Home            Past Issues            About IP            About IAP           Author Information            Subscription            Advertisement              Search  

   
clinical case letter

Indian Pediatr 2020;57: 474-475

Spontaneous Resolution of Congenital Hyperinsulinism With Octreotide Therapy


Suresh Chandran1*, Wai Lin Tun2, Phyo Thandar Htay2 and Khalid Hussain3

1Departments of Neonatology, KK Womens and Childrens Hospital, Singapore; 2Children’s Hospital, Mandalay, Myanmar and 3Pediatric Endocrinology, Sidra Medicine, Doha, Qatar.
Email: [email protected]
 


Hyperinsulinemic hypoglycemia is caused by dysregulated insulin secretion from the pancreatic b-cells. Congenital hyperinsulinism (CHI) is caused by genetic mutations in twelve known genes. Histologically, lesions can be focal or diffuse. Focal forms are often associated with paternal heterozygosity in KCNJ11/ABCC8 genes, whereas diffuse forms are seen in patients with maternal heterozygous, homozygous or compound heterozygous mutations. 18F-DOPA PET/CT imaging can precisely localize the lesion in focal forms, thereby facilitating cure by focal lesionectomy unlike diffuse form is mostly resistant to medical treatment and needs subtotal pancreatectomy [1].

We report the case of a term non-dysmorphic male baby (weight 2400 g) born to a non-consanguineous couple in Myanmar. He was born through meconium stained liquor with low Apgar scores, required resuscitation and was ventilated for ten days. Hypoglycemia (1.6 mmol/L) was noted at six hours of age, which required mini bolus followed by glucose infusion rate of 5.6 mg/kg/min. On day six, he developed seizures with hypoglycemia and GIR was gradually escalated to 19.5 mg/kg/min. Diagnosis of hyperinsulinemic hypoglycemia was made in the presence of detectable insulin (10.7 mU/L) with hypoglycemia (0.3 mmol/L) and hypoketonemia (0.3 mmol/L). Medical treatment was initiated with nifedipine while awaiting supply of diazoxide. Diazoxide was initiated at a dose of 5 mg/kg/day and was gradually increased to 15 mg/kg/day over a week with discontinuation of nifedipine. Subcutaneous octreotide (dose of 7.5 mcg/kg/day) was added as GIR continued to rise on diazoxide. With adequate response to octreotide, diazoxide was later discontinued.

DNA samples of the proband and parents were sent to UK for genetic study. A novel heterozygous KCNJ11 missense variant, c.866G>C p. (Gly289Ala) was identified in the proband. Sequencing of the ABCC8 gene was normal. Sanger sequencing of KCNJ11 gene for the familial variant indicated heterozygous mutation in father whereas the mother was negative. The clinical significance of the P. (Gly289Ala) variant is uncertain. A focal lesion was suspected with the paternal mutation and 18F-DOPA PET/CT scan was recommended.

DOPA PET/CT scan was unavailable in Myanmar and there was no funding source for overseas transfer. Treatment with octreotide was continued and GIR was successfully weaned off with feeding increments to achieve full feeds by six months of age. At nine months of age, octreotide dose was auto-tapered to 3 mcg/kg/day while maintaining normoglycemia and discontinued at 9.5 months of age. His glucose profile remained stable on follow-up but neurodevelopmental assessment at 22 months of age showed moderate mental and motor retardation. Vision and hearing tested normal. He is currently enrolled in an early intervention programme.

CHI is a heterogeneous disease caused by mutations in at least twelve known genes [1]. Loss-of-function mutations in KATP channel regulating genes constitute nearly 90% of cases of diazoxide-unresponsive CHI, of which KCNJ11 is associated in 10% [2].

The index case had diazoxide-unresponsive CHI that detected a novel paternally inherited KCNJ11 missense variant of uncertain significance at p. (Gly289Ala). A different missense variant at the same residue was previously reported by Mohnike, et al. [2] in a patient with diazoxide-responsive CHI, which was shown to have arisen de novo in the proband.

Similar spontaneous resolution has been reported at 1.6 and 1.9 year in patients with CHI [3,4]. DOPA tracer uptake may not correlate with the capacity of the pancreatic lesion to secrete insulin and the clinical remission of CHI could be a functional process without apoptosis of mutated b-cells [5]. This finding prompts long-term follow-up of our case to ensure optimal glucose regulation.

Most patients with KATP channel gene mutations do not respond to diazoxide treatment as it exerts its effects by keeping the channel open, preventing b-cell membrane depolarization and release of insulin. Octreotide reduces insulin secretion by inhibiting intracellular entry of calcium and by decreasing the insulin gene promoter activity [6]. These differences in the site action possibly explain the treatment response in the index case.

In summary, normoglycemia should be maintained to prevent brain injury with high GIR and/or high caloric enteral feeds in infants with CHI. Octreotide can be tried in diazoxide unresponsive patients and spontaneous resolution can be seen in CHI. Genetic studies help indicate the type of mutation. DOPA-PET scan confirms nature of lesion prior to surgery, which however remain poorly accessible in resource-limited settings.

Acknowledgements: Dr Sara E Flanagan PhD, Sir Henry Dale Fellow, Genetic laboratory services, University of Exeter Medical School, United Kingdom for the genetic study and assistance with the preparation of the manuscript. Dr Eddy Saputra Leman, PhD, Sr Scientific Editor, Duke-NUS Medical School, Singapore and Prof Divakaran Liginlal (Carnegie Mellon University, Pittsburgh, Pennsylvania) for editing the manuscript.

Contributors: SC: clinical management, reviewed and edited the manuscript; WLT and PTH: patient management, outline of the case report and discussion; KH: contributed to the genetic section of the case report and reviewed and finalized the manuscript.

Funding: None; Competing interest: None stated.

REFERENCES

1. Galcheva S, Demirbilek H, Al-Khawaga S, Hussain K. The genetic and molecular mechanisms of congenital hyper-insulinism. Front Endocrinol. 2019;10:111.

2. Mohnike K, Weiland L, Barthlem W, Vogelgesang S, Empting S, Mohnike W, et al. Clinical and genetic evaluation of patients with KATP channel mutations from the German registry for congenital hyperinsulism. Horm Res Paediatr. 2014;81:156-68.

3. Suchi M, Mac Mullen CM, Thornton PS, Adzick NS, Ganguly A, Ruchelli ED, et al. Molecular and immunohistochemical analyses of the focal form of congenital hyperinsulinism. Mod Pathol. 2006;19:122-9.

4. Snider KE, Becker S, Bayajian L, Shyng SL, MacMullan C, Hughes N, et al. Genotype and phenotype correlatios in 417 children with congenital hyperinsulinism. J Clin Endocrinol Metob. 2013;98:E355-63.

5. Yorifuji T1, Hosokawa Y, Fujimaru R, Kawakita R, Doi H, Matsumoto T, et al. Lasting 18F-DOPA PET uptake after clinical remission of the focal form of congenital hyperinsulinism. Horm Res Paediatr. 2011;76:286-90.

6. Katz MD, Erstad BL. Octreotide, a new somatostatin analogue. Clin Pharm. 1989;8:255-73.

 

Copyright © 1999-2020 Indian Pediatrics