search
for
 About Bioline  All Journals  Testimonials  Membership  News


Journal of Postgraduate Medicine
Medknow Publications and Staff Society of Seth GS Medical College and KEM Hospital, Mumbai, India
ISSN: 0022-3859 EISSN: 0972-2823
Vol. 51, Num. 2, 2005, pp. 140-145

Journal of Postgraduate Medicine, Vol. 51, No. 2, April-June, 2005, pp. 140-145

Review Article

Melioidosis: An emerging infectious disease

Department of Medical Microbiology, University of Malaya Medical Center, Kuala Lumpur

Correspondence Address: Department of Medical Microbiology, University of Malaya Medical Center, Kuala Lumpur, Email: rajanadeem@um.edu.my

Date of Submission: 21-Feb-2005
Date of Decision: 20-Mar-2005
Date of Acceptance: 30-May-2005

Code Number: jp05056

ABSTRACT

Infectious diseases account for a third of all the deaths in the developing world. Achievements in understanding the basic microbiology, pathogenesis, host defenses and expanded epidemiology of infectious diseases have resulted in better management and reduced mortality. However, an emerging infectious disease, melioidosis, is becoming endemic in the tropical regions of the world and is spreading to non-endemic areas. This article highlights the current understanding of melioidosis including advances in diagnosis, treatment and prevention. Better understanding of melioidosis is essential, as it is life-threatening and if untreated, patients can succumb to it. Our sources include a literature review, information from international consensus meetings on melioidosis and ongoing discussions within the medical and scientific community.

Keywords: Melioidosis, Burkholderia pseudomallei , Infection

The name melioidosis [also known as Whitmore disease] is taken from the Greek word ′melis′ meaning distemper of asses and ′eidos′ meaning resembles glanders. Melioidosis is a zoonotic disease caused by Pseudomonas pseudomallei [now known as Burkholderia pseudomallei ], a bacillus that can cause disease in horses, sheep, goats, pigs, lambs, cows, and other animals, as well as in humans.[1],[2] There have been concerns of B. pseudomallei being thought of as a potential biological warfare agent.[3]

Prevalence and epidemiology

B. pseudomallei, the causative organism of melioidosis, is a free-living gram-negative, aerobic bacillus that is mainly widespread in Southeast Asia [most cases in Thailand, Malaysia, Vietnam, Cambodia, Laos, and Myanmar] and Northern Australia.[4],[5] There are some reports of its presence in other parts of the world such as Central America, the Caribbean, China, Taiwan, Africa, the Middle East and South Asian countries.[6],[7],[8],[9],[10] In Thailand 2000 to 3000 new cases are diagnosed every year.[11] In Malaysia, reported seroprevalence in healthy individuals is 17-22% in farmers [mainly rice farmers] and 26% in blood donors.[12] In North Australia 0.6 to 16% of children have evidence of infection by B. pseudomallei .[13] The B. pseudomallei have also been isolated in America.[3] There are reports of several cases of patients with melioidosis who have immigrated into Europe and the disease has been increasingly recognized in returning travellers to Europe from endemic areas.[14] The geographic area of the prevalence of the organism is bound to increase as the awareness increases.

B. pseudomallei
is a natural inhabitant of soil and water in the tropics and subtropics but can also survive in dry atmospheric conditions. It is ubiquitous in the rice-farming areas. It is also present in rubber plantations, cleared fields, cultivated and irrigated agricultural sites as well as drains and ditches. Its true prevalence however is not known, as there is under-reporting of its incidence due to the poorly understood disease process and misdiagnosis. At the same time enough resources are not always available in some areas to carry out research and increase the awareness of the general public and to educate and familiarise the medical profession about the disease.

Melioidosis affects all ages but peak incidence is mainly between 40 to 60 years of age, with male to female ratio of 1.4:1.[15] There is a good correlation between the isolation of the organism from soil and the seroprevalence of antibodies in the population living in that region.

