Managing limitations of the LMA Classic laryngeal mask as a conduit for tracheal intubation in impending paediatric airway obstruction: a case report

© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Managing limitations of the LMA Classic laryngeal mask as a conduit for tracheal intubation in impending paediatric airway obstruction: a case report
Gareth CH Cheng, MB, ChB; Jaclyn WM Wong, FHKCA, FHKAM (Anaesthesiology)
Department of Anaesthesia and Operating Theatre Services, Kwong Wah Hospital, Hong Kong
 
Corresponding author: Dr Gareth CH Cheng (gareth@fellow.hkam.hk)
 
 Full paper in PDF
 
Case report
Peritonsillar abscess (quinsy) is the most common deep neck infection in children and adolescents, although it is less frequently seen in young children.1 Airway compromise is a feared complication. We describe the successful management of a patient with impending paediatric airway obstruction. The LMA Classic™ laryngeal mask (Teleflex Medical Ltd., Co. Westmeath, Ireland) was the only paediatric-sized supraglottic device available in our operating theatre and was modified to work around its limitations as a conduit for tracheal intubation.
 
In March 2020, an 18-month-old girl with good past health weighing 9.5 kg was admitted to our district general hospital 8 days after onset of fever up to 38.9°C, with worsening left facial swelling, inspiratory stridor during sleep, drooling and poor feeding.
 
On examination, she was pink and calm when carried upright. Her throat was swollen with bilateral grade 3 tonsils and the left cheek and submandibular region were grossly swollen (~10 cm × 6 cm). Contrast computed tomography of the neck revealed a 3.3-cm × 4.8-cm × 3.8-cm peritonsillar abscess with rightward deviation of the upper airway and significant narrowing at the larynx, with the narrowest cross-section measuring 3.5 mm (Fig 1).
 

Figure 1. Computed tomography films showing critical airway narrowing at the larynx (arrowhead) due to obstruction by peritonsillar abscess (arrow)
 
In view of the critical airway diameter and the impending progression to complete airway obstruction, and after discussion with the on-call head and neck surgeons and the patient’s mother, the decision was made to proceed with emergency incision and drainage under general anaesthesia.
 
The sizing of all our airway equipment was checked in advance. The distal aperture bars of a #1.5 LMA Classic laryngeal mask were cut to facilitate use as an intubation conduit.
 
The patient had a 24G intravenous cannula in situ on arrival in theatre. Surgeons were ready at the bedside with front-of-neck access equipment on standby. Hong Kong College of Anaesthesiologists standard monitoring was applied. Gaseous induction with spontaneous ventilation was performed using an Ayres’ T-piece and 4% sevoflurane in 100% oxygen. After an adequate depth of anaesthesia was achieved as assessed by vital parameters, intubation using video laryngoscopy was attempted. Two initial attempts were unsuccessful, with very rapid desaturation down to an SpO2 of 50% to 60%. Subsequent attempts at rescue bag mask ventilation failed despite optimisation by positioning and oropharyngeal airway. Fortunately, a final attempt to insert the laryngeal mask was successful and ventilation was maintained.
 
As a definitive airway was still necessary, a 2.2-mm fibreoptic bronchoscope was passed through the laryngeal mask and a 3.5-mm Microcuff endotracheal tube (ETT) carefully railroaded over the bronchoscope into the trachea. Correct positioning was confirmed by bilateral chest auscultation and capnography. We deemed it unsafe to leave the laryngeal mask in place as traction could risk ETT dislodgement postoperatively. The video laryngoscope was used to visualise the distal end of the ETT near the vocal cords and it was secured with Magill forceps. The laryngeal mask was slowly pulled out over the ETT, but the 15-mm connector of the laryngeal mask could not be passed over the ETT pilot balloon even when deflated. Hence, we carefully cut down the laryngeal mask with scissors, leaving only the end with the 15-mm connector still attached (Fig 2).
 

Figure 2. Endotracheal tube cuff ‘stuck’ on 15-mm connector (arrow); remnants of cut down LMA Classic laryngeal mask with scissor incision line seen (arrowhead)
 
The operation proceeded uneventfully. Subsequently the patient was transferred to the paediatric intensive care unit and kept intubated and sedated. She was discharged after a short hospital stay, with good recovery on clinic follow-up examination.
 
Discussion
Paediatric airway emergencies are rare and among the most challenging crises faced by anaesthetists. Unique paediatric considerations such as inability to cooperate during preoxygenation, intolerance to awake techniques and difficult front-of-neck access, in addition to inherent differences in paediatric physiology, significantly reduce the safe apnoeic time and greatly increase the risk of hypoxia.
 
Ideally, this case would have been managed in a specialist tertiary centre with a wider selection of equipment and expertise; unfortunately, our requests for night-time case transfer were denied. The availability and use of an intubating laryngeal mask with a larger-sized inner channel such as an air-Q™ (Salter Labs, Lake Forest [IL], United States) would have been preferable to an LMA Classic. This would have made it easier for us to remove the laryngeal mask over the ETT after intubation without having to improvise with our limited equipment in an already stressful situation. Although the Association of Paediatric Anaesthetists guidelines2 recommend leaving the laryngeal mask in place after intubation, we deemed it necessary to remove it since continued postoperative ventilation was anticipated.
 
Often, manufacturer-recommended ETT/laryngeal mask combinations take account only of the ability to pass the ETT through the laryngeal mask. They may not consider the ability to remove the laryngeal mask over the ETT when the diameter of the ETT pilot balloon cuff exceeds that of the outer diameter of the ETT. This problem was illustrated in an article by Kleine-Brueggeney et al3 who found that only the air-Q models of supraglottic airways were able to be removed over the ETT with all manufacturer-recommended size combinations, as the air-Q had the largest inner channel diameter among other equivalently sized supraglottic airways.
 
Alternatively, we could have chosen to cut off the pilot balloon and repair the cuff with an angiocatheter.4 Another option might have been to use an uncuffed tube, although it would not have been ideal in our case due to the possibility of postoperative blood or secretions tracking down and contaminating the lower airway. Furthermore, exchange of a poorly fitting uncuffed tube would have been difficult and potentially dangerous in our situation, as the airway could easily have been lost.
 
In conclusion, in addition to meticulous airway planning, testing combinations of ETT and laryngeal mask for both insertion and subsequent removal is important to avoid complications during airway management. Availability of an intubating laryngeal mask such as an air-Q may be particularly advantageous in such cases.
 
Author contributions
Concept or design: GCH Cheng.
Acquisition of data: GCH Cheng.
Analysis or interpretation of data: GCH Cheng.
Drafting of the manuscript: Both authors.
Critical revision of the manuscript for important intellectual content: GCH Cheng.
 
Both authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
Both authors have disclosed no conflicts of interest.
 
Funding/support
This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics approval
This case report was published with the written consent of the patient’s mother.
 
References
1. Ungkanont K, Yellon RF, Weissman JL, Casselbrant ML, González-Valdepeña H, Bluestone CD. Head and neck space infections in infants and children. Otolarngol Head Neck Sur 1995;112:375-82. Crossref
2. Association of Paediatric Anaesthetists. Unanticipated difficult tracheal intubation during routine induction of anaesthesia in a child aged 1 to 8 years. Available from: https://www.das.uk.com/files/APA2-UnantDiffTracInt-FINAL.pdf. Accessed 21 Jan 2021.
3. Kleine-Brueggeney M, Kotarlic M, Theiler L, Greif R. Limitations of pediatric supraglottic airway devices as conduits for intubation—an in vitro study. Can J Anaesth 2018;65:14-22. Crossref
4. Kovatsis PG, Fiadjoe JE, Stricker PA. Simple, reliable replacement of pilot balloons for a variety of clinical situations. Paediatric Anaesth 2010;20:490-4. Crossref

Intravenous iron isomaltoside (Monofer)–induced hypophosphataemia: a case report

© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Intravenous iron isomaltoside (Monofer)–induced hypophosphataemia: a case report
KY Wong, MRCP, FHKAM (Medicine); KY Yu, MRCP, FHKAM (Medicine); Maria WH Mak, MRCP, FHKAM (Medicine); KM Lee, MRCP, FHKAM (Medicine); KF Lee, FRCP, FHKAM (Medicine)
Department of Medicine and Geriatrics, Kwong Wah Hospital, Hong Kong
 
Corresponding author: Dr KY Wong (wky697@ha.org.hk)
 
 Full paper in PDF
 
Case report
In January 2019, an 85-year-old woman with a history of osteoporosis and collapsed L1 had gastric antral vascular ectasia with multiple failed attempts of argon photo-coagulation, resulting in severe iron deficiency anaemia (haemoglobin 4 g/dL). The patient had been repeatedly admitted for congestive heart failure precipitated by anaemia that required blood transfusion. In view of her severe and ongoing blood loss, intravenous iron isomaltoside (Monofer) 800 mg monthly was started in February 2019. Before commencement of iron isomaltoside, iron saturation was 5% (normal 15%-50%). She was optimally nourished with normal serum calcium (2.27 mmol/L; normal 2.15-2.55 mmol/L), phosphate (1.4 mmol/L; normal 0.8-1.5 mmol/L), alkaline phosphatase (108 IU/L; normal 53-141 IU/L) and vitamin D level (79 nmol/L; normal 50-220 nmol/L). Hypophosphataemia (0.5 mmol/L) was first noted in June 2019 (Fig). Owing to the patient’s history of collapsed vertebra, she was given a first dose of denosumab in October 2019 by a private orthopaedic surgeon but serum phosphate further worsened to 0.1 mmol/L despite aggressive oral phosphate replacement. She refused hospital admission at this time. In December 2019, the patient was hospitalised for anaemia and hypophosphataemia (0.4 mmol/L) with concomitant serum calcium 2.07 mmol/L and alkaline phosphatase 199 IU/L. Her estimated glomerular filtration rate was >90 mL/min/1.73 m2. Fractional excretion of phosphate confirmed renal phosphate wasting (FePO4 14%; normal <5%) and bicarbonate was 30 mmol/L (normal 22-26 mmol/L). Urinary protein and glucose were negative. Iron saturation was 24% and parathyroid hormone 22.6 nmol/L (normal 1.6-7.2 nmol/L). Fibroblast growth factor 23 (FGF23), measured 3 weeks after the last dose of iron isomaltoside, was 155 IU/mL (normal <188 IU/mL). Intravenous iron-induced hypophosphataemia was suspected. Iron isomaltoside was stopped and rocaltrol was commenced in January 2020 with prompt improvement in phosphate level. Another intravenous iron preparation, iron sucrose (Venofer), was started due to her severe anaemia. Attempted re-challenge with iron isomaltoside resulted in recurrent hypophosphataemia. Rocaltrol and phosphate sandoz were gradually tapered down over 6 months. Serum phosphate remained normal while on iron sucrose and denosumab.
 

Figure. Changes in serum calcium (Ca), phosphate (PO4) and alkaline phosphatase (ALP) levels before and after administration of intravenous iron and denosumab
 
Discussion
Iron deficiency anaemia is a commonly encountered problem in daily practice. Although oral iron remains the recommended route of replacement due to its low cost and availability, intravenous iron is considered superior in several respects. First, the gastrointestinal side-effects of oral iron are avoided. Second, bioavailability is improved where that of oral iron is reduced in conditions such as achlorhydria (eg, proton pump inhibitors, gastric bypass), small bowel malabsorption (eg, inflammatory bowel disease, prior small bowel resection, celiac disease) and chronic inflammation (via upregulation of hepcidin). Third, intravenous iron allows rapid repletion of iron, making it a more suitable choice when there is severe and/or ongoing blood loss.
 
The new-generation intravenous iron preparations are all stable iron-carbohydrate complexes. The three commonly used intravenous iron preparations locally are iron carboxymaltose (Ferinject), iron isomaltoside (Monofer) and iron sucrose (Venofer). They differ in the attaching carbohydrate ligands that affect the capacity, stability and immunogenicity of the complex. Hypophosphataemia is a well-described complication of iron carboxymaltose but is far less common in the other two preparations: the incidence1 of hypophosphataemia is 58%, 4% and 1% for patients with preserved renal function given iron carboxymaltose, iron isomaltoside and iron sucrose, respectively. In addition, iron carboxymaltose–induced hypophosphataemia can be severe and protracted resulting in osteomalacia and multiple fractures. Although the pathogenesis2 is not fully understood, it is believed to be mediated through iron carboxymaltose–induced production of biologically active intact FGF23. The FGF23 is a phosphaturic hormone produced by osteocytes and osteoblasts. It reduces phosphate reabsorption by downregulation of sodium-phosphate co-transporter in the proximal renal tubule. The FGF23 also inhibits 1,25-dihydroxyvitamin D synthesis, leading to vitamin D deficiency and secondary hyperparathyroidism that contribute to reduced intestinal phosphate uptake and further increased renal phosphate wasting, respectively. Iron isomaltoside also increases intact FGF23 secretion, but to a much lesser extent than iron carboxymaltose.3 Again, such difference is speculated to be due to the carbohydrate ligand, since iron carboxymaltose and iron isomaltoside are equally effective in replenishing iron store. Although other indicators of proximal renal tubular dysfunction such as fractional excretion of urate and urine amino acid were not measured in this patient, the absence of glycosuria and non-suppressed FGF23 level were not typical of a diagnosis of renal Fanconi syndrome. In addition, improved serum phosphate level after stopping iron isomaltoside supported the diagnosis of iron-induced phosphaturia in this patient.
 
In the meta-analysis by Schaefer et al,4 low baseline iron saturation and normal renal function were identified as positive predictors of iron-induced hypophosphataemia; both were present in this patient. Severe iron deficiency may cause a higher increase in FGF23 transcription and renal impairment is protective due to intrinsic kidney resistance to FGF23. Furthermore, denosumab may have worsened the pre-existing hypophosphataemia induced by iron isomaltoside in this patient since serum phosphate level fell abruptly in October 2019, 1 week after denosumab injection. Denosumab is an anti-RANKL (receptor activator of nuclear factor-κB ligand) antibody that inhibits osteoclastic activity and hence bone resorption. Severe hypophosphataemia caused by denosumab has been described in a patient with tenofovir-induced osteomalacia5 and is possibly mediated through the following two mechanisms: first, decreased bone resorption directly reduces phosphate release; second, fall in bone-derived calcium worsens secondary hyperparathyroidism that in turn enhances phosphaturia. These may explain the profound hypophosphataemia in this patient after denosumab injection.
 
There are several reasons for the normal FGF23 level in this patient. First, the commercial FGF23 assay detects both intact and cleaved FGF23; an increased intact FGF23 together with a reduced cleaved FGF23 may result in a “normal” FGF23 level. Second, FGF23 may have dropped significantly 3 weeks after the last dose of iron isomaltoside. Third, FGF23 transcription was reduced with correction of iron deficiency as reflected by the normal iron saturation.
 
In addition to phosphate replacement and switching to a less phosphaturic intravenous iron preparation, activated vitamin D is needed to increase phosphate reabsorption during the initial phase, even in a patient with preserved renal function. This is because FGF23 inhibits renal activation of 25-dihydroxyvitamin D to its active form, 1,25-dihydroxyvitamin D.
 
In conclusion, although hypophosphataemia is much less common in patients on iron isomaltoside than iron carboxymaltose, it is advisable to monitor phosphate level 1 to 2 weeks after iron isomaltoside injection in high-risk patients, such as those with severe anaemia who require repeat dosing, and patients with pre-existing vitamin D deficiency and/or hyperparathyroidism.
 
Author contributions
All authors contributed to the concept, acquisition of data, interpretation of data, drafting of the manuscript, and critical revision for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
The authors have no conflicts of interest to disclose.
 
Funding/support
This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics approval
The patient was treated in accordance with the Declaration of Helsinki. The patient provided informed consent for the treatment/procedures and publication.
 
References
1. Zoller H, Schaefer B, Glodny B. Iron-induced hypophosphatemia: an emerging complication. Curr Opin Nephrol Hypertens 2017;26:266-75. Crossref
2. Edmonston D, Wolf M. FGF23 at the crossroads of phosphate, iron economy and erythropoiesis. Nat Rev Nephrol 2020;16:7-19. Crossref
3. Wolf M, Rubin J, Achebe M, et al. Effects of iron isomaltoside vs ferric carboxymaltose on hypophosphatemia in iron-deficiency anemia: two randomized clinical trials. JAMA 2020;323:432-43. Crossref
4. Schaefer B, Tobiasch M, Viveiros A, et al. Hypophosphataemia after treatment of iron deficiency with intravenous ferric carboxymaltose or iron isomaltoside—a systematic review and meta-analysis. Br J Clin Pharmacol 2021;87:2256-73. Crossref
5. Chung TL, Chen NC, Chen CL. Severe hypophosphatemia induced by denosumab in a patient with osteomalacia and tenofovir disoproxil fumarate-related acquired Fanconi syndrome. Osteoporos Int 2019;30:519-23. Crossref

Primary pulmonary mucosa-associated lymphoid tissue lymphoma with radiological presentation of middle lobe syndrome diagnosed by bronchoscopy: a case report

© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Primary pulmonary mucosa-associated lymphoid tissue lymphoma with radiological presentation of middle lobe syndrome diagnosed by bronchoscopy: a case report
H Zhou, MD1; Z Yang, MD2; S Wu, MD1
1 Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), PR China
2 Cancer Center, Department of Pathology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), PR China
 
Corresponding author: Dr S Wu (wushengchang@126.com)
 
 Full paper in PDF
 
Case report
In March 2018, a 49-year-old woman was admitted to our respiratory medicine ward with a 2-year history of recurrent productive cough. Symptoms first developed after the patient underwent laparoscopic myomectomy and subsequently developed a fever (around 38°C). Chest computed tomography (CT) revealed patchy opacity in the right middle lobe (Fig 1a and b). Based on these symptoms, she was diagnosed with “pneumonia” for which antibiotics were prescribed. Her fever resolved but cough persisted so she was prescribed cough suppressants and antibiotics intermittently. These therapies were likewise ineffective.
 

Figure 1. Chest computed tomography (CT) scan showing patchy opacity in the right middle lobe during onset. (a) Lung window. (b) Mediastinal window. Chest CT scan showing patchy opacity persisting in the same location 2 years later. (c) Lung window. (d) Mediastinal window
 
A series of tests and examinations conducted during hospitalisation revealed no definitive cause and symptoms again persisted. A new chest CT scan revealed no change to the right middle lobe opacity compared with previous CT scan and stenosis of the middle bronchus (Fig 1c and d).
 
In the absence of any treatment response, the patient was advised to undergo bronchoscopy that revealed a narrowed lumen and rough mucosa of the middle bronchus. Pathological examination of mucosal tissue biopsy from the lesion revealed infiltration of large numbers of lymphocytes (Fig 2a and b) suggestive of diagnosis of lymphoma. Further immunohistochemistry examination confirmed that the lesion was caused by mucosa-associated lymphoid tissue (MALT) lymphoma. These lymphoma cells tested positive for CD20, CD79a and CD43 (Fig 2c-f). Further examinations including positron emission tomography–computed tomography and gastroscopy revealed no evidence of lymphoma elsewhere.
 

Figure 2. Bronchoscopic and pathological presentation of lesion in right middle lobe. (a) Bronchoscopic examination showing stenosis of the middle bronchus and nodular hyperplasia in the submucosa. (b) Hyperaemia and swelling change to the mucosa of the middle lobar bronchus. (c) Pathological examination suggested large numbers of lymphoma cells infiltrating the lesion. Immunohistochemical examination showing lymphoma cells were positive for (d) CD20, (e) CD43 and (f) CD79a
 
The patient underwent radiotherapy about 6 months after final diagnosis, with a total dose of 3960 cGy. After the last radiotherapy treatment, chest CT confirmed that the lesion in the right middle lobe had nearly disappeared. In addition, symptoms of cough and expectoration had improved significantly. The patient was followed up once every 3 months. Chest CT scan at the most recent follow-up examination indicated no sign of relapse and her symptoms had resolved.
 
Discussion
Primary pulmonary lymphoma belongs to a group of lymphoproliferative diseases, and accounts for 0.5% to 1% of all pulmonary tumours.1 Mucosa-associated lymphoid tissue lymphoma is the most common pathological type of primary pulmonary lymphoma.2 However, the disease may not be diagnosed until pathological examinations are conducted due to a lack of clinical and radiological characteristics.
 
Previous study suggests that about half of patients with MALT lymphoma are asymptomatic and some are diagnosed by accident.3 In patients with symptoms, respiratory symptoms are more frequently observed than B symptoms, including fever, fatigue and weight loss.3 Our patient presented with a transient B symptom of fever, but a chronic respiratory symptom of cough.
 
The radiological presentation of this disease is diverse and includes nodules, masses, consolidation and group glass opacity. Multiple lesions, which are often bilaterally distributed, can be detected by CT scan in most cases, but a solitary lesion is less common in patients with MALT lymphoma.4 In addition, MALT lesions may occur in any lung lobe or large airway including the trachea and main bronchus.4 5 In our patient, the right middle lobe was affected, and middle lobe syndrome (MLS) was the major radiological feature.
 
Middle lobe syndrome was first mentioned several decades ago by Graham6 to describe atelectasis of the right middle lobe. The syndrome is usually caused by extrinsic compression or intrinsic stricture of the middle lobe bronchus.6 However, there is no uniform definition of this term. Broadly speaking, MLS is defined as damage to the right middle lobe due to any cause. The most common causes are bronchiectasis, non-specific inflammation, tuberculosis and tumours. Lung cancer is the most common type of tumour affecting the middle lobe. As a haematological disease, MALT lymphoma can affect any organ or tissue although in our patient, it affected only the right middle lobe with consequent misdiagnosis for more than 2 years. It is particularly rare for MALT lymphoma to present as MLS. Only one similar case has been reported: in 2004, Toishi et al7 reported a 70-year-old male patient with a radiological change of MLS who was diagnosed with MALT lymphoma by right middle lobectomy. In our case, the final diagnosis was obtained by biopsy during bronchoscopy, not surgery.
 
Given that the clinical and radiological features of MALT are non-specific, its definitive diagnosis relies on pathology. Diffuse infiltration of small lymphocytes into the bronchiolar mucosa is a major histological characteristic. Moreover, reactive lymphoid follicles and lymphoepithelial lesions are commonly observed by microscopy.4 The molecular markers of MALT are CD20, CD79a, CD43, as observed in our case. CD5, CD10 and cyclin D are always negative and may help to discriminate other types of lymphoma. It was difficult to distinguish this disease from infectious diseases and benign lymphoproliferative diseases in the absence of pathological examination.
 
Generally, MALT lymphoma is a tumour of low-grade malignancy and patients have a rather good prognosis. The 5-year survival rate for patients with MALT lymphoma is >80%.4 The main treatments for the disease are chemotherapy, radiotherapy, target therapy and immunotherapy. Radiotherapy is the first choice in patients with localised lesions. It has been estimated that >90% of patients can achieve a complete response after radiotherapy.8 Surgery may be indicated when the lesion is isolated in certain organs, such as lung, thyroid gland, and spleen. However, if MALT lymphoma progresses to an advanced stage, or patients have a high tumour burden, systemic therapy is recommended. Anti-CD20 combined with chemotherapy is considered first-line treatment.7 In the present case, the patient refused chemotherapy and agreed to radiotherapy that relieved her symptoms with no obvious adverse effects.
 
In summary, we first report MALT lymphoma as a rare cause of MLS that was confirmed using a minimally invasive approach. Mucosa-associated lymphoid tissue lymphoma should be considered a differential diagnosis when investigating the aetiology of MLS.
 
Author contributions
Concept or design: H Zhou, S Wu.
Acquisition of data: Z Yang, S Wu.
Analysis or interpretation of data: H Zhou, Z Yang.
Drafting of the manuscript: H Zhou, S Wu.
Critical revision of the manuscript for important intellectual content: All authors.
 
All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
All authors have disclosed no conflicts of interest.
 
Ethics approval
The patient was treated in accordance with the Declaration of Helsinki. The patient provided written informed consent for all treatment and procedures and for publication of this paper.
 
References
1. Cardenas-Garcia J, Talwar A, Shah R, Fein A. Update in primary pulmonary lymphomas. Curr Opin Pulm Med 2015;21:333-7. Crossref
2. Borie R, Wislez M, Antoine M, Copie-Bergman C, Thieblemont C, Cadranel J. Pulmonary mucosa-associated lymphoid tissue lymphoma revisited. Eur Respir J 2016;47:1244-60. Crossref
3. Zhang MC, Zhou M, Song Q, et al. Clinical features and outcomes of pulmonary lymphoma: a single center experience of 180 cases. Lung Cancer 2019;132:39-44. Crossref
4. Sirajuddin A, Raparia K, Lewis VA, et al. Primary pulmonary lymphoid lesions: radiologic and pathologic findings. Radiographics 2016;36:53-70. Crossref
5. Kawaguchi T, Himeji D, Kawano N, Shimao Y, Marutsuka K. Endobronchial mucosa-associated lymphoid tissue lymphoma: a report of two cases and a review of the literature. Intern Med 2018;57:2233-6. Crossref
6. Graham EA, Burford TH, Mayer JH. Middle lobe syndrome. Postgrad Med 1948;4:29-34. Crossref
7. Toishi M, Miyazawa M, Takahashi K, et al. Mucosa-associated lymphoid tissue lymphoma; report of two cases [in Japanese]. Kyobu Geka 2004;57:75-9.
8. Lumish M, Falchi L, Imber BS, Scordo M, von Keudell G, Joffe E. How we treat mature B-cell neoplasms (indolent B-cell lymphomas). J Hematol Oncol 2021;14:5. Crossref

Primary omental pregnancy after intrauterine insemination: a case report

© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Primary omental pregnancy after intrauterine insemination: a case report
Tony PL Yuen, MB, BS1; Winnie Hui, MRCOG, FHKAM (Obstetrics and Gynaecology)1; MK Ho, MB, BS1; Richard WC Wong, FRCPA, FHKAM (Pathology)2
1 Department of Obstetrics and Gynaecology, Pamela Youde Nethersole Eastern Hospital, Hong Kong
2 Department of Clinical Pathology, Pamela Youde Nethersole Eastern Hospital, Hong Kong
 
Corresponding author: Dr Tony PL Yuen (ypl634@ha.org.hk)
 
 Full paper in PDF
 
Introduction
Ectopic pregnancy (EP), a condition in which a fertilised ovum does not implant in the endometrial cavity, occurs in 1% to 2% of all pregnancies.1 Up to 97% of EPs occur within the fallopian tube, but implantation can also occur at locations such as the cervix, ovary, uterine cornua and abdomen. Abdominal EPs are extremely rare, making up less than 1% of EPs.1 Their presentation can be non-specific and they are classified as primary or secondary abdominal pregnancies. We present a case of primary omental pregnancy with laparoscopy and omentectomy performed.
 
Case summary
In January 2020, a 33-year-old gravida 1 para 0 woman was admitted to our gynaecology unit with right-sided abdominal pain. The patient’s past health was good and she had no history of gynaecological surgery, sexually transmitted disease or pelvic inflammatory disease. She had been treated in the private sector 3 weeks before to admission with ovulation induction and subsequent intrauterine insemination (IUI) for coital problems. Serum beta-human chorionic gonadotropin (HCG) was 48 mIU/L on day 18 and 768 mIU/L on day 22 after IUI. Ultrasound of the pelvis at 5 weeks of gestation showed no intrauterine sac. She also complained of mild per-vaginal bleeding on admission. Abdominal examination revealed tenderness over the right abdomen, next to the umbilicus. Transvaginal ultrasound of the pelvis on admission showed a linear endometrial lining, with no adnexal masses or pelvic free fluid identified. Blood tests showed a haemoglobin level of 12 g/dL and beta-HCG level of 1366 mIU/mL. Diagnostic laparoscopy was offered to the patient in view of her abdominal pain but she opted for beta-HCG monitoring as she was worried about a negative laparoscopy. She subsequently complained of severe right abdominal pain about 6 hours after admission. Repeat transvaginal ultrasound revealed no adnexal masses but a moderate amount of free fluid in the pouch of Douglas. Due to the increased abdominal pain and suspicion of a ruptured EP, the patient agreed to undergo laparoscopy.
 
Laparoscopy showed haemoperitoneum of 200 mL and a normal uterus, bilateral fallopian tubes and ovaries. Survey of the peritoneal cavity revealed a 5 × 5 cm haematoma attached to the omentum at the right hepatic flexure, with mild oozing from the site of attachment (Fig 1). The rest of the abdomen was unremarkable. General surgeons were consulted and omentectomy (including the site of bleeding) was performed.
 

Figure 1. Laparoscopic view of the omental ectopic pregnancy at the omentum, inferior to the liver
 
The patient made an uneventful postoperative recovery and haemoglobin was stable. She was discharged on day 4 after surgery. Pathological examination revealed products of gestation mixed with inflammatory and reactive mesothelial cells (Fig 2). Beta-HCG monitoring after surgery showed a satisfactory drop to a non-pregnant level: 366 mIU/mL, 144 mIU/mL, and 1.7 mIU/mL on days 2, 4, and 18 after surgery.
 

Figure 2. Histological findings in the omental resection specimen. (a) Syncytiotrophoblasts (arrows) are present in fibrin exudate over inflamed omental adipose tissue. (b) Intermediate trophoblasts (arrowheads) infiltrate the fibrofatty stroma of the omentum, consistent with omental pregnancy
 
Discussion
Among EPs, abdominal pregnancy is most rare. They have been classified as either primary or secondary. Our case meets the criteria established by Studdiford2 for a primary abdominal pregnancy: normal, bilateral fallopian tubes and ovaries with no recent or remote injury; absence of any uteroperitoneal fistula; and presence of a pregnancy related exclusively to the peritoneal surface and diagnosed early enough to exclude the possibility of secondary implantation after primary nidation elsewhere.
 
Early preoperative diagnosis of an abdominal EP is very difficult in many cases. A systematic review by Poole et al3 showed that among patients with a final diagnosis of omental EP, none had a preoperative diagnosis of abdominal pregnancy. As a result of the diagnostic difficulty, there is usually a delay from presentation to definitive treatment with some cases requiring diagnosis by serial HCG monitoring supplemented with magnetic resonance imaging. A high level of vigilance is therefore vital when monitoring the symptoms and vital signs of a suspected case, and early surgical intervention should be considered if there is clinical deterioration. In our case, we elected to perform emergent laparoscopy in view of increased abdominal pain and free fluid in the pouch of Douglas.
 
Laparotomy with excision of the embryo has been the classic management for abdominal pregnancy.4 However, with its widespread availability, laparoscopy should be the modality of choice, especially when the patient is haemodynamically stable, as in our case, and the required expertise is available. Laparoscopic management is associated with fewer morbidities, reduced intraoperative blood loss and a shorter hospital stay. The importance of a general peritoneal survey is paramount; in cases of normal fallopian tubes and ovaries, extra care must be taken not to miss an EP elsewhere in the peritoneum and prematurely commit to a negative laparoscopy. If a difficult resection is encountered, the expertise of a general surgeon will be of benefit. Alternatives to surgical treatment have also been reported,3 such as intralesional methotrexate, intramuscular methotrexate, intracardiac potassium chloride injection and artery embolisation. However, the prerequisites for non-surgical treatment include reliable imaging and for the patient to be haemodynamically stable.
 
Assisted reproductive techniques are known to be associated with an increased risk of EP. Some reports state an incidence of up to 4.5% with assisted reproductive technology compared with a spontaneous pregnancy.5 With regard to IUI, the incidence of EP is reported to be 2.05% compared with 3.33% for in vitro fertilisation. A higher risk of EP is also associated with stimulated cycles (compared with natural cycles: 2.62% vs 0.99%) and use of husband sperm (compared with donor sperm: 3.54% vs 1.08%). Many postulations have been made regarding the mechanism of an abdominal EP.3 As ovarian induction was performed in this case, the risk of EP was increased. In the setting of IUI, it is possible that the fertilised embryo develops as a primary tubal pregnancy that subsequently passes through the fimbrial end and implants into the omentum.
 
Although omental EPs are extremely rare, and in our case, the first of such a condition found after IUI, clinical suspicion must be high in a patient who presents with symptoms suggestive of EP but with normal uterus and adnexa during intraoperative exploration. Clinicians should always be vigilant with regard to the patient’s clinical condition, and there should be a low threshold for surgical intervention if clinical deterioration is noted. In addition, with the rising application of assisted reproductive technology, the risk of EPs, and by extension the risk of abdominal EPs, is also increased, making the diagnosis and treatment of this potentially life-threatening condition evermore challenging.
 
Author contributions
All authors contributed to the design of the report, acquisition of data, drafting of the manuscript and critical revision for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
The authors have no conflicts of interest to disclose.
 
Funding/support
This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics approval
The patient was treated in accordance with the Declaration of Helsinki. Informed consent was obtained for all treatment involved as well as for publication of this article and accompanying images.
 
References
1. Fylstra DL. Ectopic pregnancy not within the (distal) fallopian tube: etiology, diagnosis, and treatment. Am J Obstet Gynecol 2012;206:289-99. Crossref
2. Studdiford WE. Primary peritoneal pregnancy. Am J Obstet Gynecol 1942;44:487-91. Crossref
3. Poole A, Haas D, Magann EF. Early abdominal ectopic pregnancies: a systematic review of the literature. Gynecol Obstet Invest 2012;74:249-60. Crossref
4. Yip SL, Tan WK, Tan LK. Primary omental pregnancy. BMJ Case Rep 2016;2016: bcr2016217327. Crossref
5. Bu Z, Xiong Y, Wang K, Sun Y. Risk factors for ectopic pregnancy in assisted reproductive technology: a 6-year, single-center study. Fertil Steril 2016;106:90-4. Crossref

Cold agglutinin–mediated autoimmune haemolytic anaemia associated with COVID-19 infection: a case report

Hong Kong Med J 2022 Jun;28(3):257–9  |  Epub 27 Apr 2022
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Cold agglutinin–mediated autoimmune haemolytic anaemia associated with COVID-19 infection: a case report
CY Chang, MRCP (Medicine); HH Chin, MRCP (Medicine)2; PW Chin, MMed (Medicine)2; M Zaid, MMed (Medicine)1
1 Department of Medicine, Hospital Sultanah Aminah, Johor Bahru, Johor, Malaysia
2 Department of Medicine, Hospital Enche’ Besar Hajjah Kalsom, Kluang, Johor, Malaysia
 
Corresponding author: Dr CY Chang (ccyik28@gmail.com)
 
 Full paper in PDF
 
 
Case report
In November 2020, a 70-year-old woman with diabetes mellitus, hypertension, and dyslipidaemia presented with a 3-day history of fever, cough, and rhinorrhoea. She reported no chest pain, shortness of breath, anosmia, or ageusia. Physical examination revealed that she was awake and not tachypneic. There was mild pallor present, but no jaundice. The patient’s blood pressure was 118/56 mm Hg, pulse rate 62 beats per minute, and temperature 36.5°C. Respiratory rate was 16 breaths per minute and pulse oximetry revealed oxygen saturation of 98% on ambient air. Chest auscultation revealed bibasilar crackles. There were no signs of lymphadenopathy, splenomegaly, or autoimmune disease. Physical examination was otherwise unremarkable.
 
Haematological analysis revealed haemoglobin 8.1 g/dL, white cell count 9.6 × 109/L (absolute lymphocyte count 3.1 × 109/L) and platelet count 346 × 109/L. The peripheral blood film showed moderate anaemia with occasional spherocytes and marked red blood cell agglutination that dispersed when blood was heated to 37°C, indicating cold agglutinin (Fig). The absolute reticulocyte count was raised at 2.3% and direct antiglobulin test showed presence of anti-complement (C3d) antibodies but not anti-immunoglobulin G antibodies. Due to a lack of facilities at the district hospital, we were unable to conduct the following tests: serum haptoglobin, direct antiglobulin test performed with warm-washed red blood cells, cold agglutinin titre, and thermal amplitude testing. Mild hyperbilirubinaemia was present, with indirect bilirubin predominating (total bilirubin 26.2 mol/L, direct bilirubin 4.7 mol/L, indirect bilirubin 21.5 mol/L). Liver transaminases and renal profile were within the normal range. C-reactive protein, serum ferritin, and serum lactate dehydrogenase level was 5 mg/L, 2671 ?g/L, and 321 U/L, respectively. Mycoplasma serology, blood cultures, D-dimer, and autoimmune screening were all negative, as were tests for hepatitis B, hepatitis C, and human immunodeficiency virus.
 

Figure. (a) Peripheral blood smear prior to ‘pre-warm’ method. (b) Peripheral blood smear after the application of ‘pre-warm’ method. The smears show a leucoerythroblastic picture (dashed arrows). There are extensive red cell agglutinations (black arrows) in (a) that dispersed on warming of blood to 37°C. Occasional spherocytes are seen in (b) [arrowhead]. No abnormal lymphoid cells are present
 
Chest radiograph showed ground-glass opacities in both lower zones. Coronavirus disease 2019 (COVID-19) infection was confirmed by reverse transcriptase-polymerase chain reaction for detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in nasopharyngeal and oropharyngeal swab samples (Ct value; E gene 16.09, RdRp gene 19.23). A diagnosis of cold agglutinin—mediated autoimmune haemolytic anaemia (AIHA) due to SARS-CoV-2 was made. On the seventh day of her illness, she developed hypoxaemic respiratory failure, necessitating 3 L/min supplemental oxygen administered via nasal cannula. At the time, inflammatory markers were elevated, and a new chest radiograph revealed worsening bilateral airspace opacities. The patient was prescribed intravenous methylprednisolone 500 mg as a single dose, followed by 2 mg/kg once daily for the next 5 days. She responded well and oxygen supplementation was discontinued 7 days later. Blood inflammatory marker levels (C-reactive protein 3.1 mg/L) and chest radiograph showed improved findings. The patient was prescribed a tapering dose of dexamethasone. One unit of packed cells was transfused on the third, fifth, tenth, and fourteenth day of hospitalisation due to ongoing low-grade haemolysis. In the absence of any constitutional symptoms, and no lymphadenopathy or organomegaly on physical examination, a computed tomography scan was not performed. She was discharged home on day 21 of her illness after her symptoms had resolved and she had been transfusion-independent with stable haemoglobin level for 1 week. At 1-month follow-up examination, the patient remained well: haemoglobin was 10 g/L and new peripheral blood film examination found no cold agglutinin haemolysis.
 
Discussion
This pandemic has taken the world by storm, with many new undocumented symptoms and treatment strategies. An increasing number of COVID-19-related complications involving various disciplines, particularly haematology, are being reported. Coronavirus disease 2019 is associated with prominent haematopoietic system manifestations, including leukopenia, lymphopenia, thrombocytopenia, disseminated intravascular coagulation, and prothrombotic state.1 An association between AIHA and COVID-19 infection has nonetheless been reported infrequently. The pathophysiology of this association is poorly understood with few cases reported worldwide.
 
Cold agglutinin disease (CAD) is a form of AIHA mediated by cold agglutinins that can agglutinate red blood cells at a temperature of 3°C to 4°C, resulting in complement-mediated haemolysis. Cold agglutinins arise from either primary (unknown) or secondary (when cold agglutinins are produced as a result of an underlying infection or haematological malignancy) conditions.2 The pathogenesis of CAD as a result of infectious agents is unclear. It may be the result of complement system activation, and associated with an inflammatory state, including the upregulation of pro-inflammatory cytokines.
 
In this case, our patient fulfilled the diagnostic criteria for CAD that include haemolytic anaemia, reticulocytosis, elevated lactate dehydrogenase, hyperbilirubinaemia, positive anti-C3d antibodies, and negative anti-immunoglobulin G antibodies.3 Other infections and autoimmune diseases were excluded, and no signs of malignancy were discovered. We concluded that the CAD in this case was caused by SARS-CoV-2 (COVID-19). Because of the ongoing haemolysis, our patient required packed cell transfusions on multiple occasions. We believe that her condition deteriorated due to the “cytokine storm” and complement cascade, necessitating oxygen supplementation and blood product transfusion.
 
Lazarian et al4 reported seven cases of AIHA (four cases of warm AIHA and three cases of cold AIHA) associated with COVID-19 infection. Extensive investigations into the three cases of cold AIHA revealed the presence of underlying malignancies (marginal zone lymphoma, 2 cases; prostate cancer, 1 case). No malignancy was evident in our patient. Patil et al5 reported a case of COVID-19 infection with AIHA and pulmonary embolism, and Maslov et al6 reported a patient with COVID-19 infection and cold agglutinin haemolytic anaemia complicated by stroke and bilateral upper extremity venous thrombosis. Our patient showed no signs of thromboembolism. Although patients infected with COVID-19 are at increased risk of thromboembolic complications, AIHA/CAD should be considered as a possible contributory factor.
 
Treatment of CAD is not recommended in patients who are asymptomatic with mild anaemia or compensated haemolysis and corticosteroids should not be used to treat CAD.7 However, in our patient, the use of methylprednisolone was indicated as treatment for severe COVID-19 pneumonia. Corticosteroid administration has been proposed to reduce the systemic inflammatory response that leads to lung injury and multiorgan failure in COVID-19. Prompt administration of methylprednisolone has been shown to significantly reduce mortality rate and ventilator dependence.8 The improvement of haemolysis in our patient coincided with a favourable treatment response of COVID-19 to corticosteroid. This was reflected in her need for fewer packed cell transfusions, as well as stabilisation of her haemoglobin and no need for blood transfusions for one week prior to discharge. Rituximab has also been used to treat COVID-19-associated AIHA in two reported cases following corticosteroid failure and marginal zone lymphoma, respectively.4 More research is needed to assess the safety and efficacy of these therapies in the treatment of COVID-19-associated AIHA.
 
Author contributions
Concept or design: CY Chang.
Acquisition of data: CY Chang, HH Chin.
Analysis or interpretation of data: All authors.
Drafting of the manuscript: CY Chang, HH Chin.
Critical revision of the manuscript for important intellectual content: All authors.
 
All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
All authors have disclosed no conflicts of interest.
 
Acknowledgement
The authors thank the Director General of Health Malaysia for his permission to publish this article.
 
Funding/support
This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics approval
The patients were treated in accordance with the tenets of the Declaration of Helsinki. The patient(s) provided written informed consent for all treatments and procedures and for publication of this case report.
 
References
1. Terpos E, Ntanasis-Stathopoulos I, Elalamy I, et al. Hematological findings and complications of COVID-19. Am J Hematol 2020;95:834-47. Crossref
2. Berentsen S. New insights in the pathogenesis and therapy of cold agglutinin-mediated autoimmune hemolytic anemia. Front Immunol 2020;11:590. Crossref
3. Swiecicki PL, Hegerova LT, Gertz MA. Cold agglutinin disease. Blood 2013;122:1114-21. Crossref
4. Lazarian G, Quinquenel A, Bellal M, et al. Autoimmune haemolytic anaemia associated with COVID-19 infection. Br J Haematol 2020;190:29-31. Crossref
5. Patil NR, Herc ES, Girgis M. Cold agglutinin disease and autoimmune hemolytic anemia with pulmonary embolism as a presentation of COVID-19 infection. Hematol Oncol Stem Cell Ther 2020:S1658-3876(20)30116-3. Crossref
6. Maslov DV, Simenson V, Jain S, Badari A. COVID-19 and cold agglutinin hemolytic anomie. TH Open 2020;4:e175-7. Crossref
7. Berentsen S. How I treat cold agglutinin disease. Blood 2021;137:1295-303. Crossref
8. Salton F, Confalonieri P, Meduri GU, et al. Prolonged low-dose methylprednisolone in patients with severe COVID-19 pneumonia. Open Forum Infect Dis 2020;7:ofaa421. Crossref

Fulminant necrotising amoebic colitis: a report of two cases

Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Fulminant necrotising amoebic colitis: a report of two cases
LM Tam, MB, BS1; KC Ng, MB, BS, FRCS1; CH Man, MB, BS FRCS1; FY Cheng, MB, BS2; Y Gao, LMCHK2
1 Department of Surgery, Caritas Medical Centre, Hong Kong
2 Department of Pathology, Caritas Medical Centre, Hong Kong
 
Corresponding author: Dr LM Tam (tammy520@connect.hku.hk)
 
 Full paper in PDF
 
Case report
Case 1
In January 2019, a 31-year-old man with a history of amphetamine abuse presented with a 10-day history of watery diarrhoea and abdominal pain. On admission, he had a fever of 38.1°C and tachycardia but normal blood pressure. Abdominal examination revealed tenderness and guarding over the lower abdomen. The patient was resuscitated and intravenous antibiotics prescribed since an infective cause was considered most likely. Abdominal plain radiograph showed grossly dilated small and especially large bowel (diameter up to 9 cm). White cell count was 22×109/L (neutrophil differential count 19×109/L), liver and renal function were unremarkable. Contrast computed tomography (CT) scan of the abdomen and pelvis revealed extensive colitis, suggesting pseudomembranous colitis or diffuse colitis. Stool culture for Clostridium difficile was negative and stool microscopy revealed no ova or cysts. Human immunodeficiency virus (HIV), hepatitis B and C virus serologies were non-reactive. He was treated as pseudomembranous colitis by the medical gastrointestinal team and commenced on oral vancomycin. Later sigmoidoscopy revealed inflamed mucosa with multiple ulcers, especially at the sigmoid colon (Fig 1). Biopsies were taken. The patient’s condition deteriorated and he developed septic shock. A new contrast CT scan showed pneumoperitoneum. Emergency laparotomy was performed and revealed generalised faecal peritonitis with the whole colon necrosed and communicating with the peritoneal cavity. Debridement of necrotic tissue and multiple segmental resections of bowel with end ileostomy were performed in stages. The diagnosis of fulminant amoebic colitis (FAC) was made on histopathological evaluation of the biopsy and resection specimen (Fig 2). Antibiotics were switched to metronidazole accordingly. The patient’s condition was later complicated by an intra-abdominal fluid collection and image guided drainage was performed. He was initially nursed in the intensive care unit and then transferred to a surgical ward for rehabilitation. He was discharged from hospital 4 months later.
 

