administration of targeted drugs. The most favorable therapeutic sequence for each individual patient or group of patients remains to be further elucidated as new evidence comes to light.

Author Contributions

Conceptualization, K.I.A.; methodology, O.V. and J.A.; software, O.V.; validation, K.I.A., A.S. and P.K.; formal analysis, O.V.; investigation, O.V., P.K., A.X. and P.B.K.; resources, K.I.A., S.T., D.K. and M.T.; data curation, K.I.A., O.V., A.S. and P.K.; writing—original draft preparation, P.K., O.V. and A.S.; writing—review and editing, K.I.A., M.K., G.M. and J.A.; visualization, K.I.A. and A.S.; supervision, K.I.A.; project administration, K.I.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was approved by the local institutional ethics committee with a reference number 363/13-10-2021.

Informed Consent Statement

Informed consent was obtained from the patient for the publication of this study.

Data Availability Statement

The data will be available upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA A Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
  2. Seib, C.D.; Sosa, J.A. Evolving Understanding of the Epidemiology of Thyroid Cancer. Endocrinol. Metab. Clin. N. Am. 2018, 48, 23–35. [Google Scholar] [CrossRef]
  3. Davies, L.; Welch, H.G. Current Thyroid Cancer Trends in the United States. JAMA Otolaryngol. Head Neck Surg. 2014, 140, 317–322. [Google Scholar] [CrossRef]
  4. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef]
  5. Howlader, N.; Noone, A.; Krapcho, M.; Miller, D.; Bishop, K.; Altekruse, S.F.; Kosary, C.L.; Yu, M.; Ruhl, J.; Tatalovich, Z.; et al. (Eds.) Seer Cancer Statistics Review, 1975–2013; National Cancer Institute: Bethesda, MD, USA, 2015.
  6. La Vecchia, C.; Malvezzi, M.; Bosetti, C.; Garavello, W.; Bertuccio, P.; Levi, F.; Negri, E. Thyroid cancer mortality and incidence: A global overview. Int. J. Cancer 2014, 136, 2187–2195. [Google Scholar] [CrossRef]
  7. Ahn, H.S.; Kim, H.J.; Welch, H.G. Korea’s thyroid-cancer “epidemic”—Screening and overdiagnosis. N. Engl. J. Med. 2014, 371, 1765–1767. [Google Scholar] [CrossRef]
  8. Aschebrook-Kilfoy, B.; Ward, M.H.; Sabra, M.M.; Devesa, S.S. Thyroid Cancer Incidence Patterns in the United States by Histologic Type, 1992–2006. Thyroid 2011, 21, 125–134. [Google Scholar] [CrossRef]
  9. Sanders, E.M., Jr.; LiVolsi, V.A.; Brierley, J.; Shin, J.; Randolph, G.W. An evidence-based review of poorly differentiated thyroid cancer. World J. Surg. 2007, 31, 934–945. [Google Scholar] [CrossRef]
  10. Asioli, S.; Erickson, L.A.; Righi, A.; Jin, L.; Volante, M.; Jenkins, S.; Papotti, M.; Bussolati, G.; Lloyd, R.V. Poorly differentiated carcinoma of the thyroid: Validation of the Turin proposal and analysis of IMP3 expression. Mod. Pathol. 2010, 23, 1269–1278. [Google Scholar] [CrossRef]
  11. 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]
  12. Pereira, M.M.; Williams, V.L.; Johnson, J.H.; Valderrabano, P. Thyroid Cancer Incidence Trends in the United States: Association with Changes in Professional Guideline Recommendations. Thyroid 2020, 30, 1132–1140. [Google Scholar] [CrossRef]
  13. Schneider, D.F.; Chen, H. New developments in the diagnosis and treatment of thyroid cancer. CA A Cancer J. Clin. 2013, 63, 373–394. [Google Scholar] [CrossRef]
  14. Singhal, S.; Sippel, R.S.; Chen, H.; Schneider, D.F. Distinguishing classical papillary thyroid microcancers from follicular-variant microcancers. J. Surg. Res. 2014, 190, 151–156. [Google Scholar] [CrossRef]
  15. Englum, B.R.; Pura, J.; Reed, S.D.; Roman, S.A.; Sosa, J.A.; Scheri, R.P. A Bedside Risk Calculator to Preoperatively Distinguish Follicular Thyroid Carcinoma from Follicular Variant of Papillary Thyroid Carcinoma. World J. Surg. 2015, 39, 2928–2934. [Google Scholar] [CrossRef]
  16. Oyer, S.L.; Fritsch, V.A.; Lentsch, E.J. Comparison of Survival Rates Between Papillary and Follicular Thyroid Carcinomas Among 36,725 Patients. Ann. Otol. Rhinol. Laryngol. 2014, 123, 94–100. [Google Scholar] [CrossRef]
  17. Lloyd, R.V.; Osamura, R.; Kloppel, G.; Rosai, J. WHO Classification of Tumours of Endocrine Organs, 4th ed.; International Agency for Research on Cancer: Lyon, France, 2017. [Google Scholar]
  18. Asioli, S.; Erickson, L.A.; Righi, A.; Lloyd, R.V. Papillary thyroid carcinoma with hobnail features: Histopathologic criteria to predict aggressive behavior. Hum. Pathol. 2013, 44, 320–328. [Google Scholar] [CrossRef]
  19. Asioli, S.; Erickson, L.A.; Sebo, T.J.; Zhang, J.; Jin, L.; Thompson, G.B.; Lloyd, R.V. Papillary Thyroid Carcinoma with Prominent Hobnail Features: A New Aggressive Variant of Moderately Differentiated Papillary Carcinoma. A Clinicopathologic, Immunohistochemical, and Molecular Study of Eight Cases. Am. J. Surg. Pathol. 2010, 34, 44–52. [Google Scholar] [CrossRef]
  20. Ghossein, R.A.; Leboeuf, R.; Patel, K.N.; Rivera, M.; Katabi, N.; Carlson, D.L.; Tallini, G.; Shaha, A.; Singh, B.; Tuttle, R.M. Tall Cell Variant of Papillary Thyroid Carcinoma without Extrathyroid Extension: Biologic Behavior and Clinical Implications. Thyroid 2007, 17, 655–661. [Google Scholar] [CrossRef]
  21. Ito, Y.; Hirokawa, M.; Fukushima, M.; Inoue, H.; Yabuta, T.; Uruno, T.; Kihara, M.; Higashiyama, T.; Takamura, Y.; Miya, A.; et al. Prevalence and Prognostic Significance of Poor Differentiation and Tall Cell Variant in Papillary Carcinoma in Japan. World J. Surg. 2008, 32, 1535–1543. [Google Scholar] [CrossRef]
  22. Wenig, B.M.; Thompson, L.D.; Adair, C.F.; Shmookler, B.; Heffess, C.S. Thyroid papillary carcinoma of columnar cell type: A clinicopathologic study of 16 cases. Cancer 1998, 82, 740–753. [Google Scholar] [CrossRef]
  23. Chen, J.-H.; Faquin, W.C.; Lloyd, R.V.; Nosé, V. Clinicopathological and molecular characterization of nine cases of columnar cell variant of papillary thyroid carcinoma. Mod. Pathol. 2011, 24, 739–749. [Google Scholar] [CrossRef]
  24. Chereau, N.; Giudicelli, X.; Pattou, F.; Lifante, J.-C.; Triponez, F.; Mirallié, E.; Goudet, P.; Brunaud, L.; Trésallet, C.; Tissier, F.; et al. Diffuse Sclerosing Variant of Papillary Thyroid Carcinoma Is Associated with Aggressive Histopathological Features and a Poor Outcome: Results of a Large Multicentric Study. J. Clin. Endocrinol. Metab. 2016, 101, 4603–4610. [Google Scholar] [CrossRef]
  25. Uchino, S.; Ishikawa, H.; Miyauchi, A.; Hirokawa, M.; Noguchi, S.; Ushiama, M.; Yoshida, T.; Michikura, M.; Sugano, K.; Sakai, T. Age- and gender specific risk of thyroid cancer in patients with familial adenomatous polyposis. J. Clin. Endocrinol. Metab. 2016, 101, 4611–4617. [Google Scholar] [CrossRef]
  26. Lam, A.K.-Y.; Saremi, N. Cribriform-morular variant of papillary thyroid carcinoma: A distinctive type of thyroid cancer. Endocr. Relat. Cancer 2017, 24, R109–R121. [Google Scholar] [CrossRef]
  27. Hong, J.H.; Yi, H.; Yi, S.; Kim, H.; Lee, J.; Kim, K.S. Implications of oncocytic change in papillary thyroid cancer. Clin. Endocrinol. 2016, 85, 797–804. [Google Scholar] [CrossRef]
