Skip to main content

Larotrectinib efficacy for liver metastases in papillary thyroid carcinoma patient harboring SQSTM1–NTRK1 fusion

Abstract

Background

Pooled data analysis from three phase I/II larotrectinib clinical trials revealed that larotrectinib demonstrated rapid and durable disease control and a favorable safety profile for patients with neurotrophic-tropomyosin receptor kinase (NTRK) fusion positive thyroid carcinoma. Herein, we report the case of a patient with papillary thyroid carcinoma (PTC) and liver metastases who demonstrated a durable response to treatment with larotrectinib.

Case presentation

A 50-year-old female with PTC was referred to our hospital for postoperative observation. Computed tomography (CT) scan was performed to screen for distant metastasis, since thyroglobulin concentration increased gradually, and revealed multiple distant metastases, including multiple liver metastases. Radioactive iodine was administered at a dose of 100 mCi. However, uptake was observed only in the thyroid bed, and distant metastases had no avidity. As liver metastases progressed, lenvatinib (24 mg/day) was initiated after confirmation of liver metastases by liver biopsy 9 years and 1 month after the initial referral to our hospital. Since the multiple metastases became refractory for lenvatinib, the OncoGuide™ NCC Oncopanel System was performed, and the SQSTM1–NTRK1 gene fusion was confirmed. Larotrectinib was subsequently administered at a dose of 200 mg/day. The CT before the initiation of larotrectinib showed multiple liver metastases with a maximum diameter of 48 mm. The first CT evaluation at 1 month after the initiation of larotrectinib treatment showed that the tumor volume was reduced by 28% in the RECIST 1.1 criteria. After 3 months of larotrectinib treatment, a 38% reduction in the tumor volume was achieved as the best clinical response. The only side effect was grade 1 myalgia. At 12 months after the initiation of larotrectinib treatment, none of the lesions had progressed.

Conclusions

In conclusion, larotrectinib demonstrated effective antitumor activity against liver metastases of PTC, a relatively rare site of distant metastasis. Furthermore, the efficacy of larotrectinib was maintained, even though the patient had a history of multi-tyrosine kinase inhibitor treatment and a relatively infrequent fusion gene, SQSTM1–NTRK1.

Background

Papillary thyroid carcinoma (PTC) is the most common histological type of thyroid carcinoma [1]. In general, almost all PTCs have relatively good prognoses. However, some patients with PTC develop distant metastases [2]. Subsequently, multi-tyrosine kinase inhibitors (mTKIs), such as sorafenib and lenvatinib, have been used to treat progressive radioiodine-refractory PTCs [3, 4]. The most frequent target lesions in TKI treatment are the lungs, lymph nodes, and bone [5]. In contrast, liver metastasis from PTC is rare [6].

The identification of pathways involved in the pathophysiology of carcinogenesis, metastasis, and drug resistance, as well as the emergence of technologies enabling the molecular analysis of tumors and the discovery of targeted therapies, has stimulated research focusing on the optimal use of targeted agents [7]. Cancer genomic medicine (CGM), which optimizes therapeutic interventions based on tumor genome profiling, has demonstrated clinical benefits in a wide range of cancer subtypes [8]. Japan started CGM in 2019 with two approved comprehensive genomic profiling (CGP) tests under the national health insurance umbrella [8]. CGP is appropriate for patients who have completed or are nearing the completion of standard treatment for thyroid cancers with locally advanced or metastatic disease. Larotrectinib was approved in Japan in 2021 for advanced or recurrent solid tumors harboring neurotrophic-tropomyosin receptor kinase (NTRK) fusions. Larotrectinib is an orally administered ATP-competitive inhibitor of TRKA, TRKB, and TRKC [9]. Pooled data analysis from three phase I/II larotrectinib clinical trials (NCT02576431, NCT02122913, and NCT02637687) included 29 patients with TRK-fusion-positive thyroid cancer. The analysis revealed that larotrectinib demonstrated rapid and durable disease control and a favorable safety profile [10].

Herein, we report the case of a patient with PTC and liver metastases who demonstrated a durable response to treatment with larotrectinib, along with a review of the relevant literature.