Bacteriology

When isolated from blood, sputum, pus and other body fluids, B. pseudomallei appears like safety pins [bipolar] under the microscope with methylene blue stain. It grows aerobically on ordinary media at 37°C. Colonies are wrinkled and show dry daisy-head appearance along with a distinct odour. Mucoid colonies suggest that the patient is receiving antibiotic therapy. In laboratory culture of B. pseudomallei, growth of other organisms may result in false negative result. This problem could be resolved using Ashdown′s selective medium, which contains dyes, gentamicin and Trypticase peptone.[16] Recently, more improved B. pseudomallei selective agars [BPSA] have been developed to improve the recovery of B. pseudomallei .[17]

B. pseudomallei
can survive anaerobic conditions in the presence of acidic environment, and also survive in distilled water for several years.[18] The bacterium is resistant to penicillin, aminoglycosides, rifamycins and relatively insensitive to quinolones and macrolides. Therefore the therapeutic options are limited and continuous presence of the organism in patients is not fully understood.[19],[20] B. pseudomallei is resistant to macrolide and aminoglycoside antibiotics via a multidrug efflux pump.[20] Mutations within the conserved motifs of the beta-lactamase enzyme [enzyme that hydrolyses the cyclic amide bond of beta-lactam antibiotics] also account for the resistance patterns.[21]

Modes of transmission and pathogenesis

There are several established modes of transmission within the patient population. The possible modes are inhalation, ingestion or inoculation through the skin lesions from the contaminated soil.[22] Person-to-person transmission of B. pseudomallei especially between patient and his sibling or one of their playmates is common.[23] Vertical transmission [from mother to child] is possible.[24] It can also be transmitted by direct contact with infected rodents or infected food, soil, water, excreta; person-to-person transmission is also possible through use of injection needle. B. pseudomallei can also be transmitted through sexual intercourse.[25] The link between melioidosis and consumption of Kava [ Piper methysticum ] has also been seen.[26] Heavy rains and winds may cause increased inhalation of B. pseudomallei . [27] Interestingly, a container of commercial hand-wash detergent was a source of infection in Northern Australia. [28]

B. pseudomallei
attack several eukaryotic cell lines. In both phagocytic and nonphagocytic cell lines, it can escape from the specialised endocytic vacuoles into the cytoplasm to form actin-associated membrane protrusion that is thought to contribute to cell-to-cell spreading in the infected individuals.[29],[30],[31] Capsule and a type III secretion system [TTSS-expressed mainly by pathogenic bacteria that is used to introduce deleterious proteins called effectors into host cells] facilitate B. pseudomallei to survive, escape from endocytic vesicles, facilitate bacterial invasion of epithelial cells and intracellular survival.[32],[33] The uptake of B. pseudomallei by several cell lines in culture leads to induction of cell fusion and formation of a multinucleated large cell.[34] Production of nitric oxide has bactericidal activity and failure of infected cells to successfully control the growth and subsequent survival of intracellular B. pseudomallei are due to the suppression of inducible nitric oxide synthase [iNOS] by B. pseudomallei .[35],[36] However, interferons enhance antimicrobial activity of macrophage infected B. pseudomallei by up-regulating iNOS. [36],[37]

Clinical manifestations

B. pseudomallei can cause disease in apparently healthy individuals. Clinical manifestations of melioidosis range from localised infection to acute pneumonia and fulminant septic melioidosis.[38],[39],[40] Once infected, it may remain dormant and become active after months, years or decades when host is immunocompromised by drugs [steroids] or disease [diabetes mellitus, chronic renal failure, retrovirus infections, haematological malignancies, collagen vascular disease] or social deprivation [alcoholism, drug abuse, occupational exposure].[41] The factors that provoke the reactivation of latent pathogen probably are environmental variables, stress and immunity status.[42],[43] Localized melioidosis occurs in the form of acute suppurative lesions, superficial and deep-seated abscess in the psoas muscle, parotid glands and at the root of mesentery.[44],[45] It may also present as cellulitis, chronic otitis media and sepsis after burns and trauma.[45],[46] The other manifestations are mycotic aneurysm, pericarditis, osteomyelitis epididymo-orchitis and prostatitis.[47],[48],[49] Melioidosis is also associated with systemic lupus erythematosus.[50] Melioidotic prostatic abscesses are reported very rarely and are not easy to diagnose. In endemic areas, the elderly diabetic person who presents with high-grade fever and urinary obstruction may have B. pseudomallei in the prostate gland.[51] Central nervous system involvement including brain abscess is a rare complication with high mortality.[52],[53] The immune- suppressed patients present with melioidosis septicaemia and their clinical features are similar to other gram-negative septicaemias and its prognosis is poor. Quoted mortality ranges from 40% to 75% despite rational use of anti-microbial therapy.[54] Clinical risk factors and manifestations of melioidosis are summarized in [Table - 1].

