Generalised involuntary limb twitching after ingestion of Mesobuthus martensii Karsch (Quanxie) powder

DOI: 10.12809/hkmj134091
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Generalised involuntary limb twitching after ingestion of Mesobuthus martensii Karsch (Quanxie) powder
PK Lam, FHKAM (Emergency Medicine), Dip Clin Tox1; TW Wong, FRCSEd, FHKAM (Emergency Medicine)1; YC Chan, FRCSEd, FHKAM (Emergency Medicine)2; Tony WL Mak, FRCPath, FHKAM (Pathology)3
1 Department of Accident and Emergency, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong
2 Hong Kong Poison Information Centre, United Christian Hospital, Kwun Tong, Hong Kong
3 Hospital Authority Toxicology Reference Laboratory, Princess Margaret Hospital, Laichikok, Hong Kong
 
Corresponding author: Dr PK Lam (lampkrex@hotmail.com)
 
 Full paper in PDF
Abstract
Mesobuthus martensii Karsch, commonly known as the Chinese scorpion or Manchurian scorpion, has been used in traditional Chinese medicine as Quanxie to treat chronic pain, tetanus, tremors, convulsion, and paralysis for more than a thousand years. We report a case of poisoning after ingestion of a teaspoon of Quanxie powder. The patient presented with chest pain, dizziness, diaphoresis, generalised involuntary limb twitching, and hypertonia around 15 minutes post-ingestion. The patient recovered uneventfully after supportive management. Intravenous diazepam appeared to be effective in alleviating limb twitching. Failure to accurately measure the dose and to boil before consumption may have contributed to his clinical toxicities.
 
 
Case report
A 63-year-old man complained of chest pain, dizziness, and generalised tremors 15 minutes after ingestion of a teaspoon of herbal powder with water in September 2012. He had started taking herbal decoctions prescribed by a registered Chinese medicine doctor 1 month ago because of suboptimal pain control of his trigeminal neuralgia with western medicine. He presented around 2 to 3 hours post-ingestion to our emergency department because of persistent symptoms. He was fully conscious but the limb tremor was so severe that he could barely walk. There was no numbness, headache, or any gastro-intestinal (GI) symptom. Apart from trigeminal neuralgia, he also had a history of ischaemic heart disease and hypercholesterolaemia. The medication on-hand included aspirin, famotidine, simvastatin, metoprolol, isosorbide mononitrate, diclofenac, dihydrocodeine, tramadol, and carbamazepine but he denied overdosing of any of those drugs. He had not taken any other herbs, over-the-counter medicines, or other suspicious foods such as coral reef fish and shellfish.
 
On arrival he was fully conscious with a Glasgow Coma Scale score of 15/15. His vital signs were as follows: blood pressure 126/95 mm Hg, pulse rate 66 beats/min, respiratory rate 18 breaths/min, oxygen saturation by pulse oximetry (SaO2) 99% on supplemental oxygen 2 L/min via a nasal cannula, and tympanic temperature 36.6°C. He appeared nervous with diaphoresis. Both his pupils were 2 mm in size and reactive to light. Cranial nerve examination was unremarkable but generalised involuntary limb twitching with hypertonia was evident. The muscle power in his four limbs was 5/5. Hyperreflexia and bilateral upgoing plantar reflexes were noted; there was no ankle clonus. Cardiovascular examination was unremarkable and his chest was clear. No distended urinary bladder or abnormal bowel sounds were noted. Repeated electrocardiograms showed sinus rhythm with normal axis. First-degree heart block was noted but there were no definite ischaemic changes. The QRS duration and the corrected QT interval were 89 ms and 431 ms, respectively. Chest X-ray revealed marginal cardiomegaly with clear lung field. The spot haemostix level was 6.2 mmol/L. Other blood tests including a complete blood picture, urea and electrolytes, serum calcium level, liver function tests, troponin-I, and arterial blood gas were essentially unremarkable, except a slightly raised creatine kinase (CK) level of 408 IU/L (reference range, 24-180 IU/L), which was likely due to generalised muscle twitching.
 
His chest pain decreased after administering 3 mg of intravenous (IV) morphine. Subsequently, his blood pressure dropped to 66/48 mm Hg but responded to fluid challenge with 500 mL of IV 0.9% normal saline. Less limb twitching was noted after administering 5 mg of IV diazepam. The provisional diagnosis was suspected Chinese herbal medicine (CHM)–related neurotoxicity but at the time of presentation, the formula of the herbal decoction was not available for identifying the culprit. The patient was admitted to the intensive care unit (ICU) for close monitoring in view of the unstable haemodynamic status upon presentation, which may have been the result of CHM toxicity or hypotensive effect of morphine.
 
Seven sheets of CHM prescription formula were subsequently traced back by the patient’s son 3 hours after admission. The Hong Kong Poison Information Centre was consulted for opinion. Multiple ingredients, including Quanxie (全蝎, Mesobuthus martensii Karsch), were prescribed according to the formula. However, the patient had remained free of side-effects in the previous month when he took the herbal decoctions as instructed. Further questioning revealed that the patient had found his pain control unsatisfactory even after taking the herbal decoctions. After receiving verbal advice from his Chinese medicine doctor, he took a few pieces of scorpion from the herb package and put them into a food blender. He took a teaspoon of the powder directly with water; this was the first time he took the herb in this form and fashion. He developed symptoms soon after ingestion.
 
The patient’s twitching decreased gradually after ICU admission but he remained hypertonic, which warranted administering another dose of IV diazepam 2 mg 10 hours after admission. Otherwise, he was fully conscious with a stable haemodynamic status. Computed tomography of the brain was unremarkable. His symptoms gradually resolved and he was discharged from the ICU and transferred to the Emergency Medicine Ward 16 hours after admission. His CK level peaked to 413 IU/L and normalised on day 2. He was discharged 36 hours after admission, and was totally asymptomatic on follow-up 5 days later.
 
The patient’s serum and urine samples, together with the unused herbs and herbal powder, were sent to the Hospital Authority Toxicology Reference Laboratory for further analysis. Poisoning due to other toxic herbs, such as aconitine, strychnine, and matrine was ruled out by liquid chromatography-mass spectrometry of the leftover herbal powder. However, detection of the toxic peptides of M martensii was not possible as the laboratory was not equipped to test for these toxins. The patient’s urine sample revealed the presence of diclofenac, dihydrocodeine, carbamazepine metabolite, salicylic acid and famotidine but these were the usual medications taken by the patient. The serum salicylate level was far below the toxic level. Judging from the history and absence of other toxic alkaloids to explain the symptoms, the diagnosis of this case was compatible with neurotoxicity associated with the consumption of M martensii powder, even though it could not be directly confirmed by chemical analysis.
 
Discussion
Mesobuthus martensii Karsch (synonym: Buthus martensii Karsch), commonly known as Chinese scorpion or Manchurian scorpion (東亞鉗蝎 or 馬氏鉗蝎), belongs to the Buthidae family, and is widely distributed in China. The whole body of the scorpion has been used in traditional Chinese medicine as Quanxie for more than a thousand years (Fig). It functions through the liver meridian, extinguishing wind, and stopping tremors and convulsion. According to the literature in traditional Chinese medicine, common indications include chronic pain, tetanus, tremors, acute/chronic childhood convulsions, paralysis, cerebral vascular accident, and fire toxic nodules.1 Its use is contra-indicated during pregnancy and should be avoided in cases of internal wind with blood deficiency. The recommended dose is 3 to 6 g for herbal decoction2 and 0.5 to 1 g for herbal powder.3
 

Figure. Quanxie (dried Mesobuthus martensii)
 
The venom of M martensii is complex. At least 51 long-chain peptides related to the sodium (Na+) channel toxin family and 18 peptides related to the potassium (K+) channel toxin family have been described4 and more have yet to be discovered. Neurotoxins affecting the Na+ channel consist of α (highly active in mammalian brain) and β (highly active in insects) toxins. Many β toxins, which are not noxious to mammals, were found to have analgesic properties in animal models without the risk of dependence.5 Recently, a novel peptide, BmK-YA, was found to contain an enkephalin-like sequence and can activate the mammalian δ opioid receptor.6 Novel peptides with antiepileptic (eg BmK AEP),7 antitumour (BmK AGAP),8 and antibacterial (BmKn2-7)9 effects in animal models have also been identified in the venom. These findings provide molecular evidence to support its traditional use in Chinese medicine, but relevant clinical studies are still lacking.
 
Although known to be toxic in traditional Chinese medicine, so far, literature on Quanxie poisoning has been rather limited. Many of the reported cases were due to therapeutic overdose,10 which might be related to the way the scorpions are processed. Traditionally, the captured scorpions are put into water for a few hours to allow them to spit out soil from gut, pass retained faeces, and clean the dirt over their bodies. Thereafter they are either boiled with plain water or salt water, resulting in ‘plain’ Quanxie (淡全蝎) and ‘salted’ Quanxie (鹽全蝎), respectively. Boiling kills the scorpion and decreases its toxicity. The addition of salt serves to preserve the processed scorpions for prolonged periods. The majority of the processing is undertaken in scorpion farms and there is a lack of standardised protocol.11 The details of processing vary from place to place, resulting in variable therapeutic effects, even with the same dose. Moreover, many merchants try to add more salt when processing salted Quanxie to increase its weight, for more profit in sale. The salt content in salted Quanxie could be up to 44.82% in the market,12 resulting in inadequate therapeutic effects with the recommended dose. Therefore, many traditional Chinese medicine doctors may opt to use a higher-than-recommended dose to achieve the targeted effect, thus increasing the risk of clinical toxicities.10
 
In our case, it is difficult to estimate the amount of Quanxie actually consumed as the powder had not been accurately weighed before consumption. With a standard teaspoon, the patient could have taken 5 to 6 g of the powder, which is higher than the recommended dose. Failure to boil before ingestion might also have contributed to his clinical toxicities.
 
The clinical features of M martensii (Quanxie) poisoning are summarised in the Table.10 13 14 15 There is no specific antidote, such as anti-venom, for this condition. The mainstay of treatment is supportive. The toxicokinetics of Quanxie have not yet been thoroughly studied. Judging from the rapid onset of clinical symptoms after oral ingestion, we believe that GI absorption is rapid. Gastro-intestinal decontamination with gastric lavage or activated charcoal can be considered if the patient presents within 1 hour of ingestion and has a protected airway. Such a time frame for GI decontamination has also been recommended for other toxic herbs with rapid GI absorption, such as aconitine. Clinicians may choose to consider GI decontamination in patients presenting beyond 1 hour after ingestion but the benefit and risk should be carefully weighed on a case-to-case basis. The benefit would certainly decline with time, which may not justify the risk of aspiration, especially when neurotoxic features such as generalised twitching have already set in, making the administration of activated charcoal further difficult. As Quanxie has not been shown to have enterohepatic circulation or prolonged GI absorption, multi-dose activated charcoal is not recommended. Benzodiazepines appeared effective in alleviating limb twitching muscle spasm in our case but its role in the management of M martensii poisoning remains to be elucidated.
 

Table. Clinical features of Mesobuthus martensii (Quanxie) poisoning10 13 14 15
 
Declaration
No conflicts of interests were declared by authors.
 
References
1. Li N, Yu M, Hu LN, Lin J. Collation of animal drugs — Quanxie [in Chinese]. Jilin J Tradit Chin Med 2009;29:805-6.
2. Committee of National Pharmacopoeia. Chinese pharmacopoeia [in Chinese]. Vol 1. Beijing: Chemical Industry Press; 2010: 133-4.
3. The editorial committee of “Chinese Herbals”, State Administration of Traditional Chinese Medicine. Chinese Materia Medica [in Chinese]. Vol 9. Shanghai: Shanghai Science & Technology Press; 1999: 129-35.
4. Goudet C, Chi CW, Tytgat J. An overview of toxins and genes from the venom of the Asian scorpion Buthus martensii Karsch. Toxicon 2002;40:1239-58. CrossRef
5. Shao JH, Zhang R, Ge X, Yang B, Zhang JH. Analgesic peptides in Buthus martensii Karsch: a traditional Chinese animal medicine. Asian J Tradit Med 2007;2:45-50.
6. Zhang Y, Xu J, Wang Z, Zhang X, Liang X, Civelli O. Bmk-YA, an encephalin-like peptide in scorpion venom. PLoS One 2012;7:e40417. CrossRef
7. Zhou XH, Yang D, Zhang JH, Liu CM, Lei KJ. Purification and N-terminal partial sequence of anti-epilepsy peptide from venom of the scorpion Buthus martensii Karsch. Biochem J 1989;257:509-17.
8. Liu YF, Zhang ZG, Mao YZ, et al. Production and antitumor efficacy of recombinant Buthus martensii Karsch AGAP. Asian J Tradit Med 2009;4:228-33.
9. Cao L, Dai C, Li Z, et al. Antibacterial activity and mechanism of a scorpion venom peptide derivative in vitro and in vivo. PLoS One 2012;7:e40135. CrossRef
10. Chang JM. Adverse effects of Quanxie [in Chinese]. J China Pharm 2003;14:484-5.
11. Gao ZJ. Brief review of the processing methods of Quanxie [in Chinese]. J Community Med 2006;4:59-60.
12. Shao XH, Kong XS, Fang LH. Processing of Quanxie and its clinical application [in Chinese]. Lishizhen Med Materia Medica Res 2006;17:232-3.
13. Chen XM. Analysis of adverse effects of Quanxie [in Chinese]. Lishizhen Med Materia Medica Res 2003;14:635.
14. Xiao YC. A case report of neurotoxicity induced by centipede and Quanxie [in Chinese]. China J Chin Materia Medica 1996;21:634.
15. Dai Y. A case report of free decoction for treatment of Quanxie [in Chinese]. J Jilin Med Coll 2009;3:342.

An outbreak of refrigerant-induced acute hepatitis in Hong Kong

DOI: 10.12809/hkmj134012
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
An outbreak of refrigerant-induced acute hepatitis in Hong Kong
YM Kan, MB, BS, FHKAM (Medicine); CF Lau, MRCP, FHKAM (Medicine); WC Chan, MRCP; WS Chan, MB, BS, FHKAM (Medicine); YM Tung, MRCP, FHKAM (Medicine); CK Loo, MRCP, FHKAM (Medicine)
Department of Medicine and Geriatrics, Kwong Wah Hospital, Yaumatei, Hong Kong
 
Corresponding author: Dr YM Kan (kanym@ha.org.hk)
 
 Full paper in PDF
Abstract
We report a cluster of acute hepatitis in five air-conditioning maintenance workers following accidental exposure to 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123). They presented to us with complaints of feverishness, generalised malaise, and epigastric discomfort. Their blood biochemistry tests were compatible with acute hepatitis. Viral hepatitis serology, tests for autoimmune hepatitis, and analyses for drugs and alcohol consumption were all negative. No focal hepatic lesion was detected by ultrasound imaging. Percutaneous liver biopsy samples were taken from two of them. The patients were managed with supportive treatment. All had spontaneous, but slow, recovery. Their liver function tests returned to normal after 4 months and their outcomes were favourable. Physicians should be aware of this occupational disease entity.
 
 
Case report
A 30-year-old air-conditioning maintenance worker presented to the Accident and Emergency Department in September 2010 with complaints of feverishness, dry cough, general malaise, poor appetite, and epigastric discomfort for 1 week. His initial symptoms 1 week before included feverishness and dry cough. He took paracetamol (500 mg, 4 times a day for 2 days) after consulting a general practitioner and diagnosed as having acute upper respiratory tract infection. Two days after visiting the general practitioner, he developed general malaise, nausea, and epigastric bloating. He had good past health and his family history was unremarkable. He was a social drinker and his last drink was almost 3 months ago. On physical examination, he was febrile and showed a tinge of jaundice. He was conscious and well-oriented. His pulse rate was 140 beats/min, the blood pressure was 130/75 mm Hg, and his body temperature was 39.3°C. Abdominal examination showed right upper quadrant tenderness but no other sign of acute abdomen; Murphy’s sign was negative.
 
Initial investigation showed elevated blood leukocyte count and mild thrombocytopenia (Table). His liver function tests were deranged and clotting profile was impaired. Serology for hepatitis A, B, C and E, Epstein-Barr virus, and cytomegalovirus was negative. Tests for antinuclear antibodies and anti-smooth muscle antibodies were also negative.
 