Case presentation

A 50-year-old female with PTC was referred to our hospital for postoperative observation. She had previously undergone a subtotal thyroidectomy and right-sided modified neck dissection for PTC. She developed local recurrence in the remnant right lobe of the thyroid gland, and completion thyroidectomy was performed 4 years and 6 months after her initial referral to our hospital. Three years after surgery for local recurrence, a computed tomography (CT) scan was performed to screen for distant metastasis, since the thyroglobulin (Tg) concentration increased gradually. Computed tomography (CT) revealed mediastinal lymph node, lung, left adrenal gland, splenic, and multiple liver metastases. Radioactive iodine (RAI) was administered at a dose of 100 mCi. However, uptake was observed only in the thyroid bed, and distant metastases had no avidity. Six months after the first RAI treatment, the patient underwent a whole-body scan, which showed uptake only in the thyroid bed and not at the sites of distant metastasis. Therefore, we considered the sites of distant metastases to be refractory to RAI. As liver metastases progressed, lenvatinib (24 mg/day) was initiated after confirmation of liver metastases by liver biopsy 9 years and 1 month after the initial referral to our hospital. The patient’s blood test results before the initiation of lenvatinib were as follows: aspartate aminotransferase (AST), 16 U/L; alanine aminotransferase (ALT), 14 U/L; creatinine (Cre), 0.64 mg/dL; blood urea nitrogen (BUN), 10.0 mg/dL; thyroid-stimulating hormone (TSH), 0.269 μIU/mL; free triiodothyronine (F-T3), 2.54 pg/mL; free thyroxine (F-T4), 1.39 ng/dL; Tg, 635 ng/mL; and Tg antibody (TgAb), 13 IU/mL. The CT before the initiation of lenvatininb showed multiple liver metastases with a maximum diameter of 30 mm. Although the liver metastases shrank due to lenvatinib treatment and the Tg concentration reached a nadir of 26.0 ng/mL, the medication was changed to sorafenib (400 mg/day) due to grade 3 proteinuria. After that, lenvatinib (10 mg/day) was restarted due to progressive disease under sorafenib treatment. However, the multiple metastases became refractory for lenvatinib.

Since we considered that the previous tissue specimens from the liver biopsy may not be suitable for genomic testing due to deterioration, we performed another liver biopsy. First, the Oncomine Dx Target Test (Thermo Fisher, USA) was performed to screen for RET mutations. The sampled liver tissue was negative for RET mutations. In addition, BRAF mutations were not detected. However, a SQSTM1–NTRK1 fusion was identified. Then, the OncoGuide™ NCC Oncopanel System (Sysmex Co., Ltd., Kobe, Japan) was performed to confirm the SQSTM1–NTRK1 gene fusion (SQSTM1 [chr5:179,256,832;hg19/GRCh37] joining with NTRK1 [chr1:156,843,753;hg19/GRCh37]). No other pathogenic mutations were identified.

After receiving approval from the expert panel, the decision was made to initiate larotrectinib. Larotrectinib was subsequently administered at a dose of 200 mg/day. The patient’s blood test results before the initiation of larotrectinib were as follows: AST, 12 U/L; ALT, 14 U/L; Cre, 2.08 mg/dL; BUN, 45.0 mg/dL; TSH, < 0.005 μIU/mL; F-T3, 3.13 pg/mL; F-T4, 1.80 ng/dL; Tg, 9960 ng/mL; and TgAb, < 10 IU/mL. Since patient had renal dysfunction due to proteinuria by adverse effect of lenvatinib, an enhanced CT could not be performed. The CT before the initiation of larotrectinib showed multiple liver metastases with a maximum diameter of 48 mm, and lung, mediastinal lymph node, spleen, and left adrenal gland metastases were suspected (Figs. 1 and 2ac).