Diagnosis

The diagnosis of acute or chronic melioidosis remains challenging. In endemic areas, melioidosis should be considered in the differential diagnosis of any Pyrexia of Unknown Origin [PUO], acute respiratory distress syndrome [ARDS] and acute septicaemia. The other conditions that melioidosis may present as are pneumonia, acute suppurative lesions, chronic granulomatous lesions, septic arthritis, osteomyelitis, epididymorchitis and mycotic aneurysm as well as radiological pattern of tuberculosis on the chest X-ray but not supplemented with mycobacterium tuberculosis positive sputum culture. In diabetic patients, laboratory results show leukocytosis, high level of glucose in blood and glycosylated haemoglobin, and high urea and creatinine.[55] In melioidosis, laboratory diagnosis is essential for successful patient management. C-reactive protein [CRP], an early indicator of infectious or inflammatory conditions may be elevated in melioidosis; however under normal CRP levels, melioidosis should not be ruled out.[56]

Identification of B. pseudomallei

Isolation of B. pseudomallei by culture from a clinical specimen [blood, urine, sputum, skin lesions and swab samples from throat] is the gold standard of diagnosis.[57] Correct identification of B. pseudomallei is essential for long term supportive therapy in the treatment of melioidosis. A few simple tests can be employed to identify B. pseudomallei in the endemic areas. These tests include positive oxidase test, bipolar gram staining, metallic sheen colonies on special media (Ashdown media which contains various dyes and gentamicin) and resistance to aminoglycosides. [58]

Conventional biochemical tests and API20E substrate-utilization test panel [bioMιrieux] kit is used for identification of B. pseudomallei; however, it can easily misidentify Chromobacterium violaceum ( C. violaceum ). In one study, polymerase chain reaction (PCR) results showed that isolated C. violaceum have similar repetitive extragenic pallindromic sequence (REPS) pattern with B. pseudomallei . [59]

Serological tests

Serological tests are helpful in making a provisional diagnosis in the absence of isolation of B. pseudomallei in the specimen. Culture and serological methods are cost-effective and simple to perform but require experience to interpret results. Slide agglutination test results in rapid identification of B. pseudomallei. Indirect haemagglutination test is simple to perform as it detects the antibody against B. pseudomallei that appears in the blood within 1-2 weeks after the infection and reach maximal titre in 4 to 5 months.[60] However its interpretation may be difficult because of the following points;

· False positive results due to cross-reaction with other gram negative bacteria [shares antigens (lipopolysaccharide of cell wall) particularly Burkholderia cepecia and Legionella species ],

· There may be rare false negative results,

· High antibody titre may persist for a long time after infection subsides.

Enzyme linked immunosorbent assay [ELISA] test detects specific IgG and IgM antibodies of B. pseudomallei in serum specimens. ELISA is more convincing in terms of sensitivity and specificity for antibody detection as it points to an active disease process. [61] The indirect ELISA is easy to perform and hence is recommended as a diagnostic serological test when melioidosis is in the differential diagnosis of PUO cases. Though Immunoflurorescent Antibody Assay is a rapid, highly sensitive and specific test for the identification of current infection, it requires a fluorescent microscope that is not always available in some laboratories in endemic areas.

Molecular identification techniques

During the past decade, many efforts have been made to develop new molecular procedures to identify B. pseudomallei from various specimens. Molecular biology techniques such as polymerase chain reaction [PCR], dot immunoassay, pulsed field gel electrophoresis [PFGE], restricted fragmentation length polymorphism [RFLP] and random amplification of particle of deoxyribonulease [RAPD] are also used for diagnosis. These are the recommended techniques for the rapid diagnosis of the disease and for monitoring therapy and epidemiological studies because of its high sensitivity, specificity, simplicity and speed. In recent times sensitive PCR amplification techniques for detecting the DNA of B. pseudomallei in clinical specimens, especially buffy coat specimens of acute melioidosis patients have been useful.[62],[63] Laboratory diagnostic approach is summarized in [Table - 2].

Management

The main objective of treatment is to reduce the mortality and morbidity in melioidosis. Before the advent of proper antimicrobials, the mortality of the melioidosis patients used to be around 95%. Rational use of antimicrobials has reduced it to half. Ceftazidime is the drug of choice in systemic melioidosis.[64],[65] Ceftazidime [120mg/kg/day], has shown to reduce the mortality significantly in severe melioidosis.[66] However, resistant strains are beginning to appear.[19] Carbapenem antibiotics are also suitable for the treatment of the disease .[67] Doxycycline can be used in localised infections in combination with cotrimoxazole .[68] In acute severe melioidosis, ceftazidime alone or in combination with co-trimoxazole or ciprofloxacin remains the drug of choice. Parenteral amoxyclav [160mg/kg/day for 8 weeks] is a substitute of ceftazidime and it reduces mortality.[69] Imipenem is a safe and effective treatment for acute severe melioidosis and may also be considered an alternative to ceftazidime.[70],[71] A study showed that meropenem [1g or 25 mg/kg, 8 hourly intravenously for ³14 days] can be considered as an alternative to ceftazidime and imipenem in the treatment of melioidosis but this is more expensive and more trials are required.[72]