Figure 1. Endoscopy photos showing severely inflamed mucosa in Case 1
 

Figure 2. Histological examinations from Case 1. (a) Perforated ulcer. Haematoxylin and eosin (H&E) stain ×100. (b) Amoebae labelled with arrows in the lamina propria. H&E stain ×100
 
Case 2
In February 2020, a 61-year-old man presented with a ≥1-week history of diarrhoea, abdominal pain, high fever of 39°C and tachycardia with normal blood pressure. Abdominal examination showed diffuse tenderness, but without peritoneal signs. The patient’s medical history was otherwise good, and initial investigations including blood tests and plain radiographs were all unremarkable. In view of the progressive abdominal distension, urgent contrast CT was arranged and showed a suspected perforated caecum. Emergency surgery found ischaemic colon extending from the caecum to mid sigmoid, with perforations over the proximal transverse colon and hepatic flexure. Subtotal colectomy with end ileostomy was performed. He was transferred to the intensive care unit after surgery and required vasopressor and continuous venovenous haemofiltration due to severe sepsis. He showed a good response and was weaned off vasopressor support on postoperative day 4. Pathology of the surgical specimen later confirmed amoebic colitis with extensive ulcer and perforation (Fig 3). There were no features of atherosclerosis, vasculitis, thromboembolism, or inflammatory bowel disease. Microscopic results were also available after surgery. Stool culture for C difficile, stool microscopy for amoeba, stool microscopy for ova or cysts, stool polymerase chain reaction for virus, and HIV serology test results were all negative, but Entamoeba histolytica serology test was positive. He was prescribed metronidazole for 14 days and then oral diloxanide furoate for 10 days as suggested by the microbiologist. The patient’s recovery was later complicated by wound dehiscence and abdominal cocoon. Repeat surgery for debridement was performed and skin closure changed to an ABTHERA temporary abdominal closure system. The patient recovered gradually and was transferred to a rehabilitation unit 3 months after surgery.
 

Figure 3. Histological examinations from Case 2. (a) Presence of Entamoeba histolytica, stained purple due to glycogen content, in necrotic ulcer debris. Periodic acid-Schiff ×200. (b) Deep penetrating ulcer, with extensive necrosis, down to the muscularis propria. H&E stain ×10
 
Discussion
Entamoeba histolytica is a protozoan parasite and the cause of amoebiasis in humans.1 Early diagnosis and treatment are essential to avoid progression to fulminant colitis. Amoebic dysentery is classified as a notifiable disease in Hong Kong. Prevalence of amoebiasis is higher in developing countries such as India, Mexico, and parts of Central and South America,2 mainly related to poor socioeconomic and sanitary conditions. In developed countries, where faecal-oral transmission is unusual, amoebiasis is more often seen in immigrants from or individuals with a travel history to endemic areas. Other groups at risk include male homosexuals3 with or without HIV, infants, pregnant women, and those taking immunosuppressants, especially corticosteroids.1 Neither of our two cases had a relevant travel history in the past year, and they presented several months apart, so they are unlikely to be imported cases or to have had the same source of infection. Case 1 had some risk factors for amoebiasis: he was a male homosexual living in rental housing with shared rooms. A detailed history taking from patients who present with severe diarrhoea is crucial. Prompt initiation of anti-amoebic treatment should be considered in cases where there is a high index of suspicion.
 
Although E histolytica can be cultured in vitro, this is neither routinely performed nor is it a gold standard in the diagnosis of amoebic colitis because amoebic culture is insensitive (25%). The most commonly used laboratory test is stool microscopy to identify trophozoites or cysts, although this also has low sensitivity (<60%) and specificity (10%-50%). As exemplified by our two cases, both had negative stool microscopy. In some laboratories, stool antigen detection of E histolytica might be available. However, neither stool microscopy (in the absence of ingested erythrocytes in the trophozoites) nor some antigen detection kits can distinguish between pathogenic E histolytica and commensal Entamoeba dispar. Serum antibody is commonly positive in patients with invasive amoebiasis,4 especially in endemic areas. It may be difficult to differentiate past from active infection since antibodies may persist for some time. Recent commercially available multiplex polymerase chain reaction systems offer a rapid and sensitive way of detecting E histolytica DNA in stool; however, cost and availability limit their application in most patients.
 
Another popular non-invasive investigation is different modalities of imaging, especially contrast CT scan. Some CT features specific to amoebic colitis have been reported. These include extended submucosal ulcers with intramural dissection caused by flask-shaped ulcers typical of amoebiasis and omental “wrapping” indicating adhesions with neovascularisation due to ischaemic foci of transmural amoebic colitis.5 Other non-specific findings include pancolitis with areas of target signs, discontinuous bowel necrosis and coexistence of liver abscess. None of these features were evident on CT scans in our patients. In clinical practice, CT scan is not sensitive and has a small role in diagnosing FAC. It is mainly used to distinguish the severity of colitis, looking for complications such as bowel perforation or ischaemia.
 
Sigmoidoscopy and/or colonoscopy with biopsy can also be performed as a diagnostic tool. However, there is a high risk of perforation, especially of inflamed or even ischaemic bowel. We do not recommend endoscopic investigations in cases of severe colitis or in patients with high fever.
 
Preoperative diagnosis of FAC remains a challenge and detailed history taking plays an important role in identifying high-risk cases.
 
Mild cases of amoebic colitis can be treated medically with metronidazole to control systemic invasion and diloxanide furoate, a luminal agent, in addition to eliminating luminal cysts. If the condition becomes transmural, conservative treatment is no longer appropriate. Early diagnosis and extensive surgical treatment are important to reduce morbidity and mortality.1 In both our cases, with both complicated by bowel perforation, extensive bowel resection was immediately performed.
 
Intra-operatively, skipped lesions with multiple transmural perforations of bowel were noticed. However, some gross features make differentiation from other pathology difficult, especially Crohn’s disease. In our patients, necrotic tissue was more friable with little bleeding from the necrotic bowel wall. These features are quite unique compared with other causes of bowel ischaemia. This may be related to severe necrosis with consequent poor vascular supply to the bowel. In the worst case, the necrotic bowel wall communicates with the peritoneal cavity, making bowel resection more difficult as the dissection planes may be disrupted. Therefore, extensive debridement of necrotic tissue or even staged operations are required. Special gross features of FAC are seldom mentioned in other case reports. Early identification of these features enables early commencement of empirical anti-amoebic treatment and aids the recovery of patients.
 
Author contributions
Concept or design: LM Tam, KC Ng, CH Man.
Acquisition of data: LM Tam
Analysis or interpretation of data: LM Tam, FY Cheng, Y Gao.
Drafting of the manuscript: LM Tam.
Critical revision of the manuscript for important intellectual content: KC Ng, CH Man.
 
Conflicts of interest
The authors have no conflicts of interest to disclose.
 
Funding/support
This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics approval
The patients were treated in accordance with the Declaration of Helsinki. The patients provided verbal informed consent for the treatment/procedures and consent for publication.
 
References
1. Stanley SL Jr. Amoebiasis. Lancet 2003;361:1025-34. Crossref
2. Haque R, Huston CD, Hughes M, Houpt E, Petri WA Jr. Amebiasis. N Engl J Med 2003;348:1565-73. Crossref
3. Roure S, Valerio L, Soldevila L, et al. Approach to amoebic colitis: epidemiological, clinical and diagnostic considerations in a non-endemic context (Barcelona, 2007-2017). PLoS One 2019;14:e0212791. Crossref
4. Tanyuksel M, Petri WA Jr. Laboratory diagnosis of amebiasis. Clin Microbiol Rev 2003;16:713-29. Crossref
5. Kinoo SM, Ramkelawon VV, Maharajh J, Singh B. Fulminant amoebic colitis in the era of computed tomography scan: a case report and review of the literature. SA J Radiol 2018;22:1354. Crossref

Gastric peroral endoscopic myotomy for delayed gastric conduit emptying after pharyngolaryngo-esophagectomy: a case report

Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Gastric peroral endoscopic myotomy for delayed gastric conduit emptying after pharyngo-laryngo-esophagectomy: a case report
Fion SY Chan, MB, BS, FHKAM (Surgery)1; Ian YH Wong, MB, BS, FHKAM (Surgery)1; Desmond KK Chan, MB, BS, FHKAM (Surgery)1; Claudia LY Wong, MB, BS, FHKAM (Surgery)1; Betty TT Law, MB, BS, FHKAM (Surgery)1; Velda LY Chow, MS, FHKAM (Surgery)2; Simon Law, PhD, MS1
1 Division of Esophageal and Upper Gastrointestinal Surgery, Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
2 Division of Head and Neck Surgery, Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
 
Corresponding author: Prof Simon Law (slaw@hku.hk)
 
 Full paper in PDF
 
Case report
In September 2016, a 64-year-old man with intrathoracic oesophageal cancer underwent neoadjuvant chemoradiotherapy and minimally invasive esophagectomy with no pyloroplasty. The gastric conduit was placed in the posterior mediastinum. Pathology revealed a moderately differentiated squamous cell carcinoma (ypT2N1M0) with clear margins. One year later he underwent surgery for isolated right cervical lymph node recurrence and tolerated a normal diet after surgery with no gastrointestinal symptoms. At 22 months after the second surgery, the patient developed dysphagia and a cervical oesophageal cancer was identified. Completion pharyngo-laryngo-esophagectomy (PLE) with resection of the residual cervical oesophagus, pharyngo-laryngectomy, and reconstruction with a segment of free jejunum interposed between the neopharynx and gastric conduit was performed. After surgery, the patient developed delayed gastric conduit emptying (DGCE) and reported regurgitation of undigested food soon after diet introduction. There was a persistently high nasogastric output, and non-ionic contrast study showed hold-up of contrast at the level of pylorus (Fig 1a). The patient’s symptoms persisted and he relied on nasoduodenal feeding despite prokinetic agents and pyloric balloon dilatation.
 

Figure 1. (a) Contrast study after pharyngo-laryngo-esophagectomy showing contrast hold-up at level of pylorus (yellow arrow). (b) Contrast study after gastric peroral endoscopic myotomy showing free emptying of contrast, with endoclips (red arrow) and pylorus (yellow arrow) visible
 
Gastric peroral endoscopic myotomy (G-POEM) was performed in February 2020, 4 months after completion PLE. The procedure was performed with the patient in a supine position and under general anaesthesia with endotracheal intubation via end tracheostomy. A high-definition gastroscope (GIF-H190; Olympus, Tokyo, Japan) fitted with a conical shaped transparent cap (DH-28GR; Fujifilm, Tokyo, Japan) and carbon dioxide insufflation were used. After submucosal injection of a mixture of normal saline and indigo carmine at the posterior wall of the gastric conduit, 5 cm proximal to the pylorus, a 2-cm longitudinal mucosal incision was made with DualKnife J (Olympus) using Endocut Q mode (effect 3, cut-duration 2, cut-interval 4) [VIO® 300D; Erbe, Tübingen, Germany]. The endoscope entered the submucosal space to dissect a tunnel caudally until the pyloric ring was well exposed (Fig 2a). Pyloromyotomy was performed and the circular muscle ring completely divided and flattened (Fig 2b). Haemostasis was achieved and the mucosal opening closed with repositioning clips (Single Use Hemoclip; Mednova, Zhejiang, China) [Fig 2c]. The surgery time was 120 minutes and rapid contrast passage to the duodenum was demonstrated on postoperative contrast study (Fig 1b). He resumed an oral diet thereafter.
 

Figure 2. Intra-operative photographs showing pyloric ring muscle well exposed (a) before and (b) after pyloromyotomy, and (c) mucosal incision closed with endoclips
 
Discussion
Pharyngo-laryngo-esophagectomy was first reported by Ong and Lee in 19601 and is regarded as standard treatment for hypopharyngeal and cervical oesophageal cancer. Chemoradiotherapy has gained popularity as an alternative therapeutic strategy to preserve the larynx, but salvage PLE due to incomplete response or cancer recurrence is not uncommonly required.2 Post-PLE DGCE is underreported and the incidence is unknown. Patients frequently complain of bloating, regurgitation, and poor oral intake. According to unpublished results from our prospectively collected database, DGCE was documented in five of 20 patients with PLE for cervical oesophageal cancer over the past 10 years. Of these five patients, pyloroplasty was performed in two, of whom symptoms improved with prokinetic agents alone in one, and endoscopic pyloric balloon dilation was required in the other. For those without pyloric drainage, two patients were managed by G-POEM. The remaining patient was an 83-year-old man on prolonged tube feeding who had pneumonia and died 10 weeks after the surgery.
 
The pathogenesis of DGCE after PLE may differ to that after oesophagectomy without pharyngo-laryngectomy although data are lacking. Experience in the management of DGCE after oesophagectomy (without pharyngo-laryngectomy) serves to guide treatment of post-PLE DGCE. Proposed contributing factors include gastropyloric denervation, dysfunctional gastric peristalsis and use of the whole stomach for reconstruction.3 4
 
The application of G-POEM in PLE patients has not been reported. We report a patient with prior oesophagectomy who developed DGCE only after completion PLE. Symptoms resolved after G-POEM. We postulate that removal of the upper oesophageal sphincter in PLE limits build-up of intragastric pressure, compounding DGCE. Pyloromyotomy reduces pyloric channel pressure and expedites gastric emptying, G-POEM accomplishes this as a minimally invasive method. We hypothesise that gastric conduit emptying after PLE can be viewed as a two-stage process. In the first stage, the food bolus passes passively from the proximal stomach to the antrum. In the second stage, food is evacuated from the antrum through the pylorus to the duodenum. A sufficient pressure gradient within the gastric conduit is required to overcome pyloric resistance. Resection of the pharynx and larynx results in equalisation of pressure between the gastric conduit and the atmosphere. The stomach is also exposed to negative intrathoracic pressure. The outflow resistance due to the intact pylorus assumes more importance after PLE since the paretic stomach fails to build up internal pressure. This explains why symptoms of delayed emptying in our patient emerged only after the pharyngo-laryngectomy, not after the initial oesophagectomy.
 
Pyloroplasty and pyloromyotomy have both been shown effective and safe drainage procedures for gastric conduit after oesophagectomy.5 The G-POEM disrupts the pylorus and improves gastric emptying, theoretically achieving the same outcome and serving as a salvage option for DGCE after PLE. We perform G-POEM according to the same principle applied to POEM for achalasia. The submucosal tunnel is dissected close to the muscle layer for precise pyloromyotomy. Secure mucosal closure permits early diet resumption. However, the aim of pyloromyotomy is to overcome the outlet obstruction without alleviating gastroparesis. Despite improved gastric emptying, symptoms of our patient were not completely eliminated. Patients need to make dietary adjustments to accommodate the new conduit over time while maintaining satisfactory nutrition and body weight.
 
To the best of our knowledge, this is the first report of successful management of DGCE after PLE by G-POEM. A pyloric drainage procedure is advocated since resection of the upper oesophageal sphincter, an integral part of PLE, limits pressure build-up and food emptying within the gastric conduit.
 
Author contributions
Concept or design: FSY Chan, S Law.
Acquisition of data: FSY Chan.
Analysis or interpretation of data: FSY Chan.
Drafting of the manuscript: FSY Chan.
Critical revision of the manuscript for important intellectual content: All authors.
 
All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
As an editor of the journal, VLY Chow was not involved in the peer review process. Other authors have disclosed no conflicts of interest.
 
Funding/support
This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics approval
The study was approved by the Institutional Review Board of The University of Hong Kong/Hospital Authority Hong Kong West Cluster (Ref: UW 16-2023). Consent from patient was obtained.
 
References
1. Ong GB, Lee TC. Pharyngogastric anastomosis after oesophago-pharyngectomy for carcinoma of the hypopharynx and cervical oesophagus. Br J Surg 1960;48:193-200. Crossref
2. Tong DK, Law S, Kwong DL, Wei WI, Ng RW, Wong KH. Current management of cervical esophageal cancer. World J Surg 2011;35:600-7. Crossref
3. Konradsson M, Nilsson M. Delayed emptying of the gastric conduit after esophagectomy. J Thorac Dis 2019;11:S835-44. Crossref
4. Akkerman RD, Haverkamp L, van Hillegersberg R, Ruurda JP. Surgical techniques to prevent delayed gastric emptying after esophagectomy with gastric interposition: a systematic review. Ann Thorac Surg 2014;98:1512-9. Crossref
5. Law S, Cheung MC, Fok M, Chu KM, Wong J. Pyloroplasty and pyloromyotomy in gastric replacement of the esophagus after esophagectomy: a randomized controlled trial. J Am Coll Surg 1997;184:630-6.

Pain management for painful brachial neuritis after COVID-19: a case report

Hong Kong Med J 2022 Apr;28(2):178–80  |  Epub 8 Apr 2022
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Pain management for painful brachial neuritis after COVID-19: a case report
Vivian YT Cheung, MB, BS, FHKAM (Anaesthesiology); Fiona PY Tsui, MB, BS, FHKAM (Anaesthesiology); Joyce MK Cheng, BHlthSc, FHKAN (Perioperative)
Department of Anaesthesia, Pamela Youde Nethersole Eastern Hospital, Hong Kong
 
Corresponding author: Dr Vivian YT Cheung (cyt086@ha.org.hk)
 
 Full paper in PDF
 
 
Case report
In October 2020, a 55-year-old Chinese man travelled from Hong Kong to Paris to attend a family funeral. He had psoriatic arthropathy in remission without chronic pain. In November 2020 while still in France, he and seven family members developed fever and upper respiratory symptoms, confirmed to be coronavirus disease 2019 (COVID-19). The family remained in home isolation and required no medical treatment. The patient self-treated with traditional Chinese medication: Lianhua Qingwen herbal capsules for 1 week. Two weeks later, he returned to Hong Kong after testing negative for COVID-19. In December 2020, during mandatory quarantine on re-entry to Hong Kong, the patient suddenly developed pain that extended from the neck and right interscapular region to the shoulder and down along the ulnar side of the right arm and forearm. The patient described the pain as shooting and drilling in nature, constantly severe, worst at the interscapular region, and aggravated by shoulder movement. He also reported disturbed sleep and numbness over his entire right arm and weakened right hand grip. He had no other joint pain, rash, vesicles, or fever. At this time, he was still resting alone in a quarantine hotel and performing no physical work. Most activities of daily living were manageable but some, such as bathing and dressing, were difficult. Diclofenac 100 mg daily and gabapentin 200 mg 3 times daily prescribed at a COVID-19 Clinic were ineffective. The patient’s younger sister who had recovered in France without medication reported similar symptoms in her left arm.
 
The patient presented to our pain clinic 1 month after pain onset. His motor symptoms had spontaneously improved although disturbing right shoulder and interscapular pain with paraesthesia persisted. There were no muscle wasting, scar, rash, or trophic changes. The patient’s right arm was slightly warmer than the left, and upper limb joints were not swollen or tender and there was full range of movement. He reported decreased sensation to light touch, cold and pinprick over the whole right arm, but his sense of vibration and proprioception were preserved. No touch or mechanical allodynia or hyperalgesia were noted. Apart from slightly weakened thumb opposition, other muscle strength, tendon reflexes and neck examination were unremarkable.
 
Analgesia was changed to pregabalin 75 mg twice per day and etoricoxib 90 mg daily as needed, and the patient was referred for occupational therapy for grip strengthening. Magnetic resonance imaging (MRI) in March 2021 revealed mild T2 hyperintensity at the right brachial plexus, suggestive of resolving neuritis (Fig). There was also cervical spondylosis without significant intervertebral foraminal narrowing or cord compression. Nerve conduction study in March 2021 was normal. Electromyography was not performed due to good neurological recovery.
 

Figure. Magnetic resonance imaging showing mild T2 hyperintensity at the right brachial plexus
 
At a subsequent 3-month follow-up examination in March 2021, the patient reported continued improvement with little or no pain. He reported only intermittent paraesthesia and mild weakness of his right hand and fingers. As a right-hander, he continued to have trouble turning keys and using chopsticks and pens. He coped with his office work with a speech-to-text converter to minimise keyboard usage. He could manage most household chores, including shopping for groceries, and slept well. He was calm and grieving appropriately for the loss of his mother. Pregabalin was gradually reduced, and he was weaned off etoricoxib.
 
Discussion
The Coronavirus family is known for its neurotropism with 36% of COVID-19 infected patients reporting some form of neurological manifestation.1 Mechanisms involve direct neural infection, and indirect inflammatory and immunological reactions. Other possibilities are targeting of neuronal angiotensin-converting enzyme 2, vasculitis, thrombosis and iatrogenic, such as prone position-related effects or neuropathies.2
 
The pathophysiology of acute brachial neuritis is not well understood but the pre-existence of viral infection supports immunological mechanisms. Affected subjects have more lymphocytic activity to brachial (versus sacral) plexus nerve extracts and increased antibodies to peripheral nerve myelin. A hereditary form with mutation-related deficiency in proteins from the septin family has been identified.2 Our patient took Lianhua Qingwen, a traditional Chinese medicine prepared from 13 herbs, shown to bind to angiotensin-converting enzyme 2 and shorten the course of COVID-19 infection.3 Drug-induced plexopathy, although less likely, remains possible.
 
Acute brachial neuritis is self-limiting but classically presents with excruciating pain at the shoulder, neck and interscapular region, followed by shoulder girdle weakness. Diagnosis is clinical and investigations are supportive. Cervical pathology is the major differential diagnosis but was excluded in our patient by MRI that revealed cervical spondylosis without nerve or cord compression or signal changes. Given the rheumatological history in our case, active autoimmune disease was also possible, but he had no such features.
 
To the best of our knowledge three cases of post-COVID-19 brachial neuritis4 5 6 have been reported but none in Hong Kong. Brachial neuritis is rare with an incidence of only 1.64 cases per 100 000 person-years, and underreporting is expected with isolation and restricted healthcare access during COVID-19. Compared to existing three cases, two cases similarly involved middle-aged men with delayed neuropathic symptoms 2 weeks after COVID-19 confirmation. One had similar symptoms to our patient, whereas the other two had either purely sensory components or solely proximal median nerve involvement. Our case and one existing case demonstrated classical MRI changes. Nerve conduction study in our patient did not demonstrate reduced action potential amplitude in affected nerves, which may have been related to its performance at a later course of the disease. Given the rarity of the entity and its occurrence in our patient and his sister, further research to investigate the role of genetic susceptibility to the acute form is warranted. Management of brachial neuritis is supportive and focused on pain control and functional rehabilitation with physiotherapy and occupational therapy. There is limited evidence that steroids and immunoglobulins will hasten recovery so their use should be balanced against the risk of viral replication. Currently, there are no established guidelines for pain management in patients with or recently recovered from COVID-19. Specific precautions should be taken in pain management of these cases.
 
Paracetamol has limited efficacy for neuropathic pain. Care should be taken for patients with severe COVID-19, because viral-induced cytokine storm can suppress cytochrome P450, increasing the risks of hepatotoxicity. Nonsteroidal anti-inflammatory drugs offer effective analgesia for brachial neuritis by suppressing cyclooxygenase and prostaglandin production. Although there were early concerns about ibuprofen-associated decompensation in patients with COVID-19, this has not been supported by the World Health Organization after data review. Meanwhile, cyclooxygenase-2 selective nonsteroidal anti-inflammatory drugs disturb the thromboxane A2–prostacyclin balance, potentially enhancing thrombotic tendency in patients with COVID-19. Our case illustrates the safe use of cyclooxygenase-2 inhibitors in a patient recently recovered from COVID-19. Among antineuropathic agents, gabapentinoids have relatively few adverse effects, lower cardiac toxicity, and fewer drug-drug interactions than tricyclic antidepressants and serotonin-noradrenaline reuptake inhibitors. Our patient was initially prescribed a relatively low dose of gabapentin that may account for its lack of effect. He was changed to pregabalin at a higher equivalent dose, with a better pharmacological profile with linear dose-response relationship and faster onset. Physicians should be alert to the sedative effects of analgesia that may worsen COVID-19-related ventilatory impairment. Opioids should be reserved for severe refractory pain.
 
Pain management for patients with or recently recovered from COVID-19 can be socially challenging. The need for quarantine delays presentation and management, and the associated mental stress and lack of social support may perpetuate pain. Although telemedicine enables remote medical care, controversies remain, and psychological engagement is less effective. Our patient’s appropriate grief reaction and illness coping mechanism minimises risk of chronic pain.
 
Our case report is the first to focus on the clinical management of brachial neuritis in patients with or recently recovered from COVID-19, and the first to identify a possible case series within a family. We hope our report of COVID-19-related brachial neuritis can promote awareness and understanding. Future research should focus on its pathophysiology including genetic susceptibility. Whether COVID-19 vaccination alters the course of acute brachial neuritis warrants further observation.
 
Author contributions
Concept or design: VYT Cheung, FPY Tsui.
Acquisition of data: VYT Cheung, JMK Cheng.
Analysis or interpretation of data: VYT Cheung.
Drafting of the manuscript: VYT Cheung.
Critical revision of the manuscript for important intellectual content: FPY Tsui, JMK Cheng.
 
All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
All authors have disclosed no conflicts of interest.
 
Acknowledgement
We would like to thank Dr Annie Chu for contributing to the clinical management, and Drs Mandy Au Yeung and Kendrick Tang for the investigations.
 
Funding/support
This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics approval
The patient was treated in accordance with the Declaration of Helsinki. The patient provided informed consent for the treatment/procedures, and for publication.
 
References
1. Mao L, Jin H, Wang M, et al. Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurol 2020;77:683-90. Crossref
2. Fernandez CE, Franz CK, Ko JH, et al. Imaging review of peripheral nerve injuries in patients with COVID-19. Radiology 2021;298:E117-30. Crossref
3. Alam S, Sarker MM, Afrin S, et al. Traditional herbal medicines, bioactive metabolites, and plant products against COVID-19: update on clinical trials and mechanism of actions. Front Pharmacol 2021;12:671498. Crossref
4. Mitry MA, Collins LK, Kazam JJ, Kaicker S, Kovanlikaya A. Parsonage-turner syndrome associated with SARS-CoV2 (COVID-19) infection. Clin Imaging 2021;72:8-10. Crossref
5. Siepmann T, Kitzler HH, Lueck C, Platzek I, Reichmann H, Barlinn K. Neuralgic amyotrophy following infection with SARS-CoV-2. Muscle Nerve 2020;62:E68-70. Crossref
6. Cacciavillani M, Salvalaggio A, Briani C. Pure sensory neuralgic amyotrophy in COVID-19 infection. Muscle Nerve 2021;63:E7-E8. Crossref

COVID toe in an adolescent boy: a case report

Hong Kong Med J 2022 Apr;28(2):175–7  |  Epub 17 Mar 2022
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
COVID toe in an adolescent boy: a case report
Joshua SC Wong, MB, BS, FHKAM (Paediatrics)1 †; TS Wong, MB, ChB, MRCPCH1 †; Gilbert T Chua, MB, BS, FHKAM (Paediatrics)2 †; Christy Wan, MB, BS1; SH Lau, MB, BS1; Samuel CS Ho, MB, BS1; Jaime S Rosa Duque, MD, PhD2; Ian CK Wong, PhD, FRCPCH3,4; Kelvin KW To, MD, FRCPath5; Winnie WY Tso, FHKAM (Paediatrics)2; Christine S Wong, MRCP, FHKCP6; Marco HK Ho, MD, FHKAM (Paediatrics)2; Janette Kwok, PhD, FRCPA7; CB Chow, MD, FHKAM (Paediatrics)1; Paul KH Tam, FRCS, FRCPCH8,9; Godfrey CF Chan, MD, FRCPCH,2; WH Leung, MD, PhD2; YL Lau, MD, FRCPCH2; Patrick Ip, MPH, FHKAM (Paediatrics)2; Mike YW Kwan, MSc (Applied Epidemiology) CUHK, FHKAM (Paediatrics)1
1 Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong
2 Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
3 Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
4 Research Department of Practice and Policy, UCL School of Pharmacy, University College London, United Kingdom
5 Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
6 Dermatology Division, Department of Medicine, Queen Mary Hospital, Hong Kong
7 Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong
8 Division of Paediatric Surgery, Department of Surgery, The University of Hong Kong, Hong Kong
9 Dr Li Dak-Sum Research Centre, The University of Hong Kong–Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Hong Kong
Co-first authors
 
Corresponding author: Dr Mike YW Kwan (kwanyw1@ha.org.hk)
 
 Full paper in PDF
 
 
Case report
In July 2020, a 17-year-old Pakistani boy presented with pain in his right foot unrelated to trauma or insect bite, after returning from Pakistan. The following day he tested positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. He had no previous medial history but was obese with a body mass index of 32.4 kg/m2. He denied any camping, water-trekking, or outdoor barefoot walking while in Pakistan. Physical examination revealed multiple purplish, flat, dry lesions <5 mm in diameter on his right toes and dorsum of the foot. They were tender on palpation but there was no surrounding erythema (Fig). No lesions were evident on the left foot or elsewhere and he had no symptoms or signs suggestive of any systemic autoimmune disorder. Pain associated with the lesions subsided by day 4 of illness. Some lesions spontaneously resolved but some became raised and crusted after day 3 (Fig). Topical fusidic acid for 1 week was prescribed to treat any potential bacterial infection. Of note, he developed a fever up to 39°C on day 8 and a productive cough. Vital signs remained stable with no respiratory distress or need for oxygen therapy. Chest X-ray did not show pneumonic changes and fever subsided within 24 hours. White blood cell count and differential were normal and C-reactive protein was 28 mg/L. Alanine aminotransferase was initially elevated at 131 U/L but showed a downward trend on rechecking. Clotting profile and D-dimer were normal. On day 12, SARS-COV-2 immunoglobulin G was detected and the patient was discharged from the hospital. His toe lesions resolved completely a few days later.
 

Figure. Clinical photos of COVID toes. (a) Reticular erythema and chilblain-like lesions over the dorsum of the foot and toes: multiple violaceous-erythematous macules and papules evident over the toes. (b) Some lesions were tender and crusted on day 5 of presentation. (Written consent for publication obtained)
 
Discussion
Rash is an uncommon symptom in coronavirus disease 2019 (COVID-19) infection.1 It has been described in Italy where 20% of COVID-19 patients developed cutaneous signs, including erythematous rash and widespread urticarial or vesicular lesions, at disease onset or following hospitalisation. The lesions usually subsided after a few days and there was no correlation with disease severity.2 Cutaneous manifestations included pseudo-chilblain (pernio-like), vesicular eruptions, urticarial lesions, maculopapular eruptions, and livedo or necrosis.2 3
 
Classic chilblains (or pernios) are inflammatory skin lesions that occur on the dorsal surface of the fingers and toes. They form painful and itchy erythematous and oedematous nodules that may ulcerate. They are triggered by cold and usually recur yearly during winter.3 Since March 2020, cases of acral lesions resembling chilblains have been reported across Europe, coinciding with the beginning of the COVID-19 outbreak. These lesions have differed to classic ones, showing an equal sex distribution, absence of obvious triggering factors, and involvement of the feet and distal third of the legs.3 They have been seen more commonly in previously healthy children or adolescents aged >10 years, almost always (74%-100%) on the feet but occasionally on the hands and fingers. The lesions were multiple and varied in size from a few millimetres to centimetres and were described as erythematous, violaceous, swollen, or purpuric. Itchiness and mild pain were frequently reported but required only symptomatic treatment. Lesions started to regress within 12 days to 8 weeks with complete resolution. The appearance of chilblain-like lesions was not thought to be associated with a poor disease outcome.2 3 A major limitation of these reports is that only 11% of cases hospitalised tested positive for SARS-CoV-2 by polymerase chain reaction (PCR), with the remainder untested or testing negative. Some authors have attributed this to the low sensitivity of tests or low viral load in children.3 The pathophysiological relationship between COVID-19 infection and chilblain-like lesions remains poorly understood, but has been hypothesised to be related to type 1 interferonopathies.3
 
Our patient is one of the few reported cases of laboratory-confirmed SARS-CoV-2 infection with chilblain-like lesions. To date, our patient is the only child in Hong Kong to present with SARS-CoV-2 infection as well as so-called “COVID toe”.1 Currently, there are insufficient data to determine a clear relationship between these dermatological symptoms and COVID-19. Rash is a common manifestation of many diseases and may not be associated COVID-19 infection. A recent case series of 17 adolescents in Italy who developed chilblain-like lesions during the first wave of COVID-19 screened negative on SARS-CoV-2 PCR of nasopharyngeal swabs, negative for SARS-CoV-2 immunoglobulin M and immunoglobulin G, and had no viral genome in biopsy specimens. However, this report was limited by its small sample size and did not compare data with an age- and gender-standardised background incidence of chilblains in the population.4 Most patients with dermatological manifestations were not confirmed to be infected with SARS-CoV-2. Another systematic review also concluded that some, but not all paediatric cases, who developed chilblain-like lesions during the COVID-19 pandemic had positive SARS-CoV-2 PCR, serology or viral particles confirmed in electron microscopy.5 Larger-scale epidemiological study is needed to confirm an association between these chilblain-like lesions and COVID-19 infection. Reported manifestations and histological findings were too heterogeneous to ascertain the pathophysiology. Nevertheless, physicians should remain vigilant since dermatological manifestations may be the first or only symptom in patients with COVID-19 infection,2 3 enabling a timely diagnosis of COVID-19 infection to reduce transmission. Physicians should also consider the possibility of coagulopathies and interferonopathies.
 
Author contributions
Concept or design: MYW Kwan, P Ip.
Acquisition of data: C Wan, SH Lau, SCS Ho, JS Rosa Duque.
Analysis or interpretation of data: C Wan, SH Lau, SCS Ho, JS Rosa Duque.
Drafting of the manuscript: JSC Wong, TS Wong, GT Chua.
Critical revision of the manuscript for important intellectual content: ICK Wong, KKW To, WWY Tso, CS Wong, MHK Ho, J Kwok, CB Chow, PKH Tam, GCF Chan, WH Leung, YL Lau.
 
All authors approved the final version of the manuscript and agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
 
Funding/support
This study is supported by the Collaborative Research Fund (CRF) 2020/21 and One-off CRF Coronavirus and Novel Infectious Diseases Research Exercises (Ref: C7149-20G). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
 
Ethics approval
The patient was treated in accordance with the Declaration of Helsinki, provided informed consent for the treatment/procedures, and provided consent for publication.
 
References
1. Chua GT, Wong JS, Lam I, et al. Clinical characteristics and transmission of COVID-19 in children and youths during 3 waves of outbreaks in Hong Kong. JAMA Network Open 2021;4:e218824. Crossref
2. Andina D, Belloni-Fortina A, Bodemer C, et al. Skin manifestations of COVID-19 in children: Part 2. Clin Exp Dermatol 2021;46:451-61. Crossref
3. Andina D, Belloni-Fortina A, Bodemer C, et al. Skin manifestations of COVID-19 in children: Part 1. Clin Exp Dermatol 2021;46:444-50. Crossref
4. Discepolo V, Catzola A, Pierri L, et al. Bilateral chilblain-like lesions of the toes characterized by microvascular remodeling in adolescents during the COVID-19 pandemic. JAMA Network Open 2021;4:e2111369. Crossref
5. Koschitzky M, Oyola RR, Lee-Wong M, Abittan B, Silverberg N. Pediatric COVID toes and fingers. Clin Dermatol 2021;39:84-91. Crossref

Paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2: a case report

Hong Kong Med J 2022 Feb;28(1):76–8  |  Epub 14 Feb 2022
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2: a case report
Gilbert T Chua, MB, BS, FHKAM (Paediatrics)1 †; Joshua SC Wong, MB, BS, FHKAM (Paediatrics)2 † Jaime Chung, MB, BS2; Ivan Lam, FHKCP, FHKAM (Paediatrics)2; Joyce Kwong, FHKAM (Pathology)3; Kate Leung, FRCPA, FHKAM (Pathology)3; CY Law, PhD, FHKAM (Pathology)4; CW Lam, PhD, FRCP5; Janette Kwok, PhD, FRCPA6; Patrick WK Chu, MPhil6; Elaine YL Au, FRCPA, FHKCPath7; Crystal K Lam, MB, BS7; Daniel Mak, MRCPCH, FHKAM (Paediatrics)2; NC Fong, FRCPCH2; Daniel Leung, PhD (Candidate)1; Wilfred HS Wong, PhD1; Marco HK Ho, MDM, FRCP1; Sabrina SL Tsao, MB, BS, FACC1; Christina S Wong, MRCP, FHKAM (Medicine)8; Jason C Yam, MB, BS, FCOphthHK,9; Winnie WY Tso, FHKAM (Paediatrics)1; Kelvin KW To, MD, FRCPath10; Paul KH Tam, FRCS, FRCPCH11,12Godfrey CF Chan, MD, FRCPCH1; WH Leung, MB, BS, PhD1; KY Yuen, MD, FRCPath10; Vas Novelli, FRCP, FRCPCH13,14; Nigel Klein, PhD13,14; Michael Levin, PhD, FRCPCH15; Elizabeth Whitaker, MRCPCH, PhD16; YL Lau, MD (Hon), FRCPCH1; Patrick Ip, MPH, FHKAM (Paediatrics)1; Mike YW Kwan, MRCPCH, MSc (Applied Epidemiology CUHK)2
1 Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
2 Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong
3 Haematology Laboratory, Department of Pathology, Princess Margaret Hospital, Hong Kong
4 Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong
5 Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
6 Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong
7 Division of Clinical Immunology, Department of Pathology, Queen Mary Hospital, Hong Kong
8 Division of Dermatology, Department of Medicine, Queen Mary Hospital, Hong Kong
9 Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, Hong Kong
10 Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
11 Division of Paediatric Surgery, Department of Surgery, The University of Hong Kong, Hong Kong
12 Dr Li Dak-Sum Research Centre, The University of Hong Kong–Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Hong Kong
13 Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children, London, United Kingdom
14 Institute of Child Health, University College London, London, United Kingdom
15 Section of Paediatrics, Imperial College London, London, United Kingdom
16 Paediatric Infectious Diseases Department, Imperial College Healthcare NHS Trust, London, United Kingdom
Co-first authors
 
Corresponding author: Dr Mike YW Kwan (kwanyw1@ha.org.hk)
 
 Full paper in PDF
 
Case report
A 10-year-old ethnic-Russian boy was confirmed to have severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) during the second wave of the coronavirus disease 2019 (COVID-19) outbreak in Hong Kong.1 He had a past medical history of coarctation of the aorta with corrective surgery performed at age 2 months. He returned from Russia on 6 June 2020 and his first deep throat saliva specimen saved on arrival at Hong Kong International Airport tested negative for SARS-CoV-2. Four days later, he developed fever, malaise, and headache. On 13 June 2021, he was admitted to our Paediatric Infectious Disease Unit and a new deep throat saliva specimen was positive for SARS-CoV-2. He did not require oxygen during his hospital stay. He was discharged from the hospital after being tested positive for SARS-CoV-2 anti-nucleoprotein immunoglobulin G antibodies 17 days after admission. This complied with the discharge criteria set by the Department of Health, the Government of Hong Kong Special Administrative Region.1
 
On 16 July, 16 days after being discharged, he returned to our Paediatric Infectious Disease Unit with a 2-day history of high fever and right cervical tender lymphadenopathy. Repeat nasal pharyngeal swab for SARS-CoV-2 polymerase chain reaction was negative. He was presumed to have bacterial lymphadenitis and was prescribed intravenous antibiotics but symptoms progressed. Ultrasound of the neck showed evidence of lymphadenitis but no signs of abscess formation. His fever and lymphadenitis persisted for 5 days and he also developed bilateral non-purulent conjunctivitis with peri-limbic sparing, erythematous and cracked lips, strawberry tongue and blanchable erythema over the trunk (Fig). Serial blood tests showed mild thrombocytopenia (trough 110 × 109/L), and raised erythrocyte sedimentation rate (peak 60 mm/Hr), C-reactive protein (peak 102 mg/L; range, <5.0), lactate dehydrogenase (270 U/L; range, <270), ferritin (1568 pmol/L; range, 31-279), highly sensitive troponin I (peak 643 ng/L; range, <21), N-terminal prohormone of brain natriuretic peptide (peak 3213 ng/L; range, <112), and interleukin-6 (IL-6) (peak 480.9 pg/mL; range, <4). Electrocardiogram and echocardiogram were unremarkable. His clinical presentation was compatible with Kawasaki-like disease. Since he had been infected with COVID-19 approximately 4 weeks previously, he was suspected to have PIMS-TS (paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2).2 Other differential diagnoses were excluded but included streptococcal and staphylococcal infection, Epstein–Barr virus infection and infection-related myocarditis. Owing to the rarity of PIMS-TS in East Asia, the clinical team discussed the case with experts from the United Kingdom who concurred with the diagnosis. He was treated with two doses of intravenous immunoglobulin (IVIG) at 2 g/kg/dose as his fever resurged 1 day after the first dose. Fever and other symptoms subsequently subsided after the second dose of IVIG, and serial echocardiograms did not reveal any coronary lesions. Whole exome sequencing performed to look for the possibility of an underlying monogenic immune dysregulation syndrome because of the rarity of this condition was unremarkable.
 

Figure. Clinical photographs showing (a) bilateral conjunctival injection with peri-limbic sparing and strawberry tongue, and (b) blanchable erythema over trunk. (Written consent for publication obtained from the patient’s parents)
 
Discussion
Paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2 is one of the most severe complications of COVID-19 infection in children. It was initially described in several case series in Europe and North America among children who presented with Kawasaki-like illness and were confirmed or known to have been in contact with another SARS-CoV-2–infected individual.2 3 Kawasaki disease (KD) is most prevalent among East Asians but rare in other parts of the world.3 On the contrary, paediatric cross-sectional clinical studies from East Asia have reported that PIMS-TS is rare among East Asians.1 We present the first, and so far, the only case of PIMS-TS in China. The case was an ethnic Russian boy who showed features of KD approximately 4 weeks after confirmation of SARS-CoV-2 infection. There is no consensus on the treatment regimen at present. In our patient, IVIG alone, instead of steroid or immunomodulators, was effective in treating the condition.
 
The pathophysiology of PIMS-TS remains uncertain. Studies have shown significant clinical and laboratory differences between PIMS-TS and KD, despite some similarities in clinical presentation. Patients with PIMS-TS are generally older than those with KD (median age, 8.3-9 years vs 2.7 years).2 3 They also have a higher white blood cell and neutrophil count and C-reactive protein, and a greater degree of lymphopenia and anaemia and tendency to develop thrombocytopenia in contrast to thrombocytosis in KD. In addition, fibrinogen and troponin levels are more elevated in PIMS-TS.2 These factors are associated with an increased risk of intensive care admission among children with PIMS-TS.2 These findings imply that PIMS-TS is a different entity to KD, with a greater degree of inflammation and myocardial injury. Studies have shown that certain cytokines, such as IL-6, appear to be particularly elevated in patients with PIMS-TS and may be involved in myocardial depression.2 Studies have also suggested that life-threatening COVID-19 pneumonia may be associated with monogenic inborn errors of immunity related to type 1 interferonopathies or type 1 interferon neutralising antibodies.4 Certain human leukocyte antigens, which are prevalent in East Asians but not Caucasians, have been associated with KD.4 However, no genes have been identified to cause PIMS-TS. Future studies will continue to explore the genetic factors related to PIMS-TS and the possible associated leukocyte antigen that explains the ethnic differences in PIMS-TS prevalence.
 
The treatment for PIMS-TS is similar to that for KD. A recent observational study demonstrated that patients who received IVIG and methylprednisolone together were less likely to require second-line biological agents, and were at lower risk of secondary acute left ventricular dysfunction and need for haemodynamic support with a shorter length of stay in the intensive care unit.5 Interleukin-1 and IL-6 receptor monoclonal antibodies have been used as second-line biological agents and have been shown to achieve remission when first-line therapies fail.2 5 Short-term outcomes of PIMS-TS are generally good. Immediate cardiac complications include coronary abnormalities, transient valvular regurgitation and myocardial dysfunction.2 The majority of patients recover without sequelae, but mortality has been reported.2 Data on the long-term outcomes of PIMS-TS are lacking.
 
The PIMS-TS remains a rare disease among East Asian patients.1 Nevertheless, frontline paediatricians in East Asia should remain vigilant when looking after ethnic non-East Asian children with COVID-19 infection in case they develop PIMS-TS after their initial recovery. Paediatricians should advise parents about the symptoms and signs of PIMS-TS so that timely medical consultation can be sought.
 
Author contributions
Concept or design: GT Chua, JSC Wong, P Ip, MYW Kwan.
Acquisition of data: J Chung, I Lam, J Kwong, K Leung, CY Law, CW Lam, J Kwok, PWK Chu, EYL Au, CK Lam, MYW Kwan.
Analysis or interpretation of data: D Mak, NC Fong, D Leung, WHS Wong, MHK Ho, SSL Tsao, CS Wong, JC Yam, WWY Tso, KKW To, PKH Tam, GCF Chan, WH Leung, KY Yuen, V Novelli, N Klein, M Levin, E Whitaker, YL Lau.
Drafting of the manuscript: GT Chua, JSC Wong, I Lam, J Chung.
Critical revision of the manuscript for important intellectual content: All authors.
 
All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of interest
All authors have disclosed no conflicts of interest.
 
Funding/support
This work was supported by the Collaborative Research Fund (CRF) 2020/21 and One-off CRF Coronavirus and Novel Infectious Diseases Research Exercises (Ref: C7149-20G). The funding source was not involved in the study design, collection, analysis or interpretation of data; in the writing of the manuscript; or in the decision to submit the manuscript for publication.
 
Ethics approval
The patient was treated in accordance with the Declaration of Helsinki, and the parents of the patient provided informed consent for the treatment and procedures.
 
References
1. Chua GT, Wong JS, Lam I, et al. Clinical characteristics and transmission of COVID-19 in children and youths during 3 waves of outbreaks in Hong Kong. JAMA Netw Open 2021;4:e218824. Crossref
2. Whittaker E, Bamford A, Kenny J, et al. Clinical characteristics of 58 children with a pediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2. JAMA 2020;324:259-69. Crossref
3. To KK, Chua GT, Kwok KL, et al. False-positive SARS-CoV-2 serology in 3 children with Kawasaki disease. Diagn Microbiol Infect Dis 2020;98:115141. Crossref
4. Sancho-Shimizu V, Brodin P, Cobat A, et al. SARS-CoV-2-r elated MIS-C: A key to the viral and genetic causes of Kawasaki disease? J Exp Med 2021;218:e20210446.
5. Ouldali N, Toubiana J, Antona D, et al. Association of intravenous immunoglobulins plus methylprednisolone vs immunoglobulins alone with course of fever in multisystem inflammatory syndrome in children. JAMA 2021;325:855-64.Crossref
 

Pages