Fig. 1
figure 1

Computed tomography before larotrectinib initiation. The computed tomography before larotrectinib initiation showed mediastinal lymph node (a), lung (b), liver (c, d), left adrenal gland (e), and splenic (f) metastases

Fig. 2
figure 2

Baseline and 3 months after larotrectinib computed tomography images. The computed tomography before the initiation of larotrectinib showed multiple liver metastases with a maximum diameter of 48 mm (ac). After 3 months of larotrectinib treatment, a 38% reduction in the tumor volume was achieved as the best clinical response (df)

The first CT evaluation at 1 month after the initiation of larotrectinib treatment showed that the tumor volume was reduced by 28% in the RECIST 1.1 criteria [11]. After 3 months of larotrectinib treatment, a 38% reduction in the tumor volume was achieved as the best clinical response (Fig. 2d–f). The nadir of the Tg concentration was 62.9 ng/mL during larotrectinib treatment, but the TgAb level gradually increased to 3510 IU/mL. Subsequently, the TgAb level gradually decreased (Fig. 3). During larotrectinib treatment, TSH level had been suppressed to < 0.005 μIU/mL. The only side effect was grade 1 myalgia. At 12 months after the initiation of larotrectinib treatment, none of the lesions had progressed.

Fig. 3
figure 3

Clinical course after larotrectinib. The thyroglobulin (Tg) and Tg antibody (TgAb) concentrations were 635 ng/mL and 13 IU/mL, respectively. The nadir of the Tg concentration was 62.9 ng/mL during larotrectinib treatment, but the TgAb level gradually increased to 3510 IU/mL. Subsequently, the TgAb level gradually decreased. During larotrectinib treatment, thyroid-stimulating hormone level had been suppressed to < 0.005 μIU/mL

Discussion

A rearrangement involving one of the NTRK genes, which belongs to the NTRK family, represents a significant oncogenic event in thyroid cancer [12]. The NTRK family includes three genes, NTRK1, NTRK2, and NTRK3, which encode TRKA, B, and C, respectively. Fusions involving the kinase domain of the NTRK gene lead to uncontrolled activation of the tyrosine receptor kinase and, subsequently, the MAPK, PI3K/AKT, and PLC pathways [13]. TRK fusion-positive thyroid carcinoma is more commonly associated with a younger age at diagnosis, and approximately 5–25% of pediatric PTC cases are reported to harbor NTRK gene fusions [10]. In contrast, the reported presence of NTRK fusions in predominantly adult thyroid cancer cohorts is low, ranging from 2.3 to 3.4% [14]. To date, no large-scale study has investigated the incidence of NTRK fusions in Japanese patients with thyroid cancer. As genomic testing has become increasingly common in Japanese clinical practice, the incidence of NTRK fusion in Japanese patients with thyroid cancer is likely to become clear in the near future.

Several NTRK rearrangements have been observed in TCs. Chu et al. investigated 351 primary thyroid cancers, including 186 PTCs, 54 medullary thyroid carcinomas (MTCs), 38 anaplastic thyroid carcinomas (ATCs), 24 Hurthle cell carcinomas (HCCs), 23 follicular thyroid carcinomas (FTCs), 21 poorly differentiated thyroid carcinomas (PDTCs), and five unclassifiable carcinomas [15]. The study reported that NTRK rearrangements were detected in 11 (3.1%) cases, all of which were originally diagnosed with PTC [15]. The observed NTRK rearrangements included ETV6–NTRK3 (n = 4), TPR–NTRK1 (n = 2), RBPMS–NTRK3 (n = 2), SQSTM1–NTRK1 (n = 1), SQSTM1–NTRK3 (n = 1), and EML4–NTRK3 (n = 1) [15]. In another study by Pekova et al., a total of 989 thyroid cancer tissues, including 851 PTCs, 86 MTCs, 19 FTCs, 13 ATCs, 10 PDTCs, 10 HCCs, and 30 borderline thyroid tumor tissues (17 follicular tumors of uncertain malignant potential, 13 non-invasive follicular tumors with papillary-like nuclear features, and 194 benign thyroid tissues) were analyzed [12]. The analyses revealed the NTRK fusion gene in 59 patients with thyroid cancer, 57 of 846 (6.7%) patients with PTC, and 2 of 10 (20.0%) patients with PDTC [12]. In addition, eight types of NTRK fusions were identified: ETV6–NTRK3 (n = 38), TPM3–NTRK1 (n = 5), SQSTM1–NTRK3 (n = 4), EML4–NTRK3 (n = 4), RBPMS–NTRK3 (n = 3), IRF2BP2–NTRK1 (n = 3), SQSTM1–NTRK1 (n = 1), and TPR–NTRK1 (n = 1) [12]. SQSTM1–NTRK1 was also detected in this case. Considering previous reports, the SQSTM1–NTRK1 fusion may be rare.

Larotrectinib was evaluated across 15 tumor types in 159 patients with TRK fusion cancer, and 121 (79%, 95% CI 72–85) of 153 patients had an objective response [16]. Of the 153 patients, 24 (16%) had a complete response, 97 (63%) had a partial response, 19 (12%) had stable disease, and nine (6%) had progressive disease [16]. Furthermore, Waguespack et al. reported the results of pooled data from three phase I/II larotrectinib clinical trials [10]. This study included 29 patients with TRK-fusion-positive thyroid cancer who were treated with larotrectinib. The histology was PTC in 20 (69%) patients, FTC in two (7%), and ATC in seven (24%), and the objective response rate was 71% among 28 evaluable patients [10]. The clinical responses were as follows: 2 (7%) had a complete response, 18 (64%) had a partial response, 4 (14%) had stable disease, and 3 (11%) had progressive disease [10]. The median response was 1.87 months [10]. Treatment-related adverse events (AEs) were reported in 26 patients (90%), the most common being myalgia, fatigue, dizziness, and elevated liver transaminase levels. Most treatment-related AEs were grade 1 or 2 [10]. In our case, the time to response was approximately one month. Furthermore, grade 1 myalgia, which occurred two weeks after the initiation of larotrectinib, was the only adverse effect. Therefore, larotrectinib treatment was effective and treatment-related AEs were tolerable.

To our knowledge, only three case reports of larotrectinib treatment for thyroid cancer have been published. Pitoia reported the case of a PTC patient with ETV6-NTRK3 gene fusion who had previously been treated with sorafenib and lenvatinib [17]. Surprisingly, the patient achieved a durable (sustained for 11 months) complete response after 2 months of larotrectinib treatment and a complete intracranial response in metastatic brain lesions after 7 months of larotrectinib treatment [17]. Saliba et al. reported a case of secretory carcinoma in a patient with ETV6–NTRK3 fusion [18]. Larotrectinib was administered for lung and neck metastases, and a durable response lasting 18 months was achieved [18]. Bargas et al. reported a case of PTC with SQSTM1–NTRK gene fusion [19]. Although the patient had hemoptysis and respiratory distress syndrome with marked dyspnea, all respiratory and thoracic symptoms disappeared two weeks after the initiation of larotrectinib [19]. In addition, the first CT evaluation after 2.5 months confirmed a partial response [19]. Despite our patient's history of mTKI treatment, larotrectinib was effective for distant metastases, similar to two previous case reports of patients with PTC. Therefore, larotrectinib is considered an effective treatment option even for patients with a history of mTKI treatment.

Distant metastases from differentiated thyroid carcinoma (DTC) mainly involve the lungs and bones, and metastases to other sites, such as the liver, are extremely rare [6]. Yoon et al. investigated 38,772 DTC patients and revealed that liver metastasis was observed in only five patients [6]. Although treatment outcomes have not been fully investigated among thyroid cancer patients with liver metastasis, TKIs may be beneficial [20, 21]. In our case, liver metastases were reduced by larotrectinib, which is a good treatment option for patients with TC and liver metastases.

Although the companion diagnostic test for larotrectinib is the FoundationOne CDx (Foundation Medicine Inc., Cambridge, MA, USA), the OncoGuide™ NCC Oncopanel System was performed in our case because the remaining liver biopsy specimens were small. The OncoGuide™ NCC Oncopanel System can generally be performed using fewer specimens than the FoundationOne CDx. In general, it is recommended that a specimen obtained within 3 years be used for genetic testing [22]. In this case, we performed effective genetic testing using fresh specimens.

In our case, the Tg levels decreased immediately after the initiation of larotrectinib. However, the patient was positive for TgAb, which might have affected the Tg levels. In general, an increase or reappearance of TgAb during follow-up is highly suggestive of recurrence or persistence [23]. In contrast, TgAb levels were reported to be correlated with the clinical response to lenvatinib [24]. The mechanism by which the TgAb level was positive after larotrectinib treatment is unknown. However, the TgAb level gradually decreased and liver metastases continued to shrink. Further studies are needed to investigate whether this response is specific to larotrectinib.

Conclusion

In our case, larotrectinib demonstrated effective antitumor activity against liver metastases of PTC, a relatively rare site of distant metastasis. Furthermore, the efficacy of larotrectinib was maintained, even though the patient had a history of mTKI treatment and a relatively infrequent fusion gene, SQSTM1–NTRK1.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Abbreviations

AE:

Adverse event

ALT:

Alanine aminotransferase

AST:

Aspartate aminotransferase

ATC:

Anaplastic thyroid carcinoma

BUN:

Blood urea nitrogen

CGM:

Cancer genomic medicine

CGP:

Comprehensive genomic profiling

Cre:

Creatinine

CT:

Computed tomography

DTC:

Differentiated thyroid carcinoma

FTC:

Follicular thyroid carcinoma

F-T4:

Free thyroxine

F-T3:

Free triiodothyronine

HCC:

Hurthle cell carcinoma

MTC:

Medullary thyroid carcinoma

NTRK:

Neurotrophic-tropomyosin receptor kinase

PTC:

Papillary thyroid carcinoma

PDTC:

Poorly differentiated thyroid carcinoma

RAI:

Radioactive iodine

Tg:

Thyroglobulin

TgAb:

Thyroglobulin antibody

TKI:

Tyrosine kinase inhibitor

TSH:

Thyroid-stimulating hormone

References

  1. Megwalu UC, Moon PK. Thyroid cancer incidence and mortality. Trends in the United States: 2000–2018. Thyroid. 2022;32:560–70.

    Article  CAS  PubMed  Google Scholar 

  2. Yamazaki H, Masudo K, Rino Y. Migration of risk classification between the JAES versus ATA guidelines for papillary thyroid carcinoma. World J Surg. 2023;47:1729–37.

    Article  PubMed  Google Scholar 

  3. Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet. 2014;384:319–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med. 2015;372:621–30.

    Article  PubMed  Google Scholar 

  5. Yamazaki H, Iwasaki H, Masudo K, Toda S, Matsui A, Rino Y. Prognostic significance of lung metastasis-related finding in lenvatinib treatment for differentiated thyroid cancer. Endocrine. 2022;78:543–51.

    Article  CAS  PubMed  Google Scholar 

  6. Yoon JH, Jeon MJ, Kim M, Hong AR, Kim HK, Shin DY, et al. Unusual metastases from differentiated thyroid cancers: a multicenter study in Korea. PLoS ONE. 2022;15: e0238207.

    Article  Google Scholar 

  7. Tsimberidou AM, Iskander NG, Hong DS, Wheler JJ, Falchook GS, Fu S, et al. Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res. 2012;18:6373–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kohno T, Kato M, Kohsaka S, Sudo T, Tamai I, Shiraishi Y, et al. C-CAT: the national datacenter for cancer genomic medicine in Japan. Cancer Discov. 2022;12:2509–15.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Laetsch TW, DuBois SG, Mascarenhas L, Turpin B, Federman N, Albert CM, et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol. 2018;19:705–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Waguespack SG, Drilon A, Lin JJ, Brose MS, McDermott R, Almubarak M, et al. Efficacy and safety of larotrectinib in patients with TRK fusion-positive thyroid carcinoma. Eur J Endocrinol. 2022;186:631–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47.

    Article  CAS  PubMed  Google Scholar 

  12. Pekova B, Sykorova V, Mastnikova K, Vaclavikova E, Moravcova J, Vlcek P, et al. NTRK fusion genes in thyroid carcinomas: clinicopathological characteristics and their impacts on prognosis. Cancers (Basel). 2021;13:1932.