In melioidosis, supportive therapy is an integral part of management for better prognosis as it includes management of shock and ARDS, drainage of pus, good control of diabetes in diabetics and good nursing care. The recommendation for parenteral antimicrobial therapy, when needed, is for at least two weeks before switching over to oral maintenance therapy for 12 to 20 weeks. [Table - 3]

Relapse and maintenance therapy

Appropriate treatment is imperative in order to prevent relapse and failure of treatment. Despite appropriate treatment, melioidosis has a higher relapse rate. The average time between discharge from hospital and relapse is of 21 weeks. Treated patients require long-term follow up, as B. pseudomallei remains latent for up to 26 years in the body.[73] For maintenance therapy, Co-Amoxyclav is a safe and well-tolerated antimicrobial agent [there is some concern that it may be less effective than the conventional regimen of chloramphenicol, co-trimoxazole and doxycycline]. The recommended duration for maintenance therapy is of 12 to 20 weeks.[74],[75]

It has been shown that B. pseudomallei stays intracellularly in the body where it produces biofilms and micro colonies and is sheltered from β-Lactam antimicrobial drugs [β-Lactam drugs are unable to enter intracellular sites to kill latent B. pseudomallei ].[76] It has been suggested that a combination of ciprofloxacin and macrolides is a good alternative regimen since ciprofloxacin penetrates phagocytic cells and achieves intracellular concentrations of several times higher than extracellular concentration and kills B. pseudomallei while macrolides could delay or prevent production of glycocalyx [Table - 3].[77],[78]

Prevention

Measurers for prevention require prompt cleansing of scrapes, burns, or other open wounds in endemic areas. Persons with diabetes and skin lesions should avoid contact with soil and standing-water in endemic areas. Protective clothing such as rubber boots and gloves during agricultural work can prevent infection through the feet and hands. It is important to maintain safe water through regular disinfection and safe storage of water for both human and animals bred for human consumption. Sewage wastes can attract insects and rodents and encourage the growth of B. pseudomallei. Therefore proper disposal of sewage wastes is essential in endemic areas. As dairy products can contain B. pseudomallei it is important that milk is pasteurized before consumption.[79]

There is currently no licensed vaccine available for protection against melioidosis. At present studies are underway to identify possible antigens using lipopolysacchrides of B. pseudomallei in mouse models.[80] Antibodies against B. pseudomallei flagellin reduce the motility of the bacterium and provide protection against melioidosis in animal models.[81] A recent study has shown that quicklime was able to inhibit the growth of B. pseudomallei in soil from a rice field.[82] As our understanding of the disease increases and as we move forward with the studies on the pathogenesis of the disease, new and effective vaccine against melioidosis may become a reality.

ACKNOWLEDGEMENTS

The authors would like to thank Professor Puthucheary for her tremendous support and guidance and Miss Teng Fui Enn for her valuable assistance in typing and proofreading of this article. We would also like to thank Dr Leelarasamee for assisting with [Table - 1].