Table. Progression of the patient
 
His toxicology screening was negative, except for a serum paracetamol level of 77 mmol/L (taken 6 hours after the last dose of paracetamol). Ultrasound of the abdomen was unremarkable. His liver function tests further deteriorated after admission (Table) and he remained febrile. Computed tomography of the abdomen was then performed, which revealed minimal amount of ascites and non-specific pericholecystic fluid collection.
 
He received intravenous hydration and vitamin K as supportive treatment for his acute hepatitis. He remained fully oriented, and his serum ammonia level was normal.
 
Upon further enquiry, the patient recalled that he experienced dizziness, drunk feeling, and unsteady gait after exposure to a refrigerant during maintenance of an air-conditioning system in a computer server room with an area of around 200 m2. His job was to flush the air-conditioning system and pipeline with a cleansing refrigerant. He said he was not provided with any protective gear. He was, therefore, asked to call his co-workers to see if they had developed similar symptoms.
 
Eight workers worked in rotation for 2 weeks. They were posted to clean and repair the air-conditioning system in an enclosed area without any local exhaust ventilation system. In the first week, they were not provided with any effective protective gears. They worked for 6 hours a day in the first week. In the next week, they were provided with 3M face masks and their work duration shortened to 3 hours a day because they felt dizzy during work and needed to leave the room for a rest. Thus, exposure to leaked refrigerant was estimated to be high in such an enclosed workplace in the absence of effective protective gear. All workers experienced headache, dizziness, unsteady gait, and drunk feeling 15 minutes after exposure to the chemical released. They also had symptoms of dry cough, runny nose, fever, malaise, and loss of appetite a few days later. One of them also noticed passing tea-coloured urine and two had epigastric bloating.
 
Four among his seven co-workers agreed to undertake laboratory investigations. They were all found to have deranged liver function tests compatible with acute hepatitis. All investigations including viral hepatitis serology, tests for autoimmune hepatitis, and analyses for drugs and alcohol consumption were found to be negative. The circumstantial evidence, together with the clinical and laboratory findings, made refrigerant-induced acute hepatitis highly likely in this group of air-conditioning maintenance workers.
 
Percutaneous liver biopsy in one of the affected patients revealed the presence of hepatocytic dropouts and mitotic figures in perivenular distribution. There was moderate lymphocytic infiltrate with scattered eosinophils in the portal areas with mild bile duct proliferation. Perivenular hepatocytic cholestasis was also evident (Fig). These features, coupled with the clinical history and laboratory findings, were in keeping with a diagnosis of hydrochlorofluorocarbons-related hepatitis.
 

Figure. Liver biopsy: hepatocytic dropouts and mitotic figures in perivenular distribution (H&E; original magnification, x 40)
 
However, urine analysis of volatile organic hydrocarbons, metabolites of 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123), on day 4 after admission of the index patient, turned out to be negative.
 
Over the following days, our index patient’s symptoms improved progressively. His liver function tests, serum platelet count, and prothrombin time (given by the international normalised ratio) started to improve since day 5 of admission (Table). His coworkers’ symptoms also improved gradually.
 
Four months later, the health conditions of all the workers were confirmed to be normal. Their liver function tests had normalised. All had favourable outcomes.
 
We reported the cases to the Labour Department and the Department of Health since poisoning by halogen derivatives of hydrocarbon of the aliphatic series (ie HCFC-123) is a notifiable occupational disease in Hong Kong.
 
Discussion
HCFC-123 is a common refrigerant. It is one of the major substitutes for ozone-depleting chlorofluorocarbons used mainly as a refrigerant in chillers for industrial air-conditioning and in other applications such as foam blowing, cleansing agents, and industrial solvents. It is a colourless liquid with a light ether-odour and a boiling point of 27.6°C.1
 
Acute exposure to HCFC-123 has been shown to produce severe hepatotoxicity in guinea pigs.2 A single acute exposure to 1000 ppm of HCFC-123 for 4 hours can cause increases in aspartate aminotransferase and alanine aminotransferase levels compatible with hepatocellular necrosis. Increased liver weight, focal liver necrosis, induction of peroxisomal activity and hepatocellular adenomas have been found in subchronic studies in rats and dogs.3
 
The mechanism of hepatotoxicity of HCFC-123 was believed to be similar to that of 1-bromo-1-chloro-2,2,2-trifluoroethane (halothane). Halothane is metabolised to form reactive trifluoroacetyl halide intermediates that can react with water to form trifluoroacetyl haptens which result in direct hepatotoxicity. Since HCFC-123 is metabolised in the same oxidative way as halothane, HCFC-123 exposure might result in direct hepatotoxicity. In animal studies, the relative concentrations of trifluoroacetyl-protein adducts formed in the liver after administration of halothane and HCFC-123 were found to be similar.4 Alternatively, in-vitro metabolic studies of the human liver cytochrome P450 2E1 showed that exposure of human beings to HCFC-123 might result in higher concentrations of trifluoroacetyl-adducted liver proteins than those produced by halothane. The development of autoantibodies against P450 2E1 or P58 arising from immune reactions induced by trifluoroacetyl-adducted liver protein indicates that HCFC-related hepatotoxicity might also be immune-related.3
 
Takebayashi et al1 reported a cluster of acute liver dysfunction among workers exposed to HCFC-123 for less than 5 hours. Nine out of 14 workers developed impairment of liver function tests and symptoms of poor appetite and abdominal pain. Alanine aminotransferase level went up to more than 1700 U/L in these patients, but all had a favourable outcome. By the end of 2 months after HCFC-123 exposure, their liver function tests had returned to normal. This revealed that the incidence of liver dysfunction after HCFC-123 exposure is high.
 
Hoet et al3 investigated an epidemic of liver disease in nine industrial workers who had repeated accidental exposure to a mixture of HCFC-123 and HCFC-124. The results of this study also showed that repeated exposure of humans to HCFCs can result in serious liver injury in a high proportion of the exposed population. The liver biopsy showed hepatocellular necrosis which was prominent in perivenular zone 3, and extended focally across portal tracts and to centrilobular areas. The hepatocyte dropout was well-developed. The leukocytic inflammatory infiltrates in the zones of necrosis were mononuclear. Trifluoroacetyl-adducted proteins were detected in surviving hepatocytes by immunohistochemical staining. Serum autoantibodies against P450 2E1 or P58, which are associated with halothane hepatitis, were also detected. In our index patient, the presence of hepatocytic dropouts, increased mitotic figures and eosinophils might suggest acute hepatitis and liver regeneration after a short period of refrigerant exposure.
 
At moderate levels of exposure to HCFC-123, such as in some occupational accidents or prolonged exposure in poorly ventilated areas, respiratory effects (cough, dyspnoea, and tachypnoea), central nervous system effects (dizziness, drowsiness, weakness, fatigue, numbness, and coma), and gastro-intestinal upsets are characteristic. Hepatic injury (with elevated liver enzymes) and rhabdomyolysis might also rarely occur.5 A retrospective study showed that workers who had exposure to HCFC-123 experienced symptoms related to the central nervous system, gastro-intestinal upset, and irritation of mucous membrane.6 These may include headache, dizziness, abdominal pain, nausea, vomiting, dyspepsia, irritating smell, and eye or throat irritation.6 The prevalence of these symptoms also increased in the high-exposure group. Moreover, the degree and prevalence of liver dysfunction were higher in the high-exposure group.7
 
Urinary concentration of trifluoroacetic acid (TFA), which is a major metabolite of HCFC-123, can be used to determine the degree of HCFC exposure in air. However, in our case, urinary concentration of HCFC metabolite was negative. A small-scale human study found that the concentration of TFA in the urine peaked at 20 to 30 hours, and returned to zero by 96 hours post-exposure.8 Since the urine sample of our index case was collected 4 days after exposure, a positive test would not be expected.
 
There is no specific antidotal treatment for liver injury related to HCFC exposure. Supportive treatment of liver dysfunction is recommended. Fortunately, the outcome of HCFC-related liver dysfunction is usually favourable. All our patients had spontaneous recovery after cessation of exposure. To our knowledge, there is no report of death or liver transplant due to HCFC-123–induced hepatitis.
 
Poisoning by HCFC exposure is a notifiable occupational disease in Hong Kong. We reported this outbreak to the Labour Department and the Department of Health so that the related parties could conduct investigations and recommend appropriate modifications in the relevant working environment. It is essential to implement strict measures to prevent HCFC exposure, and physicians should be aware of the potential toxicities following HCFC exposure.
 
References
1. Takebayashi T, Kabe I, Endo Y, et al. Acute liver dysfunction among workers exposed to 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123): a case report. J Occup Health 1998;40:169-70. CrossRef
2. Marit GB, Dodd DE, George ME, Vinegar A. Hepatotoxicity in guinea pigs following acute inhalation exposure to 1,1-dichloro-2,2,2-trifluoroethane. Toxicol Pathol 1994;22:404-14. CrossRef
3. Hoet P, Graf ML, Bourdi M, et al. Epidemic of liver disease caused by hydrochlorofluorocarbons used as ozone-sparing substitutes of chlorofluorocarbons. Lancet 1997;350:556-9. CrossRef
4. Harris JW, Poul LR, Martin JL, Anders MW. Tissue acylation by the chlorofluorocarbon substitute 2,2-dichloro-1,1,1-trifluoroethane. Proc Natl Acad Sci USA 1991;88:1407-10. CrossRef
5. Concise International Chemical Assessment Document No. 23. Available from: http://www.inchem.org/documents/cicads/cicads/cicad23.htm. Accessed Jun 2014.
6. Takebayashi T, Kabe I, Endo Y, et al. Exposure to 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123) and acute liver dysfunction: a causal interference. J Occup Health 1998;40:334-8. CrossRef
7. Boucher R, Hanna C, Rusch GM, Stidham D, Swan E, Vazquez M. Hepatotoxicity associated with overexposure to 1,1-dichloro-2,2,2-trifluoroethane (HCFC-123). AIHA J (Fairfax, Va) 2003;64:68-79. CrossRef
8. Tanaka S, Kabe I, Takebayashi T, et al. Environmental and biological monitoring of 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123). J Occup Health 1998;40:348-9. CrossRef

Fluoroquinolone-induced Achilles tendinitis

DOI: 10.12809/hkmj134105
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Fluoroquinolone-induced Achilles tendinitis
PK Tam, MB, BS, FHKAM (Medicine); Carmen TK Ho, MB, BS, FHKAM (Medicine)
Department of Medicine, Tung Wah Hospital, 12 Po Yan Street, Sheung Wan, Hong Kong
 
Corresponding author: Dr PK Tam (alvintam@graduate.hku.hk)
 
 Full paper in PDF
Abstract
We report a case of Achilles tendinitis after intake of ciprofloxacin for treatment of respiratory tract infection. Fluoroquinolone-induced tendinopathy is an uncommon but increasingly recognised adverse effect of this antibiotic class. Most of the cases occur in the Achilles tendon and may lead to tendon rupture. Possible predisposing risk factors include use of steroid, patients with renal impairment or renal transplant, old age, and being an athlete. The drug should be stopped once this condition is suspected. Symptomatic treatment should be given and orthopaedic referral is desirable if tendon rupture occurs.
 
 
Case report
A 59-year-old woman with good past health, except for post-radioiodine hypothyroidism on T4 replacement, had a 3-week history of cough with yellowish sputum but no fever. She visited a general practitioner and was prescribed a course of ciprofloxacin for possible respiratory tract infection. Three days after starting the drug, she noticed pain and swelling over the left heel. She was not on any other medications including steroid. She went hiking regularly and there was no history of trauma. There was no history of joint or tendon problems. On presentation in May 2012, physical examination revealed tenderness and swelling over the left Achilles tendon (Fig 1). Ultrasound revealed Achilles tendinitis with increased flow on power Doppler signal. There were no signs of tear or calcium deposition (Fig 2). Due to the temporal relationship and the absence of other obvious causes, the diagnosis was ciprofloxacin-induced Achilles tendinitis (the Naranjo Scale was 7, ie probable adverse drug reaction).1 Ciprofloxacin was stopped and the patient was advised to avoid hiking until symptoms subsided completely. She recovered fully 2 weeks later.
 

Figure 1. Swollen left Achilles tendon (arrow)
 

Figure 2. Ultrasound shows thickened Achilles tendon (arrow) with increased flow on colour Doppler examination
 
Discussion
Fluoroquinolone-induced tendinopathy was first reported in 19832 and, since then, more than 100 cases have been reported in the literature. A case-control study from Italy found that use of fluoroquinolone was associated with higher risk of tendon disorders as well as Achilles tendon rupture (odds ratios, 1.7 and 4.1, respectively) compared with the control population.3 Another case-control study done in the United Kingdom estimated the risk to be 3.2 cases per 1000 patient-years.4
 
The commonest fluoroquinolones in the reported cases included ciprofloxacin and pefloxacin but, essentially, all the commonly used fluoroquinolones such as levofloxacin, ofloxacin, and norfloxacin have been associated with this adverse event.
 
Achilles tendon is the commonest affected site, accounting for nearly 90% of the reported cases. Other tendons like quadriceps tendon, rotator cuff tendon, as well as the site of tendon insertion may also be affected (there were two cases of epicondylitis reported after quinolone use).5 Tendinitis was the commonest pathology, present in 83.7% of the cases. Tendon rupture occurred in about 40% of the patients.6 A case-control study found that patients taking fluoroquinolones had a 4-fold increased risk of Achilles tendon rupture compared with the general population.3 Other complications related to tendinitis, such as carpal tunnel syndrome, are also possible.7
 
The mean time of symptom onset was about 2 weeks after initiation of the culprit medication, although it could range from 2 hours to as long as 6 months and, in 50% of the cases, it started within 6 days of drug intake. The mean age of affected subjects was 59 years.6
 
Several risk factors have been identified in the development of tendinopathy in patients taking fluoroquinolones. Among them, steroid use was the most significant risk factor. Most of the time, this was related to long-term use of systemic steroids, although even inhaled steroids were thought to be associated with the development of quinolone-induced tendinopathy.8 Other risk factors include old age (>60 years old), haemodialysis, renal impairment, renal transplant recipients, participation in sports activities, and history of rheumatic disorders.6 9 One previous review stated that males were more likely to develop this side-effect.6 One more recent case-crossover study, however, found that the association of tendinopathy and fluoroquinolone use was stronger in females, although not statistically significant.10 Therefore, it is not sure whether a particular gender is more likely to have this problem.
 
The underlying pathophysiology of the tendinopathy is not entirely known. A number of possible mechanisms have been proposed in animal and in-vitro studies. Ciprofloxacin could affect the metabolism of fibroblasts in tendon structures by reducing collagen synthesis and increasing extracellular matrix degradation.11 The chelating property of fluoroquinolones may also disturb the physiological interaction between cells and extracellular matrix.12 There was also evidence that fluoroquinolone increased apotosis in human tenocytes.13 In renal transplant recipients, the clearance of the drug may be impaired resulting in elevated concentrations in the tendon structures. Other risk factors like age, repeated trauma due to sports activities, or steroid therapy may impair the repair process of the tendon, thus, increasing the risk of tendinopathy in this group of patients.6 14
 
The tendinopathy usually presents with acute or subacute onset of pain and swelling over the tendon. Together with a history of recent consumption of fluoroquinolones and absence of other obvious causes of the tendinopathy, the diagnosis can be established. Imaging like ultrasound or magnetic resonance imaging is not mandatory but can aid in diagnosis, especially for visualising deep structures. Typical ultrasound findings of tendinopathy include thickened tendon with increased flow on colour Doppler examination.
 
The most important step in management is to stop the culprit drug. Appropriate rest and pain control are important. Pain can be well-controlled by non-steroidal anti-inflammatory agents; non-pharmacological treatments include ice therapy and therapeutic ultrasound. In case of tendon rupture, early referral to orthopaedic surgeon is desirable. Treatment options include immobilisation with casting or operative repair.
 