    Article  CAS  PubMed  Google Scholar 

  13. Vaishnavi A, Le AT, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov. 2015;5:25–34.

    Article  CAS  PubMed  Google Scholar 

  14. Koehler VF, Achterfeld J, Sandner N, Koch C, Wiegmann JP, Ivanyi P, et al. NTRK fusion events and targeted treatment of advanced radioiodine refractory thyroid cancer. J Cancer Res Clin Oncol. 2023;149:14035–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chu YH, Dias-Santagata D, Farahani AA, Boyraz B, Faquin WC, Nosé V, et al. Clinicopathologic and molecular characterization of NTRK-rearranged thyroid carcinoma (NRTC). Mod Pathol. 2020;33:2186–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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. Lancet Oncol. 2020;21:531–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Pitoia F. Complete response to larotrectinib treatment in a patient with papillary thyroid cancer harboring an ETV6-NTRK3 gene fusion. Clin Case Rep. 2021;9:1905–12.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Saliba M, Mohanty AS, Ho AL, Drilon A, Dogan S. Secretory carcinoma of the thyroid in a 49-year-old man treated with larotrectinib: protracted clinical course of disease despite the high-grade histologic features. Head Neck Pathol. 2022;16:612–20.

    Article  PubMed  Google Scholar 

  19. Bargas S, Mc Leer A, Mondet J, Chabre O, Laramas M. An impressive response with larotrectinib in a patient with a papillary thyroid carcinoma harboring an SQSTM1-NTRK1 fusion. Eur J Endocrinol. 2022;186:K5–8.

    Article  PubMed  Google Scholar 

  20. Saito Y, Sugino K, Takami H, Matsuzu K, Uruno T, Ohkuwa K, et al. Clinical status and treatment of liver metastasis of differentiated thyroid cancer using tyrosine kinase inhibitors. World J Surg. 2018;42:3632–7.

    Article  PubMed  Google Scholar 

  21. Madani A, Jozaghi Y, Tabah R, How J, Mitmaker E. Rare metastases of well-differentiated thyroid cancers: a systematic review. Ann Surg Oncol. 2015;22:460–6.

    Article  PubMed  Google Scholar 

  22. Jennings LJ, Arcila ME, Corless C, Kamel-Reid S, Lubin IM, Pfeifer J, et al. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the association for molecular pathology and college of American pathologists. J Mol Diagn. 2017;19:341–65.

    Article  PubMed  Google Scholar 

  23. Kim WG, Yoon JH, Kim WB, Kim TY, Kim EY, Kim JM, et al. Change of serum antithyroglobulin antibody levels is useful for prediction of clinical recurrence in thyroglobulin-negative patients with differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2008;93:4683–9.

    Article  CAS  PubMed  Google Scholar 

  24. Sgrò D, Rossi P, Piaggi P, Brancatella A, Lorusso L, Bottici V, et al. Significance of thyroglobulin autoantibodies in patients with thyroid cancer treated with lenvatinib. J Endocr Soc. 2023;7:bvad084.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Japan Medical Communication (https://www.japan-mc.co.jp/) for editing this manuscript.

Funding

The authors received no funding in this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

HY contributed in writing the paper. MS participated in the patient management. AS supervised the manuscript. All authors discussed the results and contributed to the final manuscript.

Corresponding author

Correspondence to Haruhiko Yamazaki.

Ethics declarations

Ethics approval and consent to participate

Written informed consent was obtained from the patient for publication of this case report and any accompanying images. A copy of the written consent is available for review by the Editor-in-Chief of this journal.

Consent for publication

Consent for publications was obtained from the participant.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yamazaki, H., Sugimori, M. & Saito, A. Larotrectinib efficacy for liver metastases in papillary thyroid carcinoma patient harboring SQSTM1–NTRK1 fusion. surg case rep 10, 171 (2024). https://doi.org/10.1186/s40792-024-01971-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40792-024-01971-1

Keywords