REFERENCES
1.Dance DA. Melioidosis: The tip of the iceberg? Clin Microbiol Rev 1991;4:52-60.  Back to cited text no. 1  [PUBMED]  
2.Sprague LD, Neubauer H. Melioidosis in animals: a review on epizootiology, diagnosis and clinical presentations. J Vet Med 2004;51:305-20.  Back to cited text no. 2  [PUBMED]  [FULLTEXT]
3.Centers for Disease Control and Prevention (CDC). Laboratory exposure to Burkholderia pseudomallei -Los Angeles, California. MMWR Morb Mortal Wkly Rep 2004;53:988-90.  Back to cited text no. 3    
4.So SY. Melioidosis-an endemic problem in Southeast Asia. Medicine Digest Asia 1986;4:19-23.  Back to cited text no. 4    
5.Leelarasamee A. Recent development in melioidosis. Curr Opin Infect Dis 2004;17:131-6.  Back to cited text no. 5  [PUBMED]  
6.Miralles IS, Maciel Mdo C, Angelo MR, Gondini MM, Frota LH, dos Reis CM, et al. Burkholderia pseudomallei : a case report of a human infection in Ceará, Brazil. Rev Inst Med Trop Sao Paulo 2004;46:51-4.   Back to cited text no. 6  [PUBMED]  [FULLTEXT]
7.Christenson B, Fuxench Z, Morales JA, Suarez-Villamil RA, Souchet LM. Severe community-acquired pneumonia and sepsis caused by Burkholderia pseudomallei associated with flooding in Puerto Rico. Bol Asoc Med P R 2003;95:17-20.  Back to cited text no. 7  [PUBMED]  
8.Dance DA. Melioidosis as an emerging global problem. Acta Tropica 2000;74:115-9.  Back to cited text no. 8  [PUBMED]  
9.Dance DA. Melioidosis. Curr Opin Infect Dis 2002;15:127-32.  Back to cited text no. 9  [PUBMED]  
10.John TJ, Jesudason MV, Lalitha MK, Ganesh A, Mohandas V, Cherian T, et al. Melioidosis In India: the tip of the iceberg? Indian J Med Res 1996;103:62-5.  Back to cited text no. 10    
11.Leelarasamee A, Bovornkitti S. Melioidosis: Review and update. Rev Infect Dis 1989;11:413-25.  Back to cited text no. 11  [PUBMED]  
12.Vadivellu J, Puthucheary SD, Gendeh GS, Parasakthi N. Serodiagnosis of melioidosis in Malaysia. Singpore Med J 1995;36:299-302.  Back to cited text no. 12    
13.Currie BJ, Fisher DA, Howard DM, Burrow JN, Selvanayagam S, Snelling PL, et al. The epidemiology of melioidosis in Australia and Papua New Guinea. Acta Trop 2000;74:121-7.  Back to cited text no. 13    
14.Dance DA, Smith MD, Aucken HM, Pitt TL. Imported melioidosis cases in England and Wales. Lancet 1999;353:208.  Back to cited text no. 14  [PUBMED]  
15.Dance DAB. Melioidosis: Distribution and epidemiology. First international symposium on Melioidosis in Kuala Lumpur Malaysia 1994;7-8.  Back to cited text no. 15    
16.Ashdown LR. An improved screening technique for isolation of Pseudomonas pseudomallei from clinical specimens. Pathology 1979;11:293-7.  Back to cited text no. 16  [PUBMED]  
17.Howard K, Inglis TJ. Novel selective medium for isolation of Burkholderia pseudomallei . J Clin Microbiol 2003;41:3312-6.  Back to cited text no. 17    
18.Wongwanich S, Chotanachan P, Kondo E, Kanai K. Multifactorial pathogenic mechanisms of Burkholderia pseudomallei as suggested from comparison with Burkholderia cepacia . Southeast Asian J Trop Med Public Health 1996;27:111-8.  Back to cited text no. 18    
19.Dance DA, Wuthiekanun V, Chaowagul W, Suputtamongkol Y, White NJ. Development of resistance to ceftazidime and co-amoxyclav in Pseudomonas pseudomallei. J Antimicrob Chemother 1991;28:321-4.  Back to cited text no. 19    
20.Moore RA, DeShazer D, Reckseidler S, Weissman A, Woods DE. Efflux-Mediated Aminoglycoside and Macrolide Resistance in Burkholderia pseudomallei . Antimicrob Agents Chemother 1999;43:465-70.  Back to cited text no. 20    
21.Tribuddharat C, Moore RA, Baker P, Woods DE. Burkholderia pseudomallei class a beta-lactamase mutations that confer selective resistance against ceftazidime or clavulanic acid inhibition. Antimicrob Agents Chemother 2003;47:2082-7.  Back to cited text no. 21    
22.Howe C, Sampath A, Spotnitz M. The Pseudomallei group: A review. J Infect Dis 1971;124:598-606.  Back to cited text no. 22    