Prevention is also very important. One should only use fluoroquinolones when really necessary. Our patient’s symptoms did not suggest genuine lower respiratory tract infection; therefore, prescription of antibiotics was indeed not indicated. Moreover, quinolone is not the recommended first-line empirical antibiotic for treatment of chest infection in Hong Kong (it should only be considered in patients who are sensitive to penicillin or macrolide group of antibiotics). When use of antibiotics is really deemed necessary, fluoroquinolones should be avoided in patients with risk factors. Those with a history of fluoroquinolone-related tendinopathy should not be prescribed drugs of this class. We should also avoid co-prescription with steroid. If no better alternative is available, patients should be warned of this potential adverse effect and advised to stop the drug and seek medical advice if there are symptoms suggestive of tendinopathy. Athletes who are prescribed with fluoroquinolones should be advised to alter their training regimen (reduction in high-intensity and ballistic activities, decrease in total training volume) during the course of the antibiotics.14
 
References
1. Naranjo CA, Busto U, Sellers EM, et al. A method for estimating the probability of adverse drug reactions. Clin Pharmacol Ther 1981;30:239-45. CrossRef
2. Bailey RR, Kirk JA, Peddie BA. Norfloxacin-induced rheumatic disease. N Z Med J 1983;96:590.
3. Corrao G, Zambon A, Bertù L, et al. Evidence of tendinitis provoked by fluoroquinolone treatment: a case control study. Drug Saf 2006;29:889-96. CrossRef
4. van der Linden PD, Sturkenboom MC, Herings RM, Leufkens HG, Stricker BH. Fluoroquinolones and risk of Achilles tendon disorders: case-control study. BMJ 2002;324:1306-7. CrossRef
5. Le Huec JC, Schaeverbeke T, Chauveaux D, Rivel J, Dehais J, Le Rebeller A. Epicondylitis after treatment with fluoroquinolone antibiotics. J Bone Joint Surg Br 1995;77:293-5.
6. Khaliq Y, Zhanel GG. Fluoroquinolone-associated tendinopathy: a critical review of the literature. Clin Infect Dis 2003;36:1404-10. CrossRef
7. Liang VY, Ghearing GR, Zivkovic SA. Carpal tunnel syndrome after ciprofloxacin-induced tendinitis. J Clin Neuromuscul Dis 2010;11:165-6. CrossRef
8. Schwald N, Debray-Meignan S. Suspected role of ofloxacin in a case of arthralgia, myalgia, and multiple tendinopathy. Rev Rhum Engl Ed 1999;66:419-21.
9. Tsai WC, Yang YM. Fluoroquinolone-associated tendinopathy. Chang Gung Med J 2011;34:461-7.
10. Wise BL, Peloquin C, Choi H, Lane NE, Zhang Y. Impact of age, sex, obesity, and steroid use on quinolone-associated tendon disorders. Am J Med 2012;125:1228.e23-1228.e28.
11. Williams RJ 3rd, Attia E, Wickiewicz TL, Hannafin JA. The effect of ciprofloxacin on tendon, paratenon and capsular fibroblast metabolism. Am J Sports Med 2000;28:364-9.
12. Shakibaei M, Pfister K, Schwabe R, et al. Ultrastructure of Achilles tendons of rats treated with ofloxacin and fed a normal or magnesium-deficient diet. Antimicrob Agents Chemother 2000;44:261-6. CrossRef
13. Sendzik J, Shakibaei M, Schäfer-Korting M, Stahlmann R. Fluoroquinolones cause changes in extracellular matrix, signalling proteins, metalloproteinases and caspase-3 in cultured human tendon cells. Toxicology 2005;212:24-36. CrossRef
14. Hal MM, Finnoff JT, Smith J. Musculoskeletal complications of fluoroquinolones: guidelines and precautions for usage in the athletic population. PM R 2011;3:132-42. CrossRef

Post-transplantation primary central nervous system lymphoma in a patient with systemic lupus erythematosus and prolonged use of immunosuppressant

DOI: 10.12809/hkmj134095
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Post-transplantation primary central nervous system lymphoma in a patient with systemic lupus erythematosus and prolonged use of immunosuppressant
Teresa PK Tse, MB, ChB; Allan NL Chan, FHKCEM, FHKAM (Emergency Medicine); Tony KT Chan, FCSHK, FHKAM (Surgery); YC Po, FCSHK, FHKAM (Surgery)
Department of Neurosurgery, Princess Margaret Hospital, Lai Chi Kok, Hong Kong
 
Corresponding author: Dr Teresa PK Tse (teresapoki@hotmail.com), (tpk730@ha.org.hk)
 
 Full paper in PDF
Abstract
Post-transplantation primary central nervous system lymphoma is an uncommon and fatal post-transplant lymphoproliferative disorder. Such lymphomas have been described in only a few case series in the literature. The incidence of this condition is rising with improved survival after organ transplantation. A case of post-transplantation primary central nervous system lymphoma in a young Chinese woman with systemic lupus erythematosus is described here. She presented with right-sided weakness and memory loss after tooth extraction 2 weeks before admission. Contrast computed tomography of the brain demonstrated a contrast rim-enhancing lesion over the left frontal lobe. With a history of recent dental procedure, long-term immunosuppressive therapy and computed tomography findings, cerebral abscess was highly suspected. Emergency operation was performed. Histopathology showed post-transplantation primary central nervous system lymphoma, with cells positive for B-cell marker CD20. Immunosuppressant was stopped and she was treated with radiotherapy and rituximab (anti-CD20 monoclonal antibody). She remained disease-free at 16 months. Post-transplantation primary central nervous system lymphoma is rare with variable presentation and radiological features. We believe rituximab may have a role in the treatment of such lymphomas.
 
 
Introduction
Post-transplant lymphoproliferative disorder (PTLD) is a rare neoplastic complication of solid organ transplantation, affecting less than 2% of post-transplant patients. It includes a spectrum of diseases ranging from Epstein-Barr virus (EBV)–driven polyclonal lymphoid proliferation to EBV-positive or -negative malignant lymphoma. Post-transplantation primary central nervous system lymphoma (PT-PCNSL) is an uncommon and potentially fatal PTLD that develops in post-transplantation patients with the tumours confined to the brain and spinal cord, affecting 10% of patients with PTLD, which in turn affects only 1% of patients with kidney transplantation. The most common form of PCNSL is diffuse large B-cell lymphoma.1 2 To date, PT-PCNSL has been described in only case reports and a few case series in the literature.3 4 The exact incidence of PT-PCNSL is unknown, but it is expected to be rising in the future with improving survival for patients with organ transplant.5 Clinical presentation and radiological features of PT-PCNSL can vary. Here we describe a case of PT-PCNSL in a Chinese woman with systemic lupus erythematosus (SLE) and prolonged use of immunosuppressant.
 
Case report
A young woman with a known history of SLE underwent cadaveric renal transplantation for end-stage renal failure at the age of 28 years. She developed a complication of moderate cellular rejection postoperatively and was placed on mycophenolate mofetil (MMF) 750 mg every morning and 500 mg in the afternoon, and prednisolone 5 mg daily since 2000. Her renal function worsened after an episode of acute pyelonephritis in 2010 with creatinine level rising to 210 mg/dL from 150 mg/dL. She remained well afterwards until December 2011 when she was admitted to our hospital for progressive right-sided weakness and memory loss after tooth extraction 2 weeks before admission. On physical examination, she was found to have expressive dysphasia and right-sided weakness. Urgent contrast computed tomography (CT) of the brain demonstrated a 3.9 cm x 6.4 cm x 4.7 cm multilobulated contrast rim-enhancing lesion in the left frontal region with perifocal oedema and midline shift (Fig 1). With a history of recent dental procedure, long-term immunosuppressive therapy, and CT findings, cerebral abscess was highly suspected. Emergency operation was arranged and intravenous antibiotics were started immediately.
 

Figure 1. (a) Contrast and (b) plain computed tomography of the brain showing a multilobulated lesion in the left frontal region, measuring 3.9 cm x 6.4 cm x 4.7 cm, causing significant mass effect with perifocal oedema, effacement of adjacent sulcal spaces, and resultant right-sided midline shift. It has thin contrast-enhancing rims and non-enhancing central areas. No significant enhancing soft tissue component was associated
 
Operation
Burr hole for tapping of abscess was planned initially. Intra-operative ultrasound revealed an isodense lesion underneath the dura. Tapping was performed thrice but no fluid was aspirated. As frozen section was unavailable during non-office hours, we decided to perform left frontal craniotomy. Frontal lobectomy with partial excision of lesion was done. The lesion was found to be rubbery, lobulated, and non-vascular.
 
Pathological findings
Pathological examination revealed a lympho-proliferative lesion characterised by extensive infiltration by abnormal medium–to–large-sized lymphoid cells with large areas of necrosis. The abnormal lymphoid cells were monomorphic with vesicular nuclei and small nucleoli. The neoplastic cells were strongly positive for B-cell marker CD20. They were also positive for BCL2 and CD30, but negative for CD10 and T-cell marker CD3. In addition, the tumour cells were positive for EBV-encoded early RNAs (EBER) and EBV LMP-1. The Ki-67 proliferation index was estimated at 40% to 50%. The morphological findings, supported by immunohistochemical studies, were consistent with monomorphic PTLD, primary diffuse large B-cell lymphoma of the central nervous system (CNS) [Fig 2].
 

Figure 2. Various findings are shown: (a) tumour cells with prominent perivascular pattern on haematoxylin and eosin staining (x 20), (b) positive results for in-situ hybridisation for Epstein-Barr virus (x 20), and (c) aberrant expression of T-cell marker CD43 (x 20), and (d) positive staining for B-cell marker (x 20)
 
Postoperative course
Further workup showed that the patient had isolated CNS lymphoma. Bilateral bone marrow biopsies were done which showed no evidence of lymphoproliferative disease. Postoperative positron emission tomography–computed tomography (PET-CT) revealed residual hypermetabolic left frontal lymphomatous deposits but there were no hypermetabolic foci in the neck, thorax, abdomen, and pelvis. Serology was negative for EBV all along.
 
Postoperatively, the patient was continued on prednisolone and her antibiotics were discontinued. Mycophenolate mofetil was stopped and she was started on everolimus 0.25 mg daily. In view of suboptimal Karnofsky Performance Score and deteriorating renal function, she was treated with whole-brain radiotherapy (WBRT) alone (40 Gy/20 fr) followed by rituximab consolidation therapy (500 mg, once every 3 week, for 4 weeks). Five months after surgery, PET-CT showed complete resolution of the left frontal hypermetabolic foci; PET-CT 16 months after surgery showed stable disease. She is currently doing well 30 months after operation.
 
Discussion
Post-transplantation PCNSL is a rare neoplasm. Its clinical presentation and radiological features can vary. In a case series that involved 33 patients with PT-PCNSL imaged by contrast magnetic resonance imaging (MRI), 41% had homogeneously enhanced lesions, while 29% had ring enhancement and 61% had multiple lesions.6 In a review involving 221 patients with ring-enhancing lesions on MRI, 40% were gliomas, 30% were brain metastases, 12% were brain abscesses, 6% were multiple sclerosis plaques, and 2% were lymphomas.7 Imaging modalities such as magnetic resonance spectroscopy (MRS) may aid in differentiating PCNSL from brain abscess. In MRS, PCNSL typically demonstrates a lipid peak with raised choline to N-acetylacetate (NAA) ratio; while abscess typically demonstrates a lactate peak with reduced choline and NAA. Both PCNSL and abscess demonstrate restricted diffusion in diffusion-weighted imaging. Nuclear imaging such as PET scan may also help by showing high uptake in PCNSL while the uptake is low in abscess. However, urgent MRI, MRS, and PET scan were not readily available in our centre during non-office hours. In our case, the patient presented with focal neurological deficit with a history of recent dental procedure, use of long-term immunosuppressive therapy, and contrast rim-enhancing lesion on CT. The overall picture was suggestive of cerebral abscess, which warranted urgent surgical drainage.
 
Systemic lupus erythematosus is associated with an increased risk of haematological cancer, mainly non-Hodgkin’s lymphoma, while association with PCNSL is very rare with only few case reports on the condition in the literature. Moreover, most of these cases were associated with serious immunosuppressive therapy. Possible risk factors of PT-PCNSL include high-dose immunosuppressant and negative EBV serology in the transplant recipient.8 Our patient developed PT-PCNSL after kidney transplantation with prolonged use of MMF, and her EBV serology was also negative. It is postulated that EBV seronegativity and immunosuppression may predispose the transplant recipient to a novel EBV infection and, thus, the development of PT-PCNSL. However, the association of PT-PCNSL and SLE remains unclear.
 
The best treatment of PT-PCNSL has not been established. Reduction of immunosuppressive therapy, WBRT, and chemotherapy with agents like methotrexate and rituximab have been used for treating patients with PT-PCNSL. Whole-brain radiotherapy induced complete response by neuroimaging in 60% of patients with PCNSL but the median overall survival was only 12 months.9 High-dose intravenous methotrexate is now the standard of care for PCNSL with reported overall survival of up to 60 months.10 Rituximab, an anti-CD20 monoclonal antibody, has been used to treat patients with systemic PTLD. As rituximab does not penetrate the blood-brain barrier effectively, its effectiveness in treating PT-PCNSL is doubtful.11 Only three studies involving 10 patients with PT-PCNSL treated with intravenous rituximab have been reported with overall survival of at least 20 months.6 12 13 Resection of PCNSL has been discouraged as it causes significant neurological deficit without any survival benefit. In our case, after partial resection of tumour, WBRT and rituximab were used to treat PT-PCNSL. The patient remained disease-free at 16 months with MRI showing complete resolution of the lesions; she remains asymptomatic at 30 months after operation. It is believed that rituximab may have a role in the management of patients with PT-PCNSL by achieving adequate drug penetration into the brain parenchyma through leaky lymphomatous vasculature. We propose reconsidering the statement that efforts at resection of PCNSL should be discouraged, at least if resection seems safe. Yet, further studies are required to determine the best treatment for PT-PCNSL.
 
Conclusion
Post-transplantation PCNSL is a rare neoplasm with variable clinical presentation and radiological features. Possible risk factors include EBV seronegativity and prolonged use of immunosuppressive therapy. We believe rituximab and tumour resection may have a role in the treatment of PT-PCNSL.
 
Acknowledgement
We would like to express our special thanks to Dr WL Lam for the pathological examination of the specimen.
 
Declaration
No conflicts of interest were declared by authors.
 
