Skip to main content

Physiopathology, Diagnosis, and Treatment of Secondary Hypothyroidism

  • Living reference work entry
  • First Online:
Hypothalamic-Pituitary Diseases

Part of the book series: Endocrinology ((ENDOCR))

  • 728 Accesses

Abstract

Secondary (or central) hypothyroidism (CH) is characterized by decreased thyroid hormone synthesis due to absent or insufficient thyroid stimulation by pituitary thyrotropin (TSH). The disease results from anatomical and/or functional disorder of the pituitary and/or the hypothalamus. The thyroid morphology and potential of hormone synthesis are usually normal, and the thyroid insufficiency is caused by dysfunction of the upper parts of the hypothalamus-pituitary-thyroid axis.

Rarely is CH an isolated insufficiency of thyrotropic cells and, in most cases, it is a part of combined (or multiple) pituitary hormone deficiency (CPHD).

The typical biochemical test results in CH include low free thyroxine (FT4) with decreased, low normal, or simply normal serum TSH concentration. Congenital CH (CoCH) is an important diagnostic challenge since routine screening for thyroid function, usually including TSH level only, can give misleading conclusion of normal thyroid function or even of hyperthyroidism. In most cases of CoCH, the genetic basis of hypothalamic and/or pituitary pathology is undefined. Among the already known molecular causes of CoCH, rarely occurring isolated TSH deficiency is a result of mutations in genes which control TSH synthesis (TSHβ, TRHR, TBL1X). Much more often CoCH is a part of CPHD resulting from mutations in one of several transcription factors important for the entire pituitary development, such as HESX1, LHX3, LHX4, SOX2, SOX3, OTX2, GATA2, ISL1, PITX1/2, PROP1, or POU1F1.

Acquired CH or CPHD of various extent and severity can be caused by intrasellar or extrasellar tumors, with pituitary adenomas and craniopharyngiomas being the most common ones. Surgical treatment of such tumors, as well as head irradiation, may lead to CH – either isolated or coexisting with additional pituitary deficiencies. Other important potential causes of CH include brain injuries, pituitary apoplexy, empty sella syndrome, parasellar aneurysm, subarachnoid hemorrhage, lymphocytic hypophysitis, and infiltrative or infectious diseases.

In every case of CH, detailed investigation of secretion of other pituitary hormones is obligatory, as additional anterior pituitary hormone deficiencies may be present in the majority of patients. Magnetic resonance imaging of the pituitary and the hypothalamus is mandatory in every case when CH is confirmed or suspected.

Treatment of CH is challenging as TSH cannot guide levothyroxine (L-T4) dose adjustment. Similarly to primary hypothyroidism, oral daily dose of L-T4 is a treatment of choice in CH. Before starting L-T4 administration, concomitant secondary adrenal insufficiency in the course of CPHD should always be excluded. If present, secondary adrenal insufficiency should be substituted before L-T4 treatment to avoid exacerbation of cortisol deficiency, which may even precipitate adrenal crisis.

The target level of FT4 during treatment should be maintained within the upper half of the normal range. Majority of CH patients require on average 1.5–1.6 μg L-T4/kg body weight daily. Older patients need lower doses of L-T4 than the young (approximately 1.1 μg L-T4/kg body weight daily). In infants and young children with CH, higher doses of L-T4 are required comparing to adults. It is recommended to start treatment in neonates with 10–15 μg/kg body weight daily and the dose should be decreased as the child gets older.

Concomitant treatment of CPHD significantly influences L-T4 requirement, and the dose should be re-established when estrogens, androgens, or recombinant human growth hormone (rhGH) are introduced or in the patients in whom hydrocortisone therapy is introduced or modified during L-T4 treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Institutional subscriptions

Similar content being viewed by others

References

  • Asakura Y, Abe K, Muroya K, Hanakawa J, Oto Y, Narumi S, Hasegawa T, Adachi M. Combined growth hormone and thyroid-stimulating hormone deficiency in a Japanese patient with a novel frameshift mutation in IGSF1. Horm Res Paediatr. 2015;84:349–54.

    Article  CAS  PubMed  Google Scholar 

  • Biondi B, Wartofsky L. Treatment with thyroid hormone. Endocr Rev. 2014;35:433–512.

    Article  CAS  PubMed  Google Scholar 

  • Bonomi M, Busnelli M, Beck-Peccoz P, Costanzo D, Antonica F, Dolci C, Pilotta A, Buzi F, Persani L. A family with complete resistance to thyrotropin-releasing hormone. N Engl J Med. 2009;360:731–4.

    Article  CAS  PubMed  Google Scholar 

  • Collu R, Tang J, Castagné J, Lagacé G, Masson N, Huot C, Deal C, Delvin E, Faccenda E, Eidne KA, Van Vliet GA. Novel mechanism for isolated central hypothyroidism: inactivating mutations in the thyrotropin-releasing hormone receptor gene. J Clin Endocrinol Metab. 1997;82:1561–5.

    CAS  PubMed  Google Scholar 

  • Garber JR, Cobin RH, Gharib H, Hennessey JV, Klein I, Mechanick JI, Pessah-Pollack R, Singer PA, Woeber KA. American association of clinical endocrinologists and American Thyroid Association taskforce on hypothyroidism in adults. Clinical practice guidelines for hypothyroidism in adults: cosponsored by the American Association of Clinical Endocrinologists and the American Thyroid Association. Thyroid. 2012;22:1200–35.

    Article  CAS  PubMed  Google Scholar 

  • Heinen CA, Losekoot M, Sun Y, Watson PJ, Fairall L, Joustra SD, Zwaveling-Soonawala N, Oostdijk W, van den Akker EL, Alders M, Santen GW, van Rijn RR, Dreschler WA, Surovtseva OV, Biermasz NR, Hennekam RC, Wit JM, Schwabe JW, Boelen A, Fliers E, van Trotsenburg ASP. Mutations in TBL1X are associated with central hypothyroidism. J Clin Endocrinol Metab. 2016;101(12):4564–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Hermanns P, Couch R, Leonard N, Klotz C, Pohlenz J. A novel deletion in the thyrotropin Beta-subunit gene identified by array comparative genomic hybridization analysis causes central congenital hypothyroidism in a boy originating from Turkey. Horm Res Paediatr. 2014;82: 201–5.

    Article  CAS  PubMed  Google Scholar 

  • Jonklaas J, Bianco AC, Bauer AJ, Burman KD, Cappola AR, Celi FS, Cooper DS, Kim BW, Peeters RP, Rosenthal MS, Sawka AM. American Thyroid Association task force on thyroid hormone replacement. Guidelines for the treatment of hypothyroidism: prepared by the American thyroid association task force on thyroid hormone replacement. Thyroid. 2014;24: 1670–751.

    Article  PubMed  PubMed Central  Google Scholar 

  • Koulouri O, Nicholas AK, Schoenmakers E, Mokrosinski J, Lane F, Cole T, Kirk J, Farooqi IS, Chatterjee VK, Gurnell M, Schoenmakers N. A novel thyrotropin-releasing hormone receptor missense mutation (P81R) in central congenital hypothyroidism. J Clin Endocrinol Metab. 2016;101:847–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Léger J, Olivieri A, Donaldson M, Torresani T, Krude H, van Vliet G, Polak M, Butler G. ESPE-PES-SLEP-JSPE-APEG-APPES-ISPAE; congenital hypothyroidism consensus conference group. European society for Paediatric Endocrinology consensus guidelines on screening, diagnosis, and management of congenital hypothyroidism. Horm Res Paediatr. 2014;81: 80–103.

    Article  PubMed  Google Scholar 

  • Persani L. Clinical review: central hypothyroidism: pathogenic, diagnostic, and therapeutic challenges. J Clin Endocrinol Metab. 2012;97:3068–78.

    Article  CAS  PubMed  Google Scholar 

  • Persani L, Bonomi M, Radin R, Beck-Peccoz P. Diagnostic and therapeutic challenges of acquired thyrotropic deficiency. Ann Endocrinol (Paris). 2012;73:138–40.

    Article  Google Scholar 

  • Schoenmakers N, Alatzoglou KS, Chatterjee VK, Dattani MT. Recent advances in central congenital hypothyroidism. J Endocrinol. 2015;227:R51–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Tenenbaum-Rakover Y, Turgeon MO, London S, Hermanns P, Pohlenz J, Bernard DJ, Bercovich D. Familial central hypothyroidism caused by a novel IGSF1 gene mutation. Thyroid. 2016;26(12):1693–700.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Andrzej Lewiński .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG

About this entry

Cite this entry

Lewiński, A., Stasiak, M. (2018). Physiopathology, Diagnosis, and Treatment of Secondary Hypothyroidism. In: Casanueva, F., Ghigo, E. (eds) Hypothalamic-Pituitary Diseases. Endocrinology. Springer, Cham. https://doi.org/10.1007/978-3-319-38681-2_7-1

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-38681-2_7-1

  • Received:

  • Accepted:

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-38681-2

  • Online ISBN: 978-3-319-38681-2

  • eBook Packages: Springer Reference MedicineReference Module Medicine

Publish with us

Policies and ethics