23.Holland DJ, Wesley A, Drinkovic D, Currie BJ. Cystic Fibrosis and Burkholderia pseudomallei Infection: An Emerging Problem? Clin Infect Dis 2002;35:138-40.  Back to cited text no. 23    
24.Abbink FC, Orendi JM, de Beaufort AJ. Mother to child transmission of Burkholderia pseudomallei. N Eng J Med 2001;344:1171-2.  Back to cited text no. 24    
25.McCormick JB, Sexton DJ, McMurray JG, Carey E, Hayes P, Feldman RA. Human-to-human transmission of Pseudomonas pseudomallei . Ann Intern Med 1975;83:512-3.  Back to cited text no. 25    
26.Currie BJ, Fisher DA, Howard DM, Burrow JN, Lo D, Selva-Nayagam S, et al . Endemic Melioidosis in Tropical Northern Australia: A 10-Year Prospective Study and Review of the Literature. Clin Infect Dis 2000;31:981-6.  Back to cited text no. 26    
27.Munckhof WJ, Mayo MJ, Scott I, Currie BJ. Fatal human melioidosis acquired in a subtropical Australian city. Am J Trop Med Hyg 2001;65:325-8.  Back to cited text no. 27    
28.Gal D, Mayo M, Smith-Vaughan H, Dasari P, McKinnon M, Jacups SP, et al . Contamination of hand wash detergent linked to occupationally acquired melioidosis. Am J Trop Med Hyg 2004;71:360-2.  Back to cited text no. 28    
29.Jones AL, Beveridge TJ, Woods DE. Intracellular survival of Burkholderia pseudomallei. Infect Immun 1996;64:782-90.  Back to cited text no. 29    
30.Kespichayawattana W, Rattanachetkul S, Wanun T, Utaisincharoen P, Sirisinha S. Burkholderia pseudomallei Induces Cell Fusion and Actin-Associated Membrane Protrusion: a Possible Mechanism for Cell-to-Cell Spreading. Infect Immun 2000;68:5377-84.  Back to cited text no. 30    
31.Breitbach K, Rottner K, Klocke S, Rohde M, Jenzora A, Wehland J, et al . Actin-based motility of Burkholderia pseudomalle i involves the Arp 2/3 complex, but not N-WASP and Ena/VASP proteins. Cell Microbiol 2003;5:385-93.  Back to cited text no. 31    
32.Stevens MP, Galyov EE. Exploitation of host cells by Burkholderia pseudomallei . Int J Med Microbiol 2004;293:549-55.  Back to cited text no. 32    
33.Ghosh P. Process of Protein Transport by the Type III Secretion System. Microbiol Mol Biol Rev 2004;68:771-95.  Back to cited text no. 33    
34.Harley VS, Dance DA, Drasar BS, Tovey G. Effects of Burkholderia pseudomallei and other Burkholderia species on eukaryotic cells in tissue culture. Microbios 1998;96:71-93.  Back to cited text no. 34    
35.Miyagi K, Kawakami K, Saito A. Role of reactive nitrogen and oxygen intermediates in gamma interferon-stimulated murine macrophage bactericidal activity against Burkholderia pseudomallei. Infect Immun 1997;65:4108-13.   Back to cited text no. 35    
36.Utaisincharoen P, Anuntagool N, Limposuwan K, Chaisuriya P, Sirisinha S. Involvement of beta interferon in enhancing inducible nitric oxide synthase production and antimicrobial activity of Burkholderia pseudomallei -infected macrophages. Infect Immun 2003;71:3053-7.  Back to cited text no. 36    
37.Utaisincharoen P, Anuntagool N, Arjcharoen S, Limposuwan K, Chaisuriya P, Sirisinha S. Induction of iNOS expression and antimicrobial activity by interferon (IFN)-beta is distinct from IFN-gamma in Burkholderia pseudomallei -infected mouse macrophages. Clin Exp Immunol 2004;136:277-83.  Back to cited text no. 37    
38.Raja NS. Localized melioidosis. J Pak Med Assoc 2003;53:373-4.  Back to cited text no. 38    
39.Koszyca B, Currie BJ, Blumbergs PC. The neuropathology of melioidosis: two cases and a review of the literature. Clin Neuropathol 2004;23:195-203.  Back to cited text no. 39    
40.Schwarzmaier A, Riezinger-Geppert F, Schober G, Karnik R, Valentin A. Fulminant septic melioidosis after a vacation in Thailand. Wien Klin Wochenschr 2000;112:892-5.  Back to cited text no. 40    
41.Currie B, Howard D, Nguyen VT, Withnall K, Merianos A. The 1990-1991 outbreak of melioidosis in the Northern Territory of Australia: clinical aspects. Southeast Asian J Trop Med Public Health 1993;24:436-43.  Back to cited text no. 41    
42.Johnson AB, Ali N. Reactivation of latent melioidosis. Postgrad Med J 1990;66:732-3.  Back to cited text no. 42    