References
1. Castellano-Sanchez AA, Li S, Qian J, Lagoo A, Weir E, Brat DJ. Primary central nervous system posttransplant lymphoproliferative disorders. Am J Clin Pathol 2004;121:246-53. CrossRef
2. Vaglio A, Manenti L, Mancini C, et al. EBV-associated leukoencephalopathy with late onset of central nervous system lymphoma in a kidney transplant recipient. Am J Transplant 2010;10:947-51. CrossRef
3. Phan TG, O’Neill BP, Kurtin PJ. Posttransplant primary CNS lymphoma. Neuro Oncol 2000;2:229-38. CrossRef
4. Snanoudj R, Durrbach A, Leblond V, et al. Primary brain lymphomas after kidney transplantation: presentation and outcome. Transplantation 2003;76:930-7. CrossRef
5. Wolfe RA, Roys EC, Merion RM. Trends in organ donation and transplantation in the United States, 1999-2008. Am J Transplant 2010;10:961-72. CrossRef
6. Cavaliere R, Petroni G, Lopes MB, Schiff D; International Primary Central Nervous System Lymphoma Collaborative Group. Primary central nervous system post-transplantation lymphoproliferative disorder: an International Primary Central Nervous System Lymphoma Collaborative Group Report. Cancer 2010;116:863-70. CrossRef
7. Schwartz KM, Erickson BJ, Lucchinetti C. Pattern of T2 hypointensity associated with ring-enhancing brain lesions can help to differentiate pathology. Neuroradiology 2006;48:143-9. CrossRef
8. Caillard S, Dharnidharka V, Agodoa L, Bohen E, Abbott K. Posttransplant lymphoproliferative disorders after renal transplantation in the United States in era of modern immunosuppression . Transplantation 2005;80:1233-43. CrossRef
9. Nelson DF, Martz KL, Bonner H, et al. Non-Hodgkin’s lymphoma of the brain: can high dose, large volume radiation therapy improve survival? Report on a prospective trial by the Radiation Therapy Oncology Group (RTOG): RTOG 8315. Int J Radiat Oncol Biol Phys 1992;23:9-17. CrossRef
10. Thiel E, Korfel A, Martus P, et al. High-dose methotrexate with or without whole brain radiotherapy for primary CNS lymphoma (G-PCNSL-SG-1): a phase 3, randomised, non-inferiority trial. Lancet Oncol 2010;11:1036-47. CrossRef
11. Ruhstaller TW, Amsler U, Cerny T. Rituximab: active treatment of central nervous system involvement by non-Hodgkin’s lymphoma? Ann Oncol 2000;11:374-5. CrossRef
12. Traum AZ, Rodig NM, Pilichowska ME, Somers MJ. Central nervous system lymphoproliferative disorder in pediatric kidney transplant recipients. Pediatr Transplant 2006;10:505-12. CrossRef
13. Kordelas L, Trenschel R, Koldehoff M, Elmaagacli A, Beelen DW. Successful treatment of EBV PTLD with CNS lymphomas with the monoclonal anti-CD20 antibody rituximab. Onkologie 2008;31:691-3. CrossRef

Spontaneous intracranial hypotension: improving recognition and treatment strategies in the local setting

DOI: 10.12809/hkmj133996
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Spontaneous intracranial hypotension: improving recognition and treatment strategies in the local setting
Gregory KY Lee, MB, BS1; Jill M Abrigo, MD1; Tom CY Cheung, FRCR, FHKAM (Radiology)1; Deyond YW Siu, FRCR, FHKAM (Radiology)1; Danny TM Chan, FRCS, FHKAM (Surgery)2
1 Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
2 Department of Neurosurgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
 
Corresponding author: Dr Gregory KY Lee (greglee011@gmail.com)
 
 Full paper in PDF
Abstract
We report a case of spontaneous intracranial hypotension with classic symptoms of orthostatic headache and acute presentation of subdural haematoma on computed tomographic scan. Conventional approach with conservative treatment was initially adopted. The patient’s condition, however, deteriorated after 2 weeks, requiring surgical evacuation of the intracranial haemorrhage. We reviewed the clinical features of this disease and the correlated magnetic resonance imaging findings with the pathophysiological mechanisms, and described treatment strategies in the local setting. Subtle findings on initial computed tomographic scan are also reported which might improve pathology recognition. Spontaneous intracranial hypotension is not uncommonly encountered in Hong Kong, and physicians must adopt a high level of clinical suspicion to facilitate early diagnosis and appropriate management. In addition, novel therapeutic approaches may be required in those with recurrent symptoms or who are refractory to current treatment strategies.
 
 
Case report
A 54-year-old man with no history of trauma was admitted to the Prince of Wales Hospital for headache of progressive severity accompanied by dizziness in August 2012. He had consulted the emergency room 4 weeks earlier for neck pain, and had an unremarkable computed tomographic (CT) scan of the brain (CTB). Further enquiry revealed an orthostatic component within the headache (worse in upright position and relieved within minutes of assuming supine posture), while admission CTB revealed interval development of bilateral 5 mm–thick frontoparietal subacute subdural haematomas (SDHs) with disproportionate tightness of the basal cisterns.
 
Cerebral magnetic resonance imaging (MRI) additionally demonstrated compression of the midbrain, but no caudal herniation of the cerebellar tonsils beyond the foramen magnum. Contrast study showed diffuse pachymeningeal enhancement, venous sinus distension, and prominent pituitary gland. Spinal MRI was unremarkable and MRI cisternography/myelography was negative for cerebrospinal fluid (CSF) leakage.
 
The patient was advised complete bed rest with adequate hydration. His neurological status was intact all along. However, he reported persistent, severe bifrontal headache which, after 2 weeks, was accompanied with repeated projectile vomiting. Computed tomographic scan of the brain at this juncture revealed enlargement of the SDH with development of acute haemorrhage. Emergency evacuation of subdural blood was performed with development of low intracranial pressure during the evacuation process.
 
The patient’s symptoms improved markedly thereafter, and CTB reassessment showed minor residual blood. The patient was discharged in a neurosurgically stable condition, and currently remains asymptomatic.
 
Discussion
Intracranial hypotension is traditionally attributed to leakage of CSF from a dural defect along the craniospinal axis, which can occur spontaneously, such as due to rupture of Tarlov cyst1 or dural weakness in connective tissue disorder.2 Intracranial hypotension can also be precipitated by direct trauma or iatrogenic causes such as a lumbar puncture. The commonest cause, however, is a spontaneous defect of the dura (spontaneous intracranial hypotension [SIH]), though a trivial traumatic event can be elicited retrospectively in around one third of such patients.3 4 The most common sites of leakage identified were at the cervicothoracic junction and thoracic region of the spinal canal.1 5 In the absence of a dural defect, a recent alternative hypothesis proposes increased CSF absorption from negative pressure gradient in the inferior vena cava.6 In both instances, CSF hypovolaemia is the main feature and primary cause of the related clinical and imaging findings.
 
Spontaneous intracranial hypotension is an increasingly diagnosed cause of headache, with an incidence of one in 50 000 individuals.7 The female-to-male incidence ratio of SIH is 2:1, with a peak incidence occurring around the age of 40 years.3 7
 
The typical clinical feature of SIH is orthostatic headache, which, according to the International Headache Society, should occur or worsen within 15 minutes in an upright posture together with at least one feature of meningeal irritation (neck stiffness, tinnitus, hypacusia, photophobia, nausea) in addition to imaging features of SIH.8 With chronicity, the postural component may become less prominent.3 Other clinical manifestations include disappearance of or improvement in the headache within 30 minutes after lying supine, and cranial nerve palsies related to traction from caudal displacement of the brainstem.9 Severe midbrain compression may result in nigral dopaminergic dysfunction and manifest as parkinsonism.9 Coma occurs from delayed decompression of brainstem descent.4
 
Subdural haematoma is a late finding and occurs in around 10% of patients with SIH,3 commonly seen in males and those older than 35 years.10
 
Computed tomography is the frontline imaging workup for headache. Typically, SIH is not considered unless patients present with non-traumatic SDH in the setting of a normal clotting profile. The proposed mechanism involves rupture of bridging veins in expanding subdural hygromas which, in turn, results from brain sagging due to CSF hypovolaemia.
 
On CT, SIH in the absence of SDH can be easily interpreted as being unremarkable. With high level of clinical suspicion, however, subtle imaging features may suggest the diagnosis. For instance, the initial CTB of our index patient showed paucity of CSF for his age (Fig 1). On follow-up CTB, the tightness of the basal cisterns appeared rather disproportionate to the small amount of SDH. Thus, it may be possible to detect CSF hypovolaemia on CT if these subtle findings are sought and the diagnosis is borne in mind.
 

Figure 1. (a) Computed tomography of the brain (axial view) showing a normal brain with adequate space over the midbrain and basal cisterns. (b) Our patient’s scan at similar axial level showing disproportionate tightness around the midbrain and basal cistern
 
The MRI findings of SIH are well-described in the literature. This preferred modality of imaging depicts characteristic features which may obviate the need for lumbar tap.4 Additionally, the cause or site of dural defect can be investigated.
 
The MRI appearances are mainly attributed to CSF hypovolaemia, or represent secondary reactive changes following the Monro-Kellie doctrine. Briefly, decrease in CSF volume prompts an increase in dural blood flow and causes venous engorgement. The latter, when prolonged, incites surrounding fibro-proliferation that in turn accounts for diffuse dural thickening and intense enhancement with gadolinium on MRI. Such explanation is confirmed by meningeal biopsy showing proliferation of fibroblasts without inflammation.11 12
 
The primary feature of brain sagging is a very specific MRI finding in SIH. It is a collaboration of features, including decreased dimension of the suprasellar cistern, bowing of optic chiasma, flattening of the pons against the clivus, effacement of the perimesencephalic cistern and hindbrain herniation (Fig 2a).11 12 A quantitative measurement of brain sagging has been described10 although the degree of descent may be underestimated since patients are scanned in the recumbent position.
 

Figure 2. Magnetic resonance imaging of our patient showing typical features of spontaneous intracranial hypotension (SIH). (a) Brain sagging, best appreciated by the mammillary body, which normally lies on, but currently is, below the line between tentorial apex and tuberculum sella. (b) Classical diffuse dural enhancement seen in SIH
 
Secondary and less-specific signs of SIH are more readily appreciated but represent a later stage of the disease. Findings of MRI in the brain classically show diffuse pachymeningeal thickening which has a sensitivity of up to 94% (Fig 2b).9 The venous distention sign may also be seen and is best appreciated in the mid-portion of the dominant transverse sinus12; on sagittal sections, the sinus, which normally adopts a concave or straight inferior border, bulges with a convex contour. Venous engorgement at the dura mater across the sella turcica could produce reactive hyperaemia and possible increase in size of the pituitary gland. In the spine, MRI findings mirror those of the brain with diffuse dural enhancement, engorgement of venous plexus and extrathecal CSF collection.3
 
Magnetic resonance imaging cisternography or myelography can be easily added to routine MRI examination. Using a heavy T2-weighted sequence with fat signal suppression, spinal fluid outside the craniospinal axis may be detected with equal or improved accuracy than CT myelography.5 Radiological improvement lags behind clinical recovery. Meningeal enhancement resolves considerably earlier than brain sagging.12 13
 
Computed tomography myelography and radionuclide cisternography are available locally but seldom performed. These are more invasive and time-consuming to perform, and entail intrathecal injection of contrast/radioisotope label, with fluoroscopic screening or serial imaging with gamma cameras to visualise extrathecal contrast/tracer activity.
 
Conservative management of SIH includes Trendelenburg positioning, aggressive hydration, caffeine intake and abdominal binder, and has been reported to be successful in most cases. In those patients with increased persistent headache or neurological deterioration, CTB should be immediately performed to rule out expanding SDH.
 
The timing of active surgical drainage is controversial. In a particular series, surgical drainage was advised in those with focal neurological deficits, decreased level of consciousness, or subdural collection of >1 cm.4 Most surgically managed patients show transient improvement but have high likelihood of re-accumulation of subdural fluid.13
 
A more active strategy that is gaining international acceptance is epidural blood patch (EBP), which aims at sealing off the spinal leak3 but appears to be useful in those without a definite dural defect.6 The treatment involves placing 10 to 20 mL of autologous blood in the epidural space at the thoracolumbar level. As the patient is put in Trendelenburg position, the blood patch distributes along the epidural space and clots at the site of leakage.
 
The overall success rate for headache improvement is 30% to 70% after the first EBP and 30% to 50% for the remainder with repeated EBP.10 Epidural blood patch has a success rate of 85% in reverting patients who were comatose due to SIH.13 Under fluoroscopic guidance, EBP may yield a high rate of pain relief and is preferred for those with altered anatomy or failure in the initial attempt.14 A novel technique of multisite EBP via continuous infusion has been described.15
 
The newest treatment approach developed at the Stanford University recommends emergency subdural clot evacuation in the absence of improvement in Trendelenburg positioning, presence of dilated pupils, and large SDH with mass effect. Otherwise, EBP is the treatment of choice.13
 
In those with initial improvement with EBP, studies have shown concomitant spontaneous resolution of significant SDH.13 Epidural blood patch may even be performed after surgical evacuation, and has been proven to further reduce the recurrence of headache and SDH.13 16 As the brain has a tendency to sag downwards in SIH, pneumocephalus during clot evacuation may cause further downward herniation of the brain. Hence, surgical evacuation after EBP may not be advisable. Most of these patients, like our index case, have shown low-pressure subdural collection during craniotomy.
 
Conclusion
The diagnosis of SIH should be considered for any patient presenting with headache and neck pain. A high level of clinical suspicion could assist identification of subtle signs on initial CT which could facilitate early recognition and prompt treatment and, consequently, improve outcomes. However, MRI remains an important, highly sensitive, and specific method for depicting imaging markers that allow confident clinical diagnosis. Further, MRI cisternography/myelography for CSF leak localisation can be added with ease. Currently, the medical practice in Hong Kong for SIH predominantly comprises conservative treatment and symptomatic clot evacuation. More novel interventions that have been successfully employed overseas may need to be reconsidered to improve current therapeutic strategies.
 
References
1. Cheng MF, Pan MH, Wu YE, Tsai WC, Yen RF, Tzen KY. Radionuclide cisternography in diagnosing spontaneous intracranial hypotension. Ann Nucl Med Sci 2004;17:167-72.
2. Schievink WI, Gordon OK, Tourje J. Connective tissue disorders with spontaneous spinal cerebrospinal fluid leaks and intracranial hypotension: a prospective study. Neurosurgery 2004;54:65-70; discussion 70-1. CrossRef
3. Schievink WI. Spontaneous spinal cerebrospinal fluid leaks: a review. Neurosurg Focus 2000;9:e8. CrossRef
4. Chen HH, Huang CI, Hseu SS, Lirng JF. Bilateral subdural hematomas caused by spontaneous intracranial hypotension. J Chin Med Assoc 2008;71:147-51. CrossRef
5. Wang YF, Lirng JF, Fuh JL, Hseu SS, Wang SJ. Heavily T2-weighted MR myelography vs CT myelography in spontaneous intracranial hypotension. Neurology 2009;73:1892-8. CrossRef
6. Franzini A, Messina G, Nazzi V, et al. Spontaneous intracranial hypotension syndrome: a novel speculative physiopathological hypothesis and a novel patch method in a series of 28 consecutive patients. J Neurosurg 2010;112:300-6. CrossRef
7. Diaz JH. Epidemiology and outcome of postural headache management in spontaneous intracranial hypotension. Reg Anesth Pain Med 2001;26:582-7. CrossRef
8. International Headache Society, Headache Classification Subcommittee. The International classification of headache disorders, 2nd ed. Cephalalgia 2004; ICHD–II code: 7.2.3.
9. Pakiam AS, Lee C, Lang AE. Intracranial hypotension with parkinsonism, ataxia, and bulbar weakness. Arch Neurol 1999;56:869-72. CrossRef
10. Rahman M, Bidari SS, Quisling RG, Friedman WA. Spontaneous intracranial hypotension: dilemmas in diagnosis. Neurosurgery 2011;69:4-14. CrossRef
11. Metafratzi Z, Argyropoulou MI, Mokou-Kanta C, Konitsiotis S, Zikou A, Efremidis SC. Spontaneous intracranial hypotension: morphological findings and CSF flow dynamics studied by MRI. Eur Radiol 2004;14:1013-6. CrossRef
12. Farb RI, Forghani R, Lee SK, Mikulis DJ, Agid R. The venous distension sign: a diagnostic sign of intracranial hypotension at MR imaging of the brain. AJNR Am J Neuroradiol 2007;28:1489-93. CrossRef
13. Loya JJ, Mindea SA, Yu H, Venkatasubramanian C, Chang SD, Burns TC. Intracranial hypotension producing reversible coma: a systematic review, including three new cases. J Neurosurg 2012;117:615-28. CrossRef
14. Watanabe K, Hashizume K, Kawaguchi M, Fujiwara A, Sasaoka N, Furuya H. Fluoroscopically guided epidural blood patch with subsequent spinal CT scans in the treatment of spontaneous cerebrospinal fluid hypovolemia. J Neurosurg 2011;114:1731-5. CrossRef
15. Ohtonari T, Ota S, Nishihara N, et al. A novel technique of multiple-site epidural blood patch administration for the treatment of cerebrospinal fluid hypovolemia. J Neurosurg 2012;116:1049-53. CrossRef
16. Mendes FF, Gonçalces AN, Novelo B, Mariano da Rocha CR, Marques NF. Spontaneous intracranial hypotension treated with epidural blood patch. Revista Dor 2012;13:1806-13.

Digital ischaemia: a rare but severe complication of jellyfish sting

DOI: 10.12809/hkmj134155
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Digital ischaemia: a rare but severe complication of jellyfish sting
Stacey C Lam, BS, MB, ChB; YW Hung, FHKCOS, FHKAM (Orthopaedic Surgery); Esther CS Chow, FHKCOS, FHKAM (Orthopaedic Surgery); Clara WY Wong, FHKCOS, FHKAM (Orthopaedic Surgery); WL Tse, FHKCOS, FHKAM (Orthopaedic Surgery); PC Ho, FHKCOS, FHKAM (Orthopaedic Surgery)
Division of Hand and Microsurgery, Department of Orthopaedics, Prince of Wales Hospital, Shatin, Hong Kong
 
Corresponding author: Dr YW Hung (ywhung@ort.cuhk.edu.hk)
 
 Full paper in PDF
Abstract
We report a case of digital ischaemia in a 31-year-old man who presented with sudden hand numbness, swelling, and cyanosis 4 days after a jellyfish sting. This is a rare complication of jellyfish sting, characterised by a delayed but rapid downhill course. Despite serial monitoring with prompt fasciotomy and repeated debridement, he developed progressive ischaemia in multiple digits with gangrenous change. He subsequently underwent major reconstructive surgery and aggressive rehabilitation. Although jellyfish stings are not uncommon, no severe jellyfish envenomation has been reported in the past in Hong Kong and there has not been any consensus on the management of such injuries. This is the first local case report of jellyfish sting leading to serious hand complications. This case revealed that patients who sustain a jellyfish sting deserve particular attention to facilitate early detection of complications and implementation of therapy.
 
 
 
Case report
Our patient was a 31-year-old man with unremarkable past health. He was swimming in the waters of Phuket, Thailand, when he experienced a sudden, intense burning pain in his right arm and right thigh. He spotted a jellyfish in the water after the incident. After getting out of the water, he noticed immediate reddening and swelling over the right thigh and right upper limb (from elbow down to the hand). No systemic symptoms were reported. The locals gave him some water to irrigate the sting site and slathered a soothing cream on it. He was then admitted to the local hospital and given a dose of steroids.
 
The next day, he was discharged from the local hospital and returned to Hong Kong. He was admitted to the Department of Orthopaedics and Traumatology of Prince of Wales Hospital, Hong Kong, in July 2013. It was the second day after his injury, and his right arm was swollen up to the midarm, and there were multiple maculopapular lesions over his right thigh and arm (Fig 1a, 1b). The injury site in the right forearm was explored the same day, revealing healthy subcutaneous tissue, fascia, and muscle (without features of necrotising fasciitis). Wound swab culture was negative.

Figure 1. Serial photos showing progressive digital gangrene and large skin defect after repeated debridement
Typical lesion after jelly fish sting over (a) right thigh, (b) elbow, and (c) hand on day 2 after injury: linear; papular whip-like dermatitis (‘tentacle print’). (d) Finger tip with cyanotic changes on day 8 after injury. (e) Well-demarcated gangrenous change over finger tips on day 15 after injury. (f) Extensive skin defect with expose tendon after debridement on day 17 after injury. (g) Reconstruction included a two-stage operation with groin flap to cover the dorsum of hand and a second groin flap to cover gangrene tip (6 weeks after injury). (h) Outcome (attempt to make tight fist) after multiple major reconstruction (8 months after injury)
 
On the fourth day after the sting, he experienced sudden onset of numbness and pain in his right hand which rapidly progressed to almost complete loss of sensation. The swelling in the right arm and forearm also increased. The skin was cold with cyanotic change in all fingers and the thumb; capillary refill was sluggish (<4 seconds) but with preserved turgor (Fig 1c). The radial pulse was not palpable. Compartment pressure measured with Stryker needle revealed superficial flexor compartment pressure of 35 mm Hg, deep flexor compartment pressure of 25 mm Hg, and extensor compartment pressure of 22 mm Hg.
 
Immediate fasciotomy of the right forearm was done, revealing subcutaneous oedema but no evidence of myonecrosis. Arteriotomy was not performed in view of return of palpable radial and ulnar pulse. Again, wound swab culture revealed no bacterial growth. Biopsy revealed muscle necrosis and infiltrate with white cells (predominantly polymorph). Postoperatively, he was put in a warm room with adequate fluid replacement and started on subcutaneous fraxiparine.
 
Despite wound debridement and aggressive medical treatment, 2 weeks after his initial insult, his right hand circulation remained sluggish, with progressive gangrenous change in the distal phalanges of the thumb, index finger, middle finger, half of the ring finger, and dorsum of the hand (Fig 1d, 1e). Computed tomography angiogram showed that the brachial artery was patent down to the palmar arch level, suggesting distal small vessel disease. As such, his gangrene would not be amenable to surgical intervention. Finally, he had a staged reconstructive surgery with distal amputation of his distal index finger and thumb; a groin flap (distant pedicle flap) was used to cover the major skin defect over the dorsum of the hand and the thumb (Fig 1f, 1g). With careful reconstructive surgery and aggressive rehabilitation, 9 months after the incident, his right hand regained movement, although it remained functionally impaired with significant stiffness and numbness (Fig 1h).
 
Discussion
Jellyfish belong to a family of Cnidaria found all over the world. There are more than 2000 different types of jellyfish, of which approximately 70 are toxic to humans.1 The pathophysiology of jellyfish sting is a combination of toxin and immunological response (immediate allergic reaction and delayed reaction).2 The toxins are composed of a mixture of polypeptides and enzymes, leading to local or systemic inflammatory responses.2 There has also been a report on toxins causing platelet aggregation.3 Hence, the physiological response of jellyfish sting depends on the species of jellyfish and the toxins they release.
 
The toxin of Cnidaria is located in the cnidocytes, which are stinging cells composed of organelles called ‘nematocysts’. Nematocysts are present on the outer surfaces of tentacles or near the mouth. These are released when the victim’s skin is in contact with jellyfish, injecting the venom into the victim via a thread tube, sufficient to penetrate the dermis of human skin.2
 
The majority of jellyfish stings are mild with local skin reactions. The usual presentation is a painful papular-urticaria at site of contact, which looks like multiple whip-like eruptions. This is compatible with the sting on the right leg in our case, with a linear whip-like ‘tentacle print’. Lesions can last for minutes or hours. Other local reactions also include hyperhidrosis of skin, lymphadenopathy, fat atrophy, vasospasm, gangrene, and contracture. Systemic reactions—including gastro-intestinal symptoms, cardiac arrest, respiratory arrest and anaphylactic shock—are rare but not impossible.2
 
Hong Kong is a city surrounded by ocean. Many people enjoy recreational water sports and, thus, injury related to marine life is unavoidable. The latest annual report by the Hong Kong Poison Information Centre ranked venomous stings and bites as the seventh commonest cause of poisoning.4 This is likely an underestimation as most marine accidents are managed at the scene, and this figure only takes into account the ones reported. In the English literature, there have only been three case reports of serious jellyfish sting injuries leading to serious hand or foot complications,5 6 7 and our case is the first local case report. Similar to the sudden deterioration in our patient, these documented cases all reported patients who suffered from sudden oedema, cyanotic changes, and weak pulse. This occurred around 3 to 4 days after the initial insult. In addition, the case reported by Abu-Nema et al7 noted that the patient had suffered from arterial spasm complicated by thrombosis, and was subsequently treated with urokinase. Our patient shares a similar complication of vasospasm, as evidenced by patent but diminished flow in distal arteries. It may be postulated that the delayed vasospasm and possible thrombosis may have led to this rare complication of jellyfish sting. Although jellyfish injuries usually present with acute skin reactions, our patient presented with clinical deterioration of the hand on day 4 when the lower limb skin reaction was actually improving.
 
The differences between our case and the other case reports are in terms of alternative management modalities and complications. Our patient received fasciotomy, repeated debridement, and fraxiparine. Some of the other authors incorporated other surgical interventions such as cervicodorsal or thoracic sympathectomy and medical treatments such as dextran, prednisolone, reserpine, and urokinase in case of thrombosis. Our patient suffered from gangrenous thumb and fingers. In other case reports, reported complications ranged from a mere loss of superficial sensation, to amputation of necrotic digits with Volkmann’s contracture.
 
Despite the adverse effects of cnidarian stings, literature on treatment is limited and often conflicting. It is difficult to perform high-quality studies with sound methodology, owing to reasons including limited number of cases and lack of randomisation.8 The majority of cases are dealt with at the scene by the general physician or accident and emergency department. It is important for us to be well equipped with management strategies for this type of injury and maintain a high level of suspicion for major complications. Our recommendations are summarised in Figure 2a.
 

Figure 2. (a) Algorithm for prevention and treatment of jellyfish sting. (b) General measures that we recommend for limb ischaemia
Note: As ischaemia related to jellyfish injury is not well understood, management needs to be tailormade for each patient
 
The principles of the management of jellyfish injury are:
(1) Best treatment remains prevention of injury
(2) Alleviate the local effect of venom (pain and tissue damage)
(3) Prevent further discharge of nematocysts
(4) Control systemic reaction, including shock
 
Some important points need to be highlighted. After rapid resuscitation, the next step is to remove nematocysts, if technically feasible. It is also important to note that popular home remedies such as alcohol, physical rubbing by sand, or rinsing by fresh water can actually worsen symptoms for the victim, as these may lead to a massive nematocyst discharge and toxin release. Another common folk measure is the use of vinegar or urine to inactivate the venom. Unfortunately, these two methods are not applicable to all types of jellyfish stings as different species have different toxins. We do not recommend the general population to use either vinegar or baking soda if the offending organism is not well known.
 
Ice therapy is safe in general for pain relief due to an unclear mechanism; whereas the application of local heat is still debated, as it may potentially induce vasodilation and a systemic toxic reaction rather than denaturation of the venom. Anti-histamine is generally safe for local symptom control and antibiotics are also recommended for secondary infection. There are no studies to support the use of systemic corticosteroids in toxic reactions.8 Anti-venom exists for a species of jellyfish called Chironex fleckeri (sheep-derived whole immunoglobulin G). However, it has unknown cardiotoxic effects and, therefore, not approved or readily available.9
 
The underlying mechanism of local ischaemia in our case was not well understood. Toxin- and hypersensitivity-induced vasospasm and secondary thrombosis are the postulated mechanisms. The management algorithm in Figure 2b is based on this postulation. As there is no standardised management in the literature, management should be tailored to the patient and should balance the risks and benefits. Further research is needed to confirm our postulation and formulate a protocol for management of jellyfish stings.
 
References
1. Brennan J. Jellyfish and other stingers. In: World Book’s animals of the world. Chicago, IL: World Book, Inc; 2003.
2. Burnett JW, Calton GJ, Burnett HW. Jellyfish envenomation syndromes. J Am Acad Dermatol 1986;14:100-6. CrossRef
3. Azuma H, Sekizaki S, Satoh A, Nakajima T, Ishikawa M. Platelet aggregation caused by a partially purified jellyfish toxin from Carybdea rastonii. Toxicon 1986;24:489-99. CrossRef
4. Chan YC, Tse ML, Lau FL. Hong Kong Poison Information Centre: Annual Report 2010. Hong Kong J Emerg Medicine 2012;19:110-20.
5. Giordano AR, Vito L, Sardella PJ. Complication of a Portuguese man-of-war envenomation to the foot: a case report. J Foot Ankle Surg 2005;44:297-300. CrossRef
6. Drury JK, Noonan JD, Pollock JG, Reid WH. Jelly fish sting with serious hand complications. Injury 1980;12:66-8. CrossRef
7. Abu-Nema T, Ayyash K, Wafaii IK, Al-Hassan J, Thulesius O. Jellyfish sting resulting in severe hand ischaemia successfully treated with intra-arterial urokinase. Injury 1988;19:294-6. CrossRef
8. Cegolon L, Heymann WC, Lange JH, Mastrangelo G. Jellyfish stings and their management: a review. Mar Drugs 2013;11:523-50. CrossRef
9. Balhara KS, Stolbach A. Marine envenomations. Emerg Med Clin North Am 2014;32:223-43. CrossRef

Pallidal deep brain stimulation: an effective treatment in Chinese patients with tardive dystonia

DOI: 10.12809/hkmj134082
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Pallidal deep brain stimulation: an effective treatment in Chinese patients with tardive dystonia
Peter YM Woo, MB, BS, FRCS (Edin)1; Danny TM Chan, MB, ChB, FRCS (Edin)1; XL Zhu, BMed (Jinan), FRCS (Edin)1; Jonas HM Yeung, MB, ChB, FRCP (London)1; Anne YY Chan, MB, ChB, MRCP1; Angie CW Au, MB, ChB2; KM Cheng, MRCPsych, FHKAM (Psychiatry)2; KY Lau, MSc1; YK Wing, FRCPsych, FHKAM (Psychiatry)3; Vincent CT Mok, MB, BS, FRCP (Edin)1; WS Poon, MB, ChB, FRCS (Edin)1
1 Movement Disorder Group, Division of Neurosurgery, Department of Surgery and Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
2 Department of General Adult Psychiatry, Castle Peak Hospital, Hong Kong
3 Department of Psychiatry, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
 
Corresponding author: Prof WS Poon (wpoon@cuhk.edu.hk)
 
 Full paper in PDF
Abstract
Tardive dystonia is an iatrogenic complication of dopamine receptor antagonist medication such as first-generation antipsychotics. It occurs in up to 2% of patients and only 10% recover after stopping medication. Deep brain stimulation for primary dystonia has proven to be effective and its application for secondary dystonias is gaining acceptance. We report our experience in treating three ethnic Chinese schizophrenia patients with severe medically refractory tardive dystonia by globus pallidus internus deep brain stimulation. Preoperatively, all required assistance with essential activities of daily living and two were bed-bound. The mean Burke-Fahn-Marsden Dystonia Rating Scale score was 61 (range, 44-80) and mean Global Dystonia Rating Scale score was 47 (range, 40-52). No procedure-related complications were encountered. By 3 months all could return to unassisted living and walk with support with a mean of 77% and 66% improvement in the Burke-Fahn-Marsden Dystonia Rating Scale and Global Dystonia Rating Scale scores, respectively. Quality-of-life assessment performed for two patients using the EuroQol-5 dimensions visual analogue scale showed a mean improvement of 86% at 3 months. On clinical follow-up, the effect was well maintained for a period of 3 to 10 years. Pallidal deep brain stimulation is a safe and highly effective form of symptomatic treatment for patients with medically refractory tardive dystonia.
 
 
 
Click here to watch a video of showing the treatment outcome of a patient with refractory tardive dystonia by pallidal deep brain stimulation
 
Introduction
Tardive dystonia (TD) is an iatrogenic extrapyramidal movement disorder caused by the use of dopamine receptor antagonists (DRAs). Antipsychotic medications, especially belonging to the first-generation class, are typically responsible for the condition.1 The reported prevalence of this adverse drug reaction among adults varies from 1% to 2% and only 10% of patients recover after medication termination.2 The latency of onset could range from several days to 20 years.3
 
Initial management strategies include withdrawal of the offending antipsychotic, switching to a second-generation antipsychotic such as clozapine, and suppressive therapies such as benzodiazepines or tetrabenazine, but none has demonstrable efficacy.3 In recent years deep brain stimulation (DBS) of the globus pallidus interna (GPi) has proven to be effective for secondary dystonias such as TD.4 5 6 7 We report our experience with using DBS for treating three young ethnic Chinese patients with severe TD refractory to pharmacological management.
 
Case reports
Clinical assessment
Three paranoid schizophrenia patients were referred by psychiatrists for TD satisfying the proposed diagnostic criteria by Adityanjee et al.2 All three patients were managed by the Movement Disorder Group, Division of Neurosurgery, Department of Surgery and Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong. Despite withdrawal of the antipsychotic responsible for the condition, and switching to second-generation medication, they all experienced progressive TD with severe disability. Their clinical details are outlined in Table 1. Investigations excluding other movement disorders included serum levels of ceruloplasmin, copper, thyroid-stimulating hormone, thyroxine as well as syphilis, and antinuclear antibody titres were either undetected or within normal limits. Magnetic resonance imaging of the brain was also unremarkable. For objective clinical evaluation video filming, the Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS) and the Global Dystonia Rating Scale (GDS) scores were documented before the procedure, at 1 week and 3 months postoperatively.8 9 Quality of life (QoL) assessments were performed for two of the patients, preoperatively and 3 months postoperatively, using the Chinese-version validated EuroQoL-5 dimensions (EQ-5D) instrument.10 This instrument serves as a basis for comparing health outcomes using a basic ‘common core’ of health-related QoL characteristics. The dimensions covered were mobility, self-care, usual activities, pain/discomfort, and anxiety or depression.11 After targeted questioning of these five domains, the patient was required to report on a visual analogue scale (VAS) from a score of zero (worst imaginable health state) to 100 (best imaginable health state).
 

Table 1. Clinical characteristics of patients
 
Operative procedure
We performed frame-based stereotaxy using the Leksell coordinate frame and multipurpose stereotactic arc system (Elekta AB, Stockholm, Sweden). The target was set at the ventroposterior part of GPi. The target coordinates were 19 mm lateral to the inter-commissural line (from the anterior commissure to the posterior commissure [AC-PC line]), 2 mm anterior to the mid-commissural point, and 4 mm inferior to the AC-PC line. The trajectory on the coronal view was 0 degree from the mid-sagittal plane. On sagittal view, it was 60 degrees from the AC-PC line.
 
The operation was performed under local anaesthesia for patients 1 and 2 in December 2004 and March 2009, respectively. Patient 3 was operated on under total intravenous general anaesthesia in April 2011 because of uncontrolled and vigorous trunk and limb movement. For patient 3, propofol was stopped 10 minutes before commencing microelectrode recording (MER). In all patients, MER was successfully performed and bilateral pallidal discharges were recorded.
 
For patients 1 and 2, visual-evoked MERs were registered at the target point by shining a light source in their eyes. If no capsular responses were evoked during macrostimulation below a threshold of 4 mA (0.1 ms PW, 130 Hz), a permanent quadripolar electrode (Medtronic 3387; Medtronic, Minneapolis [MN], US) was implanted bilaterally. During the same operative sitting, a pulse generator (Kinetra; Medtronic, Minneapolis [MN], US) was connected and implanted subcutaneously in the left infraclavicular region. A postoperative computed tomographic scan verified the final DBS electrode positions.
 
Results
Patients’ mean age was 41 years, ranging from 28 to 49 years with a mean duration of antipsychotic exposure of 3.7 years. Patients 2 and 3 received first-generation antipsychotics and patient 1 was administered second-generation medication. Preoperatively, all patients required either assisted living or was homebound. The craniospinal regions were the most seriously affected regions and all demonstrated opisthotonus and retrocollis. Chronic rhythmic neck hyperextension movements resulted in premature cervical spondylosis, and patients 1 and 3 required nasogastric tube feeding. At the time of surgery, all were unable to walk independently. The mean preoperative BFMDRS score was 61, ranging from 44 to 80, and mean GDS score was 47, ranging from 40 to 52. There were no treatment-related complications and the procedure was well tolerated. As expected, there was no minimal response before stimulation but after pulse generator activation, marked amelioration of dystonic symptoms was observed. The stimulation settings were set at monopolar mode (anodal case and cathodal target contact) and are presented in Table 2. Notable improvement was found in patients 2 and 3 within the first week. In that week, the mean BFMDRS score decreased by over 30%. By 3 months, all patients reached a stable state with a mean of 77% and 66% improvement in the BFMDRS and the GDS scores, respectively. Patients 1 and 3 overcame dysphagia to resume oral dietary intake and all patients were able to perform basic activities of daily living without assistance. No psychiatric side-effects were detected. Patients 2 and 3 experienced an improvement in QoL as reflected by a mean increase in the EQ-5D VAS score by 86% (Table 2).

Table 2. Stimulation settings and clinical outcomes
 
Discussion
Tardive dystonia is an iatrogenic complication belonging to a group of DRA-induced movement disorders known as the tardive syndromes. Although it may co-exist with tardive dyskinesia, TD is a distinct condition with different epidemiology, clinical features, prognosis, and treatment outcome.3 Literature shows a higher prevalence of TD in men than in women, with a male-to-female ratio of 2.4:1, and a mean age of onset of 36 years.2 The first-generation antipsychotics such as chlorpromazine, flupenthixol, and haloperidol are the strongest aetiological factors although second-generation medications and antiemetics such as metoclopramide have also been implicated.1 2 Although the mean duration of medication exposure varies from 3.3 to 6.6 years, there does not appear to be a minimal ‘safe’ period and symptom onset can occur as early as within days or weeks.1
 
In this report, all patients fulfilled Adityanjee et al’s diagnostic criteria2 for TD, namely, (1) chronic dystonia characterised by sustained, stereotypical involuntary muscle contractions or posture; (2) dystonia developing during or within 2 months’ discontinuation of treatment with DRAs; (3) other secondary dystonias adequately ruled out, and (4) a negative family history for dystonia. The onset of symptoms is insidious initially, involving one body region and, typically, progressing to segmental, ie craniocervical, or generalised dystonia. Torticollis or retrocollis is characteristic of TD and chronic repetitive movements could result in spondylotic myelopathy or even fractures.12 Truncal involvement manifests as opisthotonus and, in the severest of cases, patients could become bed-bound, reduced to a state of dependency.2 In contrast to classic orobuccolingual tardive dyskinesia, TD is largely irreversible with 90% of patients failing to achieve remission at a mean follow-up of 6.6 years.13
 
The limitations of medical treatment reflect incomplete understanding of the complex pathophysiology of TD. Multiple theories have been proposed of which the most prominent describe postsynaptic dopamine receptor hypersensitivity, degeneration of striatal cholinergic neurons, and gamma-amino-butyric acid–containing neurons.13 In contrast, the success of pallidal DBS in the treatment of primary dystonias led to its adoption for secondary dystonias such as TD.5 6 7 14 15 In this series we demonstrate a significant beneficial effect for medically refractory TD where rapid remarkable improvements in motor symptoms were observed within days without exacerbation of psychiatric symptoms. Our results are in agreement with other DBS outcome trials for TD. A systematic review of 17 studies involving 50 TD patients concluded that pallidal stimulation led to a mean improvement in BFMDRS scores by 77.5% (95% confidence interval, 71.4%-83.3%).4 The motor benefits are sustainable with a documented duration of 41 months (range, 18-80 months).5 7 16 Long-term responses for 8 to 10 years have also been reported.6 17 Although most data were from case studies or small trials, our experience supports DBS as an effective and safe surgical treatment modality that can considerably improve QoL.
 
Involuntary dystonic movement imposes unique technical difficulties not only for obtaining preoperative brain scans of acceptable quality, but also for performing the actual surgery. Sedation or general anaesthesia may be needed for such procedures as was needed in our patient 3. Furthermore, correct choice of anaesthetic drug is critical for successful MER. For example, propofol was found to affect the recording quality of the subthalamic nucleus of Parkinson’s disease patients.18 To overcome this interference, lower doses of propofol were used; these proved to be equally feasible to perform MER under general anaesthesia.19 20 Due to the relatively rapid offset action of intravenous propofol, after 10 minutes of cessation, we were able to observe the characteristic mean discharge frequencies of 20 to 40 Hz at the GPi target. The discharge pattern qualities were similar to those of the other two patients for whom the procedure was performed under local anaesthesia. Our experience demonstrates that in severely dystonic patients, total intravenous general anaesthesia with propofol can produce comparable clinical outcomes.
 
In 2014, patient 1 has received DBS for 10 years. In that period, she was capable of performing daily activities at home without assistance and was considered to have dystonia remission. However, when the pulse generator battery approached its end-of-life state, 2 years after implantation, segmental dystonia reappeared over the neck and hand regions. Her BFMDRS score rapidly rose from 0 to 23 within a week. The symptoms were relieved after battery replacement. The high voltage and pulse width requirements in GPi stimulation for TD can considerably shorten battery longevity (Kinetra; Medtronic) to an average of 2 years. The introduction of a non-invasive transcutaneous rechargeable battery system (Activa; Medtronic, Minneapolis [MN], US) with a 9-year life span is an improved solution to frequent replacements. Not only is it more cost-effective in terms of overall battery costs, but also reduces the number of surgical procedures. All three patients are currently implanted with this new device. The daily recharging process was convenient and non-disruptive, and the pulse generator performance was as effective as the non-rechargeable counterparts. With these encouraging results, we have continued to provide DBS as treatment for patients with refractory TD. Two more cases were treated in 2013 and 2014, respectively, with results as good as those in the three cases reported here.
 
Conclusion
Medically refractory TD is a disabling and essentially irreversible condition that can be successfully managed by pallidal DBS. There is a need to conduct multicentre trials to reliably assess and define appropriate selection criteria for DBS as a new therapeutic option. In our series, response to stimulation can be observed as soon as within 1 week. Our patients experienced remarkable alleviation of dystonia symptoms at 3 months enabling them to return to performing daily activities without assistance, with no additional psychiatric side-effects. On clinical follow-up, the effect was well maintained for a period of 3 to 10 years.
 
Acknowledgements
The Oriental Daily News Charitable Fund, Oriental Press Group Limited subsidised the purchase of the implantable hardware devices.
 
Declaration
No conflicts of interests were declared by authors.
 
References
1. Burke RE, Fahn S, Jankovic J, et al. Tardive dystonia and inappropriate use of neuroleptic drugs. Lancet 1982;1:1299. CrossRef
2. Adityanjee, Aderibigbe YA, Jampala VC, Mathews T. The current status of tardive dystonia. Biol Psychiatry 1999;45:715-30. CrossRef
3. Kiriakakis V, Bhatia KP, Quinn NP, Marsden CD. The natural history of tardive dystonia. A long-term follow-up study of 107 cases. Brain 1998;121:2053-66. CrossRef
4. Mentzel CL, Tenback DE, Tijssen MA, Visser-Vandewalle VE, van Harten PN. Efficacy and safety of deep brain stimulation in patients with medication-induced tardive dyskinesia and/or dystonia: a systematic review. J Clin Psychiatry 2012;73:1434-8. CrossRef
5. Trottenberg T, Volkmann J, Deuschl G, et al. Treatment of severe tardive dystonia with pallidal deep brain stimulation. Neurology 2005;64:344-6. CrossRef
6. Chang EF, Schrock LE, Starr PA, Ostrem JL. Long-term benefit sustained after bilateral pallidal deep brain stimulation in patients with refractory tardive dystonia. Stereotact Funct Neurosurg 2010;88:304-10. CrossRef
7. Gruber D, Trottenberg T, Kivi A, et al. Long-term effects of pallidal deep brain stimulation in tardive dystonia. Neurology 2009;73:53-8. CrossRef
8. Burke RE, Fahn S, Marsden CD, Bressman SB, Moskowitz C, Friedman J. Validity and reliability of a rating scale for the primary torsion dystonias. Neurology 1985;35:73-7. CrossRef
9. Comella CL, Leurgans S, Wuu J, Stebbins GT, Chmura T; Dystonia Study Group. Rating scales for dystonia: a multicenter assessment. Mov Disord 2003;18:303-12. CrossRef
10. Luo N, Chew LH, Fong KY, et al. Do English and Chinese EQ-5D versions demonstrate measurement equivalence? An exploratory study. Health Qual Life Outcomes 2003;1:7. CrossRef
11. Pinto Prades JL. A European measure of health: the EuroQol [in Spanish]. Rev Enferm 1993;16:13-6.
12. Konrad C, Vollmer-Haase J, Gaubitz M, Nabavi DG, Reilmann R, Knecht S. Fracture of the odontoid process complicating tardive dystonia. Mov Disord 2004;19:983-5. CrossRef
13. Margolese HC, Chouinard G, Kolivakis TT, Beauclair L, Miller R. Tardive dyskinesia in the era of typical and atypical antipsychotics. Part 1: pathophysiology and mechanisms of induction. Can J Psychiatry 2005;50:541-7.
14. Kupsch A, Benecke R, Müller J, et al. Pallidal deep-brain stimulation in primary generalized or segmental dystonia. N Engl J Med 2006;355:1978-90. CrossRef
15. Vidailhet M, Jutras MF, Roze E, Grabli D. Deep brain stimulation for dystonia. Handb Clin Neurol 2013;116:167-87. CrossRef
16. Sako W, Goto S, Shimazu H, et al. Bilateral deep brain stimulation of the globus pallidus internus in tardive dystonia. Mov Disord 2008;23:1929-31. CrossRef
17. Boulogne S, Danaila T, Polo G, Broussolle E, Thobois S. Relapse of tardive dystonia after globus pallidus deep-brain stimulation discontinuation. J Neurol 2014;261:1636-7. CrossRef
18. Raz A, Eimerl D, Zaidel A, Bergman H, Israel Z. Propofol decreases neuronal population spiking activity in the subthalamic nucleus of Parkinsonian patients. Anesth Analg 2010;111:1285-9. CrossRef
19. Pinsker MO, Volkmann J, Falk D, et al. Deep brain stimulation of the internal globus pallidus in dystonia: target localisation under general anaesthesia. Acta Neurochir (Wien) 2009;151:751-8. CrossRef
20. Hertel F, Züchner M, Weimar I, et al. Implantation of electrodes for deep brain stimulation of the subthalamic nucleus in advanced Parkinson’s disease with the aid of intraoperative microrecording under general anesthesia. Neurosurgery 2006;59:E1138; discussion E1138.

Novel use of idebenone in Leber’s hereditary optic neuropathy in Hong Kong

DOI: 10.12809/hkmj134085
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Novel use of idebenone in Leber’s hereditary optic neuropathy in Hong Kong
SW Cheng, MB, ChB, MRCPCH1; CH Ko, FHKAM (Paediatrics), FRCP RCPS (Glasg)1; SK Yau, MB, ChB, FRCSEd2; Chloe Mak, MD, PhD3; YF Yuen, FRCSEd, FHKAM (Ophthalmology)2; CY Lee, FHKAM (Paediatrics), FRCP (Edin)1
1 Department of Paediatrics and Adolescent Medicine, Caritas Medical Centre, Shamshuipo, Hong Kong
2 Department of Ophthalmology, Caritas Medical Centre, Shamshuipo, Hong Kong
3 Department of Pathology, Princess Margaret Hospital, Laichikok, Hong Kong
 
Corresponding author: Dr SW Cheng (csw350@ha.org.hk)
 
 Full paper in PDF
Abstract
We report a case of a young Chinese male presenting with sequential, painless, bilateral visual loss in Hong Kong. He was diagnosed to have Leber’s hereditary optic neuropathy with genetic workup showing G11778A mutation with over 80% heteroplasmy. He was started on idebenone treatment 11 months after onset of the binocular disease. To our best knowledge, this is the first case of Leber’s hereditary optic neuropathy treated with idebenone in Hong Kong. The recent evidence of the diagnosis and treatment of this devastating disease is reviewed.
 
 
Introduction
Leber’s hereditary optic neuropathy (LHON) is the commonest mitochondrial disease found to affect around 1 in 30 000 people in the first population-based clinical and molecular genetic study carried out in the UK.1 It is characterised by sequential bilateral visual loss, optic nerve dysfunction, and retinal ganglion cell degeneration. Characteristic visual field defect in LHON includes either central or cecocentral scotomas. Approximately 70% of patients were found to be young adults in the UK study. Over 20 mitochondrial DNA point mutations have been reported in LHON worldwide.2 Around 95% of the LHON pedigree have one of the three mitochondrial DNA point mutations namely, G3460A, G11778A, and T14484C, which are responsible for coding of complex I subunits of the mitochondrial respiratory chain.
 
Case report
A 15-year-old Chinese male, with a history of congenital red-green colour blindness, presented with sudden blurring of right eye central vision in August 2011. He did not experience any ocular pain or photopsia. There was no family history of ocular or neurological diseases.
 
On admission, his visual acuity was 6/120 in the right eye and 6/6 in the left eye. Right-eye visual field examination showed central scotoma associated with right relative afferent papillary defect. Fundoscopy revealed right eye disc swelling without peripapillary haemorrhage or exudate (Fig a). Neurological and cardiovascular examinations were normal.
 

Figure. (a) Right optic disc swelling without haemorrhage or exudates. (b) Fundus fluorescent angiography of the left eye shows no leakage of optic disc head
 
Perimetry revealed right centrocecal scotoma. Computed tomography of brain and orbit with contrast showed normal eye globes and extra-ocular muscles but mildly thickened intra-orbital right optic nerve suggestive of right optic neuritis. Erythrocyte sedimentation rate and C-reactive protein were not elevated. Blood test for anti-nuclear antibody was negative. Serologies for toxoplasmosis, herpes virus, Lyme disease and syphilis as well as tuberculin test were negative. Visual evoked potential revealed prolonged P100 over the right eye compatible with optic neuropathy. Magnetic resonance imaging of the brain and orbit was unremarkable. The patient was diagnosed to have optic neuritis and recommended for steroid treatment according to Optic Neuritis Treatment Trial (ONTT).3 However, his parents declined the treatment due to the possible side-effects of steroids.
 
Two months later, the patient experienced sudden painless blurring of left eye central vision. Visual acuity was 6/60 in the left eye while it remained at 6/120 in the right eye. Cerebrospinal fluid examination revealed normal biochemistry and cell count, and was negative for oligoclonal bands. Serum folate and vitamin B12 levels were normal. He was started on an ONTT-recommended dose of intravenous methylprednisolone (1000 mg/day) for 3 days, followed by oral prednisolone (1 mg/kg/day) for 11 days. However, his visual function deteriorated despite treatment. In view of the atypical features of sequential visual bilateral optic neuritis without recovery, LHON was suspected and confirmed by identification of mitochondrial DNA point mutation at 11778 guanine to adenine in the blood, with high mutant load heteroplasmy (over 80%). Fundus fluorescent angiography revealed left-disc telangiectatic vessels without optic disc head leakage compatible with typical angiographic features of LHON (Fig b).
 
He was then put on high-dose oral antioxidants (vitamin C 500 mg daily and co-enzyme Q10 ubiquinone). The medication was switched to idebenone 900 mg per day in divided doses when the drug became available in the hospital.
 
Six months after treatment with idebenone, his visual acuity remained static: 1/60 and 2/60 in the right and left eye, respectively. There were no side-effects related to the treatment. He gradually adapted to school life, with improvement in using Braille. Genetic study revealed the mother to be a LHON carrier (G11778A). His sister refused genetic testing due to the possible implications on future employment and academic considerations.
 
Discussion
Optic neuritis is an inflammatory disease of the optic nerve. It is the second commonest acquired optic nerve disorder in persons under 50 years old. The typical presenting symptom includes sudden visual loss with dyschromatopsia over several hours to days. This may be associated with dull retrobulbar pain which worsens with ocular movement. Symptoms can occur in both eyes either simultaneously or sequentially. A clinical diagnosis of optic neuritis may be made based on the following signs and symptoms: sudden visual loss with progression within 1 week, dyschromatopsia, pain on extra-ocular movement, no features of uveitis, and vision improvement beginning in 3 to 4 weeks.4 According to the findings in ONTT, 80% of patients with optic neuritis will improve within the first 3 weeks.5 Over 95% will regain visual acuity of at least 20/40, regardless of treatment options at 12 months’ time from onset of symptoms.6 The initial presentation of our patient was similar to that of optic neuritis. However, failure of spontaneous recovery in the absence of other causes should alert the clinician to consider atypical optic neuritis. The differential diagnoses of atypical optic neuritis are listed in the Table. A detailed history and clinical examination helps to understand the underlying aetiology, which may be confirmed by relevant radiological, serological, bacteriological, electrophysiological, and molecular investigations.
 

Table. Differential diagnoses of atypical optic neuritis
 
Patients with LHON experience devastating visual loss, typically worse than 20/200 in both eyes. At least 97% of patients will have sequential eye involvement within 1 year.7 Over 90% of patients harbour one of the three pathogenic mitochondrial mutations (G3460A, G11778A, T14484C), which affect complex I of the mitochondrial respiratory chain.8 The pathogenesis of LHON involves a combination of decreased complex I–driven adenosine triphosphate production, increase in free radical production and, finally, retinal ganglion cell apoptosis.9 The most important prognostic factor for visual recovery is the mutation status. The T14484C mutation carries the best chance of some degree of visual improvement in 37% to 71% of patients, while the G11778A mutation is associated with only 4% chance of recovery.7
 
Kirkman et al10 suggested that visual loss occurs more often in smokers and people with high alcohol intake. It is essential to advise patients to avoid tobacco smoking, excessive alcohol intake, and medications that may have mitochondrial toxicity (eg aminoglycosides, metformin, statins), especially during the acute phase of visual loss. Directed therapies for mitochondrial disorders are very limited. To date, there is no effective treatment for LHON. The mainstay of management remains supportive, such as low visual aids to assist reading, communication, and employment.
 
Anecdotal reports have shown beneficial effects of idebenone, which is a short-chain benzoquinone structurally related to co-enzyme Q10. It is a potent antioxidant and inhibitor of lipid peroxidation. It facilitates mitochondrial electron flux in bypassing complex I.11 In a retrospective open-label study involving 28 LHON patients, Mashima et al12 reported significantly shortened onset of visual recovery (11.1 months vs 17.4 months; P=0.03) and shortened interval for recovery of vision to ≥0.3 (17.6 months vs 34.4 months; P=0.01) in the treated group versus the untreated group. Jancic13 administered idebenone to nine patients at 135 mg/day for up to 1 year. Three patients reported subjective improvement in visual acuity, one with monocular disease showed arrest of progression of visual loss to the other eye, and four demonstrated improvement in visual evoked potential. Klopstock et al14 conducted a 24-week multicentre double-blind, randomised, placebo-controlled trial in 85 patients given idebenone at a dose of 900 mg/day. There was no significant difference in the overall group for best recovery of visual acuity. However, in the subgroup with discordant visual acuities at baseline (n=30), a significant trend for improvement in visual acuity was observed in the treated group. Idebenone was well tolerated with no adverse effect. In summary, current evidence suggests that idebenone is probably beneficial in the early-stage LHON patients with discordant visual acuities, and may help to shorten the interval of visual recovery.
 
In our patient, the static eye condition in the initial 6 weeks had prompted extensive diagnostic investigations including molecular study. However, the patient developed binocular involvement 2 months after onset of the symptoms. As idebenone was not available with the local pharmaceutical company, ordering of the drug from overseas had involved time and administrative process. The poor response of visual symptoms to idebenone might be explained by the late introduction of the treatment, in this case, 11 months after binocular involvement.
 
In our patient, the interval from disease onset till binocular involvement was only 2 months. Heightened physician awareness is important to identify this devastating condition during this interval, which may represent the golden window for initiation of idebenone to prevent sequential visual loss and hasten recovery. Molecular analysis for the three common mutations, which is available locally, aids definitive diagnosis, prognostication, and detection of presymptomatic family members. Those presymptomatic family members with positive mutation require lifelong surveillance, lifestyle modification, and prompt intervention at disease onset.
 
Acknowledgement
The authors would like to thank Ms Josephine Lui, pharmacist of Caritas Medical Centre, for her arduous effort to source idebenone for this patient.
 
References
1. Man PY, Griffiths PG, Brown DT, Howell N, Turnbull DM, Chinnery PF. The epidemiology of Leber hereditary optic neuropathy in the North East of England. Am J Hum Genet 2003;72:333-9. CrossRef
2. Brown MD, Wallace DC. Spectrum of mitochondrial DNA mutations in Leber’s hereditary optic neuropathy. Clin Neurosci 1994;2:138-45.
3. Beck RW, Cleary PA, Anderson MM Jr, et al. A randomized, controlled trial of corticosteroids in the treatment of acute optic neuritis. The Optic Neuritis Study Group. N Engl J Med 1992;326:581-8. CrossRef
4. Gal RL, Vedula SS, Beck R. Corticosteroids for treating optic neuritis. Cochrane Database Syst Rev 2012;(4):CD001430.
5. Beck RW, Cleary PA, Backlund JC. The course of visual recovery after optic neuritis. Experience of the Optic Neuritis Treatment Trial. Ophthalmology 1994;101:1771-8. CrossRef
6. Beck RW, Cleary PA. Optic neuritis treatment trial. One-year follow-up results. Arch Ophthalmol 1993;111:773-5. CrossRef
7. Newman NJ, Biousse V, David R, et al. Prophylaxis for second eye involvement in Leber hereditary optic neuropathy: an open-labeled, nonrandomized multicenter trial of topical brimonidine purite. Am J Ophthalmol 2005;140:407-15. CrossRef
8. Harding AE, Sweeney MG, Govan GG, Riordan-Eva P. Pedigree analysis in Leber hereditary optic neuropathy families with a pathogenic mtDNA mutation. Am J Hum Genet 1995;57:77-86.
9. Fraser JA, Biousse V, Newman NJ. The neuro-ophthalmology of mitochondrial disease. Surv Ophthalmol 2010;55:299-334. CrossRef
10. Kirkman MA, Yu-Wai-Man P, Korsten A, et al. Gene-environment interactions in Leber hereditary optic neuropathy. Brain 2009;132:2317-26. CrossRef
11. Haefli RH, Erb M, Gemperli AC, et al. NQO1-dependent redox cycling of idebenone: effects on cellular redox potential and energy levels. PLoS One 2011;6:e17963. CrossRef
12. Mashima Y, Kigasawa K, Wakakura M, Oguchi Y. Do idebenone and vitamin therapy shorten the time to achieve visual recovery in Leber hereditary optic neuropathy? J Neuroophthalmol 2000;20:166-70. CrossRef
13. Jancic J. Effectiveness of idebenone therapy in Leber’s hereditary optic neuropathy. Proceedings of 11th European College of Neuropsychopharmacology (ECNP) Regional Meeting; 2011 Apr 14-16; St. Petersburg, Russian Federation. Amsterdam: Elsevier; 2011; 21: S175.
14. Klopstock T, Yu-Wai-Man P, Dimitriadis K, et al. A randomized placebo-controlled trial of idebenone in Leber’s hereditary optic neuropathy. Brain 2011;134:2677-86. CrossRef

Tumefactive acute disseminated encephalomyelitis complicating human swine influenza (H1N1)

DOI: 10.12809/hkmj134049
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Tumefactive acute disseminated encephalomyelitis complicating human swine influenza (H1N1)
Amanda CY Chan, FHKCP, FHKAM (Medicine)1; SH Ng, FRCP (Lond & Edin), FHKAM (Medicine)2
1 Department of Medicine and Geriatrics, Tuen Mun Hospital, Tuen Mun, Hong Kong
2 Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
 
Corresponding author: Dr Amanda CY Chan (acychan@gmail.com)
 
 Full paper in PDF
Abstract
This report illustrates an adult patient presenting with tumefactive acute disseminated encephalomyelitis complicating human swine influenza. Its presentation, diagnosis, investigation findings, course, and response to treatment are discussed herein.
 
 
Introduction
A new outbreak of influenza caused by a new strain of H1N1, also known as ‘human swine influenza’ was first described in April 2009 in Mexico. This strain was different from the rest, in that it had a propensity to infect very healthy and young subjects, and also caused severe manifestations, such as acute respiratory distress, pneumonia, and even death. Approximately 80% of affected patients were younger than the age of 25 years.1
 
Since April 2009, there have been few reports of the neurological complications of human swine influenza.2 3 4 We report a case of severe human swine influenza causing acute demyelinating encephalomyelitis of the tumefactive form.
 
Case report
A 21-year-old woman with a history of diplegic cerebral palsy and epilepsy was hospitalised for breathlessness, cough, and fever for 3 days in January 2011. The initial chest X-ray was unremarkable, the white cell count (WCC) was elevated (13 x 109 /L; reference range [RR], 3.4-9.6 x 109 /L) and showed neutrophil predominance (8.9 x 109 /L; RR, 1.27-6.2 x 109 /L). An initial nasopharyngeal swab for influenza A and influenza B was negative. She was treated empirically with amoxicillin and clavulanic acid. She developed generalised tonic-clonic convulsion and desaturation 2 days later, for which she was intubated and received intensive care. Computed tomography of the brain showed multiple patchy hypodensities at the grey/white junction and white matter of frontal, parietal and temporal lobes on both sides, suspicious of underlying white matter disease.
 
Bedside bronchoscopy showed an inflamed mucosa, a small-sized airway with a distorted right bronchus, and purulent sputum. Bronchoalveolar lavage was positive for human swine influenza. The patient was given a course of oseltamivir, and later she received treatment with piperacillin and tazobactam.
 
One week later, the patient remained comatosed despite discontinuing sedation. Physical examination showed an absent deep pain response, wandering eyes with bilateral tonic pupils, and sluggish response to light. The doll’s eye reflex was absent, and the limbs were hypotonic and areflexic.
 
Autoimmune blood testing revealed nil abnormal. Her erythrocyte sedimentation rate and C-reactive protein level were elevated at 71 mm/h (RR, 5-15 mm/h) and 92 mg/L (RR, 0-10 mg/L), respectively. The serum antibody titre for influenza type A showed a significant increase from 10 to 640 over 10 days. Other virus and atypical pneumonia titres were also negative.
 
Electroencephalogram showed alpha coma pattern, and intermittent generalised slow waves at 1-2 Hz, 50-100 µV. Periodic lateralised epileptiform discharges at 1 Hz, 40-60 µV were evident over the right frontocentral region lasting for 4 to 5 seconds. Overall, the features were supportive of severe encephalopathy and cerebral dysfunction.
 
Lumbar puncture yielded a high opening pressure, and cerebrospinal fluid (CSF) protein was elevated at 4.26 g/L (RR, 0.1-0.4 g/L), glucose 2.2 mmol/L (RR, 2.2-3.9 mmol/L), WCC 0.3 x 106 /L, red cell count 0.6 x 106 /L, oligoclonal bands and Gram stain were negative. Three serial CSF specimens were sent for influenza A viral titres and showed an upward trend. Herpes simplex virus polymerase chain reaction (PCR), tuberculosis PCR, and Cryptococcus were negative.
 
Magnetic resonance imaging (MRI) of the brain, cervical and thoracic spines showed multiple T1-weighted hypointense, and T2-weighted hyperintense lesions up to 3 cm in diameter in the cerebral white matter bilaterally, the genu and splenium of the corpus callosum, external and internal capsules, midbrain, dorsal pons, and medulla oblongata. These lesions were contrast non-enhancing. The lesions involving the cerebral white matter and corpus callosum showed ring-like peripheral restricted diffusion.
 
Long segments of T1-weighted hypointense and T2-weighted hyperintense lesions were detected along the whole spinal cord, with the cervical cord being the most severely involved. The lesions at the cervical cord were continuous with that at the medulla oblongata (Fig). Overall, the features were in favour of acute disseminated encephalomyelitis (ADEM) with tumefactive demyelination.

Figure. Magnetic resonance imaging of the (a to d) brain, and (e) cervical and (f) thoracic spines showing multiple ring-like lesions up to 3 cm in the white matter bilaterally, involving the corpus callosum, external and internal capsules, midbrain, dorsal pons, medulla oblongata (arrows), and along the whole spinal cord (in parentheses). They were (a) T1-weighted hypointense, (b and d) T2-weighted hyperintense, (c) contrast–non-enhancing, with restricted diffusion; collectively in favour of tumefactive acute disseminated encephalomyelitis
 
This patient was treated with pulsed methylprednisolone, and later given two courses of intravenous immunoglobulin. However, there was no neurological improvement and the patient finally succumbed.
 
Discussion
Since the first appearance of human swine influenza in April 2009 until now, there have been reports of neurological complications that mostly occurred in the paediatric population. A few were also reported in adults,2 3 4 but in them the presentations were not as florid and radiologically severe. We report this case of a 21-year-old, ambulatory and independent woman, with a history of cerebral palsy and epilepsy. She is one of the few adults to have ADEM as a complication of human swine influenza, and the first reported to have the tumefactive form.
 
