Open access peer-reviewed chapter

Approach and Management of Anaplastic Carcinoma Thyroid

Written By

Zuzak Sharafuddin, Rejnish Ravikumar, Malu Rafi, Lekha Madhavan Nair, John Mathew and Kainickal Cessal Thommachan

Submitted: 16 May 2022 Reviewed: 11 July 2022 Published: 09 November 2022

DOI: 10.5772/intechopen.106463

From the Edited Volume

Thyroid Cancer - The Road From Genes to Successful Treatment

Edited by Ifigenia Kostoglou-Athanassiou

Chapter metrics overview

77 Chapter Downloads

View Full Metrics

Abstract

Anaplastic carcinoma thyroid, also known as undifferentiated thyroid carcinoma, is a rare but highly aggressive malignant tumor, which accounts for 2–3% of all thyroid malignancies. It is mostly seen in elderly females in their 6th or 7th decade. It carries a very bad prognosis with an average median survival of 5 months. Patients often present with a rapidly growing, painful, woody hard lower anterior neck mass fixed to underlying structures. In addition to local invasion, patients also present with regional nodal spread and distant metastasis. Though the risk factors for anaplastic carcinoma thyroid are unknown, most of them develop in the setting of long-standing goiter, possibly in an undiagnosed, well-differentiated thyroid carcinoma. Management of anaplastic carcinoma thyroid demands a multidisciplinary approach with the involvement of surgeon, radiation oncologist, radiologist, and endocrinologist. The conventional treatment of anaplastic carcinoma thyroid includes surgery, radiation, and chemotherapy. Recently, multitarget tyrosine kinase inhibitors are also incorporated into the treatment. However, prognosis of the disease is very poor with 4 months of overall survival of 35% and overall disease-specific mortality of 98–99%. In this chapter, we discuss how to approach the condition and various treatment strategies to provide improved treatment outcomes for patients diagnosed with anaplastic carcinoma thyroid.

Keywords

  • anaplastic carcinoma thyroid
  • presentation
  • investigations
  • surgery
  • radiation
  • chemotherapy
  • targeted agents

1. Introduction

Anaplastic carcinoma thyroid (ATC) is a rare type of thyroid cancer that carries a worse prognosis. It is extremely aggressive, and the average median survival of ATC is 5 months, with less than 20% of the patients alive after 1 year of diagnosis.

Advertisement

2. Epidemiology and pathogenesis

ATC accounts for 1–2% of all thyroid malignancies. It usually affects patients in their 6th to 7th decade with a female predominance (male/female ratio: 1.5:2) [1, 2, 3]. However, it contributes up to 14–50% of the annual mortality associated with thyroid cancer [1]. The age-adjusted annual incidence is one to two per million people [4, 5].

Based on different tumor registries, the frequency of ATC is variable in different countries; however, there has been a drastic reduction in its incidence owing to better dietary iodine intake as well as timely diagnosis and treatment of DTC and MNG [1].

ATC usually originates in the background of a long-standing goiter. Dedifferentiation of differentiated thyroid cancer cells is considered the most common cause of ATC. Dedifferentiation arises due to multiple chromosomal aberrations, alteration in signal transduction pathways, and cell-cycle derangement.

BRAF, RAS oncogene, PIK3CA, PTEN, and TP53 mutations are the most commonly associated genetic alterations in ATC [6, 7]. Reduced levels of E-cadherin and beta-catenin are also associated with ATC [8].

Around 20–30% of patients have coexisting differentiated thyroid cancer with papillary being the most common (20%) [9, 10]. Serial biopsies of the thyroid gland also predispose to ATC [11]. Commonly seen mutations and their expression are listed in Table 1.

Mutations associated with ATC (frequency in %) [8]Expression
TP53 [12]Upregulated
CTNNB1 [13]Upregulated
BRAF [14]Upregulated
RAS [15]Upregulated
PI3KCA [16]Downregulated
PTEN [17]Downregulated
APC [9]Downregulated

Table 1.

Most common mutations and their expression associated with ATC.

Advertisement

3. Prognosis

ATC carries a dismal prognosis with mortality approaching 100% with 1- and 5-year survival rate less than 30 and 14%, respectively [17, 18, 19, 20]. Patients with localized disease have a better prognosis than those with regional or distant metastasis [9, 20]. Tumor size less than 6 cm in maximum dimension carries a better prognosis [9, 18]. Other favorable prognostic factors include unilateral tumor, localized with node-negative or no extrathyroidal extension [16, 20, 21].

Unfavorable factors include older age at diagnosis, male gender, airway compromise during the presentation, and distant metastasis [16, 20].

Advertisement

4. Clinical features

The majority of the patients present with a rapidly progressing neck mass with complaints of dysphagia, dyspnea, and neck pain. Approximately 90% will have regional or distant spread at the time of diagnosis [17, 18, 19]. The regional spread shows infiltration of perithyroidal fat, lymph nodes, trachea, esophagus, great vessels of the neck, and mediastinum. Based on the infiltration of adjacent neck structures, patients can also develop hoarseness (recurrent laryngeal nerve invasion), Horner’s syndrome (parasympathetic chain involvement), and thromboembolic episodes due to carotid infiltration.

Around 40% of patients present with cervical lymphadenopathy and 43% will have distant metastasis most commonly involving lungs followed by bone and brain [20].

Also, patients can present with constitutional symptoms, such as pyrexia of unknown origin, anorexia, weight loss, and rarely with features of thyroiditis [22, 23].

On examination, there will be bilateral but asymmetric enlargement of the thyroid gland with ill-defined borders. Usually, they are nodular, woody hard in consistency, and may be tender. Some nodules may be fluctuant due to focal tumor necrosis [24]. Due to adhesion with the adjacent structures, most of the time the swelling does not move with swallowing. The skin over the swelling may be erythematous and ulcerated.

During clinical examination, they should undergo an ENT evaluation to rule out vocal cord dysfunction or airway compromise.

Other findings include dilated chest wall veins due to superior vena cava obstruction (SVCO) from a retrosternal thyroid growth, stridor due to tracheal invasion, and vocal cord paralysis.

Advertisement

5. Diagnosis

ATC is a very aggressive tumor with a very short doubling time, and hence disease burden should be assessed accurately and promptly.

Laboratory investigations should include a complete blood count, a baseline evaluation of liver and renal function, thyroid function tests, albumin levels, and serum electrolytes levels. Nutritional assessment is also required as most of them will have dysphagia and reduced food intake.

Fine needle aspiration cytology from the thyroid mass can yield an early diagnosis. Morphologic patterns of ATC include spindle cells, pleomorphic giant cells, and squamoid histologies [16]. Most of them will have a mixed morphology of two or all the three patterns and they show extensive necrosis, atypical mitoses, and numerous mitotic figures. However, for immunostaining and molecular studies, a core biopsy is preferred. Routinely they will not stain positive for TTF1, PAX8, and thyroglobulin. Patterns of IHC in various thyroid malignancies are summarized in Table 2.

IHCDTCATCMTCSCCLymphoma
Pancytokeratin++++++/−++++++
Thyroglobulin+++
TTF-1+++−/++/−
BRAF v600E+/-−/+
PAX8++++/−+/−−/+
Ki67<5%>30%<20%>30%Variable
Chromogranin+++
Calcitonin+++/−
CEA+++
P53+/−+/−+/−
CD45+++

Table 2.

Patterns of IHC in various thyroid malignancies [25].

Poorly differentiated carcinoma, large cell lymphoma, and extension of laryngeal carcinoma are the most common differential diagnosis.

Radiological evaluation should be done immediately without any delay. High-resolution ultrasound can be done as it is convenient, rapid, and easy in assessing tumor extent, neck nodes, and adjacent structure involvement.

Other imaging modalities include CT/MRI of the neck and chest to assess tumor extent and infiltration to adjacent structures. FDG-PET scan has recently gained a role in imaging ATC as it helps in the accurate diagnosis of distant metastasis compared to other imaging modalities [15]. If trachea or esophagus infiltration is suspected, upper GI endoscopy or bronchoscopy is indicated [20, 25].

Molecular testing can be done if available; however, its clinical utility is not well established. Next-generation sequencing should be performed for targetable mutations under the context of a clinical trial. Rapid testing of BRAF V600E mutation is most commonly done if available in FNA or core needle biopsy specimens.

Advertisement

6. Staging

The American Joint Committee on Cancer (AJCC) 8th edition considers all anaplastic cancers as Stage IV. The T category follows the same definitions as those for differentiated thyroid cancers. Intrathyroidal anaplastic cancers form Stage IVA, whereas gross extrathyroidal extension or nodal metastasis form Stage IVB and distant metastasis form Stage IVC.

Advertisement

7. Treatment approach

Initial therapy in these patients mainly depends on the stage of disease and mutation status. Based on this, various treatment options include surgery, radiation with or without chemotherapy, systemic therapy, and palliation.

Gross resection is the main goal in patients with ATC, but the extent of resection should always outweigh the potentially devastating complications and morbidity of the procedures. Airway assessment and prompt treatment without delay are most important. Securing and maintaining a patent airway is challenging in these patients, but, routine tracheostomy is not recommended as it has not shown any improved outcome in survival or quality of life.

Tracheostomy is recommended in impending airway compromise and in those tumors that will not benefit from debulking.

7.1 Stage IVA

Stage IVa includes disease confined within the thyroid capsule. Around 2–15% of patients present with Stage IVa disease, in which, total thyroidectomy with a therapeutic central and lateral neck node dissection is recommended [25, 26]. Attaining gross negative margins have shown a significantly better prognosis than those with tumor residue (p < 0.005) [26]. Sugitani et al. reported that, although the benefit from additional therapies for completely resected Stage IVa ATC was not significant, they tend to show better survival with adjuvant radiation compared to those who underwent radical surgery alone (HR: 0.37; 95% CI: 0.121.13; p = .081) [27]. However, for completely resected ATC (R0 resection), additional therapies are not routinely indicated.

7.2 Stage IVB

Around 35% of the patients present as Stage IVb with extrathyroidal extension or cervical nodal involvement. They benefit from a combined modality approach.

For resectable tumors, total thyroidectomy with central and lateral therapeutic neck dissection followed by adjuvant chemoradiation has shown significantly prolonged cancer-specific survival compared to those who underwent surgery alone or with adjuvant RT alone (HR: 0.45; 95% CI: 0.250.81; p = .0083) [27].

The intensity-modulated RT technique is recommended to get better dose distribution and reduced toxicities [28]. Several authors have reported a dose–response relationship and showed that a dose of more than 60 Gy has shown a good outcome [14, 29, 30]. Commonly delivered radiation dose includes 70 Gy to the gross tumor or 66–70 Gy to the postoperative bed and 54 Gy to the potential microscopic spread region using a standard fractionation schedule [31, 32].

Several chemotherapy agents are used as concurrent, but mostly Doxorubicin 20 mg/m2 or Paclitaxel 50 mg/m2 weekly is given [14, 33, 34].

The role of hyperfractionation is not known, as there is poor evidence to show that it is better than conventional fractionation. Also, hyperfractionation is associated with increased toxicity [31, 35, 36].

However, careful patient selection is required as the procedure has an impact on quality of life. Hence, those who get a meaningful clinical benefit should be offered combined modality treatment. The best outcome is seen when adjuvant radiation is started as early as possible, once the patient has recovered from surgery [37].

For unresectable tumors with BRAF mutation, neoadjuvant treatment with Dabrafenib (150 mg twice daily) and Trametinib (20 mg daily) is started to downsize the tumor to facilitate a complete surgical resection [38, 39]. For poor responders of mutation-directed therapies, palliative radiation is an option.

7.3 Stage IVC

Around 55% of ATC patients present with distant metastasis. Stage IVc has no curative treatment and is fatal. One case series has reported a median survival of 4.2 months in those with distant metastasis at presentation compared to 6 months in nonmetastatic ATC [34]. Stage IVc is managed with palliative radiation or debulking with or without systemic therapies.

Palliative resection or debulking should be considered to avoid a future or to treat current airway compromise, which may prolong and improve quality of life. As discussed initially, most of the patients are in their 6th or 7th decade. Hence, airway preservation is enough for old age life preservation.

Palliative external beam radiotherapy (EBRT) has a definite role in symptom control for those who have unresectable/metastatic diseases. It helps in reducing the growth of neck mass and thereby alleviates the pressure symptoms. Various schedules are there, but commonly followed ones are 20 Gy in 5 fractions or 30 Gy in 10 fractions.

Systemic therapies include cytotoxic agents, targeted agents, and immunotherapies. If patients have another targetable mutation, such as NTRK, RET fusion, or ALK, they should be enrolled in clinical trials in mutation-directed systemic therapy.

Advertisement

8. Role of cytotoxic therapy

Chemotherapy is an important independent prognostic factor associated with improved survival [13, 40, 41]. However, data on comparing different chemotherapy regimens in these patients are very limited and underpowered due to the low incidence and aggressiveness of the tumor. In the absence of molecular abnormalities, most commonly given chemotherapy includes a combination of Paclitaxel and carboplatin, Cisplatin and Doxorubicin, Docetaxel and Doxorubicin, Paclitaxel alone or Doxorubicin alone [25]. The role of the combination of Cisplatin and Doxorubicin as well as Paclitaxel as a single agent is being studied in ATC and has shown moderate response [19]. However, all these are based on fairly small single studies that need further validation.

ATC has a very rapid doubling time of 3–12 days, hence, some authors recommend that the chemotherapy regimens should be administered in shorter intervals, on weekly basis rather giving every 3–4 weeks [3, 31]. Systemic therapy protocols are listed in Table 3.

TreatmentProtocols and dose
Chemotherapy [3]Doxorubicin (20 mg/m2) + Cisplatin 120 mg/m2) every 28 days
Paclitaxel (175 mg/m2) + carboplatin AUC 5 every 21 days
Paclitaxel 50–100 mg/m2 + Carboplatin AUC2 weekly
Docetaxel 20 mg/m2 + Doxorubicin 20 mg/m2 weekly
Paclitaxel alone 30–60 mg/m2 or Docetaxel 20 mg/m2 weekly
BRAF and MEK inhibitors [42, 43]Dabrafenib 150 mg twice daily + Trametinib 2 mg once daily
RET inhibitor [44]Selpercatinib 160 mg twice daily or 120 mg twice daily (if body weight <50 kg)
NTRK inhibitor [45]Larotrectinib 100 mg twice daily
Entrectinib 600 mg once daily
ALK inhibitor [46]Crizotinib 250 mg twice daily
Larotrectinib 100 mg twice daily

Table 3.

Systemic therapy protocols.

Advertisement

9. Role of targeted agents

Primary chemoresistance is a commonly encountered issue in ATC that often results in a bad prognosis. Hence, newer therapeutic agents are being investigated in ATC, targeting various molecular alterations.

As mentioned above, one-fourth of the ATC is associated with mutation of BRAF and RAS [47]. A phase II trial was conducted in BRAF V600E-positive tumors including 36 patients with ATC, where the patients were treated with the BRAF inhibitor Dabrafenib plus the MEK inhibitor Trametinib. The treatment was well tolerated with an ORR of 56% (95% CI, 38.1–72.1%), with three complete responses and a median PFS and OS of 6.7 and 14.5 months, respectively [42, 43]. The combination was FDA approved in 2018 as 1st line therapy for BRAF V600E-positive ATC patients.

Unfortunately, acquired resistance to BRAF inhibitors due to secondary mutation of MAPK pathway or via PI3K/AKT/mTOR pathway is common in ATC, and hence newer targeted agents are needed. Around 2–3% of ATC patients will have a mutation of NTRK, ALK, or RET fusion.

For such mutation-positive ATC, very high response rates are reported with specific inhibitors in various trials. A pooled subgroup analysis of trials investigating the role of Larotrectinib in ATC showed a response rate of 29% in two out of seven patients [45, 48]. Similarly, long-lasting responses were also noted with Selpercatinib and Crizotinib in RET fusion ATC and ALK-rearranged ATC, respectively [44, 46]. However, these are small studies, some are equivalent to case reports and need more validation.

Inhibitors of PI3K/AKT/mTOR pathway like Everolimus were tested but showed no response [49]. Combination of Sorafenib and Temsirolimus also could not demonstrate a durable response [50].

Though anti-angiogenic agents, such as Lenvatinib, are approved in radioiodine refractory thyroid cancers, their role in ATC is controversial. Based on a single-arm phase II study with 51 ATC patients, Lenvatinib showed a median PFS benefit of 7.4 months, a median OS of 10.6 months, and an ORR of 24% [51]. However, a recent prospective phase II Lenvatinib trial was stopped due to futility as the interim analysis reported a very low response rate (2.9%) and survival outcome [52].

Advertisement

10. Role of immunotherapy

ATC shows high expression of PD-1/PDL-1, and hence immunotherapy may be a promising approach in these patients. However, data available on its use in ATC are limited. Many phase II trials are ongoing with immunotherapy alone as well as in combination with other agents.

The combination of Pembrolizumab and Lenvatinib in ATC is evaluated in the phase II trial of ATLEP (Anaplastic Thyroid Carcinoma Lenvatinib Pembrolizumab) study and has shown a partial response of 37.5% [53]. Also, a single institution study by Lorch et al. has reported partial response in one-third of the ATC patients treated with a combination of Ipilimumab and Nivolumab [54].

Another PD-1 inhibitor Spartalizumab was tested by Capdevila et al. in 41 heavily pretreated ATC patients and showed a response rate of 19.6%. The median OS was 5.9 months with 40% alive at 1 year. The median PFS was 1.7 months. The response rate was higher for those who had a greater PDL-1 expression (35%) [55]. But, the drug is not FDA approved and is not commercially available.

Many newer drugs are being tested in phase II and III trials for ATC due to its aggressive nature and poorer outcomes than conventional treatment. A few are listed in Table 4.

DrugStudy designNumber of ATC patients (among total)Median OS (months)
Sunitinib [12]Phase II4/71NA
Axitinib [56]Phase II2/60NA
Pazopanib [57]Phase II15/153.7
Imatinib [58]Phase II11/11NA
Gefitinib [59]Phase II5/27NA
Vemurafenib [60]Phase II7/122NA
Fosbretabulin [61]Phase II26/264.7
Spartalizumab [55]Phase II38/425.9
Efatutazone + Paclitaxel [62]Phase 115/153.3
Fosbretabulin+ carboplatin+ Paclitaxel [63]Phase II/III80/805.2

Table 4.

Newer drugs and their clinical trials.

Stage-wise treatment approach is summarized in Figures 13. Clinical trials are encouraged and the best supportive care can be considered as an option at any point of treatment.

Figure 1.

Stage IVA treatment approach.

Figure 2.

Stage IVB treatment approach.

Figure 3.

Stage IV C treatment approach.

11. Future directions

Considering the aggressive nature of the disease and its dismal prognosis, novel therapies targeting the signal transduction pathways associated with ATC are to be investigated. Several ongoing trials are being conducted that investigate the role of combining TKIs with immunotherapy or chemotherapy (Table 5).

TrialsInvestigating drug(s)PhaseStatus
NCT03085056 [64]Trametinib + Paclitaxel in Advanced ATCPhase 1Recruiting
NCT02152137 [65]Efatutazone + Paclitaxel in Advanced ATCPhase 2Active, not recruiting
NCT04552769 [66]Abemaciclib (CDK4/6 inhibitor) in advanced ATCPhase 2Recruiting
NCT04675710 [67]Pembrolizumab + dabrafenib + Trametinib a Neoadjuvant in BRAf mutated ATCPhase 2Recruiting
NCT04400474 [68]Cabozantinib + Atezolizumab in advanced ATCPhase 2Recruiting
NCT04579757 [69]Surufatinib + Tislelizumab in advanced ATCPhase 1/2Recruiting
NCT04759911 [70]Selpercatinib as neoadjuvant in ATC with RET alterationsPhase 2Recruiting

Table 5.

Ongoing trials in ATC [3].

12. End-of-life care

Most of the patients are present in a very advanced stage due to the rapid progression of the disease and will not be eligible for any kind of local or systemic therapy. End-of-life life care and best supportive care form an integral part of management in these groups of patients [25].

13. Surveillance and follow up

Active surveillance for those who had a complete response to initial treatment is needed. CT of the chest is done within 4 weeks and a single PET–CT is done after 3 months of treatment [15].

CT of the neck/chest/abdomen is done thereafter every 1–3 months for the initial 2 years. Later on, less frequent imaging is recommended. Brain imaging is not routinely done, except in case of symptoms of brain metastasis.

There is no role for radioiodine scanning/ablation or serum thyroglobulin measurement.

Thyroid hormone replacement is required for maintaining euthyroid status. T4 should be started (1.6 mcg/kg body weight) immediately after surgery. TSH suppression to less than normal is not indicated in ATC.

14. Conclusion

ATC is a rare thyroid malignancy with an extremely grave prognosis. Diagnosis and treatment should be started quickly and should be based on a multidisciplinary approach, including surgery, radiation, chemotherapy, and targeted agents. Stage IVA tumors should undergo primary surgery with gross-negative margins. For completely resected tumors, there is no role for additional therapies. For resectable stage IVB, surgery followed by adjuvant radiation with or without chemotherapy is recommended. For unresectable tumors, mutation-directed therapies based on BRAF mutation are to be incorporated followed by surgery or radiation. For Stage IVC, palliative treatment is recommended with either palliative radiation or debulking with or without systemic therapies. Although newer mutation-directed therapies are being incorporated into the management of ATC, further validation is needed. The inclusion of novel approaches, such as targeted therapy and immunotherapy either alone or in combination with other modalities, may improve outcomes in these patients.

References

  1. 1. Remick SC, Nagaiah G, Hossain A, Mooney CJ, Parmentier J. Anaplastic thyroid cancer: A review of epidemiology, pathogenesis, and treatment. Journal of Oncology. 2011;2011:2-5
  2. 2. Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer Journal of Clinical. 2016;66(1):7-30
  3. 3. Jannin A, Escande A, al Ghuzlan A, Blanchard P, Hartl D, Chevalier B, et al. Anaplastic thyroid carcinoma: An update. Cancers (Basel). 2022;14(4):1-25
  4. 4. Burke JP, Hay ID, Dignan F, Goellner JR, Achenbach SJ, Oberg AL, et al. Long-term trends in thyroid carcinoma: A population-based study in Olmsted County, Minnesota, 1935–1999. Mayo Clinic Proceedings. 2005;80(6):753-758
  5. 5. Davies L, Welch HG. Increasing incidence of thyroid cancer in the United States, 1973–2002. Journal of the American Medical Association. 2006;295(18):2164-2167
  6. 6. Pita JM, Figueiredo IF, Moura MM, Leite V, Cavaco BM. Cell cycle deregulation and TP53 and RAS mutations are major events in poorly differentiated and undifferentiated thyroid carcinomas. Journal of Clinical Endocrinology Metabolism. 2014;99(3):E497-E507
  7. 7. Charles RP, Silva J, Iezza G, Phillips WA, McMahon M. Activating BRAF and PIK3CA mutations cooperate to promote anaplastic thyroid carcinogenesis. Molecular Cancer Research. 2014;12(7):979-986
  8. 8. Smallridge RC, Marlow LA, Copland JA. Anaplastic thyroid cancer: Molecular pathogenesis and emerging therapies. Endocrine-Related Cancer. 2009;16(1):17
  9. 9. Nel CJC, van Heerden JA, Goellner JR, Gharib H, McConahey WM, Taylor WF, et al. Anaplastic carcinoma of the thyroid: A clinicopathologic study of 82 cases. Mayo Clinic Proceedings. 1985;60(1):51-58
  10. 10. Carcangiu ML, Steeper T, Zampi G, Rosai J. Anaplastic thyroid carcinoma. A study of 70 cases. American Journal of Clinical Pathology. 1985;83(2):135-158
  11. 11. Moore JH, Bacharach B, Choi HY. Anaplastic transformation of metastatic follicular carcinoma of the thyroid. Journal of Surgical Oncology. 1985;29(4):216-221
  12. 12. Ravaud A, de la Fouchardière C, Caron P, Doussau A, Do Cao C, Asselineau J, et al. A multicenter phase II study of sunitinib in patients with locally advanced or metastatic differentiated, anaplastic or medullary thyroid carcinomas: Mature data from the THYSU study. European Journal of Cancer. 2017;76:110-117
  13. 13. Janz TA, Neskey DM, Nguyen SA, Lentsch EJ. Is the incidence of anaplastic thyroid cancer increasing: A population based epidemiology study. World Journal of Otorhinolaryngology – Head and Neck Surgery. 2019;5(1):34-40
  14. 14. Beckham TH, Romesser PB, Groen AH, Sabol C, Shaha AR, Sabra M, et al. Intensity-modulated radiation therapy with or without concurrent chemotherapy in nonanaplastic thyroid cancer with unresectable or gross residual disease. Thyroid. 2018;28(9):1180-1189
  15. 15. Poisson T, Deandreis D, Leboulleux S, Bidault F, Bonniaud G, Baillot S, et al. 18F-fluorodeoxyglucose positron emission tomography and computed tomography in anaplastic thyroid cancer. European Journal of Nuclear Medicine and Molecular Imaging. 2010;37(12):2277-2285
  16. 16. Keutgen XM, Sadowski SM, Kebebew E. Management of anaplastic thyroid cancer. Gland Surgery. 2015;4(1):44
  17. 17. McIver B, Hay ID, Giuffrida DF, Dvorak CE, Grant CS, Thompson GB, et al. Anaplastic thyroid carcinoma: A 50-year experience at a single institution. Surgery. 2001;130(6):1028-1034
  18. 18. Tan RK, Finley RK, Driscoll D, Bakamjian V, Hicks WL, Shedd DP. Anaplastic carcinoma of the thyroid: A 24-year experience. Head & Neck. 1995;17(1):41-48
  19. 19. Tennvall J, Lundell G, Wahlberg P, Bergenfelz A, Grimelius L, Åkerman M, et al. Anaplastic thyroid carcinoma: Three protocols combining doxorubicin, hyperfractionated radiotherapy and surgery. British Journal of Cancer. 2002;86(12):1848-1853
  20. 20. Kebebew E, Greenspan FS, Clark OH, Woeber KA, McMillan A. Anaplastic thyroid carcinoma: Treatment outcome and prognostic factors. Cancer. 2005;103(7):1330-1335
  21. 21. Kihara M, Miyauchi A, Yamauchi A, Yokomise H. Prognostic factors of anaplastic thyroid carcinoma. Surgery Today. 2004;34(5):394
  22. 22. Murakami T, Noguchi S, Murakami N, Tajiri J, Ohta Y. Destructive thyrotoxicosis in a patient with anaplastic thyroid cancer. Endocrinologia Japonica. 1989;36(6):905-907
  23. 23. Nishiyama RH, Dunn EL, Thompson NW. Anaplastic spindle-cell and giant-cell tumors of the thyroid gland. Cancer. 1972;30(1):113-127
  24. 24. Barr R, Dann F. Anaplastic thyroid carcinoma metastatic to skin. Journal of Cutaneous Pathology. 1974;1(5):201-206
  25. 25. Bible KC, Kebebew E, Brierley J, Brito JP, Cabanillas ME, Clark TJ, et al. 2021 American Thyroid Association Guidelines for Management of Patients with Anaplastic Thyroid Cancer. Thyroid. 2021;31(3):337-386
  26. 26. Passler C, Scheuba C, Prager G, Kaserer K, Flores JA, Vierhapper H, et al. Anaplastic (undifferentiated) thyroid carcinoma (ATC). A retrospective analysis. Langenbeck’s Archives of Surgery. 1999;384(3):284-293
  27. 27. Sugitani I, Onoda N, Ito KI, Suzuki S. Management of anaplastic thyroid carcinoma: The Fruits from the ATC Research Consortium of Japan. Journal of Nippon Medical School. 2018;85(1):18-27
  28. 28. Bhatia A, Rao A, Ang KK, Garden AS, Morrison WH, Rosenthal DI, et al. Anaplastic thyroid cancer: Clinical outcomes with conformal radiotherapy. Head & Neck. Jul 2010;32(7):829-836
  29. 29. Pezzi TA, Mohamed ASR, Sheu T, Blanchard P, Sandulache VC, Lai SY, et al. Radiation therapy dose is associated with improved survival for unresected anaplastic thyroid carcinoma: Outcomes from the National Cancer Data Base. Cancer. 2017;123(9):1653-1661
  30. 30. Mohebati A, Dilorenzo M, Palmer F, Patel SG, Pfister D, Lee N, et al. Anaplastic thyroid carcinoma: A 25-year single-institution experience. Annals of Surgical Oncology. 2014;21(5):1665-1670
  31. 31. De Crevoisier R, Baudin E, Bachelot A, Leboulleux S, Travagli JP, Caillou B, et al. Combined treatment of anaplastic thyroid carcinoma with surgery, chemotherapy, and hyperfractionated accelerated external radiotherapy. International Journal of Radiation Oncology, Biology, Physics. 2004;60(4):1137-1143
  32. 32. Kim JH, Leeper RD. Combination adriamycin and radiation therapy for locally advanced carcinoma of the thyroid gland. International Journal of Radiation Oncology, Biology, Physics. 1983;9(4):565-567
  33. 33. Baek SK, Lee MC, Hah JH, Ahn SH, Son YI, Rho YS, et al. Role of surgery in the management of anaplastic thyroid carcinoma: Korean nationwide multicenter study of 329 patients with anaplastic thyroid carcinoma, 2000 to 2012. Head & Neck. 2017;39(1):133-139
  34. 34. Ito KI, Hanamura T, Murayama K, Okada T, Watanabe T, Harada M, et al. Multimodality therapeutic outcomes in anaplastic thyroid carcinoma: Improved survival in subgroups of patients with localized primary tumors. Head & Neck. 2012;34(2):230-237
  35. 35. Dandekar P, Harmer C, Barbachano Y, Rhys-Evans P, Harrington K, Nutting C, et al. Hyperfractionated Accelerated Radiotherapy (HART) for anaplastic thyroid carcinoma: Toxicity and survival analysis. International Journal of Radiation Oncology, Biology, Physics. 2009;74(2):518-521
  36. 36. Wang Y, Tsang R, Asa S, Dickson B, Arenovich T, Brierley J. Clinical outcome of anaplastic thyroid carcinoma treated with radiotherapy of once- and twice-daily fractionation regimens. Cancer. 2006;107(8):1786-1792
  37. 37. Smallridge RC, Ain KB, Asa SL, Bible KC, Brierley JD, Burman KD, et al. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid. 2012;22(11):1104-1139
  38. 38. Cabanillas ME, Ferrarotto R, Garden AS, Ahmed S, Busaidy NL, Dadu R, et al. Neoadjuvant BRAF- and immune-directed therapy for anaplastic thyroid carcinoma. Thyroid. 2018;28(7):945-951
  39. 39. Wang JR, Zafereo ME, Dadu R, Ferrarotto R, Busaidy NL, Lu C, et al. Complete surgical resection following neoadjuvant dabrafenib plus trametinib in BRAF V600E-mutated anaplastic thyroid carcinoma. Thyroid. 2019;29(8):1036-1043
  40. 40. Lin B, Ma H, Ma M, Zhang Z, Sun Z, Hsieh IY, et al. The incidence and survival analysis for anaplastic thyroid cancer: A SEER database analysis. American Journal of Translational Research. 2019;11(9):5888
  41. 41. Kwon J, Kim BH, Jung HW, Besic N, Sugitani I, Wu HG. The prognostic impacts of postoperative radiotherapy in the patients with resected anaplastic thyroid carcinoma: A systematic review and meta-analysis. European Journal of Cancer. 2016;59:34-45
  42. 42. Subbiah V, Cabanillas ME, Kreitman RJ, Wainberg ZA, Cho JY, Keam B, et al. Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAF V600-mutant anaplastic thyroid cancer. Journal of Clinical Oncology. 2018;36(1):7-13
  43. 43. Leboulleux S, Lee EK, Bastholt L, Tahara M, Wirth LJ, Sherman SI, et al. Updated efficacy and safety data of dabrafenib (D) and trametinib (T) in patients (pts) with BRAF V600E–mutated anaplastic thyroid cancer (ATC). Annals of Oncology. 2018;29:645-646
  44. 44. Wirth LJ, Sherman E, Robinson B, Solomon B, Kang H, Lorch J, et al. Efficacy of selpercatinib in RET -altered thyroid cancers. The New England Journal of Medicine. 2020;383(9):825-835
  45. 45. Hong DS, DuBois SG, Kummar S, Farago AF, Albert CM, Rohrberg KS, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: A pooled analysis of three phase 1/2 clinical trials. The Lancet Oncology. 2020;21(4):531-540
  46. 46. Godbert Y, De Figueiredo BH, Bonichon F, Chibon F, Hostein I, Pérot G, et al. Remarkable response to crizotinib in woman with anaplastic lymphoma kinase-rearranged anaplastic thyroid carcinoma. Journal of Clinical Oncology. 2015;33(20):e84-e87
  47. 47. Cabanillas ME, Zafereo M, Williams MD, Ferrarotto R, Dadu R, Gross N, et al. Recent advances and emerging therapies in anaplastic thyroid carcinoma. F1000Research. 2018;7:511-518
  48. 48. Cabanillas ME, Zafereo M, Gunn GB, Ferrarotto R. Anaplastic thyroid carcinoma: Treatment in the age of molecular targeted therapy. Journal of Oncology Practice/ American Society of Clinical Oncology. 2016;12(6):511-518
  49. 49. Lim SM, Chang H, Yoon MJ, Hong YK, Kim H, Chung WY, et al. A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes. Annals of Oncology. 2013;24(12):3089-3094
  50. 50. Sherman EJ, Dunn LA, Ho AL, Baxi SS, Ghossein RA, Fury MG, et al. Phase 2 study evaluating the combination of sorafenib and temsirolimus in the treatment of radioactive iodine-refractory thyroid cancer. Cancer. 2017;123(21):4114-4121
  51. 51. Tahara M, Kiyota N, Yamazaki T, Chayahara N, Nakano K, Inagaki L, et al. Lenvatinib for anaplastic thyroid cancer. Frontiers in Oncology. 2017;7:3-6
  52. 52. Wirth LJ, Brose MS, Sherman EJ, Licitra L, Schlumberger M, Sherman SI, et al. Open-label, single-arm, multicenter, Phase II trial of lenvatinib for the treatment of patients with anaplastic thyroid cancer. Journal of Clinical Oncology. 2021;39(21):2359-2366
  53. 53. Dierks C, Seufert J, Aumann K, Ruf J, Klein C, Kiefer S, et al. Combination of lenvatinib and pembrolizumab is an effective treatment option for anaplastic and poorly differentiated thyroid carcinoma. Thyroid. 2021;31(7):1076-1085
  54. 54. Lorch JH, Barletta JA, Nehs M, Uppaluri R, Alexander EK, Haddad RI, et al. A phase II study of nivolumab (N) plus ipilimumab (I) in radioidine refractory differentiated thyroid cancer (RAIR DTC) with exploratory cohorts in anaplastic (ATC) and medullary thyroid cancer (MTC). Thyroid. 2020;38(15):6513
  55. 55. Wirth LJ, Eigendorff E, Capdevila J, Paz-Ares LG, Lin C-C, Taylor MH, et al. Phase I/II study of spartalizumab (PDR001), an anti-PD1 mAb, in patients with anaplastic thyroid cancer. Cancer. 2018;36(15):6024
  56. 56. Zini A, Czerninski R, Sgan-Cohen HD. Oral cancer over four decades: Epidemiology, trends, histology, and survival by anatomical sites. Journal of Oral Pathology & Medicine. 2009;39(4):299-305
  57. 57. Bible KC, Suman VJ, Menefee ME, Smallridge RC, Molina JR, Maples WJ, et al. A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. The Journal of Clinical Endocrinology and Metabolism. 2012;97(9):3179-3184
  58. 58. Ha HT, Lee JS, Urba S, Koenig RJ, Sisson J, Giordano T, et al. A phase II study of imatinib in patients with advanced anaplastic thyroid cancer. Thyroid. 2010;20(9):975-980
  59. 59. Pennell NA, Daniels GH, Haddad RI, Ross DS, Evans T, Wirth LJ, et al. A phase II study of gefitinib in patients with advanced thyroid cancer. Thyroid. 2008;18(3):317-323
  60. 60. Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay J-Y, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. The New England Journal of Medicine. 2015;373(8):726-736
  61. 61. Mooney CJ, Nagaiah G, Fu P, Wasman JK, Cooney MM, Savvides PS, et al. A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid. 2009;19(3):233-240
  62. 62. Smallridge RC, Copland JA, Brose MS, Wadsworth JT, Houvras Y, Menefee ME, et al. Efatutazone, an oral PPAR-γ agonist, in combination with paclitaxel in anaplastic thyroid cancer: Results of a multicenter phase 1 trial. The Journal of Clinical Endocrinology and Metabolism. 2013;98(6):2392-2400
  63. 63. Sosa JA, Elisei R, Jarzab B, Balkissoon J, Lu SP, Bal C, et al. Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid. 2014;24(2):232-240
  64. 64. Sherman E. Trametinib in Combination With Paclitaxel in the Treatment of Anaplastic Thyroid Cancer – Full Text View – ClinicalTrials.gov. MSKCC; 2017. Available from https://clinicaltrials.gov/ct2/show/NCT03085056
  65. 65. Smallridge R. Inolitazone Dihydrochloride and Paclitaxel in Treating Patients With Advanced Anaplastic Thyroid Cancer – Full Text View – ClinicalTrials.gov. Mayo Clinic, National Cancer Institute, Daiichi Sanyo, Inc; 2019. Available from: https://clinicaltrials.gov/ct2/show/NCT02152137
  66. 66. Khanna V. Abemaciclib in Metastatic or Locally Advanced Anaplastic/Undifferentiated Thyroid Cancer – Full Text View – ClinicalTrials.gov. Stanford Cancer Center, University of North Carolina; 2022. Available from: https://clinicaltrials.gov/ct2/show/NCT04552769
  67. 67. Zafereo M. Pembrolizumab, Dabrafenib, and Trametinib Before Surgery for the Treatment of BRAF-Mutated Anaplastic Thyroid Cancer – Full Text View – ClinicalTrials.gov. Anderson Cancer Centre; 2020. Available from: https://clinicaltrials.gov/ct2/show/NCT04675710
  68. 68. Grupo Espanol de Tumores Neuroendocrinos. Trial of Cabozantinib Plus Atezolizumab in Advanced and Progressive Neoplasms of the Endocrine System. The CABATEN Study – Full Text View – ClinicalTrials.gov; 2022. Available from: https://clinicaltrials.gov/ct2/show/NCT04400474
  69. 69. Kauh J. Surufatinib in Combination With Tislelizumab in Subjects With Advanced Solid Tumors – Full Text View – ClinicalTrials.gov. Hutchison; 2022. Available from: https://clinicaltrials.gov/ct2/show/NCT04579757
  70. 70. Zafereo M. Selpercatinib Before Surgery for the Treatment of RET-Altered Thyroid Cancer – Full Text View – ClinicalTrials.gov. Anderson Cancer Centre; 2021. Available from: https://clinicaltrials.gov/ct2/show/NCT04759911

Written By

Zuzak Sharafuddin, Rejnish Ravikumar, Malu Rafi, Lekha Madhavan Nair, John Mathew and Kainickal Cessal Thommachan

Submitted: 16 May 2022 Reviewed: 11 July 2022 Published: 09 November 2022