43.Thummakul T, Wilde H, Tantawichien T. Melioidosis, an environmental and occupational hazard in Thailand. Mil Med 1999;164:658-62.  Back to cited text no. 43    
44.Kiertiburanakul S, Sungkanuparph S, Kositchiwat S, Vorachit M. Burkholderia pseudomallei abscess in unsual site. J Postgrad Med 2002;48:124-6.  Back to cited text no. 44    
45.Lim WK, Gurdeep GS, Norain K. Melioidosis of the head and neck. Med J Malaysia 2001;56:471-7.  Back to cited text no. 45    
46.Wong PK, Ng PH. Melioidosis presenting with orbital cellulitis. Singapore Med J 1996;37:220-1.  Back to cited text no. 46    
47.Low JG, Quek AM, Sin YK, Ang BS. Mycotic aneurysm due to Burkholderia pseudomallei infection: case reports and literature review. Clin Infect Dis 2005;40:193-8.  Back to cited text no. 47    
48.Majid AA. Successful surgical management of a case of pulmonary and pericardial melioidosis. Aust N Z J Surg 1990;60:139-41.  Back to cited text no. 48    
49.Sirikulchayanonta V, Subhadrabandhu T. Melioidosis. Another etiology of granulomatous osteomyelitis. Report of 2 cases. Clin Orthop 1994;308:183-6.  Back to cited text no. 49    
50.Badsha H, Edwards CJ, Chng HH. Melioidosis in systemic lupus erythematosus: the importance of early diagnosis and treatment in patients from endemic areas. Lupus 2001;10:821-3.  Back to cited text no. 50    
51.Tan JK, Yip SK, Png DJ, Moorthy P. Primary melioidotic prostatic abscess: presentation, diagnosis and management. ANZ J Surg 2002;72:408-10.  Back to cited text no. 51    
52.Lath R, Rajshekhar V, George V. Brain abscess as the presenting feature of melioidosis. Br J Neurosurg 1998;12:170-2.  Back to cited text no. 52    
53.Currie BJ, Fisher DA, Howard DM, Burrow JN. Neurological melioidosis. Acta Trop 2000;74:145-51.  Back to cited text no. 53    
54.Chaowagul W, White NJ, Dance DA, Wattanagoon Y, Naigowit P, Davis TM, et al. Melioidosis: a major cause of community acquired septicaemia in Northerneastern Thailand. J Infec Dis 1989;159:890-9.  Back to cited text no. 54    
55.Puthucheary SD, editor. Human melioidosis. Singapore: University of Singapore Press; 2002, p. 7-46.  Back to cited text no. 55    
56.Cheng AC, Obrien M, Jacups SP, Anstey NM, Currie BJ. C-reactive protein in the diagnosis of melioidosis. Am J Trop Med Hyg 2004;70:580-2.  Back to cited text no. 56    
57.Anuntagool N, Rugdech P, Sirisinha S. Identification of specific antigens of Pseudomonas pseudomallei and their efficacies for diagnosis of melioidosis. J Clin Microbiol 1993;31:1232-6.  Back to cited text no. 57    
58.Lowe P, Engler C, Norton R. Comparison of automated and nonautomated systems for identification of Burkholderia pseudomallei . J Clin Microbiol 2002;40:4625-7.  Back to cited text no. 58    
59.Inglis TJ, Chiang D, Lee GS, Chor-Kiang L. Potential misidentification of B. pseudomallei by API 20NE. Pathology 1998;30:62-4.  Back to cited text no. 59    
60.Appassakij H, Silpapojakul KR, Wansit R, Pornpatkul M. Diagnositic value of the the indirect haemagglutination test for melioidosis. Am J Trop Med Hyg 1990;42:248-53.  Back to cited text no. 60    
61.Ashdown LR, Johnson RW, Koehler JM, Cooney CA. Enzyme-linked immunosorbent assay for the diagnosis of clinical and subclinical melioidosis. J Infect Dis 1989;160:253-60.  Back to cited text no. 61    
62.Rattanathongkom A, Sermswan RW, Wongratanacheewin S. Detection of Burkholderia pseudomallei in blood samples using polymerase chain reaction. Mol Cell Probes 1997;11:25-31.  Back to cited text no. 62    
63.Kunakorn M, Raksakait K, Sethaudom C, Sermswan RW, Dharakul T. Comparison of three PCR primer sets for diagnosis of septicemic melioidosis. Acta Trop 2000;74:247-51.  Back to cited text no. 63    
64.White NJ, Dance DA, Chaowagul W, Wattanagoon Y, Wuthiekanun V, Pitakwatchara. Halving the mortality of severe melioidosis by ceftazidime. Lancet 1989;2:679-701.  Back to cited text no. 64    