Acute disseminated encephalomyelitis is an inflammatory demyelinating disorder of the central nervous system (CNS), which is thought to be due to a T-cell hypersensitivity reaction.5 6 It is one of the many syndromes that can develop after vaccination or a microbial infection, and has a 2- to 30-day latency period.3 7
 
The typical MRI appearance is of demyelinating lesions preferentially affecting white matter tracts in a periventricular distribution. Diagnostic difficulty occurs whenever these demyelinating lesions appear to be solitary, large, or tumefactive. Tumefactive lesions are usually defined as solitary lesions, typically greater than 2 cm in diameter and imaging characteristics resembling a tumour. They tend to be circumscribed and have little in the way of mass effect or vasogenic oedema, typically involving the supratentorium, and are centred within the white matter, although they may extend to involve the cortical grey matter. The exact pathogenesis is unknown. Approximately half of tumefactive demyelinating lesions show pathological contrast enhancement, usually in the form of rings. Commonly they occur in the form of an open ring, with the incomplete portions on the grey matter side of the lesion. The enhancing portion of the ring is believed to represent the leading edge of demyelination and thus favours the white matter side of the lesion. The central non-enhancing core represents a more chronic phase of the inflammatory process.8
 
One must distinguish between infectious and post-infectious encephalitis, as all causes of the former should be excluded before concluding to the latter diagnosis. This involves systemic screening for herpes CNS infections, viruses endemic to specific regions, and other common causes of infective encephalitis. Other mimickers of ADEM include CNS lymphomas, systemic diseases like systemic lupus erythematosus, CNS vasculitis, and vascular, toxic or infectious leukoencephalopathies.5 The time course of ADEM, however, is usually hyperacute or acute, whereas the others are usually more chronic. Multiple sclerosis (MS) is also a differential diagnosis, but less likely as in the CSF oligoclonal bands were not present and protein was elevated, and radiologically there were no plaques or lesions disseminated in time. Although in most cases, ADEM is seemingly diagnosed clinically by exclusion, the definitive diagnosis of ADEM is histopathological. Lesions are usually bilateral, although not symmetrical, and mainly they involve the cerebral white matter and brainstem. Occasionally the cerebellum and spinal cord are involved. Small veins and venules in the white matter are surrounded by lymphocytes, macrophages and occasional plasma cells, whereas arteries and arterioles are relatively free of inflammation. Perivascular haemorrhages, axonal fragmentation, inflammatory cells within the leptomeninges, and subpial demyelination in the brainstem and spinal cord may be present.7
 
For our patient, the diagnosis of tumefactive ADEM was mainly made on clinical grounds and typical radiological features. However, postmortem brain biopsy was not performed. We undertook reverse-transcription (RT) PCR analysis for swine flu on CSF samples, but all results came back negative. However, paired CSF and serum samples for influenza A viral titres showed an increasing trend. For patients with suspected neurological complications of swine flu, the sensitivity and specificity of RT PCR and viral titres specifically on CSF samples have not been studied in detail and warrant further investigation. Previous reports of children with influenza A encephalitis in the US showed that CSF PCR were all negative in the three cases.9
 
The treatment of ADEM is borrowed from that of MS. First-line treatment mainly involves corticosteroids, which have been found to shorten the duration of symptoms and halt disease progression. Patients are given 6-methylprednisolone 6 to 8 g over 6 to 8 days, followed by oral prednisolone at tapering doses, but the prognosis remains variable. Approximately 80% of patients have a full recovery and ADEM is classically a monophasic disease. However, relapses have been reported in 5% to 10% of cases. If relapses occur on more than one occasion, a diagnosis of MS rather than multiphasic disseminated encephalomyelitis is probably more likely. Around 30% of patients are non-responders to steroids. Half of these non-responders benefit from treatment with intravenous immunoglobulin. Some authors recommend the use of cyclophosphamide in patients at high risk for relapse, either during the first attack or when relapse occurs. However, overall results have been disappointing.10
 
Conclusion
Herein we report one of the few adult cases of severe tumefactive demyelinating ADEM complicating human swine influenza infection. As different strains of influenza continue to spread throughout the world and in different populations, it is expected that more neurological complications will be reported. As it seems that neurological complications are more common in young age-groups with existing neurological diseases, prophylactic vaccination should be considered for such patients. In addition, resorting to early antiviral and immunomodulating therapy should also be emphasised for this patient group.
 
References
1. Novel Swine-Origin Influenza A (H1N1) Virus Investigation Team, Dawood FS, Jain S, Finelli L, et al. Emergence of a novel swine-origin influenza A (H1N1) virus in humans. N Engl J Med 2009;360:2605-15. CrossRef
2. Kimura E, Okamoto S, Uchida Y, et al. A reversible lesion of the corpus callosum splenium with adult influenza-associated encephalitis/encephalopathy: a case report. J Med Case Rep 2008;2:220. CrossRef
3. Athauda D, Andrews TC, Holmes PA, Howard RS. Multiphasic acute disseminated encephalomyelitis (ADEM) following influenza type A (swine specific H1N1). J Neurol 2012;259:775-8. CrossRef
4. Wang J, Duan S, Zhao J, Zhang L. Acute disseminated encephalomyelitis associated with Influenza A H1N1 infection. Neurol Sci 2011;32:907-9. CrossRef
5. Gupte G, Stonehouse M, Wassmer E, Coad NA, Whitehouse WP. Acute disseminated encephalomyelitis: a review of 18 cases in childhood. J Paediatr Child Health 2003;39:336-42. CrossRef
6. Ozkale Y, Erol I, Ozkale M, Demir S, Alehan F. Acute disseminated encephalomyelitis associated with influenza A H1N1 infection. Pediatr Neurol 2012;47:62-4. CrossRef
7. Love S. Demyelinating diseases. J Clin Pathol 2006;59:1151-9. CrossRef
8. Given CA 2nd, Stevens BS, Lee C. The MRI appearance of tumefactive demyelinating lesions. AJR Am J Roentgenol 2004;182:195-9. CrossRef
9. Centers for Disease Control and Prevention (CDC). Neurologic complications associated with novel influenza A (H1N1) virus infection in children—Dallas, Texas, May 2009. MMWR Morb Mortal Wkly Rep 2009;58:773-8.
10. Marchioni E, Tavazzi E, Minoli L, et al. Acute disseminated encephalomyelitis. Neurol Sci 2008;29 Suppl 2:S286-8. CrossRef

Primitive neuroectodermal adrenal gland tumour

DOI: 10.12809/hkmj134127
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
CASE REPORT
Primitive neuroectodermal adrenal gland tumour
YP Tsang, MB, ChB, MRCS(Ed); Brian HH Lang, MS, FRACS; SC Tam, MB, BS, MRCS(Ed); KP Wong, MB, BS, FRCSEd (Gen)
Division of Endocrine Surgery, Department of Surgery, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong
 
Corresponding author: Dr Brian HH Lang (blang@hku.hk)
 
 Full paper in PDF
Abstract
Ewing’s sarcoma, also called primitive neuroectodermal tumour of the adrenal gland, is extremely rare. Only a few cases have been reported in the literature. We report on a woman with adult-onset primitive neuroectodermal tumour of the adrenal gland presenting with progressive flank pain. Computed tomography confirmed an adrenal tumour with invasion of the left diaphragm and kidney. Radical surgery was performed and the pain completely resolved; histology confirmed the presence of primitive neuroectodermal tumour, for which she was given chemotherapy. The clinical presentation of this condition is non-specific, and a definitive diagnosis is based on a combination of histology, as well as immunohistochemical and cytogenic analysis. According to the literature, these tumours demonstrate rapid growth and aggressive behaviour but there are no well-established guidelines or treatment strategies. Nevertheless, surgery remains the mainstay of local disease control; curative surgery can be performed in most patients. Adjuvant chemoirradiation has been advocated yet no consensus is available. The prognosis of patients with primitive neuroectodermal tumours remains poor.
 
 
Introduction
Ewing’s sarcoma (ES), also called primitive neuroectodermal tumours (PNET), is a rare cancer of presumed neuroectodermal origin and is mostly found in children and young adults.1 Since it usually involves the diaphysis of long bones, adrenal ES/PNET is extremely rare. To our knowledge, only a handful of cases have been reported in the literature. Herein we present a patient with adult-onset adrenal PNET and discuss the diagnostic and management issues.
 
Case report
A 37-year-old woman presented to our hospital in May 2013 with progressive pain over left flank and abdomen for 1 month. Physical examination revealed a ballotable mass over the left flank. Contrast computed tomography (CT) demonstrated a 12-cm left adrenal mass with infiltration to the left crus of diaphragm and upper pole of the left kidney (Fig). On closer examination, the left renal artery was encased by the tumour but there was no evidence of distant metastases. Hormonal workup of this adrenal mass suggested that it was a non-functioning tumour. There was no excess in 24-hour urinary catecholamines and metanephrines, while 24-hour urinary free cortisol excretion was 71 (reference range [RR], 24-140) nmol. The aldosterone-renin ratio was 9.0 (reference level, <20) ng/dL per ng/(mL•h). Luteinising hormone, follicle-stimulating hormone, and testosterone levels were 3.0 (RR, 1.2-103.0) IU/L, 6.8 (1.8-22.5) IU/L, and 0.83 (0.35-2.60) nmol/L, respectively. The dehydroepiandrosterone level was 4.5 (RR, 2-11) µmol/L. Owing to increasing pain and the rapidly growing tumour, she underwent surgery, which revealed a 12-cm adrenal tumour compressing the aorta and superior mesenteric artery, and the posterior part of the diaphragm and left renal artery were invaded by the tumour bulk. Accordingly, she underwent a left adrenalectomy together with radical nephrectomy and partial resection of the diaphragm. No gross tumour was left behind. Thereafter, she made a good uneventful recovery and was discharged on the fifth day. Her back pain resolved completely after surgery.
 

Figure. Left adrenal tumour with infiltration to the left diaphragmatic crus (solid arrow) and the left kidney (dotted arrow)
 
Histology revealed a monotonous population of small round tumour cells with hyperchromatic nuclei, small nucleoli, and minimal cytoplasm arranged in sheets and cords with occasional vague rosette formation. Karyorrhexis, mitosis, and ‘starry sky’ pattern were focally observed. Immunohistochemically, the tumour cells stained positively for MIC2 antigen (CD99), vimentin, and FLI1. By contrast they stained negatively for cytokeratin, leukocyte common antigen, MPO, WT1, synaptophysin, S100, desmin, myogenin, CD68, and CD34. The reverse transcriptase–polymerase chain reaction confirmed a reciprocal translocation of t(11;22)(q24;q12) involving the EWSR1 gene on chromosome 22 and the FLI1 gene on chromosome 11 (ie EWSR1-FLI1 translocation). Overall, these findings were consistent with ES/PNET.
 
She received adjuvant chemotherapy (cyclophosphamide, adriamycin, and vincristine alternating with ifosfamide and etoposide). The latest CT at 5 months post-surgery revealed no evidence of local or distant recurrence.
 
Discussion
Ewing’s sarcoma/PNET of the adrenal gland is rarely reported.2 3 Our review of the English literature revealed only 10 reports describing 16 patients (Table).3 4 5 6 7 8 9 10 11 12 As in our patient, CD99, a highly sensitive marker for PNET, was found in all 16 of these patients. Although not routinely looked for, as in our patient over 90% of PNET cases exhibit a reciprocal translocation of t(11;22)(q24;q12) involving the EWSR1 gene on chromosome 22 and the FLI1 gene on chromosome 11.1 3 6 7 8 9 11 12 These 16 reported cases had a median age at diagnosis of 26 (range, 20-74) years, and the female-to-male ratio was 3:1. The median tumour size was 10.5 (range, 3-17) cm.
 

Table. Summary of case reports regarding clinical conditions3 4 5 6 7 8 9 10 11 12
 
Nine of these patients underwent curative surgery despite rapid growth and aggressive tumour behaviour.4 7 8 12 13 Nevertheless, in a few patients the tumour behaves indolently or has a relatively quiescent period before rapid growth. A case of incidental PNET at autopsy has been reported in a 26-year-old man who committed suicide by hanging himself; his latent PNET measured 8 cm.5 Similarly, another patient aged 53 years underwent a laparoscopic adrenalectomy for a presumed non-functional incidental adrenocortical adenoma, but turned out to be a 3-cm PNET.8
 
Diagnostic issues
It is difficult to make a definitive diagnosis of ES/PNET before surgery because currently it is based on a combination of histology, immunohistochemistry, and cytogenetic analysis.4 8 Although needle biopsy is a possible approach to making a definitive diagnosis without resection, it is rarely performed unless the tumour is considered not resectable or adrenal metastases are suspected. Since our patient had a potentially resectable tumour with no evidence of distant metastases, needle biopsy was not considered. Furthermore, she was clearly suffering pain arising from the tumour and therefore surgical resection was clearly indicated.
 
Management issues
Due to its rarity, there are no well-established guidelines or treatment strategies for PNET. Surgical resection remains the mainstay for local disease control.4 7 8 Since the tumour is believed to be both chemo- and radio-sensitive, adjuvant chemotherapy and radiotherapy have often been advocated for better local and distant control, but there is no consensus.13 A standard regimen of chemoirradiation is not yet established due to the sporadic nature of cases.13 Most chemotherapy regimens for adult-onset PNET are akin to those for ES in children as both share the same origin.4 14 Chemotherapeutic agents such as cyclophosphamide, adriamycin, vincristine, ifosfamide, or etoposide have been used.4 7 8 Recently, it has been noted that chemotherapy might be effective only for the first few cycles, and that the tumours develop resistance very quickly.4 Adjuvant radiotherapy had been used for local recurrences as well as unresectable or incompletely resected PNETs.4 12 From our review, adrenalectomies were performed for 11 patients, while chemotherapy and radiotherapy was also given to six and three of them, respectively. Follow-up revealed that these tumours were aggressive; three of the 16 patients died within 1 year of diagnosis or surgery. Regarding the six patients who were still surviving when this report was submitted, one had distant metastasis and another two had local recurrence. Because of this high rate of recurrence, intensive follow-up with regular CT scans (every 6 months) has been advocated even after seemingly curative surgery.12
 
Conclusion
Herein we report a rare case of adult adrenal PNET. The clinical presentation is often vague and non-specific and a definitive diagnosis depends on a combination of histology, immunohistochemistry, and cytogenetic analysis. Surgical resection remains the mainstay of treatment coupled with adjuvant chemoirradiation. Nevertheless, the prognosis appears poor.
 
References
1. Grier HE. The Ewing family of tumors. Ewing’s sarcoma and primitive neuroectodermal tumors. Pediatr Clin North Am 1997;44:991-1004. CrossRef
2. Quezado M, Benjamin DR, Tsokos M. EWS/FLI-1 fusion transcripts in three peripheral primitive neuroectodermal tumors of the kidney. Hum Pathol 1997;28:767-71. CrossRef
3. Renshaw AA, Perez-Atayde AR, Fletcher JA, Granter SR. Cytology of typical and atypical Ewing’s sarcoma/PNET. Am J Clin Pathol 1996;106:620-4.
4. Zhang Y, Li H. Primitive neuroectodermal tumors of adrenal gland. Jpn J Clin Oncol 2010;40:800-4. CrossRef
5. Yamamoto T, Takasu K, Emoto Y, et al. Latent adrenal Ewing sarcoma family of tumors: a case report. Leg Med (Tokyo) 2013;15:96-8. CrossRef
6. Matsuoka Y, Fujii Y, Akashi T, Gosehi N, Kihara K. Primitive neuroectodermal tumour of the adrenal gland. BJU Int 1999;83:515-6. CrossRef
7. Pirani JF, Woolums CS, Dishop MK, Herman JR. Primitive neuroectodermal tumor of the adrenal gland. J Urol 2000;163:1855-6. CrossRef
8. Komatsu S, Watanabe R, Naito M, et al. Primitive neuroectodermal tumor of the adrenal gland. Int J Urol 2006;13:606-7. CrossRef
9. Ahmed AA, Nava VE, Pham T, et al. Ewing sarcoma family of tumors in unusual sites: confirmation by rt-PCR. Pediatr Dev Pathol 2006;9:488-95. CrossRef
10. Kim MS, Kim B, Park CS, et al. Radiologic findings of peripheral primitive neuroectodermal tumor arising in the retroperitoneum. AJR Am J Roentgenol 2006;186:1125-32. CrossRef
11. Abi-Raad R, Manetti GJ, Colberg JW, Hornick JL, Shah JG, Prasad ML. Ewing sarcoma/PNET arising in the adrenal gland. Pathol Int 2013;63:283-6. CrossRef
12. Lena M. Retroperitoneal primitive neuroectodermal tumour (PNET). A case report and review of the literature. Rep Practical Oncol Radiother 2009;14:221-4. CrossRef
13. Malpica A, Moran CA. Primitive neuroectodermal tumor of the cervix: a clinicopathologic and immunohistochemical study of two cases. Ann Diagn Pathol 2002;6:281-7. CrossRef
14. Bisogno G, Carli M, Stevens M, et al. Intensive chemotherapy for children and young adults with metastatic primitive neuroectodermal tumors of the soft tissue. Bone Marrow Transplant 2002;30:297-302. CrossRef

Pages