  28. Nilubol, N.; Kebebew, E. Should small papillary thyroid cancer be observed? A population-based study. Cancer 2014, 121, 1017–1024. [Google Scholar] [CrossRef]
  29. Landa, I.; Cabanillas, M.E. Genomic alterations in thyroid cancer: Biological and clinical insights. Nat. Rev. Endocrinol. 2023, 20, 93–110. [Google Scholar] [CrossRef]
  30. Brose, M.S.; Cabanillas, M.E.; Cohen, E.E.W.; Wirth, L.J.; Riehl, T.; Yue, H.; Sherman, S.I.; Sherman, E.J. Vemurafenib in patients with BRAFV600E-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: A non-randomised, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 1272–1282. [Google Scholar] [CrossRef]
  31. Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.-K.; Attar, N.; Sazegar, H.; et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010, 468, 973–977. [Google Scholar] [CrossRef]
  32. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  33. Tuttle, R.M.; Haugen, B.; Perrier, N.D. Updated American Joint Committee on Cancer/Tumor-Node-Metastasis Staging System for Differentiated and Anaplastic Thyroid Cancer (Eighth Edition): What Changed and Why? Thyroid 2017, 27, 751–756. [Google Scholar] [CrossRef]
  34. Spyroglou, A.; Kostopoulos, G.; Tseleni, S.; Toulis, K.; Bramis, K.; Mastorakos, G.; Konstadoulakis, M.; Vamvakidis, K.; Alexandraki, K.I. Hobnail Papillary Thyroid Carcinoma, A Systematic Review and Meta-Analysis. Cancers 2022, 14, 2785. [Google Scholar] [CrossRef]
  35. Busaidy, N.L.; Konda, B.; Wei, L.; Wirth, L.J.; Devine, C.; Daniels, G.A.; DeSouza, J.A.; Poi, M.; Seligson, N.D.; Cabanillas, M.E. Dabrafenib Versus Dabrafenib + Trametinib in BRAF-Mutated Radioactive Iodine Refractory Differentiated Thyroid Cancer: Results of a Randomized, Phase 2, Open-Label Multicenter Trial. Thyroid 2022, 32, 1184–1192. [Google Scholar] [CrossRef]
  36. Jeon, Y.; Park, S.; Lee, S.-H.; Kim, T.H.; Kim, S.W.; Ahn, M.-J.; Jung, H.A.; Chung, J.H. Combination of Dabrafenib and Trametinib in Patients with Metastatic BRAFV600E-Mutated Thyroid Cancer. Cancer Res. Treat. 2024. [Google Scholar] [CrossRef]
  37. Brose, M.S.; Robinson, B.G.; Sherman, S.I.; Jarzab, B.; Lin, C.; Vaisman, F.; Hoff, A.O.; Hitre, E.; Bowles, D.W.; Sen, S.; et al. Cabozantinib for previously treated radioiodine-refractory differentiated thyroid cancer: Updated results from the phase 3 COSMIC-311 trial. Cancer 2022, 128, 4203–4212. [Google Scholar] [CrossRef]
  38. Abdellaoui, W.; Assarrar, I.; Benyakhlef, S.; Tahri, A.; Messaoudi, N.; Haloui, A.; Rouf, S.; Bennani, A.; Latrech, H. Insular thyroid carcinoma in a young Moroccan man: Case report and review of the literature. Ann. Med. Surg. 2022, 77, 103592. [Google Scholar] [CrossRef]
  39. Abu Rumman, A.M.; Alsoudi, M.A.; Qasimeh, H.M.; Alnajada, W.A.; Alshunnaq, R.Y. Isolated poorly differentiated cancer (insular) in a thyroglossal cyst: A case report. Pan. Afr. Med. J. 2021, 39. [Google Scholar] [CrossRef]
  40. Agarwal, A.; George, N.; Kumari, N.; Krishnani, N.; Mishra, P.; Gupta, S. Poorly differentiated thyroid cancer: Clinical, pathological, mutational, and outcome analysis. Indian J. Pathol. Microbiol. 2024. [Google Scholar] [CrossRef]
  41. Ahmadian, S.S.; Jones, D.; Wakely, P., Jr.; Lott Limbach, A.A. Thyroid poorly differentiated carcinoma metastatic to pancreas diagnosed by fine-needle aspiration and demonstrating a novel BRAF fusion. Cytopathology 2024, 35, 408–411. [Google Scholar] [CrossRef]
  42. Alshehri, K.; Alqurashi, Y.; Merdad, M.; Samargandy, S.; Daghistani, R.; Marzouki, H. Neoadjuvant lenvatinib for inoperable thyroid cancer: A case report and literature review. Cancer Rep. 2021, 5, e1466. [Google Scholar] [CrossRef]
  43. Altiner, H.I.; Keskin, H.; Günel, C. Low Differential Thyroid Cancer Metastasizing to the Sphenoid Sinus and Orbital Apex: A Case Report. Eur. J. Rhinol. Allergy 2023, 6, 34–36. [Google Scholar] [CrossRef]
  44. Alzahrani, A.S.; Alswailem, M.; Alghamdi, B.; Al-Hindi, H. Fumarate Hydratase is a Novel Gene for Familial Non-Medullary Thyroid Cancer. J. Clin. Endocrinol. Metab. 2022, 107, 2539–2544. [Google Scholar] [CrossRef]
  45. Ambre, S.; Sultania, M.; Biswal, S.; Mitra, S.; Sahoo, B.; Muduly, D.K.; Kar, M. Poorly differentiated “insular” thyroid carcinoma with solitary vascular mandibular metastasis–A rare histology and management. Oral Oncol. 2021, 124, 105416. [Google Scholar] [CrossRef]
  46. Atif, Z.; Morante, J.; Curiel, L.; Doshi, K.; Babury, M. Hemoptysis as an Initial Presentation of Poorly Differentiated Thyroid Carcinoma: An Ominous Presentation. Am. J. Respir. Crit. Care Med. 2018, 197, A4152. [Google Scholar]
  47. Bellini, M.I.; Biffoni, M.; Patrone, R.; Borcea, M.C.; Costanzo, M.L.; Garritano, T.; Melcarne, R.; Menditto, R.; Metere, A.; Scorziello, C.; et al. Poorly Differentiated Thyroid Carcinoma: Single Centre Experience and Review of the Literature. J. Clin. Med. 2021, 10, 5258. [Google Scholar] [CrossRef]
  48. Bertrand, A.-S.; Iannessi, A.; Peyrottes, I.; Lacout, A.; Thyss, A.; Marcy, P.-Y. Myoma Hot Spot: Tumor-to-Tumor Metastasis of Thyroid Origin into Uterine Leiomyoma. Eur. Thyroid. J. 2019, 8, 273–277. [Google Scholar] [CrossRef]
  49. Beute, J.E.; Silberzweig, A.M.; Gold, B.S.; Shaari, A.L.; Kapustin, D.A.; Matloob, A.; Dowling, E.M.; Starc, M.T.; Fan, J.; Khorsandi, A.S.; et al. Thyroid cancer necrosis not evident on imaging: A cautionary case series on poorly differentiated thyroid carcinoma diagnosed only on final pathology. Am. J. Otolaryngol. 2024, 45, 104054. [Google Scholar] [CrossRef]
  50. Bicho, R.A.; Mishra, A.; Kumari, N.; Krishnani, N.; Chand, G.; Agarwal, G.; Agarwal, A.; Mishra, S.K. Poorly differentiated thyroid carcinoma and poorly differentiated area in differentiated thyroid carcinoma: Is there any difference? Langenbecks Arch. Surg. 2019, 404, 45–53. [Google Scholar] [CrossRef]
  51. Elsheikh, M.; Hemmings, C.B.; Rastogi, N.; Schultz, A.; Brijmohan, S.H. Unusual Clinical Manifestations of Thyroid Carcinoma. Cureus 2023, 15, e37474. [Google Scholar] [CrossRef]
  52. Ching, D.; Leslie, C. Atypical Features Resembling Poorly Differentiated Thyroid Carcinoma Presenting Entirely within a Follicular Adenoma. Case Rep. Pathol. 2018, 2018, 7290343. [Google Scholar] [CrossRef]
  53. Choi, J.H.; Hong, Y.O.; Kim, H.-J.; Jung, A.R. Poorly differentiated thyroid carcinoma arising from a lithium-induced goiter in a patient with schizophrenia: A case report. Thyroid. Res. 2021, 14, 1–5. [Google Scholar] [CrossRef] [PubMed]
  54. Choi, S.W.; Lee, J.-H.; Kim, Y.Y.; Chung, Y.S.; Choi, S.; Kim, N.R.; Kang, J.M.; Park, H.K.; Chun, Y.S. Poorly differentiated thyroid carcinoma: An institutional experience. Korean J. Clin. Oncol. 2020, 16, 25–32. [Google Scholar] [CrossRef] [PubMed]
  55. Colombo, C.; Pogliaghi, G.; Tosi, D.; Muzza, M.; Bulfamante, G.; Persani, L.; Fugazzola, L.; Cirello, V. Thyroid cancer harboring PTEN and TP53 mutations: A peculiar molecular and clinical case report. Front. Oncol. 2022, 12, 949098. [Google Scholar] [CrossRef] [PubMed]
  56. Corean, J.; Furtado, L.V.; Kadri, S.; Segal, J.P.; Emerson, L.L. Cribriform-Morular Variant of Papillary Thyroid Carcinoma with Poorly Differentiated Features: A Case Report with Immunohistochemical and Molecular Genetic Analysis. Int. J. Surg. Pathol. 2018, 27, 294–304. [Google Scholar] [CrossRef] [PubMed]
  57. Das, J.; Ghosh, J.; Zameer, L.; Ray, S. 18F-fluorodeoxyglucose positron emission tomography/computed tomography finding in a rare case of follicular carcinoma of thyroid with rhabdoid morphology. Indian J. Nucl. Med. 2021, 36, 56–58. [Google Scholar] [CrossRef] [PubMed]
  58. de la Fouchardière, C.; Decaussin-Petrucci, M.; Berthiller, J.; Descotes, F.; Lopez, J.; Lifante, J.-C.; Peix, J.-L.; Giraudet, A.-L.; Delahaye, A.; Masson, S.; et al. Predictive factors of outcome in poorly differentiated thyroid carcinomas. Eur. J. Cancer 2018, 92, 40–47. [Google Scholar] [CrossRef] [PubMed]
  59. Dettmer, M.S.; Hürlimann, S.; Scheuble, L.; Vassella, E.; Perren, A.; Wicke, C. Cribriform Morular Thyroid Carcinoma-Ultimobranchial Pouch-Related? Deep Molecular Insights of a Unique Case. Endocr. Pathol. 2023, 34, 342–348. [Google Scholar] [CrossRef] [PubMed]
  60. 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] [PubMed]
  61. Elshafie, O.; Jain, A.; Bichpuria, S.; Rassou, Y.; Hashmi, S.F.; Khalil, A.B. Calcaneus metastasis: A rare presentation of poorly differentiated thyroid cancer. Endocrinol. Diabetes Metab. Case Rep. 2023, 2023, 23-0103. [Google Scholar] [CrossRef]
  62. Farahmandfar, F.; Shakeri, S.; Askari, E.; Jafarian, A.H.; Jashmidi, S.T.; Shafiri, S.; Zakavi, S.R. Parotid metastasis as the first presentation of papillary thyroid carcinoma. Iran. J. Nucl. Med. 2020, 28, 42–45. [Google Scholar]
  63. Feffer, J.B.; Usera, G.L.; Schulman, R.C. Unilateral Exophthalmos Due to Metastasis of Poorly Differentiated Thyroid Carcinoma to The Left Sphenoid Wing with Intra-Orbital Extension. AACE Clin. Case Rep. 2017, 3, e89–e92. [Google Scholar] [CrossRef]
  64. Gay, S.; Monti, E.; Antonelli, C.T.; Mora, M.; Spina, B.; Ansaldo, G.; Teliti, M.; Comina, M.; Conte, L.; Minuto, M.; et al. Case report: Lenvatinib in neoadjuvant setting in a patient affected by invasive poorly differentiated thyroid carcinoma. Futur. Oncol. 2019, 15 (Suppl. S24), 13–19. [Google Scholar] [CrossRef] [PubMed]
  65. Gazeu, A.; Lopez, J.; Guyetant, S.; Sobrinho-Simoes, M.; Lifante, J.C.; Cugnet-Anceau, C.; Decaussin-Petrucci, M. Poorly differentiated thyroid carcinoma with pleomorphic giant cells—A case report. Virchows Arch. 2020, 477, 597–601. [Google Scholar] [CrossRef] [PubMed]
  66. Gill, S.M.; Hassan, A.; Bashir, H.; Shafiq, W. I-131 Avid Tumor Thrombus in a Case of Poorly Differentiated Thyroid Cancer. Mol. Imaging Radionucl. Ther. 2023, 32, 178–180. [Google Scholar] [CrossRef] [PubMed]
  67. Goto, H.; Kiyota, N.; Otsuki, N.; Imamura, Y.; Chayahara, N.; Suto, H.; Nagatani, Y.; Toyoda, M.; Mukohara, T.; Nibu, K.-I.; et al. Successful treatment switch from lenvatinib to sorafenib in a patient with radioactive iodine-refractory differentiated thyroid cancer intolerant to lenvatinib due to severe proteinuria. Auris Nasus Larynx 2018, 45, 1249–1252. [Google Scholar] [CrossRef] [PubMed]
  68. Grawe, F.; Cahya, A.; Fabritius, M.P.; Beyer, L.; Wenter, V.; Ruebenthaler, J.; Geyer, T.; Burgard, C.; Bartenstein, P.; Ilhan, H.; et al. Course of Disease and Clinical Management of Patients with Poorly Differentiated Thyroid Carcinoma. Cancers 2021, 13, 5309. [Google Scholar] [CrossRef] [PubMed]
  69. Gubbiotti, M.A.; Andrianus, S.; Sakhi, R.; Zhang, Q.; Montone, K.; Jalaly, J.B.; Baloch, Z. Does the presence of capsule influence prognosis in poorly differentiated thyroid carcinoma? Hum. Pathol. 2023, 136, 96–104. [Google Scholar] [CrossRef] [PubMed]
  70. Gülbahar Ateş, S.; Demiral, B.B.; Ucmak, G. Poorly differentiated thyroid cancer with an extensive tumor thrombus in superior vena cava on 18F-FDG PET/CT: A case report. Med. Nucl. 2024, 48, 161–164. [Google Scholar] [CrossRef]
  71. Hu, J.; Xu, X.; Wang, S.; Dong, F.; Zhang, X.; Ming, J.; Huang, T. Case Report: Implantation of Dedifferentiated to Poorly Differentiated Thyroid Carcinoma After Endoscopic Thyroid Surgery. Front. Oncol. 2022, 12, 896942. [Google Scholar] [CrossRef]
  72. Ieni, A.; Fadda, G.; Alario, G.; Pino, A.; Ficarra, V.; Dionigi, G.; Tuccari, G. Metastatic thyroid carcinoma mimicking as a primary neoplasia of the kidney: A case report. Mol. Clin. Oncol. 2021, 15, 268. [Google Scholar] [CrossRef]
  73. Iravani, A.; Solomon, B.; Pattison, D.A.; Jackson, P.; Kumar, A.R.; Kong, G.; Hofman, M.S.; Akhurst, T.; Hicks, R.J. Mitogen-Activated Protein Kinase Pathway Inhibition for Redifferentiation of Radioiodine Refractory Differentiated Thyroid Cancer: An Evolving Protocol. Thyroid 2019, 29, 1634–1645. [Google Scholar] [CrossRef] [PubMed]
  74. Isaev, P.A.; Polkin, V.V.; Severskaya, N.V.; Ilyin, A.A.; Plugar, A.K.; Ivanov, S.A.; Kaprin, A.D. Results of treatment of patients with poorly differentiated carcinoma of the thyroid gland. Head Neck Tumors 2023, 12, 17–24. [Google Scholar] [CrossRef]
  75. Kalshetty, A.; Basu, S. Thyroglobulin "Nonsecretor" Metastatic Poorly Differentiated Thyroid Carcinoma with Noniodine Concentrating Disease and Aggressive Clinical Course: A Clinical Case Series. Indian J. Nucl. Med. 2018, 33, 218–223. [Google Scholar] [PubMed]
  76. Kersting, D.; Rischpler, C.; Plönes, T.; Aigner, C.; Umutlu, L.; Herrmann, K.; Hautzel, H. Atypical bilateral ventilation/perfusion mismatches in an asymptomatic patient suffering from metastatic thyroid cancer. Eur. J. Hybrid Imaging 2021, 5, 1–5. [Google Scholar] [CrossRef] [PubMed]
  77. Khetrapal, S.; Rana, S.; Jetley, S.; Jairajpuri, Z. Poorly differentiated carcinoma of thyroid: Case report of an uncommon entity. J. Cancer Res. Ther. 2018, 14, 1142–1144. [Google Scholar] [CrossRef] [PubMed]
  78. Kim, K.M.; Ahn, A.R.; Hong, Y.T.; Chung, M.J. Primary small cell thyroid carcinoma combined with poorly differentiated thyroid carcinoma, evidence for a common origin: A case report. Oncol. Lett. 2023, 25, 233. [Google Scholar] [CrossRef] [PubMed]
  79. Kim, Y.; Ahn, J.; Kim, Y.S. Subcutaneous implantation of thyroid carcinoma and benign tissue after thyroidectomy: Report on two cases and review of the current literature. Gland Surg. 2023, 12, 1305–1312. [Google Scholar] [CrossRef] [PubMed]
  80. Kunte, S.; Sharett, J.; Wei, W.; Nasr, C.; Prendes, B.; Lamarre, E.; Ku, J.; Lorenz, R.R.; Scharpf, J.; Burkey, B.B.; et al. Poorly Differentiated Thyroid Carcinoma: Single Institution Series of Outcomes. Anticancer. Res. 2022, 42, 2531–2539. [Google Scholar] [CrossRef] [PubMed]
  81. Kut, C.; Liang, A.; Kiess, A.P. A Case of Fistula After Adjuvant External Beam Radiotherapy and Lenvatinib for High-Risk Follicular Thyroid Cancer. In Thyroid Cancer: A Case-Based Approach; Grani, G., Cooper, D.S., Durante, C., Eds.; Springer: Cham, Switzerland, 2020; pp. 249–260. [Google Scholar]
  82. Laforga, J.B.; Cortés, V.A. Oncocytic poorly differentiated (insular) thyroid carcinoma mimicking metastatic adenocarcinoma. A case report and review of the literature. Diagn. Cytopathol. 2019, 47, 584–588. [Google Scholar] [CrossRef]
  83. Leboulleux, S.; Dupuy, C.; Lacroix, L.; Attard, M.; Grimaldi, S.; Corre, R.; Ricard, M.; Nasr, S.; Berdelou, A.; Hadoux, J.; et al. Redifferentiation of a BRAF(K601E)-Mutated Poorly Differentiated Thyroid Cancer Patient with Dabrafenib and Trametinib Treatment. Thyroid 2019, 29, 735–742. [Google Scholar] [CrossRef]
  84. Lee, C.-S.; Miao, E.; Das, K.; Seetharamu, N. Clinical efficacy with dabrafenib and trametinib in a T599_V600insT poorly differentiated metastatic thyroid carcinoma. BMJ Case Rep. 2021, 14, e243264. [Google Scholar] [CrossRef]
  85. Lukovic, J.; Petrovic, I.; Liu, Z.; Armstrong, S.M.; Brierley, J.D.; Tsang, R.; Pasternak, J.D.; Gomez-Hernandez, K.; Liu, A.; Asa, S.L.; et al. Oncocytic Papillary Thyroid Carcinoma and Oncocytic Poorly Differentiated Thyroid Carcinoma: Clinical Features, Uptake, and Response to Radioactive Iodine Therapy, and Outcome. Front. Endocrinol. 2021, 12, 795184. [Google Scholar] [CrossRef]
  86. Molinaro, E.; Viola, D.; Viola, N.; Falcetta, P.; Orsolini, F.; Torregrossa, L.; Vagli, P.; Ribechini, A.; Materazzi, G.; Vitti, P.; et al. Lenvatinib Administered via Nasogastric Tube in Poorly Differentiated Thyroid Cancer. Case Rep. Endocrinol. 2019, 2019, 6831237. [Google Scholar] [CrossRef]
  87. Morvan, J.-B.; Boudin, L.; Metivier, D.; Delarbre, D.; Bouquillon, E.; Thariat, J.; Pascaud, D.; Marcy, P.-Y. Internal Jugular Vein Tumor Thrombus: A Tricky Question for the Thyroid Surgeon. Curr. Oncol. 2022, 29, 9235–9241. [Google Scholar] [CrossRef] [PubMed]
  88. Nagaoka, R.; Saitou, M.; Nagahama, K.; Okamura, R.; Akasu, H.; Igarashi, T.; Yokoshima, K.; Ohashi, R.; Sugitani, I. Downhill Varices in the Hypopharynx of a Patient with a Large Thyroid Tumor: A Case Report. J. Nippon. Med. Sch. 2023, 90, 408–413. [Google Scholar] [CrossRef] [PubMed]
  89. O’Donohue, P.; Lisewski, D. Intra-abdominal ectopic metastatic poorly differentiated follicular thyroid cancer. ANZ J. Surg. 2022, 92, 3053–3054. [Google Scholar] [CrossRef] [PubMed]
  90. Oh, Y.; Park, J.H.; Djunadi, T.A.; Shah, Z.; Chung, L.I.-Y.; Chae, Y.K. Deep response to a combination of mTOR inhibitor temsirolimus and dual immunotherapy of nivolumab/ipilimumab in poorly differentiated thyroid carcinoma with PTEN mutation: A case report and literature review. Front. Endocrinol. 2024, 15, 1304188. [Google Scholar] [CrossRef]
  91. Panchangam, R.B.; Puthenveetil, P.; Mayilvaganan, S. Prognostic Impact of Focal Poorly Differentiated Areas in Follicular Differentiated Thyroid Cancer: Is It a Distinct Entity from Poorly Differentiated Thyroid Cancer? Indian J. Surg. Oncol. 2022, 13, 157–163. [Google Scholar] [CrossRef]
  92. Peng, X.; Lei, J.; Li, Z.; Zhang, K. Case report: Visibly curative effect of dabrafenib and trametinib on advanced thyroid carcinoma in 2 patients. Front. Oncol. 2023, 12, 1099268. [Google Scholar] [CrossRef]
  93. Pinto, A.; Drake, T.; Cayci, Z.; Burmeister, L.A. Thyroid Storm with Coma in a Patient with Metastatic Thyroid Carcinoma and Graves Disease: Won the Battle but Lost the War. AACE Clin. Case Rep. 2019, 5, e7–e12. [Google Scholar] [CrossRef]
  94. Prete, A.; Pieroni, E.; Marrama, E.; Bruschini, L.; Ferrari, M.; Scioti, G.; Aprile, V.; Guarracino, F.; Ambrosini, C.E.; Molinaro, E.; et al. Management of patients with extensive locally advanced thyroid cancer: Results of multimodal treatments. J. Endocrinol. Investig. 2023, 47, 1165–1173. [Google Scholar] [CrossRef]
  95. Purbhoo, K.; Vangu, M.; Bayat, Z.; Daya, R. A rare case of pituitary gland metastases of poorly differentiated thyroid carcinoma. J. Nucl. Med. 2023, 64, P208. [Google Scholar]
  96. Raffaelli, M.; Sessa, L.; De Crea, C. Total thyroidectomy with central and lateral neck dissection for poorly differentiated thyroid carcinoma (with video). J. Visc. Surg. 2023, 160, 76–77. [Google Scholar] [CrossRef]
  97. Roque, J.; Silva, T.N.; Regala, C.; Rodrigues, R.; Leite, V. Outcomes of lenvatinib therapy in poorly differentiated thyroid carcinoma. Eur. Thyroid. J. 2023, 12, e230003. [Google Scholar] [CrossRef]
  98. Temperley, T.S.; Temperley, H.C.; O’Sullivan, N.J.; Corr, A.; Brennan, I.; Kelly, M.E.; Prior, L. Tracheoesophageal fistula development following radiotherapy and tyrosine kinase inhibitors in a patient with advanced follicular thyroid carcinoma: A case-based review. Ir. J. Med. Sci. 2023, 193, 1143–1147. [Google Scholar] [CrossRef] [PubMed]
  99. Sato, H.; Tsukahara, K.; Motohashi, R.; Wakiya, M.; Serizawa, H.; Kurata, A. Thyroid Carcinoma on the Side of the Absent Lobe in a Patient with Thyroid Hemiagenesis. Case Rep. Otolaryngol. 2017, 2017, 4592783. [Google Scholar] [CrossRef]
  100. Schopper, H.K.; Stence, A.; Ma, D.; Pagedar, N.A.; Robinson, R.A. Single thyroid tumour showing multiple differentiated morphological patterns and intramorphological molecular genetic heterogeneity. J. Clin. Pathol. 2016, 70, 116–119. [Google Scholar] [CrossRef] [PubMed]
  101. Shakeri, S.; Jahanpanah, P.; Divband, G.; Massoudi, T.; Aryana, K. 99m Tc-Octreotide-Avid Brain Mass in A Patient with Poorly Differentiated Papillary Thyroid Carcinoma, Hope in Despair. Nucl. Med. Rev. Cent. East Eur. 2020, 23, 49–50. [Google Scholar]
  102. Sonavane, S.N.; Upadhye, T.; Basu, S. Sellar-Parasellar and Petrous Bone Metastasis from Differentiated Thyroid Carcinoma: Imaging Characteristics and Follow-Up Profile Post Radioiodine Therapy. World J. Nucl. Med. 2023, 22, 144–149. [Google Scholar] [CrossRef]
  103. Suehiro, A.; Nagahara, K.; Moritani, S.; Omori, K. Axillary lymph node metastases from thyroid carcinoma: Report of seven cases. Auris Nasus Larynx 2021, 48, 718–722. [Google Scholar] [CrossRef]
  104. Sugawara, E.; Shibata, Y.; Katsumata, K. Werner syndrome associated with poorly differentiated thyroid carcinoma and systemic sclerosis-like skin manifestations: A case report. Mod. Rheumatol. Case Rep. 2023, 8, 95–100. [Google Scholar] [CrossRef]
  105. Sukrithan, V.; Kim, L.; Sipos, J.A.; Goyal, A.; Zhou, Y.; Addison, D.; Shah, M.; Konda, B.; Vallakati, A. Coronary Artery and Peripheral Vascular Disease in a Patient with Poorly Differentiated Thyroid Cancer Treated with the Tyrosine Kinase Inhibitor Lenvatinib. Case Rep. Endocrinol. 2023, 2023, 8841696. [Google Scholar] [CrossRef]
  106. Suman, S.; Basu, S. Solitary Metacarpophalangeal Metastasis from Poorly Differentiated Thyroid Carcinoma: Excellent Tumor Marker and Scan Response to Two Fractions of Radioiodine Therapy. Indian J. Nucl. Med. 2018, 33, 362–363. [Google Scholar] [PubMed]
  107. Thiagarajan, S.; Yousuf, A.; Shetty, R.; Dhar, H.; Mathur, Y.; Nair, D.; Basu, S.; Patil, A.; Kane, S.; Ghosh-Laskar, S.; et al. Poorly differentiated thyroid carcinoma (PDTC) characteristics and the efficacy of radioactive iodine (RAI) therapy as an adjuvant treatment in a tertiary cancer care center. Eur. Arch. Oto-Rhino-Laryngol. 2020, 277, 1807–1814. [Google Scholar] [CrossRef] [PubMed]
  108. Thompson, L.D.R. High Grade Differentiated Follicular Cell-Derived Thyroid Carcinoma Versus Poorly Differentiated Thyroid Carcinoma: A Clinicopathologic Analysis of 41 Cases. Endocr. Pathol. 2023, 34, 234–246. [Google Scholar] [CrossRef]
  109. Toyoshima, M.T.K.; Domingues, R.B.; Soares, I.C.; Danilovic, D.L.S.; Amorim, L.C.; Cavalcante, E.R.C.; Antonacio, F.F.; Roitberg, F.S.R.; Hoff, A.O. Thyroid collision tumor containing oncocytic carcinoma, classical and hobnail variants of papillary carcinoma and areas of poorly differentiated carcinoma. Arq. Bras. Endocrinol. Metabol. 2021, 65, 495–499. [Google Scholar] [CrossRef]
  110. Tsuji, H.; Yasuoka, H.; Nakamura, Y.; Hirokawa, M.; Hiroshima, T.; Sakamaki, Y.; Miyauchi, A.; Tsujimoto, M. Aggressive cribriform-morular variant of papillary thyroid carcinoma: Report of an unusual case with pulmonary metastasis displaying poorly differentiated features. Pathol. Int. 2018, 68, 700–705. [Google Scholar] [CrossRef] [PubMed]
  111. Uchida, T.; Yamaguchi, H.; Nagamine, K.; Yonekawa, T.; Nakamura, E.; Shibata, N.; Kawano, F.; Asada, Y.; Nakazato, M. Rapid pleural effusion after discontinuation of lenvatinib in a patient with pleural metastasis from thyroid cancer. Endocrinol. Diabetes Metab. Case Rep. 2019, 2019. [Google Scholar] [CrossRef]
  112. Wan, Z.; Wang, B.; Yao, J.; Li, Q.; Miao, X.; Jian, Y.; Huang, S.; Lai, S.; Li, C.; Tian, W. Predictive factors and clinicopathological characteristics of outcome in poorly differentiated thyroid carcinoma: A single-institution study. Front. Oncol. 2023, 13, 1102936. [Google Scholar] [CrossRef]
  113. Xu, B.; Lubin, D.J.; Dogan, S.; Ghossein, R.A.; Viswanathan, K. Significance of oncocytic features in poorly differentiated thyroid carcinoma—A bi-institutional experience. Virchows Arch. 2022, 482, 479–491. [Google Scholar] [CrossRef]
  114. Xue, F.; Li, D.; Hu, C.; Wang, Z.; He, X.; Wu, Y. Application of intensity-modulated radiotherapy in unresectable poorly differentiated thyroid carcinoma. Oncotarget 2016, 8, 15934–15942. [Google Scholar] [CrossRef]
  115. Yasuoka, H.; Nakamura, Y.; Hirokawa, M.; Yoshida, K.-I.; Anno, K.; Tori, M.; Tsujimoto, M. A rare case of poorly differentiated thyroid carcinoma probably arising from a nodular goiter. BMC Clin. Pathol. 2017, 17, 9. [Google Scholar] [CrossRef] [PubMed]
  116. Yin, L.; Hou, S.; Hou, L.-L.; Pu, C.-C. Clinical characteristics and prognostic nomogram for patients with insular thyroid carcinoma: A population-based analysis. Endocrine 2022, 79, 331–341. [Google Scholar] [CrossRef] [PubMed]
  117. Yu, M.G.; Rivera, J.; Jimeno, C. Poorly Differentiated Thyroid Carcinoma: 10-Year Experience in a Southeast Asian Population. Endocrinol. Metab. 2017, 32, 288–295. [Google Scholar] [CrossRef] [PubMed]
  118. Yuang, K.; Al-Bahadili, H.; Chang, A. An Unexpected Finding of Poorly Differentiated Thyroid Carcinoma in a Toxic Thyroid Nodule. JCEM Case Rep. 2023, 1, luad052. [Google Scholar] [CrossRef] [PubMed]
  119. Zhang, L.; Zheng, S.; Qin, Y.; Chen, X.; Zhou, S. Combination therapy including immune checkpoint inhibitors for anaplastic and poorly differentiated thyroid carcinoma: A promising protocol. J. Biol. Regul. Homeost. Agents 2023, 37, 3115–3122. [Google Scholar]
  120. Walczyk, A.; Kopczyński, J.; Gąsior-Perczak, D.; Pałyga, I.; Kowalik, A.; Chrapek, M.; Hejnold, M.; Góźdź, S.; Kowalska, A. Poorly differentiated thyroid cancer in the context of the revised 2015 American Thyroid Association Guidelines and the Updated American Joint Committee on Cancer/Tumor-Node-Metastasis Staging System (eighth edition). Clin. Endocrinol. 2018, 91, 331–339. [Google Scholar] [CrossRef]
  121. Chernock, R.D.; Rivera, B.; Borrelli, N.; Hill, D.A.; Fahiminiya, S.; Shah, T.; Chong, A.-S.; Aqil, B.; Mehrad, M.; Giordano, T.J.; et al. Poorly differentiated thyroid carcinoma of childhood and adolescence: A distinct entity characterized by DICER1 mutations. Mod. Pathol. 2020, 33, 1264–1274. [Google Scholar] [CrossRef] [PubMed]
  122. Lim, H.; Devesa, S.S.; Sosa, J.A.; Check, D.; Kitahara, C.M. Trends in Thyroid Cancer Incidence and Mortality in the United States, 1974–2013. J. Am. Med. Assoc. 2017, 317, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  123. Durante, C.; Haddy, N.; Baudin, E.; Leboulleux, S.; Hartl, D.; Travagli, J.P.; Caillou, B.; Ricard, M.; Lumbroso, J.D.; De Vathaire, F.; 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, 2892–2899. [Google Scholar] [CrossRef]
  124. Lee, J.; Soh, E.Y. Differentiated thyroid carcinoma presenting with distant metastasis at initial diagnosis clinical outcomes and prognostic factors. Ann. Surg. 2010, 251, 114–119. [Google Scholar] [CrossRef] [PubMed]
  125. Hannallah, J.; Rose, J.; Guerrero, M.A. Comprehensive Literature Review: Recent Advances in Diagnosing and Managing Patients with Poorly Differentiated Thyroid Carcinoma. Int. J. Endocrinol. 2013, 2013, 317487. [Google Scholar] [CrossRef] [PubMed]
  126. Walczyk, A.; Kowalska, A.; Sygut, J. The clinical course of poorly differentiated thyroid carcinoma (insular carcinoma)–Own observations. Endokrynol. Pol. 2010, 61, 467–473. [Google Scholar] [PubMed]
  127. Morandi, L.; Righi, A.; Maletta, F.; Rucci, P.; Pagni, F.; Gallo, M.; Rossi, S.; Caporali, L.; Sapino, A.; Lloyd, R.V.; et al. Somatic mutation profiling of hobnail variant of papillary thyroid carcinoma. Endocr. Relat. Cancer 2017, 24, 107–117. [Google Scholar] [CrossRef] [PubMed]
  128. Ito, Y.; Hirokawa, M.; Kihara, M.; Takamura, Y.; Kobayashi, K.; Miyauchi, A. The prognostic value of PDTC in a series of PTC patients of the Japanese Society of Thyroid Surgerymade a comparision with risk classification system in Kuma Hospital. Endocr. J. 2012, 59, 817–821. [Google Scholar] [CrossRef]
  129. Riesco-Eizaguirre, G.; Santisteban, P. ENDOCRINE TUMOURS: Advances in the molecular pathogenesis of thyroid cancer: Lessons from the cancer genome. Eur. J. Endocrinol. 2016, 175, R203–R217. [Google Scholar] [CrossRef]
  130. Capdevila, J.; Mayor, R.; Mancuso, F.M.; Iglesias, C.; Caratù, G.; Matos, I.; Zafón, C.; Hernando, J.; Petit, A.; Nuciforo, P.; et al. Early evolutionary divergence between papillary and anaplastic thyroid cancers. Ann. Oncol. 2018, 29, 1454–1460. [Google Scholar] [CrossRef] [PubMed]
  131. Agrawal, N.; Akbani, R.; Aksoy, B.A.; Ally, A.; Arachchi, H.; Asa, S.L.; Auman, J.T.; Balasundaram, M.; Balu, S.; Baylin, S.B.; et al. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690. [Google Scholar] [CrossRef]
  132. Landa, I.; Ibrahimpasic, T.; Boucai, L.; Sinha, R.; Knauf, J.A.; Shah, R.H.; Dogan, S.; Ricarte-Filho, J.C.; Krishnamoorthy, G.P.; Xu, B.; et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J. Clin. Investig. 2016, 126, 1052–1066. [Google Scholar] [CrossRef]
  133. Pozdeyev, N.; Gay, L.M.; Sokol, E.S.; Hartmaier, R.; Deaver, K.E.; Davis, S.; French, J.D.; Borre, P.V.; LaBarbera, D.V.; Tan, A.C.; et al. Genetic analysis of 779 advanced differentiated and anaplastic thyroid cancers. Clin. Cancer Res. 2018, 24, 3059–3068. [Google Scholar] [CrossRef]
  134. Zou, M.; Baitei, E.Y.; Alzahrani, A.S.; BinHumaid, F.S.; Alkhafaji, D.; Al-Rijjal, R.A.; Meyer, B.F.; Shi, Y. Concomitant RAS, RET/PTC, or BRAF mutations in advanced stage of papillary thyroid carcinoma. Thyroid 2014, 24, 1256–1266. [Google Scholar] [CrossRef] [PubMed]
  135. Huang, M.; Yan, C.; Xiao, J.; Wang, T.; Ling, R. Relevance and clinicopathologic relationship of BRAF V600E, TERT and NRAS mutations for papillary thyroid carcinoma patients in Northwest China. Diagn. Pathol. 2019, 14, 1–10. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, S.E.; Hwang, T.S.; Choi, Y.L.; Han, H.S.; Kim, W.S.; Jang, M.H.; Kim, S.K.; Yang, J.H. Prognostic Significance of TERT Promoter Mutations in Papillary Thyroid Carcinomas in a BRAF(V600E) Mutation-Prevalent Population. Thyroid 2016, 26, 901–910. [Google Scholar] [CrossRef] [PubMed]
  137. Liu, X.; Qu, S.; Liu, R.; Sheng, C.; Shi, X.; Zhu, G.; Murugan, A.K.; Guan, H.; Yu, H.; Wang, Y.; et al. TERT promoter mutations and their association with BRAFV600E mutation and aggressive clinicopathological characteristics of thyroid cancer. J. Clin. Endocrinol. Metab. 2014, 99, E1130–E1136. [Google Scholar] [CrossRef] [PubMed]
  138. Vuong, H.G.; Altibi, A.M.A.; Duong, U.N.P.; Hassell, L. Prognostic implication of BRAF and TERT promoter mutation combination in papillary thyroid carcinoma-A meta-analysis. Clin. Endocrinol. 2017, 87, 411–417. [Google Scholar] [CrossRef] [PubMed]
  139. Xing, M.; Alzahrani, A.S.; Carson, K.A.; Viola, D.; Elisei, R.; Bendlova, B.; Yip, L.; Mian, C.; Vianello, F.; Tuttle, R.M.; et al. Association Between BRAF V600E Mutation and Mortality in Patients with Papillary Thyroid Cancer. JAMA 2013, 309, 1493–1501. [Google Scholar] [CrossRef] [PubMed]
  140. Tufano, R.P.; Teixeira, G.V.; Bishop, J.; Carson, K.A.; Xing, M. BRAF mutation in papillary thyroid cancer and its value in tailoring initial treatment: A systematic review and meta-analysis. Medicine 2012, 91, 274–286. [Google Scholar] [CrossRef] [PubMed]
  141. Liu, J.; Liu, Y.; Lin, Y.; Liang, J. Radioactive Iodine-Refractory Differentiated Thyroid Cancer and Redifferentiation Therapy. Endocrinol. Metab. 2019, 34, 215–225. [Google Scholar] [CrossRef] [PubMed]
  142. Jonklaas, J.; Sarlis, N.J.; Litofsky, D.; Ain, K.B.; Bigos, S.T.; Brierley, J.D.; Cooper, D.S.; Haugen, B.R.; Ladenson, P.W.; Magner, J.; et al. Outcomes of Patients with Differentiated Thyroid Carcinoma Following Initial Therapy. Thyroid 2006, 16, 1229–1242. [Google Scholar] [CrossRef]
  143. Jonklaas, J.; Cooper, D.S.; Ain, K.B.; Bigos, T.; Brierley, J.D.; Haugen, B.R.; Ladenson, P.W.; Magner, J.; Ross, D.S.; Skarulis, M.C.; et al. Radioiodine Therapy in Patients with Stage I Differentiated Thyroid Cancer. Thyroid 2010, 20, 1423–1424. [Google Scholar] [CrossRef]
  144. Cracolici, V.; Cipriani, N.A. High-Grade Non-Anaplastic Thyroid Carcinomas of Follicular Cell Origin: A Review of Poorly Differentiated and High-Grade Differentiated Carcinomas. Endocr. Pathol. 2023, 34, 34–47. [Google Scholar] [CrossRef] [PubMed]
  145. Huang, J.; Sun, W.; Zhang, Q.; Wang, Z.; Dong, W.; Zhang, D.; Lv, C.; Shao, L.; Zhang, P.; Zhang, H. Clinicopathological Characteristics and Prognosis of Poorly Differentiated Thyroid Carcinoma Diagnosed According to the Turin Criteria. Endocr. Pr. 2021, 27, 401–407. [Google Scholar] [CrossRef] [PubMed]
  146. Ibrahimpasic, T.; Ghossein, R.; Shah, J.P.; Ganly, I. Poorly Differentiated Carcinoma of the Thyroid Gland: Current Status and Future Prospects. Thyroid 2019, 29, 311–321. [Google Scholar] [CrossRef] [PubMed]
  147. Brose, M.S.; Nutting, C.M.; Jarzab, B.; Elisei, R.; Siena, S.; Bastholt, L.; de la Fouchardiere, C.; Pacini, F.; Paschke, R.; Shong, Y.K.; et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: A randomised, double-blind, phase 3 trial. Lancet 2014, 384, 319–328. [Google Scholar] [CrossRef] [PubMed]
  148. Schlumberger, M.; Tahara, M.; Wirth, L.J.; Robinson, B.; Brose, M.S.; Elisei, R.; Habra, M.A.; Newbold, K.; Shah, M.H.; Hoff, A.O.; et al. Lenvatinib versus Placebo in Radioiodine-Refractory Thyroid Cancer. N. Engl. J. Med. 2015, 372, 621–630. [Google Scholar] [CrossRef] [PubMed]
  149. 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]
  150. Bible, K.C.; Kebebew, E.; Brierley, J.; Brito, J.P.; Cabanillas, M.E.; Clark, T.J., Jr.; Di Cristofano, A.; Foote, R.; Giordano, T.; Kasperbauer, J.; et al. American Thyroid Association Guidelines for Management of Patients with Anaplastic Thyroid Cancer. Thyroid 2021, 31, 337–386. [Google Scholar] [CrossRef]
  151. Study of Efficacy and Safety of Dabrafenib Plus Trametinib in Previously Treated Patients with Locally Advanced or Metastatic, Radio-Active Iodine Refractory BRAFV600E Mutation-Positive Differentiated Thyroid Cancer. Available online: https://www.novartis.com/clinicaltrials/study/nct04940052 (accessed on 1 May 2024).
  152. Wirth, L.J.; Tahara, M.; Robinson, B.; Francis, S.; Brose, M.S.; Habra, M.A.; Newbold, K.; Kiyota, N.; Dutcus, C.E.; Mathias, E.; et al. Treatment-Emergent Hypertension and Efficacy in the Phase 3 Study of (E7080) Lenvatinib in Differentiated Cancer of the Thyroid (SELECT). Cancer 2018, 124, 2365–2372. [Google Scholar] [CrossRef]
Figure 1. Invasive front with a solid pattern and nuclear atypia (HE × 250).
Figure 1. Invasive front with a solid pattern and nuclear atypia (HE × 250).
Jpm 14 00654 g001
Figure 2. Increased 18-F FDG uptake in the left sphenoid sinus. First image: CT scan, second image: merged CT and PET scan, third image: FDG uptake.
Figure 2. Increased 18-F FDG uptake in the left sphenoid sinus. First image: CT scan, second image: merged CT and PET scan, third image: FDG uptake.
Jpm 14 00654 g002
Figure 3. Response to cabozantinib treatment: differentiation of right supraclavicular lymph block before (B) and after (A) treatment; metastatic foci size reduction before (D) and after (C) treatment, respectively.
Figure 3. Response to cabozantinib treatment: differentiation of right supraclavicular lymph block before (B) and after (A) treatment; metastatic foci size reduction before (D) and after (C) treatment, respectively.
Jpm 14 00654 g003
Figure 4. Patient timeline. Arrows depict the time frame of each treatment and the cumulative treatment. The respective PFS is depicted in months (m). anti-Tg: anti-thyroglobulin antibodies, G: grade, LND: lymph node dissection, mono: monotherapy, PFS: progression-free survival, PPES: palmar–plantar erythrodysesthesia syndrome, PTC: papillary thyroid carcinoma, RAI: radioactive iodine, RT: radiotherapy, Tg: thyroglobulin, TKI: tyrosine kinase inhibitor.
Figure 4. Patient timeline. Arrows depict the time frame of each treatment and the cumulative treatment. The respective PFS is depicted in months (m). anti-Tg: anti-thyroglobulin antibodies, G: grade, LND: lymph node dissection, mono: monotherapy, PFS: progression-free survival, PPES: palmar–plantar erythrodysesthesia syndrome, PTC: papillary thyroid carcinoma, RAI: radioactive iodine, RT: radiotherapy, Tg: thyroglobulin, TKI: tyrosine kinase inhibitor.
Jpm 14 00654 g004
Figure 5. PRISMA flow diagram.
Figure 5. PRISMA flow diagram.
Jpm 14 00654 g005
Table 1. Characteristics of the included studies.
Table 1. Characteristics of the included studies.
StudynM/FAgeHistological CombinationsETEDmLnSurgeryRAISystematic TreatmentOutcomeMolecular
Abdellaoui, 2022 [38]11/027Insular, solid000Rt HTmy (first), TTmy10ANDNA
Abu Rumman, 2021 [39]11/023Insular, solid, trabecular
(thyroglossal cyst)
*00Sistrunk procedure (first), TTmy10ANDNA
Agarwal, 2024 [40]391/2.955 (median)Insular (32), solid (25), trabecular (18), PTC (2), FTC (14)67.7%57%45.1%TTmy (+lateral dissection 10)25.6% (multiple)073% survival (12 m)BRAF (5), NRAS (9)
Ahmadian, 2024 [41]11/071PTC (thyroid), PDTC (pancreas, liver)0Liver, pancreas, peritoneum, (adrenal glands)NATTmy (+ND)1NANAAGAP3: BRAF fusion
Alshehri K, 2022 [42]10/156PDTC1Lung, sternal osseous0TTmy (+central, Rt ND)1EBRT (15) + (paclitaxel, carboplatin, doxorubicin, sorafenib, lenvatinib)AWDHRAS-BCORL1
Altiner, 2023 [43]10/170PDTC0Lung, rib, orbital0TTmy, rid resection1PaclitaxelAWDNA
Alzahrani, 2022 [44]10/143Insular1Lung, (adrenal)1PTmy0LenvatinibAWDKRAS+ PIC3CA (missense), PICR1 (insertion), FH (nonsense)
Ambre, 2022 [45]10/174Insular, trabecular0Mandible0TTmy, hemi-mandibulectomy00ANDNA
Atif, 2018 [46]10/156PDTC0Lung, bones1TTmy (+CND)1 KRAS
Bellini, 2021 [47]83/554–85PDTC, oncocytic 5/6NA3/8TTmy (6), nodule removal (2), lymphadenectomy (5)NANANANA
Bertrand, 2019 [48]10/153PTC (thyroid), PDTC–trabecular (uterus)0Lung, bones, uterus0TTmy (+CND)2NAAWDNA
Beute, 2024 [49]73/430–82PDTC (7/7), hobnail (1/7), tall-cell (1/7)5/72/72/3TTmy (4/7), STTmy (2/7), lymphadenectomy (6/7)1/1EBRT (1/1)NABRAF (1/1)
Bichoo, 2019 [50]1421/2.150.9 (mean)PDTC (27/142), PTC with PDA (27/142), FTC with PDA (88/142)51/14275/14243/142TTmy (136/142), near total (1/142), HTmy (1/142), debulking (4/142), lymphadenectomy (53/142), resection of metastasis (20/142)110/142EBRT (30/142), Sorafenib (3/142)64% (G1), 85% (G2), 62% (G3) DOD. From those alive: 66% (G1), 50% (G2), 39% (G3) AWDNA
Brijmohan, 2023 [51]11/068PDTC (insular, solid, trabecular), PTC1/1Liver, bones, scalp1/1TTmy (+ND)1/1NANANA
Ching, 2018 [52]10/144PDTC (insular, trabecular)000Rt Tmy (first), TTmy NANAANDNA
Choi, 2021 [53]10/161PDTC (insular, solid, trabecular)000TTmy1NAANDTERT promoter point mutation
Choi, 2020 [54]217/1415–78PDTC (insular, trabecular), FC (15/21), PTC (5/21),21/214/21 (lung, bones)0TTmy (8/21), CTmy (10/21)19/210AND (16/21) AWD (5/21)NA
Colombo, 2022 [55]11/035PDTC, FTC1Lung, liver, adrenal1TTmy, thoracic surgery1SorafenibDODPTEN, p53
Corean, 2019 [56]10/129CMVPTC, PDTC000TTmy0NAANDAPC
Das, 2021 [57]11/058Poorly differentiated follicular carcinoma with rhabdoid phenotype1/1LungNATracheostomy onlyNACisplatin, endoxan, doxorubicinAWDNA
de la Fouchardière, 2018 [58]10440/6412–91Insular (93/104), solid (11/104), trabecular (17/104), PTC (59/102), FTC (29/102), Hurthle (14/102)40/10017/9811/98TTmy (101/104), ND (36/104)99/104 (1–6 cycles)EBRT (9/104), TKIs (21/104)AND (36/104), AWD (33/104), DOD (35/104)TERT promoter (24/63), BRAF (3/38), RAS (10/38)
Dettmer, 2023 [59]10/144CMTC, PDTC000Rt Tmy (first), TTmy 10ANDAPC, TERT promoter, PIC3CA
Dierks, 2021 [60]2NA49/63PDTC2/22/2 (bone, lung, liver, kidney)2/2TTmy (2/2), ND(1/2)1/2 (2 cycles)EBRT (1/2), cisplatin/doxorubicin and carboplatin/paclitaxel (1/2), lenvatinib + pembrolizumab (2/2)DOD (2/2)TERT promoter and PTEN (2/2)
Elshafie, 2023 [61]10/160PDTCNABones, lung1/1TTmy (+centra/left lateral ND)1EBRTAWDNA
Farahmandfar, 2020 [62]11/070PTC, PDTCNAParotid, lung, pericardium1/1TTmy (+CND)1EBRT, chemo (NA)DODNA
Feffer, 2017 [63]10/155PDTC (insular)NASphenoid bone000EBRTAWDNA
Gay, 2019 [64]10/181PDTC with focal squamous differentiation1/100TTmy1EBRT, lenvatinibANDNA
Gazeu, 2020 [65]10/143PDTC (solid), oncocytic, follicular component000Rt Tmy (first), TTmy 10AND0
Gill, 2023 [66]10/146PDTC0SVC tumor thrombus1TTmy3EBRTAWDNA
Goto, 2018 [67]10/175PDTC, PTCNALungNATTmy, ND1Lenvatinib, sorafenibAWDNA
Grawe, 2021 [68]4721/2657 ± 19PDTC9/4718/4714/47TTmy1 (0–3)Lenvatinib/sorafenib (2/47), EBRT AND (18/47), AWD (15/47), DOD (14/47)NA
Gubbiotti, 2023 [69]6529/3621–85PDTC, PTC (52/65), FTC(13/65)31/6519/65 (lung, bones, brain)11/65TTmy (53/65), lobectomy first (11/12)42/65Chemo (6/65), EBRT (4/65)DOD (11/65)HRAS (2/8), NRAS (2/8), BRAF (1/8), TERT promoter (1/8), p53 (2/8), PAX8: PPARy rearrangement (2/8), NSD3:: NUTM1 fusion(1/8)
Gülbahar Ateş, 2024 [70]10/166PDTC (solid, insular, trabecular)1/1Lung, bones, tumor thrombus0Neck surgery0EBRTAWDNA
Hu, 2022 [71]10/135PTC, PDTCNABreast, neck1/1Endoscopic Tmy (first), TTmy (+bilateral CND), local tumor excision, modified radical ND, partial mastectomy 0Adjuvant therapy (?)AND0
Ieni, 2021 [72]11/069FTC, PDTC0Kidney0Partial nephrectomy, TTmy00ANDNA
Iravani, 2019 [73]30/345–61FTC, PDTC3/33/3 (lung, neck, muscle, bone)1/3TTmy (3/3), lymphadenectomy (1/3)3/3Sunitinib,
sorafenib (1/3), trametinib (3/3), lenvatinib(2/3)
AWD (3/3)NRAS (3/3)
Isaev, 2022 [74]9135/5616–93PDTC63/9138/9137/91TTmy (55/91), extended TTmy (25/91), lobectomy (9/91), subtotal (2/91), ND (59/91)67/85EBRT (7/85), systemic therapy (11/89)AWD (40/91), DOD (27/91)NA
Kalshetty, 2018 [75]32/132–51PDTC (3/3), PTC (1/3), focal cribriform (1/3)2/33/3(bone, pleura, neck, lung)1/3TTmy (3/3), lymphadenectomy (2/3), tumor resection2/3EBRT (2/3), sorafenib (1/3)DOD (1/3), AWD(2/3)NA
Kersting, 2021 [76]11/038PDTC0Lung1/1NA, metastasectomyNATKIsAWDNA
Khetrapal, 2018 [77]10/142PDTC1/1NA1/1TTmy, CNDNANANANA
Kim, 2023 [78]11/034PDTC (solid), SCC1/1Lung (SCC)NATTmy, ND, lobectomyNAPaclitaxel, cisplatin, and etoposideDODATRX (c.6793G>T), TP53 (c.377A>G) MYCL (c.332G>T)
im, 2023 [79]10/167PDTC (neck)000lobectomy + CND (for benign lesion), neck excisionNANAAWDNA
Kunte, 2022 [80]2314/939–89PDTC (23/23), PTC(7/23), FTC(7/23)18/137/238/23TTmy (19/23), lymphadenectomy (8/19)13/23EBRT (2/19), TKIs (sorafenib, lenvatinib, sorafenib, and pazopanib)(6/23)DOD (12/23)NRAS, TERT promoter (1/2)
Kut, 2020 [81]11/060FTC, PDTC (insular)1/1Lung1/1TTmy, lymphadenectomy, neck surgery1EBRT, TKIs (lenvatinib, pazopanib), pembrolizumabAWDTERT promoter
Laforga and Cortés, 2019 [82]11/076OV–PDTCNALung, spleen, bones, liver1/1TTmy, bilateral ND1NADODNA
Leboulleux, 2019 [83]10/159PDTC1/1Lung, bone1/100Dabrafenib and trametinibAWDBRAF-K601E mutation
Lee, 2021 [84]11/076PDTC1/1Lung1/100Carboplatin, EBRT, pembrolizumab, lenvatinib, rametinib, dabrafenibAWDBRAF T599_V600insT, CDKN2A/B loss, loss of MTAP exons 2–8 and TERT promoter mutation
Lukovic, 2021 [85]4524/2135.5–83.6OV–PDTC6/4516/453/16TTmy (21/45), staged Tmy (24/45)39/45EBRT (4/45), VEGFNANA
Molinaro, 2019 [86]10/165PTC, PDTC with squamous cells1/100TTmy1LenvatinibDODBRAF V600E
Morvan, 2022 [87]10/158PDTC1/1Bone, IJV tumor thrombus0TTmy + removal infiltrated portion IJV, ND10ANDNA
Nagaoka, 2023 [88]11/070PDTC1/1Bones0TTmy10AWDNA
O’Donohue, 2022 [89]11/079PDTC0Liver (port hepatis), bones0TTmy0LenvatinibDOD
Oh, 2024 [90]10/150PDTC1/1Lung, liver, bones1/1TTmy1EBRT, lenvatinib, nivolumab and ipilimumab, cabozantinib, temsirolimus, nivolumab + relatinibDODPTEN L194fs and TP53 F270S
Panchangam, 2022 [91]611/1.3–1.616–81PDTC, PTC with PDA59–73%19–41%50–55%TTmy (52/61), near Tmy (5/61), debulking (4/61)29–65%EBRT (7/61), chemo(1/61)NANA
Peng, 2022 [92]20/272–73PDTC, poorly differentiated squamous cell carcinoma2/21/2(lung)1/2TTmy + ND(Hx PTC)3(Hx PTC)Anlotinib (1/2), dabrafenib, and trametinib (2/2)DOC (1/2), AWD (1/2)BRAFV660E (1/2), TERT promoter (1/2)
Pinto, 2019 [93]10/171PDTC with a focal papillary and follicular patternNALung, bones0TTmy00DODNA
Prete, 2024 [94]22/068–69PDTC1/2Lung, IJV + SVC tumor thrombus1/2TTmy (1/2), thrombectomy (1/2), lymphadenectomy(2/2)0Lenvatinib (1/2)DOC (1/2), AWD(1/2)NA
Purbhoo, 2023 [95]10/151PDTC (solid, insular)0Bone, lung, pituitary gland0TTmy + CNDNANAAWDNA
Raffaelli, 2023 [96]10/172PDTC1/101/1TTmy + central/
lateral ND
NANAANDNA
Roque, 2023 [97]83/534–67PDTCNA7/8NA Surgery (6/8), metastasis excision (2/8)6/8EBRT (4/8), paclitaxel and carboplatin (1/8), other TKIS(3/8), lenvatinib(8/8)AWD (5/8), DOD(3/8)NA
Temperley, 2023 [98]11/069PDTC1/11/1(liver, bones, hilar LN)000Lenvatinib, cabozantinib, EBRTDODNA
Sato, 2017 [99]10/164PTC, PDTC1/100total Rt thyroid lobectomy isthmectomy, resection + CND1NAANDNA
Schopper, 2017 [100]11/049Conventional PTC, follicular variant of papillary carcinoma, columnar cell carcinoma, clear cell papillary
carcinoma, PDCT
1/101/1TTmy + L NDNANANAKRAS, BRAF (PDTC)
Shakeri, 2020 [101]10/158PDTCNALung, brain1/1TTmy + ND1/1NANANA
Sonavane, 2023 [102]11/039PDTC1/1Brain, bones, lung1/1TTmy + CND3EBRTAWDNA
Suehiro, 2021 [103]61/521–67PDTC (6/6), PTC (2/6), squamous differentiation (1/6), signet ring differentiation (1/6)NAAxillary lymph nodesNATTmy (5/6), CTmy (1/6), ND (6/6), axillary node dissection (6/6)NANAAWD (2/3), AND (1/3)NA
Sugawar, 2023 [104]10/136PDTC (trabecular)NANANAleft lobectomy (first), TTmy10ANDNA
Sukrithan, 2023 [105]11/070 sPDTCNA1/1NATTmy1EBRT, lenvatinibAWDNA
Suman and Basu, 2018 [106]10/177PDTC, FTC (bones)0Bones0TTmy + Rt CND20ANDNA
Thiagarajan, 2020 [107]3511/2422–77PDTC (solid (10/35), insular (11/35), trabecular (1/35),
mixed (6/35))
12/3518/359/35completion Tmy (7/35), TTmy (27/35), thyroid bed exploration (1/35), ND(21/53)35/35EBRT (16/35)AND (23/35), AWD (10/35), DOD (2/35)NA
Thompson, 2023 [108]2411/1358(mean)PDTC (24/24), PTC (13/24), metastatic SCC to thyroid gland (1/24)9/245/246/24Surgery (lobectomy, Tmy, and/or CTmy) (24/24)18/24EBRT (5/24), chemo (1/24)AND (15/24), AWD (3/24), DOD (5/24), DOC(1/24)NRAS (3/13) TERT+ NRAS (1/13), TERT + NRAS + KAT6B (1/13) NRAS + MUTYH (1/13); NRAS + PALB2 (1/13) PTENdeletion (1/13), WHSC1L1::NUTM1 (1/13), PAX8::PPARγ (1/13)
Toyoshima, 2021 [109]10/163Oncocytic carcinoma, PTC (classic and hobnail component), PDTC0Lung, liver1/1TTmy, bilateral LN dissection1EBRT, sorafenibDODNA
Tsuji, 2018 [110]10/126CMV–PTC (thyroid), CMV–PTC, PDTC (lung)NALung0TTmy, cervical lymph ND, Rt partial lobectomy1SorafenibAWDNA
Uchida, 2019 [111]10/173PDTCNALung, pleura1/101/1LenvatinibDODNA
Wan, 2023 [112]9434/558–85PDTC (94/94), PTC only (20/94), FTC only (17/94), other (34/94)83/9423/9478/94Tmy (73/95), ND(29/94)23/94EBRT (26/94), chemo (22/94)Median OS (m) (min–max) 33(1–170)NA
Xu, 2023 [113]210101/1095–87PDTC, oncocytic component (79/210)132/210NA39/210lobectomy/HTmy (38/210), TTmy/STmy (172/210)157/200EBRT (90/199), TKIs (67/209), chemo (37/199)NABRAF (6/87), NRAS (35/87), HRAS (4/87), KRAS (3/87), RET (1/87), PPARG (1/60), ALK (1/87), TERT (43/87), PTEN (15/87), TP53 (15/87), EIF1AX (11/87), NF 1(6/87), RBM10 (5/87), ATM (4/87), DNMT3A (4/87), STK11 (4/87), ARID1A (4/87)
Xue, 2017 [114]52/343–76PDTC5/52/5 (lung)5/5unilateral Tmy (1/5)NAIMRT (5/5), gemcitabine (1/5), cisplatin (1/5) paclitaxel and cisplatin (4/5)AWD (2/5), DOD (3/5)NA
Yasuoka, 2017 [115]10/135PDTC (solid, trabecular, microfollicular)1/100L HTmy (+ND), completion Tmy (+CND)1EBRTANDNRASgene
Yin, 2023 [116]20698/10857.9 ± 15.9Insular158/200 (T3 + T4)53/20266/192193/204155/206Chemo (22/206)90.3% (1year-OS), 82.0% (2year-OS), 62.2% (5 year-OS), and 42.5%(10 year-OS)NA
Yu, 2017 [117]185/1340–75PDTC insular (8/18), trabecular (7/18), solid (3/18)8/185/18 (lung (3/5), bone (2/5))5/18TTmy (+neck dissection) (5/18), TTmy only (5/18), subtotal + completion Tmy (2/18), lobectomy completion (1/18), tumor debulking (4/18)8/180AND (6/18), AWD (9/18), DOD (3/18)NA
Yuang, 2023 [118]11/066PDTC (solid/trabecular), FTCNA00Rt lobectomy + isthmectomy00ANDNA
Zhang, 2023 [119]21/1<60 (1/2), >60 (1/2)PDTC, poorly differentiated squamous cell carcinoma1/21/2 (lung, bone, brain)2/20EBRT (1/2)nab-paclitaxel + carboplatin (2/2), toripalimab (2/2)AWD (1/2), DOD (1/2)NA
PRESENT CASE11/045PDTC, hobnail, tall-cell1/11/1 (brain, bone, sinus)1/1TTmy (+neck dissection, completion ND)2Lenvatinib, sorafenib, trametinib/dabrafenibDODBRAF
Note: AND: alive without disease; AWD: alive with disease, CMTC: Cribriform—morular thyroid carcinoma; CMV-PTC: Cribriform-morular variant of papillary thyroid carcinoma; CND: central neck dissection; CTmy: completion thyroidectomy; Dm: distant metastases; DOC: died of other causes; DOD: died of disease; EBRT: external beam radiation therapy; ETE: extrathyroidal extension; FTC: follicular thyroid carcinoma, HTmy: hemithyroidectomy; IJV: internal jugular vein; IMRT: Intensity-modulated radiation therapy; L: left; Ln: lymph nodes; OV-PDTC: oncocytic variant poorly differentiated thyroid carcinoma, PDA: poorly differentiated areas, M/F: male-to-female, N: number of patients, NA: not applicable or, specifically, in molecular section, BRAF negative when performed; PDTC: poorly differentiated thyroid carcinoma, PTC: papillary thyroid carcinoma; PTmy: partial thyroidectomy; RAI: radioactive iodine; Rt: right; SVC: superior vena cava, TKI: tyrosine kinase inhibitor; Tmy: thyroidectomy; TTmy: total thyroidectomy, * thyroglossal cyst.
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

Violetis, O.; Konstantakou, P.; Spyroglou, A.; Xydakis, A.; Kekis, P.B.; Tseleni, S.; Kolomodi, D.; Konstadoulakis, M.; Mastorakos, G.; Theochari, M.; et al. The Long Journey towards Personalized Targeted Therapy in Poorly Differentiated Thyroid Carcinoma (PDTC): A Case Report and Systematic Review. J. Pers. Med. 2024, 14, 654. https://doi.org/10.3390/jpm14060654

AMA Style

Violetis O, Konstantakou P, Spyroglou A, Xydakis A, Kekis PB, Tseleni S, Kolomodi D, Konstadoulakis M, Mastorakos G, Theochari M, et al. The Long Journey towards Personalized Targeted Therapy in Poorly Differentiated Thyroid Carcinoma (PDTC): A Case Report and Systematic Review. Journal of Personalized Medicine. 2024; 14(6):654. https://doi.org/10.3390/jpm14060654

Chicago/Turabian Style

Violetis, Odysseas, Panagiota Konstantakou, Ariadni Spyroglou, Antonios Xydakis, Panagiotis B. Kekis, Sofia Tseleni, Denise Kolomodi, Manousos Konstadoulakis, George Mastorakos, Maria Theochari, and et al. 2024. "The Long Journey towards Personalized Targeted Therapy in Poorly Differentiated Thyroid Carcinoma (PDTC): A Case Report and Systematic Review" Journal of Personalized Medicine 14, no. 6: 654. https://doi.org/10.3390/jpm14060654

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