65.Sookpranee M, Boonma P, Susaengrat W, Bhuripanyo K, Punyagupta S. Multi center prospective randomised trial comparing ceftazidime and cotimoxazole with chloramphenicol plus doxycycline for treatment of melioidosis. Antimicrob Agents Chemoth 1992;36:158-62.  Back to cited text no. 65    
66.Suputtamongkol Y, Chaowagul W. Management and therapy of melioidosis. First international symposium on Melioidosis in Kuala Lumpur Malaysia. 1994;7-8.  Back to cited text no. 66    
67.Smith MD, Wuthiekanun V, Walsh AL, White NJ. In-vitro activity of carbapenem antibiotics against beta-lactam susceptible and resistant strains of Burkholderia pseudomallei . J Antimicrob Chemother 1996;37:611-5.  Back to cited text no. 67    
68.Chau PY, Ng WS, Leung YK, Lolekha S. In vitro susceptibilty of strains of Pseudomonas pseudomallei isolated in Thailand and Hong Kong to some newer β -lactam antibiotics and quinolone derivatives. J Infect Dis 1986;153:167-70.  Back to cited text no. 68    
69.Suputtamongkol Y, Chaowagul W, Chetchotisakd P, Lertpatanasuwun N, Intaranongpai S, Ruchutrakool T, et al . Risk factors for melioidosis and bacteraemic melioidosis. Clin Infect Dis 1999;29:408-13.  Back to cited text no. 69    
70.Simpson AJ, Suputtamongkol Y, Smith MD, Angus BJ, Rajanuwong A, Wuthiekanun V, et al . Comparison of imipenem and ceftazidime as therapy for severe melioidosis. Clin Infect Dis 1999;29:381-7.  Back to cited text no. 70    
71.Lumbiganon P, Panamonta O. Imipenem therapy for septicemic melioidosis in a child with penicillin and cephalosporin adverse reaction. Southeast Asian J Trop Med Public Health 2000;31:89-90.   Back to cited text no. 71    
72.Cheng AC, Fisher DA, Anstey NM, Stephens DP, Jacups SP, Currie BJ. Outcomes of patients with melioidosis treated with meropenem. Antimicrob. Agents Chemother 2004;48:1763-5.  Back to cited text no. 72    
73.Mays EE, Ricketts EA. Melioidosis: Recrudescence associated with bronchogenic carcinoma twenty-six year following initial geographic exposure. Chest 1975;68:261-3.  Back to cited text no. 73    
74.Rajchanuvong A, Chaowagul W, Suputtamongkol Y, Smith MD, Dance DA, White NJ. Aprospective comparison of co-amoxiclav and the combination of chloramphenicol, doxycycline and cotimoxazole for the oral treatment of melioidosis. Trans R Soc Trop Med Hyg 1995;89:546-9.  Back to cited text no. 74    
75.Chetchotisakd P, Chaowagul W, Mootsikapun P, Budhsarawong D, Thinkamrop B. Maintenance therapy of melioidosis with ciprofloxacin plus azithromycin compared with cotrimoxazole plus doxycycline. Am J Trop Med Hyg 2001;64:24-7.  Back to cited text no. 75    
76.Pruksachartvuthi S, Aswapokee N, Thankerngpol K. Survival of Pseudomonas pseudomallei in human phagocytes. J Med Microbiol 1990;31:109-14.  Back to cited text no. 76    
77.Chateau MT, Caravano R. Rapid fluorometric measurement of the intra-cellular concentration of ciprofloxacin in mouse peritoneal macrophages. J Antimicrob Chemother 1993;31:281-7.  Back to cited text no. 77    
78.Vorachit M, Chongtrakool P, Arkomsean S, Boonsong S. Antimicrobial resistance in Burkholderia pseudomallei . Acta Tropica 2000;74:139-44.  Back to cited text no. 78    
79.Choy JL, Mayo M, Janmaat A, Currie BJ. Animal melioidosis in Australia. Acta Trop 2000;74:153-8.  Back to cited text no. 79    
80.Nelson M, Prior JL, Lever MS, Jones HE, Atkins TP, Titball RW. Evaluation of lipopolysaccharide and capsular polysaccharide as subunit vaccines against experimental melioidosis. J Med Microbiol 2004;53:1177-82.  Back to cited text no. 80    
81.Brett PJ, Mah DC, Woods DE. Isolation and characterization of Pseudomonas pseudomallei flagellin proteins. Infect Immun 1994;62:1914-9.  Back to cited text no. 81    
82.Nangam N, Angkititakul S, Noimay P, Thamlikitkul V. The effect of quicklime (calcium oxide) as an inhibitor of Burkholderia pseudomallei. Trans R Soc Trop Med Hyg 2004;98:337-41.  Back to cited text no. 82    

Copyright 2005 - Journal of Postgraduate Medicine


The following images related to this document are available:

Photo images

[jp05056t2.jpg] [jp05056t3.jpg] [jp05056t1.jpg]
Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil