Categories
Review Articles

Glibenclamide therapy as tertiary prevention of melioidosis for Type 2 diabetics.

Background: Melioidosis is a relatively high-incidence, high-mortality tropical infectious disease caused by Burkholderia pseudomallei. To date, the few prevention and management strategies in practice have failed to reduce mortality from septic shock in patients with severe melioidosis. Up to 60.9% of patients also have pre-existing type 2 diabetes mellitus (T2DM), the most significant risk factor for melioidosis. An effective tertiary prevention strategy against melioidosis for these diabetic individuals would impact significantly on the burden of disease. Glibenclamide, a drug belonging to the sulfonylurea class and commonly used as an antidiabetic, may have the potential to be one such strategy.

Aim: To examine the underlying inflammatory cause of morbidity and mortality in melioidosis and to assess the potential for glibenclamide to ameliorate these causes.

Results: Bacteraemia with B. pseudomallei leads to widespread infection. Multi-organ damage and loss of function results from both direct bacterial damage and an excessive inflammatory response, the latter of which also causes potentially fatal septic shock in 21% of cases. Massive cellular infiltration of the lungs is particularly damaging. The immunomodulatory effects of T2DM further exacerbate the deleterious immune response into a dysregulated, hyperinflammatory state. On the other hand, the several anti-inflammatory effects of glibenclamide have been demonstrated to significantly reduce mortality from septic shock in melioidosis.

Conclusion: For diabetics living in regions where melioidosis is endemic, choosing glibenclamide over other sulfonylureas and metformin for antidiabetic treatment could be a promising tertiary prevention measure against melioidosis.

Introduction

Glibenclamide therapy as tertiary prevention of melioidosis for Type 2 diabeticsMelioidosis, a tropical infectious disease, is a significant cause of death and illness in northern Australia and Southeast Asia. Its non-specific prodrome and clinical presentation makes early detection difficult.[1] Yet, timely diagnosis is critical because responsiveness to treatment declines with progression of the disease over time.[2] Morbidity and mortality remains high despite aggressive antibiotic treatment. As of yet, no vaccine is available for primary prevention and novel developments such as granulocyte colony-stimulating factor (G-CSF) and corticosteroid therapy lack evidence as good measures of tertiary prevention. Considering the significant population health threat, any effective prevention strategy would be of value. This article suggests the potential use of glibenclamide, an antidiabetic sulfonylurea, for tertiary prevention of melioidosis in diabetics. Because diabetics are approximately 13 times more likely to contract melioidosis,[3,4] this strategy may have widespread application.

One of the major and dangerous end-stage complications of melioidosis is septic shock. Especially in settings such as Northeast (NE) Thailand where melioidosis is endemic but intensive care is unavailable, morbidity and mortality from septic shock are unacceptably high. Systemically-spread B. pseudomallei stimulates massive release of cytokines into the blood, this event being instrumental in the immunopathogenesis of septic systemic vasodilation, hypotension, vascular hyperpermeability, and disseminated intravascular coagulation (DIC). Furthermore, poor glycaemic control in diabetics increases the risk and severity of these events by precipitating a compensatory, dysregulated, and hyperinflammatory response to bacterial immunogenic stimuli. Demanding particular attention is the stimulated production of cytokine IL-1β because it promotes excessive neutrophilic recruitment to the lungs and bacterial persistence inside cells. Fortunately, there is strong evidence that glibenclamide acts protectively against IL-1β-mediated damage by targeting IL-1β production directly as well as multiple upstream components in the IL-1β production pathway. The result of glibenclamide use for diabetics is a significantly reduced morbidity and mortality from melioidosis septic shock compared with diabetics not taking the drug.

Epidemiology of Melioidosis

Melioidosis is endemic in Southeast Asia, northern Australia, India, Hong Kong, southern China and Taiwan.[5,6] Isolated sporadic cases have also appeared in Central America, the Caribbean, New Caledonia, Mauritius, Africa, and the Middle East.[7] Of particular concern are the hyperendemic hotspots – NE Thailand and the Top End of the Northern Territory of Australia. A 2006 NE Thailand study that gathered results from four large hospitals reported the region experienced an estimated minimum annual incidence of 21.3 cases per 100,000 people and case fatality rate of 40.5%, melioidosis locally being the third highest infectious cause of death and second most common pathogenic cause of community-acquired bacteraemia.[8,3] From 2004 to 2009, according to data collected at Royal Darwin Hospital (RDH), the annual incidence in the Top End was 21.6 per 100,000 people and average case fatality rate plateaued at 9% having decreased from 30% since 1989. The lower fatality rate in Darwin is attributed to greater access to intensive care unit (ICU) supportive care and early sepsis management that prolongs life in the event of fatal septic shock, a complication that develops in at least 21% of cases.[9]

Type 2 diabetes mellitus (T2DM) is the largest risk factor for melioidosis. 48% and 60.9% of individuals with melioidosis were confirmed to have T2DM at RDH and in NE Thailand respectively.[10,11] The 2006 Thai study reported that the relative risk (RR) of melioidosis in adult diabetics compared with non-diabetics was 12.4.[3] In the Top End region, combining diabetes with the additional risk factor of Indigenous ethnicity raised the RR from 13.1 to 20.6. Other notable risk factors associated with a significant RR are chronic lung disease (RR 4.3), age ≥ 45 years (RR 4.0), chronic renal disease (RR 3.2), Indigenous ethnicity (RR 3.0), male sex (RR 2.4) and hazardous alcohol consumption (RR 2.1).[6]

The presence of any of these risk factors in infected individuals also dramatically increases their chance of developing septic shock (RR 4.5) and death (RR 9.2), the risk factors with highest independent association with death being age ≥ 50 years (RR 2.1) and chronic lung disease (RR 1.5). Malignancy (RR 1.9) and rheumatic heart disease and/or congestive cardiac failure (RR 1.7) are statistically insignificant risk factors for death. Clinical presentations most likely to result in death are septic shock (RR 11.2) and, out of the non-septic shock cases, the presence of neurological infection foci (RR 4.7).[9] Additionally, case fatality during the peak rainfall months of December through February is 1.6 times greater than it is during other months.[9]

Immunopathogenesis and pathophysiology

Melioidosis is caused by Gram-negative bacterium B. pseudomallei. Bacteria, found in contaminated soil, rodents, food, water, and excretions, are transmitted via inhalation, ingestion, or percutaneous inoculation – usually direct contact with open skin lesions.[12,13] Post-inoculation, B. pseudomallei can invade most human cell types. Employing Type 3 Secretion System (TTSS) clusters, bacteria enter non-phagocytic cells. If bacteria are phagocytosed, TTSS enables exit from intracellular phagocytic endosomes such that degradation is evaded. Once escaped into the cytoplasm of cells both phagocytic and non-phagocytic, bacteria replicate and self-induce polarised actin filamentation that confers motility, facilitating spread to neighbouring cells by forcing host-cell membrane protrusion and fusion.[14]

B. pseudomallei possesses highly immunogenic factors that trigger a strong host immune reaction essential to the host for early bacterial containment.[15] B. pseudomallei expresses pathogen-associated molecular patterns including lipopeptides, peptidoglycan, lipopolysaccharide, flagellin, TTSS, and DNA. They are recognised by host cell toll-like receptors (TLR) and NOD-like receptors (NLR). TLRs and NLRs are expressed by immune cells both professional – macrophages and dendritic cells – and non-professional – epithelial cells, endothelial cells, and fibroblasts.[16]

Activation of NLRC4 or NLRP3 induces binding of caspase-1, Asc (apoptosis-associated speck-like protein containing CARD) and NLRC4 or NLRP3 to form an inflammasome. The NLRC4 inflammasome in infected macrophages triggers pyroptosis, serving to limit intracellular B. pseudomallei growth and proliferation, while the NLRP3 inflammasome performs proteolytic activation of pro-IL-1β and pro-IL-18 into their mature forms for secretion.[17] IL-1β recruits neutrophils to infection site(s). However, excessive recruitment is often deleterious because neutrophils, lacking NLRC4, fail to pyroptose and instead provide a favourable intracellular environment that sustains chronic bacterial persistence. In the lungs, persistence leads to damaging pulmonary abscess formation and acute respiratory distress syndrome.[17,18] In contrast, elevated IL-18 production correlates with survival and immunoprotection because IL-18 induces IFN-γ production.[17,19] IFN-γ activates macrophages, stimulating their direct antimicrobial processes – phagolysosomal fusion and toxic reactive nitrogen species synthesis that produces nitrosative stress.[20] The cytokine also facilitates macrophage antigen processing and presentation, recruits leukocytes to infection site(s), upregulates Th1 CD4+ cell population, enhances natural killer cell function, regulates B cell anti-lipopolysaccharide antibody production and isotype switching.[21]

Glibenclamide therapy as tertiary prevention of melioidosis for Type 2 diabetics
Figure 1. NLRC4 inflammasome leads to immunoprotective effects while NLRP3 inflammasome produces both protective and deleterious responses. Glibenclamide works by inhibiting NLRP3 inflammasome formation and its deleterious effects.

TLR activation upregulates secretion of principal inflammatory mediators including type 1 interferon, chemokines, antimicrobial proteins, and pro-inflammatory cytokines.[22] Elevated expression of TLR1, TLR2, TLR3, TLR4, TLR5, TLR8, and TLR10 has been demonstrated in patients with septic melioidosis.[16] In particular, stimulation of the TLR2-mediated signalling pathway is a principal step in recognising the immune challenge of B. pseudomallei and initiating early inflammatory processes.[23] TNF-α and IL-6, along with IL-1β, increase vascular permeability, induce acute phase protein production, and recruit leukocytes to the site of infection.[24] TNF-α and IL-6 also activate the complement and coagulation cascades that are key defense mechanisms of the innate response.[25] However, excessive inflammatory cytokine production induced by widespread bacteraemia often leads to septic shock characterised by systemic vasodilatory hypotension, vascular hyperpermeability causing major cellular and fluid leakage from the intravascular to extravascular compartments, DIC, and death in the absence of immediate treatment.[26]

Through the establishment of bacteraemia, B. pseudomallei spreads from primary foci of infection to other body tissues, usually the lungs, genitourinary tract, skin, joints, bones, liver, spleen, skeletal muscles, prostate, parotid gland, and nervous system. Multi-organ spread leading to impaired organ function and failure is the main source of morbidity and mortality in melioidosis.[27] Thus, a rapid but non-deleterious inflammatory response is critical before B. pseudomallei establishes an intracellular niche that enables its persistence and protection against eradication by a subsequent immune attack. Paradoxically, subsequent immune attacks may cause further damage to the host.

Effect of type 2 diabetic state on host response

Diabetics, due to their generally immunosuppressed state, compared with non-diabetics, are more susceptible to developing sepsis from most organisms,[28] with the most common source of systemic spread being respiratory, followed by urinary and abdominal.[29] Gram-positive bacteria have become the leading cause of sepsis since the 1980s, and sepsis due to causes other than B. pseudomallei actually affects diabetics more commonly than melioidosis.[29,30] However, it is still of significance that diabetics are particularly vulnerable to pathogens such as B. pseudomallei and Mycobacterium tuberculosis because immune defence against these intracellular bacteria is highly macrophage-mediated – a function that is critically impaired in poorly-controlled T2DM.[31]

T2DM, especially if poorly controlled, causes marked immunomodulation that produces B. pseudomallei-induced immune responses that are damaging and yet insufficient to offer host protection. Morris et al. created an ex vivo whole-blood assay using human peripheral blood to compare inflammatory responses to B. pseudomallei between diabetics and non-diabetics. Immune insufficiency was worst in poorly controlled diabetics, the assay detecting: elevated serum IL-10 (an anti-inflammatory cytokine); reduced CD11b on polymorphonuclear leukocytes (PMNs) leading to decreased PMN function with reduced activation, adhesion, transmigration, and migratory capacity towards IL-8; reduced endotoxaemia-induced upregulation of ICAM-1; and defects in phagocytic detection and response to the bacteria.[32] In vivo and in vitro models using streptozotocin (STZ)-induced, leptin deficiency, leptin receptor deficiency, and diet-induced diabetic mice found similar immune impairments.[31,33-35] Transcriptional analysis of STZ-diabetic mice responses over the first 42 hours of B. pseudomallei exposure attributed delayed defence and splenic dysfunction (permitting uncontrolled bacterial replication intracellularly causing susceptibility to sepsis) to diminished TLR2 recognition of the bacterium.[34]

Incompetence of the early inflammatory response to contain an initial infection precipitates a compensatory, dysregulated, hyperinflammatory response to the spreading infection. The intensity of this ensuing reaction is exacerbated by chronic low-grade inflammation and increased oxidative stress, both characteristic of T2DM. Morris et al. found poorly controlled diabetics to have the most elevated levels of pro-inflammatory markers ESR, CRP, TNF-α, IL-1β, IL-6, IL-8, IL-12p70, MCP-1, and MPO (which indicates increased oxidative burst activity in PMNs). At focal infection sites, there was extensive infiltration with PMNs and greater risk of tissue damage, as well as generalised endothelial dysfunction and vascular inflammation.[32]

Hyperglycaemia is a major link between T2DM, immune derangement, and susceptibility to sepsis. The supra-physiological hyperosmolarity of hyperglycaemic blood directly potentiates TLR4-mediated cytokine production, as well as dampening phagocytic responses and granulocyte oxidative burst.[36] Activation of the RAGE pathway by advanced glycation end-products formed secondary to hyperglycaemia perpetuates inflammation and worsens survival in septic mice.[37] Furthermore, in the event of sepsis, poor glycaemic control allowing for periods of hyperglycaemia in both diabetics and non-diabetics correlates with longer hospital stay, greater morbidity, and possibly decreased survival.[38] In addition to immunomodulatory effects, hyperglycaemia exacerbates hypotension in septic shock by promoting glycosuric diuresis and cardiac hyperresponsiveness to cholinergic stimulation and the baroreflex.[39] Elevated risk of myocardial infarction, stroke, and venous thromboembolism is associated with the prothrombotic effect of acute hyperglycaemia that is further potentiated by hyperinsulinaemia or an inflammatory stress state.[40]

Prevention and management

No human vaccine is available.[41] CDC prevention guidelines recommend avoiding contact with soil and stagnant water in endemic regions.[42] High-risk individuals should stay indoors during heavy wind and rain in endemic regions due to possible bacterial aerosolisation that greatly facilitates transmission.[43]

Treatment involves two weeks of intravenous antibiotics – ceftazidime, meropenem, or imipenem, in combination with trimethoprim-sulfamethoxazole – followed by 3 months of oral eradication therapy to prevent recrudescence.[44] However, even with early antibiotic administration, case fatality from septicaemia remains at 37% in Thailand.[45] The development of septic shock is an indication for ICU supportive care usually involving invasive monitoring, fluid resuscitation, renal replacement therapy, and vasopressor, inotropic, and mechanical ventilatory support.[46,47]

Adjunctive use of recombinant G-CSF holds no clinical utility for improving sepsis from melioidosis and other aetiologies. Both animal and human trials – including prospective, multicentre, randomised, double-blind, and placebo-controlled studies – confirm this.[47-49] In fact, G-CSF may cause increased hepatic dysfunction and higher peak troponin I levels.[50] Other immunomodulatory adjuncts such as corticosteroids do not improve survival.[46] In light of the paucity of effective evidence-based prevention strategies and adjunct therapies for underserved populations, we turn to a novel potential for tertiary prevention.

Glibenclamide and its protective effect

Glibenclamide is a sulfonylurea used in the management of diabetes. It antagonises ATP-dependent potassium (KATP) channels in pancreatic β-cells, stimulating insulin release. The salient risk associated with glibenclamide is accidentally developing hypoglycaemia, an event of varying seriousness in patients who are not critically ill but a great predictor of mortality in acute sepsis. Furthermore, recent studies comparing glibenclamide with metformin and other sulfonylureas elucidated its significant adverse effects in addition to severe hypoglycaemia, such as increased risk of malignancy and mortality from cardiovascular disease due to interference with ischaemic preconditioning.[51,52] Despite glibenclamide still being a popular choice, a trend in medical opinion has arisen against its use in favour of alternative sulfonylureas.[53,54]

However, for T2DM individuals living in regions endemic with melioidosis, choosing glibenclamide over other antidiabetic drugs has an additional benefit that clinicians must consider. In a 5-year prospective cohort study from 2002 to 2006, Koh et al. followed 1160 adult patients with culture-confirmed melioidosis for 28 days during and after their admission to a major NE Thailand hospital. 71.3% of diabetic individuals taking glibenclamide survived from melioidosis after 28 days, while only 50.7% of diabetics not taking glibenclamide and 47.0% of non-diabetics survived (survival of patients discharged from hospital within 28 days were assumed to have survived). Using a logistics regression model, glibenclamide treatment reduced case fatality with an adjusted odds ratio (AOR) of 0.34 when compared to diabetics not taking glibenclamide and an AOR of 0.47 compared to non-diabetics. Incidences of hypotension (AOR 0.48) and respiratory failure (AOR 0.50) were also reduced in these patients.[55] Therefore, this protective effect must be factored into the drug’s risk-benefit comparison with metformin and with other sulfonylureas. If it informs clinicians’ drug choices for glycaemic control in management of T2DM, glibenclamide can contribute to tertiary prevention of melioidosis.

Although preventing hyperglycaemia in sepsis improves patient outcomes, glibenclamide does not rely on its glucose-lowering effect.[56] Using a mouse model, Koh et al. attributed the mechanism of glibenclamide’s protective effect to two main anti-inflammatory pathways. Firstly, glibenclamide reduces IL-1β secretion, attenuating IL-8 production and neutrophilic and monocytic influx into the lungs (without completely ablating the neutrophilic response).[55,57] In vitro evidence suggests that glibenclamide does this by partially blocking NLRP3 inflammasome formation and/or by directly inhibiting the secretion of mature IL-1β (Figure 1).[57-59] Glibenclamide may also attenuate transcription and translation of caspase-1, NLRP3, and IL-1β genes.[57] Secondly, by reducing systemic vasodilation and maintaining normal peripheral vascular resistance, glibenclamide restores systemic mean arterial pressure in the event of septic shock. KATP channels in vascular smooth muscle preferentially open during sepsis.[60] When they do, they hyperpolarise the cell, inhibiting Ca2+ influx (via voltage-dependent Ca2+ channels) and causing muscular relaxation. Glibenclamide opposes this effect by blocking the KATP channels. However, the only evidence for this is found in animal models.[61,62]

Conclusion

Melioidosis remains a significant public health concern due to its high incidence, mortality and case fatality rates particularly in the Top End of Northern Territory and NE Thailand. T2DM is the largest risk factor for melioidosis. The antidiabetic drug, glibenclamide, reduces morbidity and mortality from melioidosis-induced septic shock and so plays a potential role in tertiary prevention of melioidosis. In the debate over glibenclamide use, consideration of the drug’s protective effect for diabetics living in melioidosis-endemic regions, particularly those that are resource-limited, is critical.

Acknowledgements

None

Conflict of Interests

None declared

References

1. Loveleena, Chaudry R, Dhawan B. Melioidosis; the remarkable imitator: recent perspectives. J Assoc Physicians India. 2004;52:417-20.

2. Suputtamongkol Y, Hall AJ, Dance DA, Chaowagul W, Rajchanuvong A, Smith MD, et al. The epidemiology of melioidosis in Ubon Ratchatani, northeast Thailand. Int J Epidemiol. 1994;23(5):1082-90.

3. Kanoksil M, Jatapai A, Sharon PJ, Limmathurotsakul D. Epidemiology, microbiology and mortality associated with community-acquired bactaremia in Northeast Thailand: a multicenter surveillance study. PLoS One. 2013 Oct;8(1):e54714.

4. Suputtamongkol Y, Chaowagul W, Chetchotisakd P, Lertpatansuwun N, Intaranongpai S, Ruchutrakool T, et al. Risk factors for melioidosis and bacteraemic melioidosis. Clin Infect Dis. 1999;29(2):408-13.

5. Blaney DD, Gee JE, Theresa SL. Chapter 3: infectious diseases related to travel: melioidosis. Centers for Disease Control and Prevention 2011.

6. Currie BJ, Dance DA, Cheng AC. The global distribution of Burkholderia pseudomallei and melioidosis: an update. Trans R Soc Trop Med Hyg. 2008;102 Suppl 1:S1-4.

7. Raja NS, Ahmed MZ, Singh NN. Melioidosis: an emerging infectious disease. J Postgrad Med. 2005;51(2):140-5.

8. Limmathurotsakul D, Wongratanacheewin S, Teerawattanasook N, Wongsuvan G, Chaisuksant S, Chetchotisakd P, et al. Increasing incidence of human melioidosis in Northeast Thailand. Am J Trop Med Hyg. 2010;82(6):1113-7.

9. Currie BJ, Ward L, Cheng AC. The epidemiology and clinical spectrum of melioidosis: 540 cases from the 20 year Darwin prospective study. PLoS Negl Trop Dis. 2010;4(11):e900.

10. Parameswaran U, Baird RW, Ward LM, Currie BJ. Melioidosis at Royal Darwin Hospital in the big 2009-2010 wet season: comparison with the preceding 20 years. Med J Aust. 2012;196(5):345-8.

11. Currie BJ, Jacups SP, Cheng AC, Fisher DA, Anstey NM, Huffam SE, et al. Melioidosis epidemiology and risk factors from a prospective whole-population study in northern Australia. Trop Med Int Health. 2004;9(11):1167-74.

12. Currie BJ, Jacups SP. Intensity of rainfall and severity of melioidosis, Australia. Emerg Infect Dis. 2003;9:1538-42.

13. Howe C, Sampath A, Spotnitz M. The Pseudomallei group: A review. J Infect Dis. 1971;124(6):598-606.

14. Allwood EM, Devenish RJ, Prescott M, Adler B, Boyce JD. Strategies for intracellular survival of burkholderia pseudomallei. Front Microbiol. 2011;2:170.

15. Easton A, Haque A, Chu K, Lukaszewski R, Bancroft GJ. A critical role for neutrophils in resistance to experimental infection with Burkholderia pseudomallei. J Infect Dis. 2007;195(1):99-107.

16. Wiersinga WJ, Catharina WW, Mark DC. Toll-like receptor 2 impairs host defense in Gram-Negative sepsis caused by Burkholderia pseudomallei (melioidosis). PLoS Med. 2007;4(7):e248.

17. Ceballos-Olvera I, Sahoo M, Miller MA, Del Barrio L, Re F. Inflammasome-dependent pyroptosis and IL-18 protect against Burkholderia pseudomallei lung infection while IL-1β is deleterious. PLoS Pathog. 2011;7(12):e1002452.

18. Koh GC, Schreiber MF, Bautista R, Maude RR, Dunachie S, Limmathurotsakul D, et al. Host responses to melioidosis and tuberculosis are both dominated by interferon-mediated signaling. PLoS ONE. 2013;8(1):e54961. doi:10.1371/journal.pone.0054961.

19. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med. 2000;342(18):1334-49.

20. Flynn JL, Chan J. Immune evasion by Mycobacterium tuberculosis: living with the enemy. Curr Opin Immunol. 2003;15(4):450-5.

21. Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to interferon-gamma. Annu Rev Immunol. 1997;15:749-95.

22. Wiersinga WJ, van der Poll T, White NJ, Day NP, Peacock SJ. Melioidosis: insights into the pathogenicity of Burkholderia pseudomallei. Nat Rev Microbiol. 2006;4(4):272-82.

23. Chin CY, Monach DM, Nathan S. Genome wide transcriptome profiling of a murine acute melioidosis model reveals new insights into how Burkholderia pseudomallei overcomes host innate immunity. BMC Genomics. 2011;11:672.

24. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;149(6):805-20.

25. Wiersinga WJ, Currie BJ, Peacock SJ. Melioidosis. N Engl J Med. 2012;367(11):1035-44.

26. Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AM, Knaus WA, et al. Definitions of sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644-55.

27. Meumann EM, Cheng AC, Ward L, Currie BJ. Clinical features and epidemiology of melioidosis pneumonia: results from a 21-year study and review of the literature. Clin Infect Dis. 2012;54:362-9.

28. Muller LMAJ, Gorter KJ, Hak E, Goudzwaard WL, Schellevis FG, Hoepelman AIM, et al. Increased risk of common infections in patients with type 1 and type 2 diabetes mellitus. Clin Infect Dis. 2005;41:281-8.

29. Martin GS. Sepsis, severe sepsis and septic shock: changes in incidence, pathogens and outcomes. Expert Rev Anti Infect Ther. 2012;10(8):701-6.

30. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med. 2003;348(16):1546-54.

31. Hodgson KA, Morris JL, Feterl ML, Govan BL, Ketheesan N. Altered macrophage function is associated with severe Burkholderia pseudomallei infection in a murine model of type 2 diabetes. Microbes Infect. 2011;13(14-15):1177-84.

32. Morris J, Williams N, Rush C, Govan B, Sangla K, Norton R, et al. Burkholderia pseudomallei triggers altered inflammatory profiles in a whole-blood model of type 2 diabetes-melioidosis comorbidity. Infect Immun. 2012;80(6):2089-99.

33. Williams NL, Morris JL, Rush C, Govan BL, Ketheesan N. Impact of streptozotocin-induced diabetes on functional responses of dendritic cells and macrophages towards Burkholderia pseudomallei. FEMS Immunol Med Microbiol. 2011 Mar;61(2):218-27.

34. Chin CY, Monack DM, Nathan S. Delayed activation of host innate immune pathways in streptozotocin-induced diabetic hosts leads to more severe disease during infection with Burkholderia pseudomallei. Immunology. 2012;135(4):312-32.

35. Hodgson KA, Govan BL, Walduck AK, Ketheesan N, Morris JL. Impaired early cytokine responses at the site of infection in a murine model of type 2 diabetes and melioidosis comorbidity. Infect Immun. 2013;81(2):470-7.

36. Wade CE. Hyperglycemia may alter cytokine production and phagocytosis by means other than hyperosmotic stress. Crit Care. 2008;12(5):182.

37. Bopp C, Bierhaus A, Hofer S, Bouchon A, Nawroth PP, Martin E, et al. Bench-to-bedside review: The inflammation-perpetuating pattern-recognition receptor RAGE as a therapeutic target in sepsis. Crit Care. 2008;12(1):201.

38. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, et al. Intensive insulin therapy in the medical ICU. N Engl J Med. 2006;354(5):449-61.

39. Liu IM, Chang CK, Juang SW, Kou DH, Tong YC, Cheng KC, et al. Role of hyperglycaemia in the pathogenesis of hypotension observed in type-1 diabetic rats. Int J Exp Pathol. 2008;89(4):292-300.

40. Lemkes BA, Hermanides J, Devries JH, Holleman F, Meijers JC, Hoekstra JB. Hyperglycemia: a prothrombotic factor? J Thromb Haemost. 2010;8(8):1663-9.

41. Patel N, Conejero L, De Reynal M, Easton A, Bancroft GJ, Titball RW. Development of vaccines against Burkholderia pseudomallei. Front Microbiol. 2011;2:198.

42. Centers for Disease Control and Prevention (CDC). Melioidosis: prevention. Atlanta: CDC; 2012.

43. Australian Centre for Disease Control. Melioidosis. Darwin: ACDC; 2012.

44. Currie B, Anstey N. Treatment and prognosis of melioidosis. UpToDate; 2012.

45. White NJ, Dance DA, Chaowagul W, Wattanagoon Y, Wuthiekanun V, Pitakwatchara N. Halving of mortality of severe melioidosis by ceftazidime. Lancet. 1989;2(8665):697-701.

46. Panomket P, Chetchotisakd P, Sermswan RW, Pannengpetch P, Wongratanacheewin S. Use of a low-dose steroid as an adjunct in the treatment, in mice, of severe sepsis caused by Burkholderia pseudomallei. Ann Trop Med Parasitol. 2009;103(7):635-46.

47. Mohammad RA. Use of granulocute colony-stimulating factor in patients with severe sepsis or septic shock. Am J Health Syst Pharm. 2010;67(15):1238-1245.

48. Cheng AC, Stephens DP, Anstey NM, Currie BJ. Adjunctive granulocyte colony-stimulating factor for treatment of septic shock due to melioidosis. Clin Infect Dis. 2004;38(1):32-7.

49. Cheng AC, Limmathurotsakui D, Chierakul W, Getchalarat N, Wuthiekanun V, Stephens DP, et al. A randomized controlled trial of granulocyte colony-stimulating factor for the treatment of severe sepsis due to melioidosis in Thailand. Clin Infect Dis. 2007;45(3):308-14.

50. Stephens DP, Thomas JH, Higgins A, Bailey M, Anstey NM, Currie BJ, et al. Randomized, double-blind, placebo-controlled trial of granulocyte colony-stimulating factor in patients with septic shock. Crit Care Med. 2008;36(2):448-54.

51. Monami M, Balzi D, Lamanna C, Barchielli A, Masotti G, Buiatti E, et al. Are sulphonylureas all the same? A cohort study on cardiovascular and cancer-related mortality. Diabetes Metab Res Rev. 2007;23(6):479-84.

52. Schramm TK, Gislason GH, Vaag A, Rasmussen JN, Folke F, Hansen ML, et al. Mortality and cardiovascular risk associated with different insulin secretagogues compared with metformin in type 2 diabetes, with or without a previous myocardial infarction: a nationwide study. Eur Heart J. 2011;32(5):1900-8.

53. WHO Expert Committee. The selection and use of essential medicines. World Health Organ Tech Rep Ser. 2008;(950):backcover, vii-174.

54. Riddle MC. Editorial: sulfonylureas differ in effects on ischaemic preconditioning – is it time to retire glyburide? J Clin Endocrinol Metab. 2003;88(2):528-30.

55. Koh GC, Maude RR, Schreiber MF, Limmathurotsakul D, Wiersinga WJ, Lee SJ, et al. Glyburide is anti-inflammatory and associated with reduced mortality in melioidosis. Clin Infect Dis. 2011;52(6):717-25.

56. Koh GC, Weehuizen TA, Breitbach K, Krause K, de Jonh HK, Kager LM, et al. Glyburide reduces bacterial dissemination in a mouse model of melioidosis. PLoS Negl Trop Dis. 2013;7(10):e2500.

57. Kewcharoenwong C, Rinchai D, Utispan K, Suwannasaen D, Bancroft GJ, Ato M, et al. Glibenclamide reduces pro-inflammatory cytokine production by neutrophils of diabetes patients in response to bacterial infection.

58. Lamkanfi M, Mueller JL, Vitari AC, Misaphi S, Fedorova A, Deshayes K, et al. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J Cell Biol. 2009;187(1):61-70.

59. Hamon Y, Luciani MF, Becq F, Verrier B, Rubartelli A, Chimini G. Interleukin-1beta secretion is impaired by inhibitors of the Atp binding cassette transporter, ABC1. Blood. 1997;90(8):2911-5.

60. Sorrentino R, d’Emmanuele di Villa Bianca R, Lippolis L, Sorrentino L, Autore G, Pinto A. Involvement of ATP-sensitive potassium channels in a model of a delayed vascular hyporeactivity induced by liposaccharide in rats. Br J Pharmacol. 1999;127(6):1447-53.

61. Vanelli G, Hussain SN, Aguggini G. Glibenclamide, a blocker of ATP-sensitive potassium channels, reverses endotoxin-induced hypotension in pig. Exp Physiol. 1985;80(1):167-70.

62. Landry DW, Oliver JA. The ATP-sensitive K+ channel mediates hypotension in endotoxaemia and hypoxic lactic acidosis in dog. J Clin Invest. 1992;89(6):2071-4.

Categories
Review Articles

A review of early intervention in youth psychosis

Early intervention in youth psychosis has been a topic of contentious discussion. In particular, there is a lack of consensus regarding how early to treat patients with a psychotic disorder. There has been a recent push to provide treatment early in the development of psychosis, specifically to patients in an ultra-high risk or prodromal stage. There is also debate about the types of interventions that should be used, such as psychoeducation, psychotherapy and pharmacotherapy. In Australia, these uncertainties have been reflected by the production of conflicting guidelines by key stakeholders in this area. There are significant arguments both for and against the practice of early intervention. This article explores these arguments and reviews current practices in Australia. A number of updated recommendations are also set out in accordance with the findings of this article.

Introduction

A review of early intervention in youth psychosisPsychotic disorders are characterised by the presence of symptoms that reflect an excess or distortion of normal functions. For example, hallucinations, delusions, thought disorder and disorganised behaviour are symptoms characteristic of psychosis. Patients diagnosed with schizophrenia must demonstrate positive symptoms or severe negative symptoms (e.g. flattened affect, social withdrawal) in addition to deterioration in their social and vocational functioning. [1] Hence, the diagnosis is typically made after the onset of significant symptomology.

McGorry et al. [2] argue that late-stage diagnosis of a psychotic illness leads to delayed and inconsistent management of these patients. The concept of “early intervention” refers to appropriately managing patients in the early stages of psychotic disease, to minimise long-term negative social and psychological outcomes. As such, it represents a secondary prevention strategy and a paradigm shift in the way schizophrenia and other psychotic disorders are viewed; rather than being seen as illnesses with an inevitably poor social and functional outcome, they are viewed as conditions whose course can be altered
by recognition of the early warning signs and application of timely intervention. [2] The proponents of early intervention argue that many of the recognised risk factors for the development and progression of a psychotic disorder (e.g. disrupted peer and family networks, substance A review of early intervention in youth psychosis use, depression) are recognisable in advance and can be acted upon. [2]

The clinical staging model [3] proposes that psychiatric illnesses should be viewed as a sequence of stages that increase in disease severity. Employing the appropriate treatment modality at a particular stage would allow regression of the disease to an earlier stage. The clinical stages of early psychosis include the ‘ultra-high risk’ stage, the ‘first psychotic episode’ stage and the ‘first 5 years after diagnosis’ stage. [2]

The ‘ultra-high risk’ stage is the stage preceding the first psychotic episode. Although the first psychotic episode is often the first recognised sign of a psychotic illness, retrospective analysis reveals many changes occur in an individual’s thoughts and behaviour in the period preceding the psychotic episode. This is known as the ‘prodromal phase’. To intervene at this stage, it is clearly necessary to be able to identify this period in advance, and a considerable research effort is being focused on developing prospective criteria for this purpose. Two tools currently in use are the Positive and Negative Syndrome Scale (PANSS) or Attenuated Positive Symptoms (APS) approach and the Basic Symptoms (BS) approach. [4] The PANSS is a 30-point questionnaire with a 7-point rating for each question. It covers positive symptoms (e.g. delusions, hallucinations), negative symptoms (e.g. social withdrawal, blunted affect) and general symptoms of psychopathology (e.g. depression, poor insight, feelings of tension). [5] The Basic Symptoms approach focuses on subtler, self-experienced subclinical symptoms such as thought interference, disturbance of receptive language, inability to divide attention between tasks and derealisation. [6]

Intervention at the ‘first psychotic episode’ stage is largely aimed at reducing the duration of untreated psychosis (DUP), as a high DUP has been shown to result in poorer outcomes. Some authors have argued that untreated psychosis can lead to irreversible brain damage. [7,8] Although this theory has yet to receive widespread support, the personal, social and societal consequences of untreated psychosis can have a tremendous impact on the patient’s ability to recover from the episode. [2] Functional MRI brain imaging studies have shown decreased memory encoding in patients with schizophrenia and interestingly, decreased posterior cingulate activity in patients with ongoing first-episode psychosis compared to those showing remission at one year. [9] Such alterations in brain activity in patients more likely to proceed to a significant psychotic illness has exciting implications for the use of fMRI as a tool in screening for patients most likely to benefit from early intervention.

The ‘first 5 years after diagnosis’ stage is a crucial period that determines a patient’s long-term outcome. It is the time most likely to result in suicide, disengagement, relapse, [2] long-term treatment resistance and the break down and accumulation of disabilities in personal, social and occupational settings. [10] Mason et al. [11] suggest that the level of disability accumulated in the first 2 years of psychosis may in fact ‘set a ceiling for recovery in the long term’. Hence, intervention at this period is important. Maintaining a steady support structure especially tailored towards young people receiving a diagnosis of psychosis is likely to maximise chances of engagement with mental health care, life-style modifications, and adequate family involvement. [2]

Current Practice
There are currently a number of different practices/guidelines in Australia relating to early intervention in youth psychosis. The Royal Australian and New Zealand College of Psychiatrists (RANZCP) has produced clinical practice guidelines for schizophrenia, which include recommendations for patients at ultra-high risk (UHR). [12] Orygen Youth Health and headspace have also developed guidelines, called ‘The Australian Clinical Guidelines for Early Psychosis’, which are now in the second edition. [13]

Australia has established the first clinical and research clinic in the world for individuals considered to be at imminent risk of psychosis. The Personal Assessment and Crisis Evaluation (PACE) clinic was established by Orygen in Melbourne in 1994. [14] The clinic receives referrals from general practice, school counsellors and various health services. [14] They facilitate case management and provide a variety of in-house support services to families and carers including group programs, vocational and educational assistance, and occupational therapy. [15] Orygen, in conjunction with the Australian General Practice Network, the Australian Psychological Society and the Brain and Mind Research Institute also established headspace, which is a national youth mental health foundation. [16] The aim of headspace was to facilitate early intervention by increasing community awareness, clinician training and taking a youth-specific approach to management, as well as utilising multidisciplinary care. [16,17] Another service available is the Early Psychosis Prevention and Intervention Centres (EPPIC). In the 2010-11 and 2011-12 budgets, the Federal Government allocated $247m to the establishment of a network of 16 of these centres across Australia, modelled upon Orygen’s EPPIC centre in Melbourne. [18] A more detailed summary of the current guidelines/practices existing in Australia for youth psychosis is listed in Table 1.

Table 1. Current practice (guidelines and health services) in Australia for youth psychosis.

Guideline
Recommendation
RANZCP Clinical Practice Guidelines for the Treatment of Schizophrenia and Related Disorders (2005) [12] Assessment and close monitoring every 2-4 weeks along with the provision of information to the patient and their family about the risk and likelihood of progression. Other techniques such as cognitive behavioural therapy (CBT), stress management and vocational rehabilitation should be employed depending on any concurrent psychosocial difficulties. Antipsychotics are only to be prescribed when the patient has been frankly psychotic for over a week, or in cases when milder symptoms are associated with a risk of self-harm or aggression (however, patients without such a history are often treated regularly with antipsychotics and the primary concern here is that they may have a delirium or physical illness, which should be excluded first). [12]
The Australian Clinical Guidelines for Early Psychosis [13] Commencement of CBT for all patients identified as being at ultra-high risk is recommended. Family, vocational, educational and accommodation support should also be provided as required in a low stigma setting. Antipsychotic medication should only be considered once full threshold psychotic symptoms have been sustained for over a week, or if there is rapid deterioration accompanied by psychotic-like symptoms. [13]
Health Service
Nature of service provided
The Personal Assessment and Crisis Evaluation (PACE) clinic PACE provides information to individuals and their families about what it means to be at risk of psychosis. [14] They facilitate case management and provide a variety of in-house support services to families and carers including group programs, vocational and educational assistance and occupational therapy. [15] Specific treatment is largely in the form of voluntary participation in clinical trials, such as those looking at antipsychotic use or CBT in ultra-high risk individuals. [14]
Headspace These centres for 12-25 year olds combine specialist mental health, drug and alcohol and primary care services, vocational services and training, and employment support within a youth and family-friendly environment. [16,17] Headspace centres are also tasked with developing awareness campaigns for their local community and providing training for primary care and other workers using an evidence-based approach. [16]
Early Psychosis Prevention and Intervention Centres (EPPIC) Provide comprehensive in-patient and mobile components and aim to identify patients as early as possible and deliver phase-specific bestpractice interventions to psychotic individuals between the ages of 15 to 24. [19] This model has also been adopted widely around the world, including in the UK [20] and the US. [21]

 

The early intervention model has also been subject to some criticism. The major basis for this is a lack of evidence, especially with regard to the use of anti-psychotics in the prodromal stages of psychotic illness and the significant cost associated with creating a clinical infrastructure for patients who may never proceed to a long-term psychotic illness.

Results and Discussion

Evidence for early intervention

There is evidence from several small studies that psychotherapy such as CBT [22] and pharmacotherapy [3,23] can reduce the progression of ultra-high risk individuals to first episode psychosis.

Wyatt et al. [8] reviewed 22 studies, of varying study designs, which included contemporaneous control group studies, cohort studies, mirror image studies and early intervention studies. In these studies, patients with schizophrenia were either given or not given neuroleptics at a specific time during the course of their illness. 19 of the studies, in particular, looked at patients who were experiencing their first psychotic episode. After re-analysing the data, Wyatt et al. [8] showed that early intervention with a neuroleptic in first-break schizophrenic patients improved the long-term course of the illness, commonly assessed based on re-hospitalisation and relapse rates. It was also shown that with the use of neuroleptics, the length of the initial psychotic period was reduced. In addition, when neuroleptics were discontinued, it resulted in poorer outcomes as the patients were not able to return to their previous level of functioning and relapses occurred more frequently. Neuroleptic medication has the strongest support for relapse prevention in schizophrenia and is the basis of most interventions.

It has been suggested that the duration of untreated psychotic episodes directly correlates with less complete recovery, a higher rate of relapse and increased levels of compromised functioning, since these episodes have a toxic effect on the brain. [7,8,24-26] These studies, both retrospective and prospective, suggest that a longer DUP in the early stage of schizophrenia is associated with a longer time to remission, a lower level of recovery, a greater likelihood of relapse and a worse overall outcome.

Studies have shown that raising public awareness and using mobile outreach detection teams to identify candidate patients [27] has significantly reduced DUP, leading to beneficial outcomes. In particular there has been a reduction in negative symptoms in schizophrenic patients.

Arguments against early intervention

There are certain groups who are against early intervention. One of the arguments against early intervention relates to whether it is cost effective, as resources may be diverted from treatment programs for patients who already have an established diagnosis of psychosis. In addition, they argue that the great majority of high-risk patients do not in fact progress to frank psychosis. There is also the argument that some patients seeking early intervention may not have ‘true prodromal’ features, thus inflating the numbers of those who actually require early intervention. These arguments are discussed in more detail below.

Economic cost of early intervention may be infeasible

Those against early intervention believe the increased attention and funding given to early intervention diverts funding away from treatment in those with established psychosis. [28-30] They also argue that proponents of early intervention have touted the cost-effectiveness of early intervention as such programs utilise more outpatient resources compared to inpatient resources, thus reducing overall healthcare costs (with outpatient services being much cheaper than inpatient treatment). However, critics of early intervention have pointed out that implementation of a cost-effective treatment actually increases total costs [31,32] since cheaper treatment would have a much higher uptake compared to an expensive alternative, thus raising the total cost of treatment. In addition, Amos argues that total healthcare costs are further increased since in-patient costs are not reduced with early intervention. [33] This is because 80% or more of hospital costs are fixed costs and by shifting psychosis treatment to largely outpatient settings in the community, community costs increase but hospital costs are not reduced. [33] This is corroborated by previous studies, which show an increase in total costs when hospitalisation rates had been reduced. [34,35]

Most high-risk patients do not progress to frank psychosis

One possible explanation for this is that a subset of adolescents whom are identified as being UHR may just be odd adolescents that become odd adults with few progressing to a frank psychosis. The prominent child psychiatrist Sula Wolff was the first to describe these odd adolescents in her book, Loners: The Life Path of Unusual Children. [36] Her research has shown that while odd qualities such as those found in schizoid and schizotypal disorders are found pre-morbidly in patients with schizophrenia, very few children with such personality traits/disorders go on to develop schizophrenia. For example, in 1995 Wolff undertook a records survey of all psychiatric hospital admissions in Scotland. Overall, 5% of schizoid young people were affected by schizophrenia in adulthood compared to a population prevalence rate in the UK of 0.31-0.49%. [36] These numbers suggest that while the risk for schizophrenia in schizoid children is higher than that of the general population, it is still low. To reiterate, there may be a proportion of patients who are flagged as being prodromal but whom actually have qualities consistent with schizoid personality disorder that will never progress to psychosis.

Recently, there has been a decline in the proportion of patients at high risk of psychosis actually progressing to frank psychosis

This decline has important ramifications for the practice of early intervention. A decline in the transition rate of patients identified as UHR has been reported within the PACE clinic (Melbourne, Australia) and in other UHR clinics as well. [37,38] As an example, The PACE clinic has reported that each successive year between 1995-2000 had a rate equal to 0.8 of the previous year. [38] The reported decline in transition rate was not due to differing patient characteristics across the years, such as gender, age, family history, baseline functioning and degree of psychopathology and psychiatric symptoms. [38] Additionally, the UHR criteria remained unchanged in the PACE clinic between 1995-2000. [38]

There are a number of possible explanations for the declining transition rate to psychosis. Firstly, UHR patients are being detected more quickly than in the past (the duration of symptoms prior to detection is getting shorter). [38] However, it is unclear whether the resulting decline in transition rate is due to earlier treatment (which may be more effective than delayed treatment), the identification of increased numbers of false positives (those who are not going to progress to psychosis) or a combination of both. [38] There may also be an effect from clinicians becoming better at managing UHR patients. [38] Additionally, it has been noted that the decline in transition rate was more prominent for patients who met two of the UHR inclusion criteria simultaneously compared to those who met only one of the criteria. [38] This could have been due to the increased emphasis which was placed on detection of patients who met both criteria, both in the UHR clinic and from referrers, thereby leading to earlier detection and treatment. [38] This is also in keeping with the wider community shift and preoccupation towards early psychosis and its recognition, and the increase in available referral pathways.

The decline in transition rate also raises questions about the validity of intervention approaches, such as pharmacotherapy and psychosocial treatment, on patients who may not ultimately transition to psychosis. [38] Such intervention may be harmful and therefore unjustified in this context. The UHR concept, which is used extensively in psychosis research, may also have to be re-visited if many of the identified patients are not transitioning. [38]

Due to the uncertainties regarding the basis for the declining transition rate, a review of the role of UHR clinics may be warranted. [38] It may be necessary to initially monitor patients and treat conditions such as depression, substance use problems and anxiety disorders while withholding antipsychotic treatment until features suggestive of transition occur, such as worsening of sub-threshold psychotic symptoms. [38] This may be prudent in the context of detecting increasing numbers of patients who were never destined to transition to psychosis. In any case, further research is needed to clarify the ongoing uncertainties in this area.

Bias in patient selection

Specialised teams set up to treat early psychosis engage with anyone who is seeking help. However, Castle [39] believes that this would skew the treatment group, as it would engage those with help-seeking behaviours rather than prodromal psychosis. Furthermore, it also raises the issue that those seeking help may have signs and symptoms of what is a normal developmental process or a ‘psychosis proneness’, which is part of a normal distribution within the general population. [40] Thus, these individuals may not require treatment for psychosis at all as they would either grow out of ‘psychotic proneness’ or would
stabilise and never develop psychosis.

Prescribing anti-psychotics to a population that is not psychotic: An ethical implication

The potential dangers of psychotropic drugs on young people are outlined in the United Nations Convention on the Rights of the Child, where children are recognised as being particularly deserving of protection from unnecessary exposure to psychotropic substances. [41] However, much of the research into early intervention includes administration of a low dose of antipsychotics as a crucial and efficacious treatment option. [42] Furthermore, antipsychotics are known to have serious side effects including sedation, weight gain, mild sexual dysfunction and disconcerting extrapyramidal symptoms (EPS) such as pseudoparkinsonism, akathisia, acute dystonia, and tardive dyskinesia. [43] While these effects have a stronger association with first generation antipsychotics, there is increasing evidence suggesting that second generation antipsychotics (SGA) are associated with significant side effects such as weight gain, hyperprolactinemia and EPS in the adolescent population.

Summary and recommendations

In view of the currently available literature, the authors make the following summary and recommendations with regards to early intervention in psychosis.

  • Psychosis is a highly disabling condition with detrimental impacts on patients’ relationships and occupational and social functioning
  • Possible interventions that delay or prevent transition from the prodromal period to psychosis are important, both clinically and economically
  • A systematic review by the Cochrane Database found limited evidence about interventions to prevent psychosis. Despite this, early intervention facilities such as headspace are widespread in Australia

Our recommendations

  1. We do not recommend the use of antipsychotics in children and adolescents who have been identified as at increased risk but who have not yet progressed to frank psychosis. Exposing children and adolescents to the serious side effects of antipsychotics is both unethical and inappropriate considering a proportion of these patients will not progress to psychosis.
  2. We recommend more research into safer, less harmful interventions such as omega-3 fatty acids and psychotherapy. For omega-3 fatty acids, evidence suggests a beneficial effect on transition rates compared to placebo. [44] However, this evidence comes from a single trial with few participants. A replication study with a larger sample size is needed to more definitively ascertain the merit of this intervention
  3. As previously discussed, preliminary evidence shows that CBT may reduce the transition rate to psychosis. Further research should be undertaken to conclusively establish the benefit of psychotherapy in high-risk individuals. Further research should
    include investigation of the cost-effectiveness of psychotherapy as an early intervention for youth psychosis. In addition, research should aim to identify any detrimental effects associated with providing psychotherapy to patients who do not progress to psychosis.
  4. Patients identified as being at risk of developing psychosis should be monitored closely by a multi-disciplinary team. Team members may include a general practitioner, social worker, psychiatrist and psychologist. By closely monitoring at-risk patients, their progression into frank psychosis can be detected earlier and appropriate treatment given in a timely manner. Prompt detection and treatment of psychosis is crucial, as delayed untreated psychosis has been shown to result in poorer outcomes.

Table 2. Summary of the evidence supporting and arguments against early intervention in psychosis.

Evidence supporting early intervention Evidence from small studies showing psychotherapy such as CBT and pharmacotherapy can reduce the progression
of ultra-high risk individuals to first episode psychosis.Studies show that raising public awareness and using mobile outreach detection teams to identify candidate patients significantly reduces the duration of psychosis.
Arguments not in favour of early intervention The economic cost of early intervention may be infeasible.Most patients identified as being high risk do not progress to frank psychosis.Treatment teams for early psychosis may disproportionately target patients with “help
seeking behaviour” and thereby treat more patients who simply display signs and symptoms of a normal developmental process or “psychosis proneness”.The negative ethical implications associated with prescribing antipsychotics to a population that is not psychotic.

Acknowledgements

The authors would like to thank Professor Jeff Cubis and Professor David Harley for their guidance and expert opinion on the matter.

Conflict of interest

None declared.

Correspondence

H C Y Yu: u4788941@anu.edu.au

Categories
Review Articles

The α-5 subunit-containing GABA-A receptor: a target for the treatment of cognitive defects

Amnesic effects of benzodiazepines are in part the result of the activity of α5-subunit containing GABAA receptors (GABRA5). Negative modulators at this receptor could improve cognition. In order to explore this beneficial effect, this article reviews the evidence on the effects of GABRA5 negative modulators and searches potential uses for such drugs. A literature search found a number of GABRA5 negative modulators. These drugs generally improve hippocampal-dependant learning via an increase in longterm potentiation (LTP) in the hippocampus. Passive avoidance learning was also improved. In addition, the compounds examined demonstrated minimal side effects partly due to lack of binding to different alpha subunit-containing GABAA types. Due to its beneficial properties, there is potential for such a drug in treating Alzheimer’s, alcohol-related amnesia and Down syndrome. Despite the myriad animal studies that utilised GABRA5 negative modulators, only three human studies were found. Due to its cognitive enhancing properties and minimal side effects, further human trials should be conducted in order to ascertain the potential of such drugs in treating cognitive deficits.



Introduction
The α5 subunit-containing GABAA receptor: a target for the treatment of cognitive defectsγ-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the brain and is responsible for regulating neuronal excitability. There are at least three different receptors that it targets — GABAA, GABAB and GABAC. [1] The GABAA receptor is the main target for the popular class of drugs, the benzodiazepines. This receptor is an ionotropic membrane receptor, which facilitates the movement of chloride into cells. In neurons, this increases the threshold needed to excite them.[2] Benzodiazepines are positive modulators at this receptor and exert their effects by binding to the interface between the γ2 and α subunits on the GABAA receptor. [3] Since they are modulators and not agonists, they do not work in the absence of GABA. [4] They only bind to the receptors containing the α5, α3, α2, and α1 subunits. [5] Each of these subunits mediates different effects. In knockout mice, the α1 subunit has shown hypnotic/sedative effects, whilst the α3, α2 and α5 subunits have anti-anxiolytic effects and these have been exploited for therapeutic use.[6]

Despite their uses in treating various conditions, traditional benzodiazepines have numerous side effects. The main side effects associated with therapeutic use are amnesia, confusion, impaired coordination and dizziness. There may be tolerance due to rapid escalation in the dose needed to provide the required effect. There are also long-term issues with dependence. In acute overdose the most life-threatening effect is respiratory depression, especially when combined with alcohol. [7]

Interestingly, it is thought that positive modulators at the α5 subunit-containing GABAA receptors (GABRA5) produce the anterograde amnesia associated with benzodiazepine use. [7] Most of these receptors are found in the hippocampus (a brain region associated with memory) and provide tonic inhibition in this region. [8] The exploitation of this receptor has led to the increasing use of the infamous ‘date rape’ drug flunitrazepam, which is a positive modulator at the GABRA5 in addition to its other functions. Because of its amnestic effects, victims are unable to recall events following intoxication and this provides a major challenge for prosecutors. [7] Despite these negative properties, by using a GABRA5 negative modulator the opposite effect might The α5 subunit-containing GABAA receptor: a target for the treatment of cognitive defects be achieved and cognition improved. The use of such a drug could potentially improve the quality of life for those living with cognitive defects and could also counteract drug-induced amnesia (for example, alcoholic ‘blackout’). However, this must be balanced with the inverse activity of non-selective negative modulators which could produce
convulsant or anxiogenic effects. [4]

Based on the premise that a GABRA5 negative modulator could improve cognition, the aim of this literature review was to review the evidence on (1) the effects of GABRA5 negative modulators on cognition; and (2) investigate the potential of GABRA5 negative modulators in managing conditions involving cognitive defects.

Effects of GABRA5 selective negative modulators

A number of GABRA5 negative modulator compounds were examined. Many of the studies used animals as subjects. Studies in animals provide a solid starting platform for understanding the various physiological changes that a drug induces. [9] Of particular importance is the avoidance of potential side effects as a result of non-selective actions at other GABAA receptors. Side effects could include an increase in anxiety, aggressiveness, motor impairment, inability to sleep and proconvulsant effects. [7] Ultimately, the knowledge gained from animal experimentation can be used to conduct safe and effective clinical trials.

Dawson et al. [10] examined a compound named α51A (3-(5-Methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-yl)methyloxy]-1,2,4-triazolo[3,4-a]phthalazine), which has selective negative modulator effects at GABRA5. The authors found that α51A reversed the inhibiting effects of GABRA5 in the hippocampus in rats and mice. This resulted in an increase in performance in a memory test named the “delayed matching-to-position version of the Morris water maze”, which is a hippocampus-dependant cognitive test. [11] In addition, ‘long-term potentiation’ (LTP), which is thought to underlie the synaptic changes that take place during memory formation, was found to be enhanced in the hippocampus. [11,12] Benzodiazepine (agonist) effects and non-selective GABAA negative modulator effects were also examined. The authors found no anxiogenic, convulsant, withdrawal or motor-impairing effects from the drug. [10]

Only certain components of memory have shown to be improved by GABRA5 negative modulators. Collinson et al. [13] extrapolated on the results obtained by Dawson et al. [10] and looked at the effect of a modified version of α51A, α51A-II. They separated memory into three components — encoding, consolidation (conversion into long-term memory) and recall. Results were obtained by measuring performance in the delayed matching-to-position (DMTP) version of the Morris water maze in rats. The authors found that the compound improved encoding and recall but not consolidation in this hippocampaldependant memory test. [13]

The effect on cognition by another selective GABRA5 negative modulator was examined by Ballard et al. [14] This was done by the use of an imidazo-triazolo-benzodiazepine compound named RO4938581. The effect of this compound on cognition was examined in rats. [15] This compound demonstrated similar effects to those found by Dawson et al. [10] in that they found no convulsant or anxiogenic effects. In addition, similar to Dawson et al. [10], there was an increase in hippocampal LTP. The authors also found that working memory was enhanced since RO493881 reversed scopolamine-induced working memory impairment. [14] This was shown by an increase in performance in the DMTP task, which is used to assess spatial working memory. [14,15] It also reversed diazepam-induced spatial impairment. This was demonstrated by an increase in performance in the Morris water maze task. [14]

‘Moderate’ GABRA5 negative modulators improve passive avoidance learning but generally have no effect on active learning. [16] This was shown by an experiment conducted by Savic et al. [16] in which they examined effects of PWZ-029 (a ‘moderate’ GABRA5 negative modulator) on passive and active learning avoidance in rats. The result was obtained through various shuttle-box based behavioural experiments. This experiment proved that even at ‘moderate’ efficacy a GABRA5 negative modulator can induce memory formation. The compound also had no effect on muscle tension and anxiety (nonselective side effects). [16] Although promising, this study was limited by the fact that the compound only had ‘moderate’ negative modulator activity at GABRA5 so using a more efficient compound may display different effects on avoidance learning. Despite this limitation, this shows that the use of a ‘moderate’ GABRA5 negative modulator would be beneficial in the treatment of disease due to its limited side effects and its memory-enhancing properties. [16]

Application in management

The compounds examined in this review show that selective GABRA5 negative modulators have nootropic effects without any serious side effects, which are seen in non-selective negative modulators at the alpha subunit of the GABAA receptor. [17] Thus, there is strong potential for the use of GABRA5 negative modulators in healthcare settings. One major limitation is that most of the data obtained for this review was from animals. Further human trials need to be conducted to ascertain the potential of this drug. Drawing on the literature, possible future uses for a GABRA5 negative modulator are detailed below.

GABRA5 negative modulators could be used to treat Alzheimer’s disease since GABRA5 is preserved in Alzheimer’s disease patients.
[18] Alzheimer’s disease is commonly characterised by the gradual worsening of ability to remember new information. [19] Administration of a GABRA5 negative modulator could help with the ‘encoding’ and ‘recall’ of this information. [8] It could also be used to treat mild cognitive impairment (MCI), which is a risk factor for later developing the disease. [20] Administration of such a drug to patients may provide relief to older caregivers, who often show signs of sleep detriment. [21]

A review by Attack [22] in 2010 found two human trials on the GABRA5 negative modulator α5Ia and one trial on MRK-016. Since then no human trials were found and this could be an area of future research. The first study found that a potential application of GABRA5 negative modulators is the treatment of alcohol-induced amnesia. Nutt et al. [23] found that pre-treatment reduces alcohol’s amnestic effects in humans. This was measured by word list learning which is linked to hippocampal processing. Alcohol-induced amnesia has been shown to predict future alcohol-related injury. [24] Therefore, the use of a GABRA5 negative modulator may help in reducing this risk. In addition, it may also reduce alcohol-related stress since it has been found that amnestic episodes related to alcohol have resulted in moderate psychological stress. [25]

Unfortunately, GABRA5 does not improve age-related cognitive defects. In fact it has been found that α5IA significantly impairs cognition in the elderly despite having positive effects on the young. Attack [22] found that young subjects (mean age 22 years) performed much better than older subjects (mean age 72 years) on the paired associates learning test, which is sensitive to age-related cognitive decline. Therefore, this trial showed no potential in reversing age-related cognitive decline. This demonstrates that careful consideration based on age should be taken in to account when using this drug.

MRK-016 (3-tert-butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d]-[1,2,4]triazine) is another negative modulator and showed greater LTP in rat hippocampal slices than α51A. It also enhanced performance in the DMTP and Morris water maze tasks, which are used to test spatial memory. In humans, it was well tolerated in young adults with a maximum tolerated dose of 5 mg with 75% occupancy. In elderly subjects, however, it was poorly tolerated even at 10% of the maximum dosage in young adult males. Therefore, this particular drug has been precluded for development. [26]

Recent trials of GABRA5 agonists, in particular L-655,708 and MRK-016, have focused on restoring post-anaesthetic cognitive deficits. Lecker et al. [27] found that L-655,708 and MRK-016 reduced the potentiation of GABRA5 post-inhalation of isoflurane and sevoflurane. A further study by Zureck et al. [28] found that short-term memory assessed by the novel object recognition task was fully reversed by L-655,708 after isoflurane anaesthesia. This demonstrates the potential use of L-655,708 in reducing post-anaesthetic amnesia. However, further studies which include those performed on humans are needed to validate the potential of MRK-016 and other GABRA5 negative modulators in reducing post-anaesthetic amnesia.

The use of GABRA5 negative modulators could help with treating cognitive deficits related to Down syndrome. A recent review by Martínez-Cué et al. [29] investigated this specific application. It identified two studies that examined the effects of a GABRA5 inverse modulator on a Down syndrome mouse model (Ts65Dn). Braudeau et al. [30] found that acute treatment with the GABRA negative modulator α5IA improved learning deficits in the Morris water maze task. The second study also showed that chronic administration of a similar drug, RO4938581 has also been shown to have memory-promoting effects in the Morris water maze task on Ts65Dn mice. [31] In a practical sense, administration of such a drug could improve performance in learning a wide range of functional skills in those living with Down syndrome. For example, in children this may include learning how to use the toilet and administering self-care. [32]

Conclusion

The literature supporting the use of a GABRA5 negative modulator in the treatment of cognitive deficits is promising. GABRA5 negative modulators exert their actions by enhancing hippocampal dependant memory formation. There are minimal side effects as no withdrawal symptoms, convulsant, anxiogenic or motor-impairing effects were found. There is great potential for the use of GABRA5 negative modulators as they have been shown to reduce alcohol-related amnesia and may have potential in the treatment of Alzheimer’s disease. They could also treat cognitive deficits in Down syndrome patients, increasing the speed at which they learn functional skills. Due to these favourable findings, there is an increased need for human clinical trials in order to validate the potential for this important receptor target.

Acknowledgements

A/Prof Zoltan Sarnyai, for reviewing this article when I submitted it as an assignment.

Conflict of interest

None declared.

Correspondence

A Bhandari: abhishta.bhandari@my.jcu.edu.au

References

[1] Lonstein JS, Maguire J, Meinlschmidt G, Neumann ID. Emotion and mood adaptations in the peripartum female: complementary contributions of gamma-aminobutyric acid and oxytocin. J Neuroendocrinol. Forthcoming 2014. DOI: 10.1111/jne.12188.

[2] D’Hulst C, Atack JR, Kooy RF. The complexity of the GABAA receptor shapes unique pharmacological profiles. Drug Discov Today. 2009;14(17-18):866-75.

[3] Sigel E, Buhr A. The benzodiazepine binding site of GABAA receptors. Trends Pharmacol Sci. 1997;18(11):425-9.

[4] Rudolph U, Knoflach F. Beyond classical benzodiazepines: novel therapeutic potential of GABAA receptor subtypes. Nat Rev Drug Discov. 2011;10(9):685-97.

[5] Atack JR. GABAA receptor subtype-selective modulators. II. α5-selective inverse agonists for cognition enhancement. Curr Top Med Chem. 2011;11(9):1203-14.

[6] Mohler H. GABAA receptor diversity and pharmacology. Cell Tissue Res. 2006;326(2):505-16.

[7] Rang HP, Dale M. Rang and Dale’s Pharmacology: Churchill Livingstone; 2007.

[8] Collinson N, Atack JR, Laughton P, Dawson GR, Stephens DN. An inverse agonist selective for α5 subunit-containing GABAA receptors improves encoding and recall but not consolidation in the Morris water maze. Psychopharmacology (Berl). 2006;188(4):619-28.

[9] Ferreira LM, Hochman B, Barbosa MVJ. Modelos experimentais em pesquisa. Acta Cirurgica Brasileira. 2005;20:28-34.

[10] Dawson GR, Maubach KA, Collinson N, Cobain M, Everitt BJ, MacLeod AM, et al. An inverse agonist selective for α5 subunit-containing GABAA receptors enhances cognition. J Pharmacol Exp Ther. 2006;316(3):1335-45.

[11] Nakazawa K, Sun LD, Quirk MC, Rondi-Reig L, Wilson MA, Tonegawa S. Hippocampal CA3 NMDA receptors are crucial for memory acquisition of one-time experience. Neuron. 2003;38(2):305-15.

[12] Cooke SF, Bliss TV. Plasticity in the human central nervous system. Brain. 2006;129(Pt 7):1659-73.

[13] Collinson N, Kuenzi FM, Jarolimek W, Maubach KA, Cothliff R, Sur C, et al. Enhanced learning and memory and altered GABAergic synaptic transmission in mice lacking the alpha 5 subunit of the GABAA receptor. J Neuro Sci. 2002;22(13):5572-80.

[14] Ballard TM, Knoflach F, Prinssen E, Borroni E, Vivian JA, Basile J, et al. RO4938581, a novel cognitive enhancer acting at GABAA α5 subunit-containing receptors. Psychopharmacology (Berl). 2009;202(1-3):207-23.

[15] Goto K, Kurashima R, Watanabe S. Delayed matching-to-position performance in C57BL/6N mice. Behav Process. 2010;84(2):591-7.

[16] Savic MM, Clayton T, Furtmuller R, Gavrilovic I, Samardzic J, Savic S, et al. PWZ-029, a compound with moderate inverse agonist functional selectivity at GABAA receptors containing α5 subunits, improves passive, but not active, avoidance learning in rats. Brain Res. 2008;1208:150-9.

[17] Navarro JF, Buron E, Martin-Lopez M. Anxiogenic-like activity of L-655,708, a selective ligand for the benzodiazepine site of GABAA receptors which contain the α5 subunit, in the elevated plus-maze test. Prog Neuropsychopharmacol Biol Psychiatry. 2002;26(7-8):1389-92.

[18] Howell O, Atack JR, Dewar D, McKernan RM, Sur C. Density and pharmacology of α5 subunit-containing GABAA receptors are preserved in hippocampus of Alzheimer’s disease patients. Neuroscience. 2000;98(4):669-75.

[19] Daulatzai MA. Early stages of pathogenesis in memory impairment during normal senescence and Alzheimer’s disease. J Alzheimers Dis: JAD. 2010;20(2):355-67.

[20] Grundman M, Petersen RC, Ferris SH, Thomas RG, Aisen PS, Bennett DA, et al. Mild cognitive impairment can be distinguished from Alzheimer disease and normal aging for clinical trials. Arch Neurol. 2004;61(1):59-66.

[21] Rowe MA, McCrae CS, Campbell JM, Benito AP, Cheng J. Sleep pattern differences between older adult dementia caregivers and older adult noncaregivers using objective and subjective measures. J Clin Sleep Med. 2008;4(4):362-9.

[22] Atack JR. Preclinical and clinical pharmacology of the GABAA receptor α5 subtypeselective inverse agonist α5IA. Pharmacol Ther. 2010;125(1):11-26.

[23] Nutt DJ, Besson M, Wilson SJ, Dawson GR, Lingford-Hughes AR. Blockade of alcohol’s amnestic activity in humans by an α5 subtype benzodiazepine receptor inverse agonist. Neuropharmacology. 2007;53(7):810-20.

[24] Mundt MP, Zakletskaia LI, Brown DD, Fleming MF. Alcohol-induced memory blackouts as an indicator of injury risk among college drinkers. Inj Prev. 2012;18(1):44-9.

[25] Buelow G, Koeppel J. Psychological consequences of alcohol induced blackout among college students. J Alcohol Drug Educ. 1995.

[26] Atack JR, Maubach KA, Wafford KA, O’Connor D, Rodrigues AD, Evans DC, et al. In vitro and in vivo properties of 3-tert-butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)- pyrazolo[1,5-d]-[1,2,4]triazine (MRK-016), a GABAA receptor α5 subtype-selective inverse agonist. J Pharmacol Exp Ther. 2009;331(2):470-84.

[27] Lecker I, Yin Y, Wang DS, Orser BA. Potentiation of GABAA receptor activity by volatile anaesthetics is reduced by α5-GABAA receptor-preferring inverse agonists. Bri J Anaesth. 2013;110 Suppl 1:i73-81.

[28] Zurek AA, Bridgwater EM, Orser BA. Inhibition of α5 γ-Aminobutyric Acid Type A Receptors Restores Recognition Memory After General Anesthesia. Anesth Analg. 2012;114(4):845-55 DOI: 10.1213/ANE.0b013e31824720da.

[29] Martínez-Cué C, Delatour B, Potier M-C. Treating enhanced GABAergic inhibition in Down syndrome: Use of GABA α5-selective inverse agonists. Neurosci Biobehav Rev. Forthcoming 2014. DOI: 10.1016/j.neubiorev.2013.12.008.

[30] Braudeau J, Delatour B, Duchon A, Pereira PL, Dauphinot L, de Chaumont F, et al. Specific targeting of the GABAA receptor α5 subtype by a selective inverse agonist restores cognitive deficits in Down syndrome mice. J Psychopharmacol. 2011;25(8):1030-42.

[31] Martinez-Cue C, Martinez P, Rueda N, Vidal R, Garcia S, Vidal V, et al. Reducing GABAA α5 receptor-mediated inhibition rescues functional and neuromorphological deficits in a mouse model of down syndrome. J Neurosci. 2013;33(9):3953-66.

[32] Dolva A-S, Coster W, Lilja M. Functional performance in children with Down syndrome. Am J of Occup Ther. 2004;58(6):621-9.

Categories
Articles Review Articles

Factors that influence Australian medical graduates to become General Practitioners

Aim: To determine the factors that influence Australian medical graduates to become general practitioners. Method: A literature review was conducted. Medline, PubMed and Cochrane Library were searched using the terms; “Australia”, “medical”, “graduates”, “interns”, “students”, “choice”, “specialty”, “general”, “practice”, “factors” and “influencing”. Results: The factors were grouped into intrinsic (age, gender, personality and skill set,) and extrinsic influences (lifestyle, income, stress, location and role models), with extrinsic influences regarded as the most influential. Most importantly, 72% of the Australian medical graduates viewed work culture as important, while 56% prioritised flexibility of working arrangements and hours of work. Conclusion: There are a variety of both intrinsic and extrinsic factors influencing medical graduates to choose General Practice over others. This can be seen as an opportunity for Australian workforce planners and policy makers to target the extrinsic factors with the aim of balancing the medical workforce to combat the shortage of rural general practitioners.

Introduction

In the field of medicine, a specialty is simply a specific study of medical science. [1] Dermatology, Obstetrics and Gynaecology, Cardiology, Neurosurgery and General Practice are just a few of the vast array of medical specialties that medical graduates must decide between before embarking on a long, strenuous but nevertheless, highly rewarding journey. Students endure four to six years of medical school, only to begin a new journey as junior doctors. Internship is followed by residency, and the pathway after this depends upon the choice of specialty. [2] Medical students and junior doctors are faced with the tough challenge of selecting a specialty. This review seeks to precisely identify the factors influencing medical graduates to undertake General Practice.

This narrative review aims to explore the intricate complexities that invade the mind of medical graduates faced with the dilemma of choosing a specialty; in particular, what influences them to choose General Practice. The aims of this literature review are to highlight the spectrum of factors that play a role in medical students and interns choosing to undertake General Practice, and present medical colleges, recruitment agencies, workforce planners and national organisations with a platform upon which they can correct the imbalances in the medical workforce.

Methodology

This literature review covered recent literature that has focused on the factors influencing choice of medical specialisation (in particular General Practice) in Australia. Medical and social science databases were searched for publications from 1990-2013. Medline, PubMed and Cochrane Library were searched using these terms; “Australia”, “medical”, “graduates”, “interns”, “students”, “choice”, “specialty”, “general”, “practice”, “factors” and “influencing”. 7670 papers were identified through the database searches. These were then reviewed to only include studies conducted in Australia and concerning Australian medical graduates, which narrowed it down to 25 papers. 9 of these papers were excluded because they were not completely relevant to the topic. In addition, the bibliographies of articles were searched for further relevant publications. Studies referred to in this review vary widely and include both qualitative and quantitative studies.

Results

The primary influential factors involved in the selection of a particular specialisation can be separated into intrinsic and extrinsic factors.

Intrinsic Factors

Intrinsic factors include age, personality and gender. Individuals have little or no control over such factors. [8]

Age is an intrinsic factor that plays a role in the selection of a particular specialty. The majority of medical students in Australian universities are under the age of 24. [9] Despite this, there has been an increase in the number of ‘mature age’ students over the past two decades. One Australian study, that compared the career choices of medical graduates, found that older students were more likely to specialise in a primary care field such as General Practice. [3]

An individuals’ personal set of skills and the satisfaction they receive from using these skills are intrinsic factors which play a large role in influencing medical graduates to undertake General Practice. In 2005, Harris et al examined the factors influencing the choice of specialty of 4259 Australian medical graduates. [3] The study showed that 79% of graduates considered their own skills and aptitude to be of importance when selecting General Practice. Personal satisfaction was also linked to choosing General Practice as a specialty. Laurence and Elliot supported this in their 2007 study which concluded that personal satisfaction arises from procedural skills, activities involved and patient contact. [4] All of the 54 Pre-Registration Junior Medical Officers (PMJOs) interviewed in South Australia, agreed with this however, this was only a small sample size.

Gender has been shown by studies to be a vital influencing factor for Australian medical graduates when choosing General Practice as a speciality. Whilst the responsibilities of raising children have evolved over the past few decades, Prideaux et al found that Australian female medical graduates are more likely to become specialists in General Practice due to child bearing responsibilities and family commitments. [10] It has been noted the Australian literature that female doctors tend to work shorter hours and have a preference for working shorter hours due to family commitments. [11] Despite this, there has been a rise in male doctors choosing to work fewer hours due to family reasons. [10] This reflects the fact that both partners now commonly work.

Extrinsic Factors

Extrinsic factors include stress, work hours, family commitments, lifestyle and mentors. They are variables that may be controlled. [8]

Lifestyle plays the greatest role in influencing medical graduates to choose particular specialties over others. Laurence and Elliot found that 100% of the 54 South Australian PMJOs interviewed regarded lifestyle as a vital factor in choosing General Practice as a specialty. [4] This included hours worked, stress, career potential and potential for travel (55%). Most participants described their ideal job as having shorter working hours and less time on call. Most PMJOs also wanted a certain amount of control over hours and hence chose anaesthesia and GP practice. Wanting a life outside of medicine (85%) for example, spending more time with family and friends was also important. [4] Similarly, Harris et al also rated extrinsic factors as the most influential factors of choosing a medical specialty in Australia. [3] Seventy two percent of the Australian medical graduates viewed work culture as important, while 56% prioritised flexibility of working arrangements and hours of work. In contrast, Thomas concluded that only 28% of Australian medical graduates saw work life balance and lifestyle as important to selecting General Practice as their specialty. [7] However, this study had a small sample size and focused on only one speciality (General Practice).

Location is of importance when choosing a medical specialty. Stagg et al found that key influences on choosing a rural pathway specialty were mentors and undergraduate rural exposure. [6] In contrast, Ward et al, in a longitudinal study that followed 229 UWA medical graduates, showed that a rural background is the most important predicator of rural general practice. [5] Clearly, there factors that influence an individual to undergo a rural generalist pathway are multifactorial and more research is needed in this area.

The studies used in this literature review all stressed the importance of role models in influencing Australian medical graduates to choose particular specialties. Laurence and Elliot studied when, what and how SA pre-registration junior medical officers made their career choice. [4] Fifty four percent of the 54 graduates perceived the role of mentors, supervisors and consultants to be of importance in selecting General Practice as a specialty. Their interaction with ‘mentors’ was through observing and asking questions. Role models were seen to demonstrate specific characteristics admired by the students. [12]

Discussion

The results confirm that choosing General Practice as a specialty is a complex decision strongly influenced by personal qualities (intrinsic factors), individual experiences and opportunities (extrinsic factors), and domestic circumstances. Whilst parenting dynamics have changed over the last century, there is still a trend for females to choose General Practice over other specialties due to flexibility and option of part time employment, which may be helpful when choosing to start a family. [13]

The most important extrinsic factors include lifestyle, work experience since graduation, flexible hours, influence of mentors and hours of work. [3] In Australia, it was concluded that factors relating to lifestyle and job satisfaction were the most important influencing factor. [4] This is consistent with the belief that recent graduates regard lifestyle factors as more important than income. [14] The younger generation of graduates also prioritise potential for travel. These graduates are influenced by their experience of General Practice and confirm that work experience is helpful for developing knowledge within medical. [3]

The results also suggested that trainees in different specialties prioritised certain influencing factors over others. Surgical trainees viewed mentors and role models as more important than trainees in other specialties. Additionally, General Practice trainees were more likely to prioritise flexibility, whilst this factor was of less importance to trainees in Adult Medicine. [15] Compounding this notion is the fact that surgical and emergency trainees found it extremely important to do procedural work compared to trainees in other programs. [3]

Clearly, there is not one single factor that influences an individual to undertake a particular career path, rather a vast array of factors. A medical graduate’s choice of career is dependent upon a wide range of intrinsic and extrinsic factors. [3] Whilst there is not much chance of altering intrinsic factors, the nature of extrinsic factors allows for an interventionist approach.  [16]

The main goal of medical workforce agencies and groups is to ensure a balance of doctors across a vast array of specialties to provide equal, effective and holistic medical care to the community. For 80% of doctors, the decision about choice of specialty has to be made by the end of the third postgraduate year (PGY3). [3] As such, training programs, teaching facilities and recruitment agencies involved in medical workforce planning should aim to educate medical students and graduates up until PGY3 and allow them to make an informed decision. Given the importance of extrinsic factors, there should be a review of the work culture typical of specialties that are under-represented. [13] Training providers can therefore implement strategies that attempt to increase entry to less well-represented specialties. [4]

This literature review has a number of limitations. Firstly, this review was limited to articles concerning the Australian medical workforce. This review excluded international medical graduates, which may have given greater insight into the factors influencing the specialty choice of graduates. A comparison study could be done in the future, comparing the mindset of Australian medical graduates to overseas graduates. Secondly, whilst certain conclusions can be made concerning the factors which influence choice of General Practice as a specialisation, this review did not focus on the factors that influence doctors to change specialties or even the percentage of medical students that graduate without having made a decision about their future career. Research on the factors influencing General Practice specialty choice could be improved by including a larger number of schools and students, studying trends over several years, and using validated measures and outcomes.

Conclusion

The main factors which were identified as influencing medical graduates to choose General Practice included both intrinsic and extrinsic factors. Additionally, a career choice that is made when an individual is young may not represent how they will feel as they progress through life and their priorities will often change. While there remains a continuous need for valuable research in the area of factors affecting medical specialisation, it appears that we need to use this information to prevent imbalances and skews in medical workforce planning. There is a great opportunity for governments, health authorities and the medical profession to influence extrinsic determinants of choice of specialty. [13]

Acknowledgements

I was given the opportunity to conduct this research through the GPSN First Wave Scholarship and Tropical Medical Training (TMT). I am thankful to Dr. Aileen Traves who provided me with support, encouragement and feedback throughout the course of this review.

Conflict of interest

None declared.

Correspondence

K Singh: karan.singh@my.jcu.edu.au

References

[1] Ellsbury, K. E., & Stritter, F. T.. A study of medical students’ specialty-choice pathways: trying on possible selves. Acad Med. 1997; 72, 534-541.

[2] Zurn, P., Dal Poz, M. R., Stilwell, B., & Adams, O. Imbalance in the health workforce. Hum Resour Health. 2004; 2(1), 13.

[3] Harris, M. G., Gavel, P. H., & Young, J. R. Factors influencing the choice of specialty of Australian medical graduates. Med Educ. 2005; 183(6), 295.

[4] Laurence, C. and Elliott, T. When, what and how South Australian pre-registration junior medical officers’ career choices are made. Medical Educ. 2007; 41: 467–475. doi: 10.1111/j.1365-2929.2007.02728.x

[5] Ward, A. M., Kamien, M. and Lopez, D. G. Medical career choice and practice location: early factors predicting course completion, career choice and practice location. Medical Educ. 2004; 38: 239–248. doi: 10.1046/j.1365-2923.2004.01762.x

[6] Stagg, P., Greenhill, J., & Worley, P. A new model to understand the career choice and practice location decisions of medical graduates. Rural Remote Health. 2009; 9(4):1245.

[7] Thomas, T. Factors affecting career choice in psychiatry: a survey of RANZCP trainees. Australas Psychiatry. 2008; 16(3), 179-182.

[8] Belfer, B. Stress and the medical practitioner. Stress Medicine. 1989; 5(2), 109-113.

[9] Joyce C, McNeil J & Stoelwinder J, ‘More doctors, but not enough: Australian medical workforce supply 2001–2012’, Med J Australia 2006; 185(3):182

[10] Prideaux D, Saunders N, Schofield K, Wing, L, Gordon J, Hays R, Worley P, Martin A, Paget N, ‘Country report: Australia’, Med Educ. 2001;35:495-504.

[11] Firth-Cozens J. and Lema VC. ‘Specialty choice, stress  and  personality:  Their relationships over time’, Hosp Med. 1999; 60(10):751-55

[12] Dunbabin, J., & Levitt, L. (2003). Rural origin and rural medical exposure: their impact on the rural and remote medical workforce in Australia. Rural Remote Health, 3(1), 212.

[13] Australian Medical Workforce Advisory Committee. Career decision making by doctors in their postgraduate years — a literature review. Sydney: AMWAC, 2002. (AMWAC Report 2002.1.)

[14] Joyce, C. M., & McNeil, J. J. Fewer medical graduates are choosing general practice: a comparison of four cohorts, 1980-1995. Med J Australia. 2006;185(2):102.

[15] Mowbray, R. Research in choice of medical  speciality: A review of the literature 1977-87. Aust NZ J Med. 1989;19(4):389-399.

[16] Joyce, C. M., Stoelwinder, J. U., McNeil, J. J., & Piterman, L. Riding the wave: current and emerging trends in graduates from Australian university medical schools. Med J Australia. 2007;186(6): 309.

Categories
Review Articles Articles

Sugammadex – the solution to our relaxant problems?

Sugammadex is the first of a class of selective relaxant binding agents. It acts by binding with high affinity to steroidal non-depolarising neuromuscular blockade drugs terminating neuromuscular blockade (NMB) through 1:1 encapsulation. Reversal of NMB has traditionally been performed by acetylcholinesterase inhibitors however these drugs have their drawbacks and are therefore not ideal.  This review examines the indications and advantages of sugammadex as well as the potential risks and shortcomings associated with its use.  Sugammadex is a relatively new drug that has been shown to be efficacious with an improved side effect profile as compared to its alternatives however several factors associated with its use have yet to be determined. These shortcomings have relevance on a therapeutic level as well as on a health economics level.

Introduction

NMB has been an important development in anaesthetic practice improving operative scenarios through patient paralysis. Muscle relaxation facilitates endotracheal intubation, ensures patient immobility and improves conditions for laparoscopic abdominal surgery. [1] Broadly speaking, the two classes of agents used are the depolarising NMB agents, of which there is only one in use, and the non-depolarising NMB agents. One of the significant problems with the non-depolarising NMB agents is their propensity to cause post operative residual blockade. This side effect of the drug has both patient safety implications and economic implications. The perfect solution to post operative residual blockade is absolute reversal of a non-depolarising NMB agent. This is routinely performed by cholinesterase inhibitors. These drugs however are less than perfect, as will be discussed and come with their own side effects. [11] A relatively new drug that has appeared on the marked is sugammadex, a selective reversal agent that is considered far superior. Given the recent arrival of sugammadex to the market, its use is yet to be perfected and its risks are yet to be fully understood. Furthermore it is a very costly drug raising questions regarding cost effectiveness. This review article will look at the extent to which sugammadex is the solution to the problems associated with muscle relaxant in anaesthesia.

Method

The study was performed through review of existing literature on sugammadex and its use.  Searches were performed using Ovid MEDLINE and the Cochrane Database of Systematic Reviews using the following terms: sugammadex, rocuronium, pancuronium, neostigmine, vecuronium, neuromuscular block, neuromuscular blockade, post operative residual block, post operative residual curarisation, post operative residual paralysis and economic assessment. Titles and abstracts were read and assessed for relevance to the paper. Bibliographies of the identified articles were hand searched to find additional relevant studies. Searches were limited to: humans and the years 2000 to current.

Results

The Ovid MEDLINE search identified 1832 articles. Of these, 15 articles were identified as pertinent to this review. The Cochrane Database of Systematic Reviews identified one systematic review. A remaining six articles were identified from bibliographies. Therefore, a total of 21 articles were included in the final analysis.

Discussion

Neuromuscular Blockade

Neuromuscular blocking agents are used on certain patients undergoing anaesthesia in addition to an anaesthetic agent and an analgesic agent. The drugs have significant risks. They pose the hazard of post-operative residual blockade which will be discussed. They are also the most common cause of anaphylaxis during anaesthesia accounting for between 60% and 70% of cases. The most commonly offending agents are rocuronium and suxamethonium. [5]

Neuromuscular blocking agents aim to totally paralyse the surgical patient by creating a blockade at the neuromuscular junction. This is not a therapeutic intervention but is rather used to facilitate endotracheal intubation, to eliminate spontaneous ventilation and to provide abdominal muscle relaxation for laparoscopic surgery. [4]

There are two classes of neuromuscular blocking drugs; depolarising agents and non-depolarising agents. Depolarising agents work by binding to nicotinic receptors causing depolarisation. They are not metabolised by acetylcholinesterase unlike acetylcholine thus prolonged activation of the receptor is produced causing paralysis. The only clinically approved depolarising agent is suxamethonium, a very short acting non-reversible drug. [22]

The other class is the non-depolarising agents. These are competitive antagonists that bind to post-synaptic nicotinic receptors preventing access and depolarisation by acetylcholine. [22] There are numerous agents under this class, notably pancuronium, rocuronium, vecuronium and mivacurium. These drugs are categorised by their length of action; pancuronium is long acting, rocuronium and vecuronium are intermediate acting and mivacurium is short acting. They are used in different scenarios depending upon procedural requirements.

Rocuronium

Rocuronium is a commonly given non-depolarising neuromuscular blocking agent and is the primary target agent of sugammadex. It has a quick onset of action of 1-2 minutes and if given in high doses can mimic the rapid onset of suxamethonium. This is useful when considering rapid sequence induction for Caesarean section. If given in such high doses however its duration of action is lengthened behaving in a manner similar to pancuronium increasing the risk of postoperative residual blockade. It has a good side effect profile and has a 30 to 50% quicker recovery rate than pancuronium. [2,4] The problem with non-depolarising NMBDs is the risk of postoperative residual curarisation or residual NMB and the significant but small risk of anaphylaxis.

Post-operative residual neuromuscular blockade

Post-operative residual NMB presents a very real risk to surgical patients.  It is a potentially reversible condition and should be avoided where possible.  It has the potential to impair the integrity of an airway and can contribute to patient death. [6] Classic signs include airway obstruction, inadequate ventilation and hypoxia. Evidence suggests the incidence of adverse respiratory events is from 1.3 to 6.9% with one study suggesting the figure as high as 88% during the post anaesthetic care period. [7,8]  The reason for such great variability in figures is in part due to the different definitions and methods of detection.  In addition to patient risk, there is also evidence to suggest residual NMB has economic consequences contributing to operating theatre congestion and a bottleneck in patient flow. [9]

Postoperative residual blockade can be minimised through two strategies: 1) pharmacological reversal of NMBD effects and 2) optimisation of NMBD dosing through careful monitoring and titration of the relaxant. [11]

Neuromuscular Blockade Monitoring

Neuromuscular monitoring is routinely practiced, most commonly with train of four (TOF) ratios1. Classically a TOF of <0.7 was the criteria for residual NMB.  This, however, has been discredited by Murphy et al. (2009) with evidence suggesting a TOF <0.9 is required to ensure a recovery. [7] Despite increasing stringency of neuromuscular monitoring the methods are not sufficiently objective or accurate.  Naguib et al. [10] found in their meta-analysis the difference in residual NMB between TOF monitored and non-monitored patients with intermediate acting NMB agents was not statistically significant (P=0.314); however, incidence was increased with long acting NMB agents as compared with intermediate NMB agents. [10] Further methods of NMB monitoring include tidal volume, vital capacity, sustained tetanus, head lift and hand grips however all are considered inferior to TOF. [2]

Neuromuscular Blockade Reversal Agents

The other strategy for the prevention of residual paralysis is the use of pharmacological measures. Kovac et al. (2009) postulated that

“An ideal NMB reversal agent would; (1) have rapid onset; (2) be 100% effective and predictable; (3) reverse any degree of NMB; (4) be effective in the presence of potent anaesthetics; and (5) have minimal or no side effects.” [1]

Neostigmine

The common class of drug for NMB reversal agents are cholinesterase inhibitors, the most commonly used being neostigmine. [1,2] Cholinesterase inhibitors prevent the breakdown of acetylcholine in the neuromuscular junction, increasing neuromuscular transmission. [12] Neostigmine does not have a rapid onset, with the mean time to muscle recovery being 50.4 minutes. [16] The drug cannot reverse deep NMB with TOF<0.1. [13] The drug also has a ceiling dose and can only reverse drugs of certain potencies and of certain doses. [2] Duration of action is limited and consequently residual paralysis may still be evident or paralysis may reappear post administration. [3] The drug also has significant parasympathetic side effects due to excessive stimulation of muscarinic receptors. Side effects include bradycardia, arrhythmias, nausea, vomiting, increased GIT motility, bronchospasm and excessive secretions. To prevent these side effects, anticholinergic drugs are co-administered, notably glycopyrrolate or atropine, which have their own side effects, notably tachycardia, altered cardiac conduction, dysrhythmias and urinary retention. [1,12] In addition to the side effects, anticholinesterase drugs have further limitations including their lack of predictability and unreliability. [13]

As discussed, there are significant issues with residual NMB that are clinically underappreciated. The standard reversal agents that are routinely used are not without their drawbacks; their onset is slow, their side effect profile is significant and their efficacy is insufficient in particularly deep NMB. Furthermore, monitoring methods for residual blockade are inaccurate and technically difficult.

Sugammadex

Due to the limitations of the current class of NMB agents, sugammadex has become of interest. It is a modified cyclodextrin that has a high affinity with steroidal NMB agents (rocuronium>vecuronium>>pancuronium). [1,12] Cyclodextrins are oligosaccharides arranged in a circular shape surrounding a central cavity that can be used to bind molecules within the cavity, eliminating the target’s pharmacological action. In the case of sugammadex, cyclodextrins are modified to have a rocuronium inclusion complex. It will bind to all non-depolarising NMB agents, although with a decreased affinity. [23]

One of the major benefits of sugammadex is that unlike the anticholinesterase inhibitors, it does not interfere with the receptor systems but rather acts on the NMB agent itself, meaning there are little to no muscarinic side effects. The drug binds to the respective NMB agent rendering it unavailable at the neuromuscular junction. [12] A high dose can be given if required without a high risk of cardiovascular effects, as with neostigmine. Furthermore it does not need to be given with a muscarinic agonist, unlike anticholinesterase agents, eliminating the potential for further adverse events.

The drug is currently approved for use in Australia and the European Union; however, it is yet to be approved by the FDA in the United States. In August 2008, a not-approvable letter was issued not due to lack of efficacy but rather due to the risk of hypersensitivity and allergic reactions that had not been adequately determined. Further studies are currently being performed by Schering-Plough. [1]

The efficacy of sugammadex is well established by several significant studies. It has been shown to be a very effective NMBD reversal agent of non-depolarising NMB. Puhringer et al. (2010) reported an improvement in NMB reversal from rocuronium and vecuronium as compared with placebo, however these results represented trends and were not statistically significant. Mean rocuronium reversal times were 96.3 min with placebo and 1.5 min with sugammadex. Mean vecuronium reversal times were 79min and 3 min respectively. [20] One study by Lee et al. (2009) found that reversal of profound high dose rocuronium induced NMB with sugammadex reversal, and was substantially quicker than the use of the short acting suxamethonium. [18] Jones et al. (2008) found in a randomised comparison that sugammadex reverses profound rocuronium induced NMB significantly faster than that of neostigmine. [16] Alvarez-Gomez et al. (2007) made a similar finding in their study comparing the two drugs. [19] Sugammadex is also thought to halt relaxant induced anaphylaxis as it encircles the relaxant drugs theoretically preventing further immune reactions. However, this has not be sufficiently studied to confirm. [5] The drug has also been used successfully to reverse rocuronium induced NMB in a ‘can’t intubate can’t ventilate’ scenario. [21]

That being said there are adverse events as have been reported in 30 studies looking at 2000 patients. The most frequently reported side effects with an incidence greater than 2%, were hypotension, bronchospasm, QTc prolongation greater than 400msec, constipation, hyperactivity and altered taste sensation. Less common side effects included cough, dry mouth, temperature changes, parasthesia, parasomia, mild erythemia, abdominal discomfort, increased creatinine phosphokinase, bradycardia and dizziness. These adverse events did not appear to have a dose-response relationship. [1] While generally well tolerated, the adverse events one ought to be aware of are procedural pain, nausea and vomiting. [3]

Sugammadex can serve a purpose in rapid sequence induction. Traditionally, suxamethonium was used due to its quick speed of onset and short duration of action. However, this drug comes with a substantial list of side effects. [4] Instead, rocuronium can be given in high doses to quicken onset and can be quickly reversed at the close of the operation with sugammadex, although this is still considered second line.

The risks of residual NMB, as discussed previously, can be eliminated with the use of sugammadex. There are still some concerns for its regular use. Many studies have been conducted on the drug, looking at factors such as side effects and suitable dose ranges; however, more studies need to be conducted with larger cohorts to fully appreciate the risks. Patients with poorer health and who are more predisposed to adverse events have yet to be studied in great detail. [3]

While the cost of sugammadex is of no therapeutic relevance it needs to be taken into account from a health economics point of view. The cost is significant with a 200mg/2mL vial costing AUD188.90 and a 500mg/5mL vial costing AUD477.80 (cost sourced from FRED Dispense®, accessed 9th August 2013). It is not covered by the PBS and must be bought privately. Two systematic reviews have been performed in the UK on the cost benefit of sugammadex, both published in the British Journal of Anaesthesia. Both studies acknowledge that there could be cost benefit both from mortality and morbidity reduction point of views and with regards to optimisation of theatre time and post-anaesthetic care. However, the studies conclude that it would not be feasible to make an accurate economic assessment due to a lack of evidence. [14,15] It should be noted that these studies are UK relevant and apply differently to Australian practice. Zhang et al. (2008) found in their preliminary study of cost benefit that there is an appreciable decrease in postoperative time spent in an operating theatre improving cost efficiency; however, this failed to take into account the drug cost itself. Furthermore the study is applicable to the US health system and again may lack relevance to the Australian health system. [17]

The impetus for this paper came from an episode that occurred in theatre. A middle aged female due to receive a cholecystectomy was in an extremely anxious state before entering the operating theatre. She was convinced to go ahead with the procedure, which was uneventful. She was paralysed with rocuronium which was reversed with sugammadex. Upon reversal, the patient had a sudden severe reflexive episode going into a tonic-clonic contracture causing her jaw to occlude the endotracheal tube, in turn causing her oxygen saturation levels to fall. She had to be re-paralysed with suxamethonium to allow for manual respiration with bag and mask.

An episode as described above is not an uncommon event and can occur during the emergence from anaesthesia; however the episodes are rarely so severe. It is very possible the sugammadex can be partly blamed for the reflexive episode, with a sudden return of muscle tone increasing afferent input through the muscle and tendon stretch receptors causing the biting. Because the standard reversal agents are not as effective as sugammadex, similar reflexive episodes that have taken place will have not had the severity seen here. The drug is still very new and anaesthetists are perhaps yet to fully understand its use. With experience such events will become increasingly rare through improved use.

It has been shown convincingly that sugammadex is a superior NMB reversal agent to the cholinesterase inhibitors in terms of efficacy, although it has a significant side effect profile. Despite the considerable research that has been performed on the benefits and risks of the drug’s use, there are still many gaps in the literature which require further research.

There was no case report or evidence of similar cases to that in the clinical scenario discussed earlier. A case report of this incident may be of value. The patient’s response may have been due to incorrect dosing or indeed a rare reaction that is yet to be clinically identified.

Conclusion

This paper examined the use of sugammadex and its role in anaesthetic, focussing both on the risks and benefits of use. Having studied the available literature, there is a clear therapeutic benefit in the reduction of postoperative residual NMB, a preventable event that poses significant risk to patients. It presents a superior alternative to the current first line anticholinesterase NMB reversal agents. The benefit of the drug from a health economics point of view is yet to be determined, having regard to its high cost. Furthermore, the potential adverse effects and hypersensitivity reactions have not been adequately studied. The true side effect profile may require a very long period of testing or long term routine use before there is a good understanding. Sugammadex does have a role in very specific anaesthetic scenarios, however, given its significant cost and gaps in the literature, it cannot be recommended suitable for routine use.

Conflict of interest

None declared.

Correspondence

H Badgery: hebad2@student.monash.edu

References

[1] Kovac AL. Sugammadex: the first selective binding reversal agent for neuromuscular block. Journ Clin Anes.; 2009;21(6):444 – 453.

[2] Miller RD, Pardo M 2011 Basics of Anaesthesia. Sixth edition, pp 224-226

[3] Yang LPH, Keam SJ. Sugammadex: A review of its use in Anaesthetic Practice. Drugs; 2009;69(7):919 – 942.

[4] Rang HP, Dale MM, Ritter JM, Moore PK. 2003 Pharmacology, Fifth Edition. pp 149-154.

[5] McDonnell NJ, Pavy TGP, Green LK, Platt PR. Sugammadex in the management of rocuronium-induced anaphylaxis. Brit Journ Anaes; 2011;106(2):119-201.

[6] Eikermann M, Peters J, Herbstreit F. Impaired upper airway integrity by residual neuromuscular blockade: increased airway collapsibility and blunted genioglossus muscle activity in response to negative pharyngeal pressure. Anesthesiol; 2009;110(6):1253-1260.

[7] Murphy GS, Szokol JW, Marymont JH, Greenberg SB, Avram MJ, Vender JS. Residual neuromuscular blockade and critical respiratory events in the postanesthesia care unit. Anesth Analg; 2009;107(1):130-7.

[8] Mathias LAST, de Bernadis RCG. Postoperative Residual Paralysis. Rev Bras Anesthesiol; 2012;62(3):439-450.

[9] Butterly A, Bittner EA, George E, Sandberg WS, Eikermann M, Schmidt U. Postoperative Residual Curarization from intermediate-acting neuromuscular blocking agent delays recovery room discharge. Brit Journ Anaes; 2010;105(3):304-309.

[10] Naguib M, Kopman AF, Ensor JE. Neuromuscular monitoring and postoperative residual curarisation: a meta-analysis. Brit Journ Anaes; 2007;98(3):302-316.

[11] Maybauer DM, Geldner G, Blobner M, Pühringber, Hofmockel R, Rex C, Wulf HF, Eberhart L, Arndt C, Eikermann M. Incidence and duration of residual paralysis at the end of surgery after multiple administrations of cisatracurium and rocuronium. Anaes; 2007;62(1):12-17.

[12] Abrishami A, Ho J, [12] Abrishami A, Ho J, Wong J, Yin L, Chung F. Sugammadex, a selective reversal medication for preventing postoperative residual neuromuscular blockade. Cochrane Database Syst Rev [Internet] 2009 [cited 15 Sept 2012]. Available from http://onlinelibrary.wiley.com.ezproxy.lib.monash.edu.au/doi/10.1002/14651858.CD007362.pub2/pdf

[13] Magorian T, Lynam DP, Caldwell JE, Miller RD: Can early administration of neostigmine in single or repeated doses alter the course of neuromuscular recovery from a vecuronium-induced neuromuscular blockade? Anesthesiol; 1990; 73(3):410-414.

[14] Chambers D, Paulden M, Paton F, Heirs M, Duffy S, Hunter JM, Sculpher M, Woolacott N. Sugammadex for reversal of neuromuscular block after rapid sequence intubation: a systematic review and economic assessment. Brit Journ Anaes; 2010;105(5):568-575.

[15] Paton F, Paulden M, Chambers D, Heirs M, Duffy S, Hunter JM, Sculpher M, Woolacott N. Sugammadex compared with neostigmine/glycopyrrolate for routine reversal of neuromuscular bloc: a systematic review and economic evaluation. Brit Journ Anaes; 2010;105(5):558-567.

[16] Jones RK, Caldwell JE, Brull SJ, Soto RG. Reversal of profound rocuronium-induced blockade with Sugammadex. A randomized comparison with neostigmine. Anesthesiol; 2008;109(5):816-824.

[17] Zhang B, Menzin J, Tran MH, Neumann PJ, Friedman M, Sussman M, Hepner D. The potential savings in operating room time associated with the use of sugammadex to reverse selected neuromuscular blocking agents: findings from a hospital efficiency model. Val Health; 2008;11(3):244.

[18] Lee C, Jahr JS, Candiotti KA, Warriner B, Zornow MH, Naguib M. Reversal of Profound Neuromuscular Block by Sugammadex Administered Three Minutes After Rocuronium: A Comparison with Spontaneous Recovery from Succinylcholine. Anesthesiol; 2009;110(5):1020-1050.

[19] Alvarez-Gomez JA, Wattiwill M, Vanacker B, Lora-Tamayo JI, Khunl-Brady KS. Reversal of vecuronium-induced shallow neuromuscular blockade is significantly faster with Sugammadex compared with neostigmine. Euro Journ Anaes; 2007;24(suppl.39):124-125.

[20] Puhringer FK, Gordon M, Demeyer I, Sparr HJ, Ingimarsson J, Klarin B, van Duijnhoven W, Heeringa M. Sugammadex rapidly reverses moderate rocuronium- or vecuronium- induced neuromuscular block during sevoflurane anaesthesia a dose-response relationship. Brit Jour Anaes; 2010;105(5):610-619.

[21] Curtis R, Lomax S, Patel B. Use of Sugammadex in a ‘can’t intubate, can’t ventilate’ situation. Brit Journ Anaes; 2012;108(4):612-614.

[22] Appiah-Ankam J, Hunter JM. Pharmacology of neuromuscular blocking drugs Contin Educ Anaesth Crit Care Pain; 2004;4(1):2-7.

[23] Baldo BA, McDonnell NJ, Pham NH. Drug-specific cyclodextrins with emphasis on sugammadex, the neuromuscular blocker rocuronium and perioperative anaphylaxis: implications for drug allergy. Clin Exp Allergy; 2011;41(12):1663-1678.

 

Categories
Review Articles Articles

Examining the pathological nature of Hepatitis C and current drug therapies used in an Australian general practice context

Aim: This review aims to examine the pathological nature of Hepatitis C and review current drug therapies relevant to Australian health practitioners. Methods: Terms hepatitis C, Australia, pathogenesis and current treatment were searched using MEDLINE and CHINAL databases to identify research articles and systematic reviews. Constraints were used when researching drug developments to include only full-length papers, on humans published between 2009 and 2013.  Literature was analysed to identify shared themes. Sixty-eight articles were analysed and fifty-two chosen based on relevance to objective, reputable data sources and current information. Two websites and five books were included upon cross referencing data to journal articles. Four Australian guideline publications were included due to relevance to topic and general practitioners. Results: The aetiology, clinical significance and molecular pathogenesis of hepatitis C virus were examined to provide Australian practitioners with a basis of knowledge for presentation of both acute and chronic stages of hepatitis C infection. This understanding was further linked to current drug treatments available in Australia and potential future therapeutic options. Conclusion: The consequences of Hepatitis C infections will burden the Australian healthcare system in the next few decades as the chronic nature of HCV infection leads to complications of liver failure, cirrhosis and hepatocellular carcinoma in many patients. Practitioners must equip themselves with knowledge of HCV pathogenesis which forms the basis of current and future treatments in order to provide best quality care at all levels of prevention and management.

Introduction

The recognition of viral hepatitis can be dated as far back as the fifth century BC to Babylonian records. [1] Our understanding of Hepatitis C gained remarkable ground when previously non-A non-B hepatitis infections were attributed to the hepatitis C virus discovered by Choo et al. in 1989. [1,2] Since then efforts have been made to develop drug treatments to combat the virus which progresses to chronic infection in up to 80% of patients, increasing their risk of cirrhosis, liver failure and hepatocellular carcinoma. [3,4,5] Chronic hepatitis C infection is currently the leading cause of liver transplantation in Australia. [6,7,8]

Hepatitis C is a major health concern for Australian practitioners with 260 000 Australians infected in 2010 and an estimated 12 000 new infections occurring annually. [4,7] The dominant mode of transmission of the hepatitis C virus (HCV) is parenteral exposure to infected blood and thus the epidemic of HCV infection in Australia continues to escalate predominantly through people who inject drugs (PWID). [7]

This review aims to summarise the aetiology, transmission and life cycle of HCV as well as examine the most recent literature regarding current and future drug therapies to provide the Australian general practitioner with a contemporary understanding in emerging hepatitis treatments.

Aetiology of Hepatitis C

Transmission in the Australian context

Hepatitis C is a blood-borne viral infection and is most commonly spread in Australia via shared injecting equipment (up to 80% acquiring the infection via this route). [7] Other means of transmission include unsterile tattooing, needle-stick injuries and vertical transmission from mother-to-infants from trauma during pregnancy and/or birth. [9,10] About 5% of all cases in Australia arise from HCV contaminated blood transfusions and blood products prior to screening introduced in February 1990. [7]

Virological Structure

Hepatitis C is caused by a small, positive-stranded RNA virus of the Flaviviridae family. The RNA strand is enveloped by a protein capsid which is further surrounded by a lipid bilayer envelope studded with E1 and E2 heterodimer proteins. The genome contains a 5’ noncoding region required for viral translation, followed by an open reading frame terminated by a 3’ noncoding region necessary for replication. The open reading frame translates into a 3000 amino acid polyprotein which is cleaved into structural (core, E1, E2) and non-structural (p7, NS3, NS4A, NS4B, NS5A, and NS5B) proteins. [11-15]

The NS5B protein is a RNA-dependent RNA-polymerase which lacks proofreading function. This combined with a high replication rate (1012 virions/day) results in rapid mutations driving genetic diversity. [16] Thus within a host, HCV circulates as a population of extremely closely related, but not identical variants called quasispecies. [17] This feature has contributed to difficulty in developing a vaccine as well as implications for pharmacological therapies. HCV is classified into seven major genotypes which differ genetically by at least 30% with over 100 subtypes. [11,13] The prevalence of genotypes differ with geographical distribution. Genotype 1 mostly dominates Australia, the Americas, Japan and Europe with genotypes 2 and 3 also prevalent in these areas. Genotype 7 was only recently discovered in a small proportion of people in Central Africa. Disease association is largely similar across genotypes, however genotype 3 has been correlated with a higher risk of hepatic steatosis and progressive liver disease. [13,18]

Life Cycle

The main stages of the HCV replication cycle are binding and entry, uncoating, translation and replication of RNA, assembly into new particles, maturation and secretion. [11,19] Several host factors have been identified aiding entry of HCV including heparan sulphate and low-density lipoprotein receptor. Other host factors CD81, scavenger receptor B1 and tight junction proteins claudin-1 and occludin allow for clathrin-dependent endocytosis which delivers the virus to early endosomes, which become acidified causing fusion of the viral envelope, uncoating and release of the viral RNA into the cytoplasm. [12,19] HCV replication induces a membranous web concentrating lipid-rich structures that aid the replication process. Hepatocyte -specific microRNA-122 has been shown to bind to target sites on the 5’ untranslated region of the HCV genome which forms a complex that protects the HCV genome from nucleolytic degeneration and innate host immune responses. [20] Lipid droplets interact with the core and NS5A viral proteins allowing viral assembly. The newly synthesised viral proteins can then exit the cell in a manner similar to the hosts’ very low-density lipoprotein (VLDL) export pathway by utilising cofactor ApoB and microsomal triglyceride transfer protein to form low-density viral particles termed lipo-viral particles. [11,12,18] ApoE is involved in HCV particle morphogenesis and infectivity. HCV particles exist in the serum as a mixture of complete low-density infectious lipo-viral particles and an excess of apoB-associated empty non-infectious particles complexed with anti-HCV envelope antibodies. [18]

The mechanism responsible for onset and progression of chronic hepatitis are not fully understood but it is currently believed that HCV establishes persistent infection by impairing host innate and adaptive immunity. [1,21] The infected hepatocytes recognise Pathogen Associated Molecular Patterns (PAMPs) through receptors known as Pattern Recognition Receptors (PRRs) which include Toll like receptors (TLRs) and RIG-1 like receptors (RLRs). Upon sensing HCV via TLR3 and RIG-1, intracellular signalling cascades result in the induction of type I and type III interferon and pro-inflammatory cytokines which establish an antiviral state in infected and neighbouring cells. [21,22] Resident antigen presenting cells, such as dendritic cells residing in the liver migrate from infected tissue to lymph nodes where they prime T and B cell activation to induce adaptive immunity. [11]

Recent studies have established multiple routes whereby HCV impairs immune functioning so as to coexist and replicate in the host. [23] NS3/4A protease cleaves intracellular pathway protein TRIF and CARDIF to impair TLR3 and RIG-1 receptors. [11] Furthermore, the HCV enveloped particle is not detected by TLR-2 and TLR-4 which also contribute to antiviral states. HCV has been shown to up regulate MHC Class I molecules on infected hepatocytes which suppresses Natural Killer cell activity. [23] Finally generation of quasispecies carry mutations to evade B and T-cell recognition as well induce hypermutation in B cell receptors to lower affinity allowing the virus to escape immune surveillance. [11]

Clinical Implications

The World Health Organisation estimates 170 million people are infected with Hepatitis C globally. [1,24] Hepatitis C is thus  the leading cause of chronic liver disease worldwide and is a growing burden on healthcare systems,  including within Australia. [3,25] Infection is characterised by a wide range of clinical manifestations and propensity to develop into chronicity. Up to 80% of infected patients will develop a chronic infection. [1,3] Persistent infection and chronic hepatitis are the hallmarks of HCV infection with severity varying widely from asymptomatic chronic infection with normal liver function tests to severe cases leading to cirrhosis and hepatocellular carcinoma. [2,14]

Acute Hepatitis

Acute HCV infection is often asymptomatic due to a mild immune response and reversible cellular injury seen at the microscopic level. [9] Where symptoms occur they tend to be minimal involving jaundice and flu-like malaise. [5] Strong immune responses during the acute infection are associated with clearance of the virus however in the majority of cases milder initial infections lead to chronic viral persistence. [5,26]

Chronic Hepatitis

Cirrhosis develops in as many as 20% of chronic HCV patients and is associated with hepatocellular failure. Patients may present with portal hypertension manifested as splenomegaly, variceal bleeding or ascites. [9,26] Primary hepatocellular carcinoma is thought to result from the continual division of infected hepatocytes attempting to regenerate in the presence of injury. [5] Once cirrhosis has established, patients have a 5% annual risk of developing hepatocellular carcinoma. [27] Extra-hepatic diseases such as mixed cryoglobulinemia and glomerulonephritis are also believed to be caused by HCV induced antibody complex depositions in small vessels causing vasculitis, however the pathogenesis of this is not fully understood providing an area for investigation in the future. [28,29]

Current drug therapies

Current standard of care treatment of HCV genotype 1 is triple therapy with pegylated interferon-α (cytokine), ribavirin (antiviral) and a direct acting antiviral (NS3/4A protease inhibitor) – either telaprevir or boceprevir. [2,8,30] The combination of pegylated interferon (PEG-IFN) and ribavirin remain the recommended treatment for HCV infection with genotypes 2, 3, 4, 5 and 6. [19] The aim of treatment is a sustained virological response (SVR), defined as the absence of detectable HCV RNA for 6 months after treatment cessation. [31] SVR is associated with crucial end points, particularly survival and protection from the complications of chronic hepatitis C such as cirrhosis and hepatocellular carcinoma. [19,30]

For reasons that remain elusive, interferon-based therapies result in a SVR of 80% in genotype 2 and 3 infections but only 45% in genotype 1 and 4 infections. [4,13] With the approval of boceprevir and telaprevir in 2011 by the US Food and Drug Administration, triple therapy has enabled the SVR to increase in patients with genotype 1 from 45% in 2010 to ~66% in 2011 and is expected to be >75% by 2014. [4,26]

The SVR is also influenced by a myriad of host factors such as ethnicity, gender, age and insulin resistance. [13,32] Furthermore, new biomarkers such as serum IP10 levels and genetic tests to determine polymorphisms in the gene encoding IFNL3 (formerly known as IL28B or IFN-λ3) and recently discovered IFNL4 show strong value with respect to interferon-based therapy as predictors of treatment outcome. [1,13,30]

In Australia, hepatitis C treatment is available for all eligible patients over 18 years of age who have chronic HCV infection with compensated liver disease and are using effective forms of contraception. Treatment is subsidised by the government under the Highly Specialised Drugs (HSD) program, section 100 (S100) of the National Health Act 1953 (Cwlth). [8]

Pegylated Interferon-α

Interferons are naturally produced by immunological cells in response to tumour or infectious organism. They are glycoproteins with antiviral, anti-proliferative and immunomodulatory functions. [31]

Upon administration of IFN- α, the type I interferon binds to IFNAR-1 and IFNAR-2 receptors on cell surfaces initiating a complex intracellular signalling pathway resulting in activation of genes coding for proteins which inhibit intracellular viral replication. Proteins include RNA-dependant protein kinase (PKR) which inhibits RNA translation and oligoadenylate synthetase (OAS) which mediates RNA degradation. IFN- α also stimulates TH1 cell production while reducing suppressor TH2 cells as part of its immunomodulation. [31,33]

Pathogenesis of HCV occurs as a result of the virus’ ability to prevent host cells from responding to natural levels of interferon. As previously discussed, HCV blocks TLR3 and RIG-1 receptors reducing type I IFN production. [5,11] Thus overwhelming host cells with high levels of injected IFN allow normal cellular mechanisms to control the virus. [5,13] Replacement of standard interferon with pegylated interferon (interferon-α conjugated to polyethylene glycol) improves pharmacokinetics and efficacy and has allowed its administration as a once weekly subcutaneous injection. [30]

Ribavirin

Unlike pegylated interferon-α, whose function was unravelled due to developments in cell culture models, the mechanism of action of ribavirin against HCV is unknown. [33] Ribavirin was originally synthesised as a guanosine analogue that could inhibit viral polymerases by chain termination. The process by which this is thought to occur is when the polymerase incorporates the nucleotide but cannot add more after inserting the analogue, hence preventing viral replication and transcription. [5]

However there is much debate about the mechanism of ribavirin activity in chronic hepatitis C.  Despite showing in vitro activity against some RNA and DNA molecules, studies conducted with ribavirin as a monotherapy against HCV reflect no effect on HCV RNA levels or improvement of hepatic histology following 12 months of therapy. [33] Yet analysis of the current literature shows multiple studies where combination therapy of IFN- α and ribavirin is significantly more effective then IFN- α alone. [34-36] Furthermore, the anti-HCV activity of ribavirin occurs at much lower doses then expected for the chain termination theory to occur. [5] This suggests other mechanisms of action are at work.

Greenblatt presents two possibilities. [5] One involves ribavirin as depleting the cell’s reservoir of normal guanosine to interfere with viral RNA synthesis. Secondly she proposes a mutagenic theory in which ribavirin incorporation into viral genomes renders them funtionless. [5] Other theories propose that ribavirin induces IFN-stimulated genes or may have immunomodulatory functions which like IFN- α, push patient cytokine profiles towards TH1 types which are more effective against viral infections then type 2 Helper T cells. [13,31]

Telaprevir and Boceprevir

Telaprevir and boceprevir are first generation peptidomimetic, reversible inhibitors of NS3/4A protease. [30] The HCV NS3/4A serine protease is essential for viral replication by cleaving polyproteins into mature non-structural proteins. [13] Thus by inhibiting this protease, telaprevir and boceprevir are the first direct-acting antivirals (DAAs) approved for use against HCV genotype 1.

Despite both drugs having similar mechanisms of action and thus sharing most clinically relevant strengths and weaknesses, there are discrepancies between telaprevir-based regimens and boceprevir-based regimens. [1,30] These differences are in the timing and duration of combined therapy. Typically, telaprevir is given in triple therapy with PEG-IFN and ribavirin for the first 12 weeks of therapy, PEG-IFN and ribavirin are then continued for the remainder of treatment (either 24 or 48 weeks) without the protease inhibitor. Duration of treatment is dependent on virological response (response-guided therapy). Boceprevir, however is started 4 weeks after commencement with PEG-IFN and ribavirin and is continued for the remaining treatment duration of 28 or 48 weeks depending on response. [19] Telaprevir-based regimen is stopped in patients with a HCV RNA level greater than 1000 IU/ml at week 4 or 12 and all three drugs should be discontinued. For the boceprevir-based regimen, patients with HCV RNA levels greater than 100 IU/ml at week 12 should discontinue treatment. For both treatments, if HCV RNA is detectable at 24 weeks of therapy, all three drugs should be stopped. [19]

Side effects profile – pegylated interferon-α and ribavirin

These can be quite distressing and contribute to low tolerance and compliance in patients. The major effects of interferon include depression, constant flu-like symptoms, thrombocytopenia, leucopenia, thyroid dysfunction, retinopathy and alopecia. [25,37] Ribavirin is highly teratogenic and can lead to haemolytic anaemia and autoimmune disorders. [37]

Psychiatric status as well as full blood count, kidney and liver function tests should be monitored continuously throughout therapy. Furthermore, precautions should be taken with patients with depressive histories, thyroid dysfunctions, diabetes, autoimmune disorders and renal impairment. [38] Finally pregnancy in female patients or the partners of male patients must be avoided during treatment, and owing to the long half life of ribavirin, also 6 months after cessation of treatment. [39]

Side effects profile – telaprevir and boceprevir

Although triple therapy is more efficacious in HCV genotype 1 infections, there are additional side effects compared to traditional dual therapy and thus management of hepatitis C patients has become more complex. Common side effects of telaprevir include rash and anorectal discomfort while dysgeusia (altered taste sensation) and neutropaenia are associated with boceprevir. The most challenging side effect of both drugs is marked anaemia (haemoglobin level < 10 g per decilitre) occurring in 36-50% of patients. [19,30] Erythrocyte-stimulating agents have some success in managing this complication however are not approved for routine use in chronic hepatitis C patients due to serious side effects and cost. Some studies have shown that reduction in the dose of ribavirin can effectively manage anaemia in this setting and this is the current recommended first line approach. [19]

Due to the highly variable nature of HCV with the error-prone RNA polymerase, drug resistance is also an issue with these protease inhibitors and can develop as early as day 4 upon use in monotherapy. Consequently, these drugs are not to be used in isolation. Because of the similar mechanism of action, resistance to one protease inhibitor can result in other drugs within the same class to be ineffective.  Once the drug is stopped, the frequency of resistance-associated variants within the quasispecies slowly decreases until they disappear, most likely because they do not replicate as effectively as the wild-type virus. [19,30] General practitioners can play a crucial role in patient education to ensure adherence to the prescribed regimen in order to limit the development of resistance-associated variants.

The third major consideration with these new drugs is the issue of drug-drug interactions. Both telaprevir and boceprevir are inhibitors of the cytochrome P450 3A (CYP3A). CYP3A enzymes are involved in the metabolism of numerous drugs such as statins, antidepressants, antiarrhythmics, anticonvulsants, analgesics and sedatives. [19] As such, these are all contraindicated in patients undergoing treatment with telaprevir and boceprevir. This has important implications for general practitioners who are frontline prescribers of such agents. Efforts are being made to make such complex information widely available to the medical community through platforms such as the ‘Hepatitis Drug Interactions’ website from the University of Liverpool, UK. [30,40]

The future of hepatitis C

With the exponential increase in knowledge of life cycle and replication of HCV due to breakthroughs in cell culture systems in 2005, there is fierce competition to develop medicines that will replace PEG-IFN, ribavirin and first generation protease inhibitors. [30] About two-thirds of agents in Phase II and III trials are directed against the NS3/4A and NS5B viral proteins called second generation protease inhibitors and polymerase inhibitors respectively. [19,30] The current challenges in drug development are decreasing side effects and drug interactions, exploring combinations for genotypes 2-6, exploring individualised drugs to specific genetic polymorphisms, and eradicating the need for interferon and ribavirin in treatment.

A number of drugs are currently being developed for genotypes 2-6. A preliminary phase 2a study in New Zealand involved combining sofosbuvir, an oral nucleotide inhibitor of HCV polymerase, and ribavirin in various interferon and interferon-sparing regimens for 12 weeks. [41] Patients with HCV genotype 1, 2 and 3 were investigated. In this early trial sofosbuvir showed a promising result with 100% rate of SVR among patients with genotype 2 or 3 infection. [19,26,41] However phase 3 studies of sofosbuvir fall short of the results produced by the phase 2a study. [42,43,44] One noninferiority trial looked at sofosbuvir plus ribavirin compared to standard peginterferon alfa-2a plus ribavirin in 499 patients with HCV genotype 2 or 3 infection. The results revealed the same SVR rate of 67% in both the sofosbuvir-ribavirin and peginterferon-ribavirin group at 12 weeks after cessation of therapy. [43] Two further phase 3 trials (POSITRON and FUSION studies) investigated sofosbuvir-ribavirin therapy in patients for whom peginterferon treatment was not an option (due to pre-existing psychiatric or autoimmune disorders) and in those who did not have a response to previous interferon treatment. The POSITRON trial was a randomized, blinded, placebo-controlled study that compared 12 weeks of sofosbuvir-ribavirin treatment with matching placebo in patients who had previously discontinued interferon therapy due to adverse events or concurrent medical condition. In this group, 78% had a SVR at 12 weeks after treatment compared to 0% in the placebo group. [42] The FUSION study looked at patients who had failed a sustained response to interferon-based therapy and compared 12 and 16 week regimens of sofosbuvir-ribavirin therapy. Results showed that four additional weeks of therapy made a difference with an increase in the SVR from 86% to 94% in patients with genotype 2 infection and from 30% to 63% in genotype 3 infections. [42,44] All three phase 3 studies with sofosbuvir-ribavirin showed better SVR rates in genotype 2 infections compared to genotype 3 infections. [42,43,44]

Nonstructural 5A (NS5A) protein is also a target of recent drug development. [45] The functions of the NS5A protein are not yet fully understood with in vitro studies suggesting is has a role in viral replication and assembly and release of infectious particles. [45] Daclatasvir is a potent NS5A inhibitor which has shown early promising results for use in interferon-free combinations with rapid decline of extracellular HCV titres upon administration. [45] In a phase 2a trial, patients who had not had a response to previous therapy received daclatasvir and a protease inhibitor (asunaprevir) for 24 weeks. [46] Four out of eleven patients had a SVR at 12 and 24 weeks after treatment ended, suggesting a cure may be possible with an all-oral interferon-ribavirin free treatment. [45,46]

Another group of host targeting antiviral agents are arising. Miravirsen is a drug undergoing development which targets miR-122. Liver specific miR-122, as discussed previously is a microRNA which all strains of HCV use to survive and replicate in liver cells. [11,12] A recent phase 2a study by Janssen et al, dose-dependent reductions in HCV RNA levels were found without viral resistance. [20] The study was limited by a small sample of 36 patients and only moderate levels HCV RNA reduction which rebounded once miravirsen was stopped in patients who had not begun interferon and ribavirin. [20,47] Normally miR-122 is involved in controlling cholesterol levels independent of its effect on HCV. In the study, there was a sustained decrease of serum cholesterol levels by ~25% which lasted 14 weeks after the final injection. [47] Given statins are contraindicated in the current triple therapy treatment of HCV genotype 1 infections, there is potential in the future to develop liver-targeting nucleic acid drugs which can be used intermittently for both HCV treatment and other co-morbid conditions.

Furthermore, the future of HCV treatment is trending towards highly individualised regimens which consider not only the viral genotypes but also the patient’s genetic polymorphisms. For instance, easy-to-treat patients have been identified as treatment-naïve, IL28B CC. [30] Patients with a favourable interlukin-28B genotype (CC variant as opposed to CT or TT) have shown sustained virologic responses up to 80%. [19] It is theorised that these patients could receive PEG-IFN and ribavirin first, minimising the adverse effects of triple therapy. [19] Although testing for the IL28B genotype is not currently the approved standard of care, in the future Australian general practitioners may be managing care of these ‘easy-to-treat’ patients while more complex cases, such as patients with IL28B TT with decompensated cirrhosis are managed at tertiary centres using a cocktail of tailor made drug regimens. [30]

Implications for Australian health practitioners

Accessing and treating hepatitis C infection in PWID – the role of the general practitioner

Advances made in the development of better tolerated interferon free HCV treatment will remain negligible as long as access to therapy cannot be expanded to the most affected and underserved risk groups. [48,49,50] People who inject drugs act as a virus reservoir, as the burden of HCV-related liver disease in this group is increasing but treatment uptake remains low. [49,50] There are a number of barriers to accessing care at the level of the patient, practitioner and system. [48,50,51] New guidelines have been published with recommendations for the management of HCV infection among PWID which aim to overcome these barriers by providing evidence-based treatment recommendations.[50] Analysis of the literature revealed a common theme supported by high quality evidence which was the use of multidisciplinary care teams in enhancing treatment uptake in PWID. [49,50] General practitioners can play a crucial role in co-ordinating multidisciplinary care between specialists, drug and alcohol support services, psychiatric services, social work and other social supports such as peer-based groups. [49] In an Australian community-based study, hepatitis C positive patients who had seen a general practitioner about HCV in the last 6 months were four times more likely to be assessed for therapy by a specialist. [50] Furthermore, a prospective cohort study using telehealth technology in supporting and training GPs was compared to HCV treatment provided at a tertiary centre. Similar rates of treatment success were achieved in both groups. [49,51] From these studies, it was seen that general practitioners not only co-ordinated care but provided a more patient-centred approach necessary in dealing with the complex psychiatric and substance abuse co-morbidities which required individualised models of care. Enhanced personal contact provides an ideal environment for pre-therapeutic assessment of housing, education, cultural and social issues, supports, finances, nutrition, drug and alcohol use and psychiatric evaluation. [50] Merging different disciplines into one general practice model may be a simple and effective model in the future for a sub-population of PWID with HCV but will require commitment by motivated and supported GPs who have undergone further training in addiction and HCV medicine. [49]

The role of general practitioners in assessment and management of HCV infection

As stated previously, hepatitis C may be present in patients unknowingly for decades before symptoms of liver failure prompt a seeking of treatment. At this stage of irreversible cellular damage, treatment options are limited, often associated with distressing side effects and yield less efficient results in certain genotypes. The onus is on health practitioners, armed with an understanding of the pathogenesis of HCV, to identify high risk patients and test for anti-HCV antibodies and HCV RNA levels as a secondary preventative strategy to provide early detection and referral. There are a number of useful international guidelines which can assist general practitioners in managing newly diagnosed hepatitis C patients in regards to indications for treatment as well as first-line treatment recommendations. [3,24] Furthermore, primary prevention strategies whereby educating the public about transmission, symptoms and progression of the disease can be effectively implemented in a consultation setting.

In regards to treating chronic hepatitis C patients, this review aims to equip the general practitioner with an up-to-date understanding of the molecular and immunological aspects of HCV pathogenesis to aid in diagnostic tools as well as provide a platform of knowledge for future pharmaceuticals. We are entering an exciting new era of hepatitis C treatment where interferon-free therapies are likely to dominate the therapeutic landscape within the next 5 years [19] and so an understanding of their mechanism of action in hepatitis C is crucial for continuing treatment and management.

Conclusion

From the ancient observations to the discovery of hepatitis C virus 24 years ago, up until recent advances in cell model systems, our understanding of the pathological nature of hepatitis C has grown exponentially. With this growth have come parallel developments in treatment, both in understanding mechanisms behind current drug therapies but also providing a platform for future pharmaceuticals to target aspects of HCV pathogenicity. These developments come at a timely point, as the burden of an escalating epidemic of hepatitis C in Australia will have major impacts on our healthcare system within the next few decades as the chronic nature of the disease will come into play.

Conflict of interest

None declared.

Correspondence

J Aslanidis: jaimie.aslanidis@gmail.com

References

[1] Boyer N, Marcellin P. Pathogenesis, diagnosis and management of hepatitis C.  J Hepatol. 2000;32(1 Suppl):98-112.

[2] Liang TJ. Current progress in development of hepatitis C virus vaccines. Nat Med. 2013;19(7):896-78.

[3] EASL Clinical Practice Guidelines: Management of hepatitis C virus infection. J Hepatol. 2011;55:245-64.

[4] Boonwaat L, Haber PS, Levy MH, Lloyd AR. Establishment of a successful assessment and treatment service for Australian prison inmates with chronic hepatitis C. Med J Aust. 2010;192(9):496-500.

[5] Greenblatt R. Hepatitis C: Progress and Challenges. Clinical Microbiology Newsletter. 2006;28(15):113-118.

[6] Hepatitis Australia. Hepatitis C burden set to explode in decades to come if government doesn’t act [Internet]. 2010 [updated 2010 April 1; cited 2011 April 4]. Available at: http://www.hepatitisaustralia.com/media-centre/media-releases/media-releases-items/hepatitis-c-burden-set-to-explode-in-decades-to-come-if-government-doesnt-act

[7] Dore GJ, Law M, MacDonald M, Kaldor JM. Epidemiology of hepatitis C virus infection in Australia. J Clin Virol. 2003;26(2):170-184.

[8] Gidding HF, Law MG, Amin J, Macdonald GA, Sasaduesz JJ, Jones TL et al. Predictors of deferral treatment for hepatitis C infection in Australian clinics. Med J Aust. 2011;194(8):398-402.

[9] Bisceglie A. Hepatitis C. Lancet. 1998;351(9099):351-5.

[10] Sompayrac L. How Pathogenic Viruses Work. London: Jones and Barlett Publishers; 2002. 56-59.

[11] Georgel P, Schuster C, Zeisel MB, Stoll-Keller F, Berg T, Bahram S et al. Virus-host interactions in hepatitis C virus infection: implications for molecular pathogenesis and antiviral strategies. Trends Mol Med. 2010;16(6):277-86.

[12] Fischer R, Baumert T, Blum HE. Hepatitis C virus infection and apoptosis. World J Gastroenterol. 2007;13(36):4865-72.

[13] Scheel TK, Rice CM. Understanding the hepatitis C virus life cycle paves the way for highly effective therapies. Nat Med. 2013;19(7):837-49.

[14] Pawlotsky JM. Pathophysiology of hepatitis C virus infection and related liver disease. Trends Microbiol. 2004;12(2):96-102.

[15] Kumar V, Abbas AK, Aster JC, Fausto N. Robbins and Cotran Pathological Basis of Disease. 8th ed. Philadelphia: Saunders Elsevier; 2010. p. 847-53.

[16] Wyles DL. Moving beyond interferon alfa: Investigational drugs for hepatitis C virus infection. Top HIV Med. 2010;18(4):132-6.

[17] Fishman SL, Branch AD. The quasispecies nature and biological implications of the hepatitis C virus. Infect Genet Evol. 2009;9(6):1158-67.

[18] Bassendine MF, Sheridan DA, Bridge SH, Felmlee DJ, Neely RDG. Lipids and HCV. Semin Immunopathol. 2013;35:87-100.

[19] Liang TJ, Ghany MG. Current and future therapies for hepatitis C virus infection. N Engl J Med. 2013;368(20):1907-17.

[20] Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Keyur P et al. Treatment of HCV infection by targeting microRNA. New Engl J Med. 2013;368(18):1685-95.

[21] Saito T, Gale MJ. RIG-1 mediated hepatic innate immune signalling that controls HCV infection. Uirusu. 2008;58(2):105-15.

[22] Barth H. Insights into the role of interferon lambda in hepatitis C virus infection. J Hepatol. 2011;54:844-847.

[23] Rehermann B. Hepatitis C virus versus innate and adaptive immune responses: a tale of coevolution and coexistence. J Clin Invest. 2009;119(7):1745-54.

[24] Ghany MG, Strader DB, Thomas DL, Seeff LB. American Association for the Study of Liver Diseases: Diagnosis, management, and treatment of hepatitis C: an update. Hepatology. 2009;49:1335-74.

[25] Suzuki T. A Hepatitis C virus-host interaction involved in viral replication: toward the identification of antiviral targets. Jpn J Infect Dis. 2010;63(5):307-11.

[26] Thomas DL. Global control of hepatitis C: where challenge meets opportunity. Nat Med. 2013;19(7):850-8.

[27] Bode JG, Brenndorfer ED, Haussinger D. Subversion of innate host antiviral strategies by the hepatitis C virus. Arch Biochem Biophys. 2007;462(2):254-65.

[28] Eisen-Vandervelde AL, Yao ZQ, Hahn YS. The molecular basis of HCV-mediated immune dysregulation. Clin Immunol. 2004;111(1):16-21.

[29] Craxi A, Laffi G, Zignego AL. Hepatitis C virus (HCV) infection: a systemic disease. Mol Aspects Med. 2008;29(1-2):85-95.

[30] Manns MP, von Hahn T. Novel therapies for hepatitis C – one pill fits all? Nat Rev Drug Discov. Forthcoming 2013 June.

[31] Vezali E, Aghemo A, Colombo M. Interferon in the treatment of chronic hepatitis C: a drug caught between past and future. Expert Opin. Biol. Ther. 2011;11(3):301-13.

[32] Cua IHY, Hui JM, Kench JG, George J. Genotype-specific interactions of insulin resistance, steatosis, and fibrosis in chronic hepatitis C. Hepatology. 2008;48:723–731.

[33] Clark V, Nelson DR. The role of ribavirin in direct acting antiviral drug regimens for chronic hepatitis C. Liver Int. 2012 Feb;32(Suppl 1):103-7.

[34] Berg T, Hoffmann RM, Teuber G, Leifeld L, Lafrenz M, Baumgarten R et al. Efficacy of a short-term ribavirin plus interferon alpha combination therapy followed by interferon alpha alone in previously untreated patients with chronic hepatitis C: a randomised multicenter trial. Liver. 2000;20(6):427-36.

[35] Tsubota A, Fujise K, Namiki Y, Tada N. Peginterferon and ribavirin treatment for hepatitis C virus infection. World J Gastroenterol. 2011;17(4):419-32.

[36] Foster GR. Pegylated interferons for the treatment of chronic hepatitis C: pharmacological and clinical differences between peginterferon-alpha-2a and peginterferon-alpha-2b. Drugs. 2010;70(2):147-65.

[37] Murtagh J. Murtagh’s general practice companion handbook. 4th ed. Sydney. McGraw-Hill Australia. 2008. p. 262-3.

[38] MIMS Australia. St Leonards NSW: UBM Medica Pty. Ltd: 2009 Dec. Chap. Immunomodifiers.

[39] Chapter 5, Anti-infectives. In: Australian Medicines Handbook [online]. 2011. [cited 2011 April 11]. Available from: http://amh.hcn.net.au.elibrary.jcu.edu.au/view.php?page=chapter5/monographribavirin-interferon.html#ribavirin-interferon

[40] University of Liverpool Drug Interaction Charts [Internet] 2013 [updated 2013 April 17; cited 2013 July 14]. Available from: http://www.hep-druginteractions.org

[41] Gane EJ, Stedman CA, Hyland RH, Ding X, Svarovskaia E, Symonds WT et al. Nucelotide polymerase inhibitor sofosbuvir plus ribavirin for hepatitis C. New Engl J Med. 2013;368(1):34-44.

[42] Jacobson IM, Gordon SC, Kowdley KV, Yoshida EM, Rodriguez-Torres M, Sulkowski MS, et al. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. N Engl J Med. 2013;368:1867–1877.

[43] Lawitz E, Mangia A, Wyles D, Rodriguez-Torres M, Hassanein T, Gordon SC, et al. Sofosbuvir for previously untreated chronic hepatitis C infection. N Engl J Med. 2013;368:1878–1887.

[44] Drenth JPH. Editorial: HCV treatment–no more room for interferonologists? N Engl J Med. 2013;368:1931–1932.

[45] Guedj J, Dahari H, Rong L, Sansone ND, Nettles RE, Cotler SJ et al. Modeling shows that the NS5A inhibitor daclatasvir has two modes of action and yields a shorter estimate of the hepatitis C virus half-life. Proc Natl Acad Sci. 2013;110(10):3991-6.

[46] Lok AS, Gardiner DF, Lawitz E, Martorell C, Everson GT, Ghalib R et al. Preliminary study of two antiviral agents for hepatitis C genotype 1. N Engl J Med. 2012;366:216-224.

[47] Lieberman J, Sarnow P. Micromanaging hepatitis C virus. New Engl J Med. 2013;368(18):1741-3.

[48] Bruggmann P. Accessing hepatitis C patients who are difficult to reach: it is time to overcome barriers. J Viral Hepat. 2012;19(12):829-35.

[49] Bruggmann P, Litwin AH. Models of care for the management of hepatitis C virus among people who inject drugs: one size does not fit all. Clin Infect Dis. 2013;57(Suppl 2):S56-61.

[50] Robaeys G, Grebely J, Mauss S, Bruggmann P, Moussalli J, de Gottardi A et al. Recommendations for the management of hepatitis C virus infection among people who inject drugs. Clin Infect Dis. 2013;57(Suppl 2):S129-137.

[51] Grebely J, Dore GJ. An expanding role for primary care providers in the treatment of hepatitis C virus in the community. Hepatology. 2011;54(6):2258-2260.

[52] Batey R. Managing hepatitis C in the community. Aust Prescr. 2006;29(2):36-9.

Categories
Review Articles Articles

Is plasmapheresis the optimal treatment option for acute pancreatitis secondary to hypertriglyceridemia? A systematic review

Background: Hypertriglyceridemia is an uncommon cause of acute pancreatitis, which is a life-threatening illness. Conventional management involves fasting, lipid-lowering medication, insulin and heparin. Plasmapheresis is an approach which is used occasionally to achieve rapid lowering of triglyceride levels in patients where conventional management is unsuccessful. It is currently unclear whether plasmapheresis improves outcome in patients with hypertriglyceridaemia-induced pancreatitis. Aim: A literature review and critical analysis was conducted to assess the effectiveness of plasmapheresis in improving patient outcomes in patients with acute pancreatitis secondary to hypertriglyceridemia Methods: The PICO model (Population, Intervention, Comparator, Outcomes) was used to synthesise a research question. Thereafter, a search was conducted through the Scopus database (includes complete MEDLINE coverage) applying the terms ‘plasmapheresis’ OR ‘plasma exchange’ OR ‘lipid apheresis’ AND ‘pancreatitis’ AND ‘hypertriglyceridemia’ OR ‘hyperlipidaemia’ OR ‘hyperlipidemia’. Article titles and/or abstracts were screened for relevance to the topic. Original research articles assessing the efficacy of plasmapheresis in hypertriglyceridaemia-induced pancreatitis were included. Results: To date, no randomised controlled trials have been published assessing the efficacy of plasmapheresis in this population. Two retrospective primary research studies were identified. Both studies demonstrated a rapid reduction in triglyceride levels following plasmapheresis in the magnitude of 65.8-80%. The studies showed no significant clinical benefit in terms of mortality and morbidity, but were limited by small sample size and study design. Conclusion: Current evidence demonstrates that plasmapheresis in the setting of hypertriglyceridemia-induced pancreatitis reduces triglyceride levels by 46-80%. [1] However there is insufficient data to suggest a beneficial effect on clinical outcomes. Well-designed prospective studies with adequate follow-up are required to elucidate whether plasmapheresis is associated with reduced morbidity and mortality in this population.

Introduction

Hypertriglyceridemia is an uncommon cause of acute pancreatitis, accounting for 1.3-3.8 % of cases with an incidence of 18/100,000 per year in the United States of America. [1,2] Primary (genetic) and secondary causes, such as uncontrolled diabetes mellitus, hypothyroidism, alcohol, obesity, certain medications, and pregnancy, are associated with hypertriglyceridemia-induced pancreatitis. [1,3] The mechanism for severe hypertriglyceridemia-inducing pancreatitis remains unclear, [3] although triglyceride levels exceeding 10 mmol/l (1000 mg/dl) can trigger a bout of pancreatitis. [3,4] One postulated theory involves the idea that pancreatic lipase hydrolyses excess triglycerides to produce free fatty acids around the pancreas. These free fatty acids can damage the pancreatic acinar cells and pancreatic vascular endothelium, resulting in ischaemia and inflammation. The acidic environment can further amplify the free fatty acid toxicity in a vicious cycle. [3,4]

Hypertriglyceridemic pancreatitis is a life-threatening illness with a mortality rate of 7-30%. [5] Complications include sepsis, pancreatic necrosis, abscess formation and renal insufficiency; which account for the high mortality seen in this disease. [3] Optimal management of hypertriglyceridemia-induced pancreatitis is essential to reduce morbidity and mortality. Current management includes fasting, lipid-lowering medication (such as fenofibrate), and insulin and heparin, used to ‘accelerate lipoprotein lipase activity’. [2,4] These interventions have shown limited efficacy in reducing inflammation and life-threatening complications associated with severe acute pancreatitis. Novel therapies are needed to improve patient outcomes. [5]

Plasmapheresis is defined as ‘removing the plasma and replacing it with donor plasma or a plasma substitute’. [6] The term plasmapheresis is used interchangeably with the term ‘therapeutic plasma exchange’. The use of plasmapheresis in patients with hypertriglyceridemia can be traced back to a case report in 1978. [3] However, it is not widely utilised in this patient population at present. Given that plasmapheresis provides rapid removal of the triglycerides responsible for underlying inflammation in hypertriglyceridemia-induced pancreatitis, it is expected that this intervention may prove highly effective in reversing this sub-type of acute pancreatitis. The aim of this review was to assess the effectiveness of plasmapheresis in achieving positive patient outcomes (as per Table 1) in patients with acute pancreatitis secondary to hypertriglyceridemia.

Methods

The PICO model was used to synthesise the research question. [7]

Search Methodology

The literature search was conducted on Scopus, which is one of the largest databases of abstracts and citations of research literature, and includes complete coverage of the MEDLINE database. [8] The following search terms were used: (“plasmapheresis” OR “plasma exchange”) AND “pancreatitis” AND (“hypertriglyceridemia” OR “hyperlipidaemia” OR “hyperlipidemia”). The initial database search yielded a total of 139 documents, of which 110 were written in English. After further limiting the search query to include only ‘articles’ or ‘reviews’, a total of 86 documents were found. The documents were then sorted by relevance, and titles and abstracts were screened to identify articles that reported on the use of plasmapheresis in the management of hypertriglyceridemia-induced acute pancreatitis. A total of 28 relevant articles were identified: one primary research study, one review paper, one guideline (the 2010 American Society for Apheresis (ASFA) guideline), eleven case-series (62 patients), and fourteen individual case studies (Figure 1). Review of the references and citations of these studies yielded an additional two articles: one original retrospective study and one review. Focused searches revealed no additional relevant articles. The inclusion criterion for this review was limited to primary research studies only.

Results

Two cohort studies meeting the pre-specified inclusion and exclusion criteria were included. No randomised-controlled trials were identified, and are probably not feasible given the low incidence of hypertriglyceridemia-induced pancreatitis.

Chen et al. [9] conducted a retrospective cohort study to compare the mortality and morbidity in patients with hyperlipidaemic pancreatitis before and after the introduction of plasmapheresis in Shin kong Wu-Ho-Su Memorial Hospital, Taiwan, in August 1999. This study separated the patients into two cohorts: group I (pre-August 1999) and group II (post-August 1999). There were 34 patients in group I and 60 patients in group II, of which twenty patients received plasmapheresis. The cohort was recruited appropriately with all patients fitting the time-frame criteria (pre- or post-August 1999). There was no statistical difference between the demographics of group I and group II, including mean age, gender distribution, initial mean triglyceride level, diabetes mellitus, alcohol consumption, Ranson’s score ≥ 3 and Balthazar grade D or E. [9]

Furthermore, patients with severe hypertriglyceridemic pancreatitis (defined by Ranson’s score ≥ 3) were analysed separately, comparing Group A (those who received plasmapheresis) and Group B (those who did not receive plasmapheresis). There were ten patients in Group A and nineteen patients in Group B. Morbidity was defined in terms of systemic complications, including acute renal failure, upper gastrointestinal bleeding, shock and acute respiratory distress syndrome; and local complications, in particular abscess and pseudocyst formation.

The results were analysed using a t-test and chi-square test. There was no statistical significance (defined as p<0.05) between the mortality and complications of patients with severe pancreatitis who received plasmapheresis, compared to those who received conventional therapy. Similarly, there was no significant difference (defined as p<0.05) between the clinical outcomes of pre-August 1999 and post-August 1999 samples. Interestingly, Chen et al. found that the mean serum concentration of triglycerides and lipase were markedly reduced after plasmapheresis, with a 65.8% reduction of triglyceride levels and 88.8% reduction of lipase levels. [9]

Gubensek et al. [4] also carried out a retrospective cohort study. They looked at two sets of patients. The first sample consisted of 50 patients who were treated with plasmapheresis between 1992 and 2008 at a tertiary-care hospital (University Medical Center Ljubljana, Slovenia), and the triglyceride and total cholesterol levels before and after plasmapheresis were compared. The demographic characteristics of these patients revealed a gender bias, with 92% of the sample being male. The second set of patients included 40 patients treated between 2003 and 2008 with plasmapheresis. The Acute Physiology and Chronic Health Evaluation II (APACHE II) score was used as a prognostic tool, with a score of < 8 indicating mild pancreatitis and a score of ≥ 8 indicating severe pancreatitis. A comparison of mortality rates was made between these two groups. The mortality rate was 4% and 42% respectively, with statistically significant differences between the two groups (p<0.001).

The results of the first cohort in the study by Gubensek et al. [4] showed a statistically significant (defined as p<0.001) reduction in triglyceride and cholesterol levels after plasmapheresis within 24 hours. On average, a reduction in serum triglyceride levels of approximately 80% was achieved by plasmapheresis. The analysis of the second cohort showed a significantly higher mortality in patients who had an APACHE II score of ≥ 8 (42% vs. 4%). The overall mortality was 15%, which the authors acknowledged as ‘considerable’.

Discussion

The efficacy of plasmapheresis in hypertriglyceridemia-induced pancreatitis has been unclear. In an attempt to clarify this, we conducted a systematic review of published studies and guidelines investigating the benefits of this therapy. Studies by Chen et al. and Gubensek et al. reported on different populations; however both highlighted that a significant reduction in serum triglyceride levels can be achieved by plasmapheresis in patients with hypertriglyceridemic pancreatitis. Chen et al. [9] showed a reduction in triglyceride levels by 65.8%, and Gubensek et al. [4] demonstrated an average reduction of 80% in triglyceride levels. It should be highlighted that a rapid reduction in triglyceride and cholesterol levels does not necessarily imply a clinical benefit to the patient. The exact pathophysiology of hypertriglyceridemia-induced pancreatitis remains unclear. During a prolonged episode of acute pancreatitis cellular injury can occur which may remain irreversible regardless of lowering of triglyceride levels, although further pancreatic destruction and recurrent episodes may be reduced. [5,10-11] Admission triglyceride levels have not been associated with severity, complication rates or clinical course. [5] Therefore more research is required to look at the effect of the rapid lowering of triglyceride levels on patient outcomes.

The weaknesses of both studies are the small sample size and sub-optimal study design. Chen et al. only looked at a small number of patients from a single hospital, and no definitive conclusions could be reached based on the limited statistical power of the study. Moreover, Chen et al. [9] compared group I and group II outcomes, despite group II comprising of 40 patients who had not received plasmapheresis in addition to the twenty patients who had received plasmapheresis. Therefore, since group II comprised of a mixed sample of patients receiving plasmapheresis and those receiving conventional treatment, the outcomes do not give a true picture of the effect of plasmapheresis only. A statistical comparison between group I and group II only accentuates that there was no benefit in terms of mortality and morbidity between patients presenting with acute pancreatitis secondary to hypertriglyceridemia before August 1999 or after August 1999 at that specific hospital. Thus, any conclusion on the effectiveness of plasmapheresis at reducing patient morbidity and mortality using these statistical results is invalid.

On the other hand, Chen et al. [9] should be commended on their detailed explanation of the apheresis procedure performed, which was well controlled. However, replacement fluid was either fresh frozen plasma (N=8) or isovolumetric 5% albumin solution (N=12). The authors did not adjust their results for this potentially confounding variable. The mean serum concentration of triglycerides and lipase were markedly reduced after plasmapheresis, with a 65.8% reduction of triglyceride levels and 88.8% reduction of lipase levels. However, the time period in which these reductions occurred post-procedure is unclear, and no comparisons were made with the reduction in serum lipase and triglycerides in patients presenting pre-August 1999. The authors did not follow the patients over a considerable time period and were unable to assess recurrence of acute pancreatitis or mortality between patient samples.

The research study by Chen et al. [9] is essentially the only study comparing the use of plasmapheresis and conventional treatment in acute pancreatitis due to hypertriglyceridemia. This study found no differences in mortality and morbidity between conventional therapy and plasmapheresis, but the results need to be carefully evaluated, as mentioned above. A review of this study by the 2010 ASFA Guidelines highlight that ‘adequate information was not provided to ascertain the comparability of the two groups’. [1] Chen et al. stated that earlier intervention might provide positive outcomes for plasmapheresis, given that the median time for starting plasma exchange was three days for their patients. [9] This is a possible explanation for the results, and further studies are needed to evaluate the relationship between the time of initiating plasmapheresis and patient outcomes. A review by Tsuang et al. reported that ‘early initiation of treatment for hypertriglyceridemic pancreatitis is likely to be beneficial’, [3] based on findings from the retrospective case series by Kyriakidis et al., who reported positive patient outcomes in eight out of nine patients treated with plasmapheresis within 48 hours of diagnosis. [2]

The study by Gubensek et al. is also limited, in that it did not compare the effect of plasmapheresis with conventional treatment options.  No comparison was made with the mortality rate of patients who were treated conventionally. The study makes a strong point on the effectiveness of plasmapheresis in acutely reducing serum triglyceride and cholesterol levels, but does not determine whether there is any clinical benefit of this to the patient.

The indications and criteria for applying plasmapheresis was not consistent across the literature. Plasmapheresis is commonly used in settings where there is inadequate outcomes achieved using conventional management. [5] However, the patients treated with plasmapheresis in Chen et al. [9] and Gubensek et al. [4] include those with mild to moderate pancreatitis as well as severe pancreatitis (evaluated using Ranson’s scores or APACHE II score), but no information was given about any conventional treatment prior to the initiation of plasmapheresis. Therefore, it is unclear whether any prior conventional treatment had an effect on patient outcomes.

Neither study reviewed the potential symptomatic relief from plasmapheresis. A possible reason could be due to the subjectivity in measuring this outcome and the retrospective nature of the studies. Evaluation of potential symptomatic relief from plasmapheresis may be useful in minimising the severe pain associated with hypertriglyceridaemic pancreatitis.

ASFA [1] conducted a literature review to evaluate the rationale for plasmapheresis in patients with hypertriglyceridemic pancreatitis. This review is consistent with the findings of studies by Chen et al. and Gubensek et al., and reported no randomised controlled trial evaluating the effectiveness of plasmapheresis in these patients.  Their search on PubMed yielded twelve case-series and 28 case reports, with sample sizes ranging from 100-300. The ASFA have given a Category III indication to the use of plasmapheresis in patients with hypertriglyceridemic pancreatitis, which implies that the optimum role of apheresis therapy in these patients is not established. ASFA highlight the role of clinicians in making individualised decisions due to the weak evidence in this area. [1]

There is clearly a need for stronger evidence to determine the effectiveness of plasmapheresis in patients with hypertriglyceridemic pancreatitis. However, the low incidence of this disease means that randomised controlled trials may not be feasible. Gubensek et al. [4] argue that it would be questionable to perform a randomised controlled trial, given the large reduction in triglyceride levels by plasmapheresis. The ethical issues and feasibility of performing a randomised controlled trial on a small sample are barriers in answering this question. Nevertheless, large cohort studies with sufficient follow-up and appropriate adjustments for population stratification should provide immense support either in favour or against the use of plasmapheresis. Furthermore, appropriate studies assessing the effectiveness of rapidly lowering triglyceride levels on patient outcomes should be conducted.

Conclusion

There is insufficient evidence to confirm that plasmapheresis is a beneficial treatment option for patients with acute pancreatitis secondary to hypertriglyceridemia. Current literature shows that plasmapheresis promotes a rapid reduction in triglyceride levels of 46-80% [1], however its effect on patient outcomes remains unclear. Adequately powered prospective studies with long term follow-up are recommended to elucidate whether plasmapheresis is associated with reduced morbidity and mortality in this population.

Acknowledgements

Thank you to Dr Channa Perera, Endocrinologist at Campbelltown Hospital, for the inspiration to research and write up on this topic.

Conflict of interest

None declared.

Correspondence

M Rehmanjan: mrehmanjan@gmail.com

References

[1] Szczepiorkowski ZM, Winters JL, Bandarenko N, Kim HC, Linenberfer ML, Marques MB, et al. Guidelines on the use of therapeutic apheresis in clinical practice. Journal of Clinical Apheresis 2010; 25:83-177.

[2] Kyriakidis AV, Raitsiou B, Sakagianni A, Harisopoulou V, Pyrgioti M, Panagopoulou A, et al. Management of Acute Severe Hyperlipidemic Pancreatitis. Digestion 2006; 73(4):259-64.

[3] Tsuang W, Navaneethan U, Ruiz L, Palascak JB, Gelrud A. Hypertriglyceridemic Pancreatitis: Presentation and Management. The American Journal of Gastroenterology 2009;104:984-91

[4] Gubensek J, Buturovic-Ponikvar J, Marn-Pernat A, Kovac J, Knap B, Premru V, et al. Treatment of hyperlipidemic acute pancreatitis with plasma exchange: A single-center experience. Therapeutic Apheresis and Dialysis 2009;13(4):314–17.

[5] Lebenson J, Oliver T. Acute pancreatitis [Internet]. Rijeka, Croatia: InTech; 2012. Chapter 16, Hypertriglyceride Induced Acute Pancreatitis [cited 2013 Aug 25]. Available from: http://www.intechopen.com/books/acute-pancreatitis/hypertriglyceride-induced-acute-pancreatitis

[6] Brooker C. Medical Dictionary. Philadelphia. Churchill Livingstone Elsevier; 2008.

[7] University of Warwick Library. The Pico Method [Internet]. 2010 Sep 1 [cited 2012 Aug 6]. Available from: http://www2.warwick.ac.uk/services/library/tealea/sciences/medicine/evidence/pico/

[8] Elsevier. Scopus Database [Internet]. 2012 [cited 2012 Aug 6]. Available from: http://www.scopus.com/home.url?null

[9] Chen JH, Yeh JH, Lai HW, Liao CS. Therapeutic plasma exchange in patients with hyperlipidemic pancreatitis. World Journal of Gastroenterology 2004; 10(15):2272-74.

[10] Banks PA, Conwell DL, Toskes PP. The Management of Acute and Chronic Pancreatitis. Gastroenterol Hepatol 2010; 6(2 Suppl 5):1–16.

[11] Piolot A, Nadler F, Cavallero E, Coquard JL, Jacotot B. Prevention of recurrent acute pancreatitis in patients with severe hypertriglyceridemia: value of regular plasmapheresis. Pancreas 1996; 13(1):96-9.

Categories
Review Articles Articles

A systematic review evaluating non-invasive techniques to diagnose genetic disorders in a human fetus and the ethical implications of their use

Introduction: Genetic disorders are a significant cause of neonatal morbidity and mortality. [1] Diagnosing a genetic disorder currently involves invasive tissue sampling which carries an increased risk of miscarriage. The discovery of cell-free fetal DNA (cffDNA) in maternal plasma has enabled the development of non-invasive prenantal diagnostic tests (NIPD). [2,3] The scientific and ethical implications are examined. Methods: Medline, PubMed and Cochrane Library were searched for original research articles, review articles and meta-analyses focussed on screening and diagnosis of fetal genetic disorders. Results: 422 original research and review articles were assessed using processes in the Cochrane Handbook for Systematic Reviews of Interventions. [4] Using maternal plasma obtained during the second trimester, researchers were able to sequence the fetal genome with up to 98% accuracy. Clinicians reported the test will improve prenatal screening uptake, and reduce morbidity and mortality associated with genetic disorders. Ethicists argue it has implications for informed consent, rates of termination, reliability of future applications, inadvertent findings in clinical settings, commercial exploitation and inconsistent use of the technology internationally. Conclusions: Once NIPD tests utilising cffDNA are refined and costs reduced it is likely its implementation will affect both specialist genetic and routine antenatal services. However, given the complex set of ethical, legal and sociocultural issues raised by NIPD, professional education, public engagement, formal evaluation and the development of international standards are urgently needed. Health systems and policy makers must prepare to respond to cffDNA technology in a responsible and effective manner.

Introduction

Most pregnant women wish to be reassured that their unborn baby is healthy. [5] The aim of antenatal care is therefore to select screening and diagnostic tests that are accurate, safe and can be performed sufficiently early to allow parents to plan ahead or terminate the pregnancy in the event that fetal abnormality is diagnosed. [6] Genetic disorders are a significant cause (20%) of neonatal mortality. [1] At present, maternal serum screening, alone or in combination with ultrasound, is used to identify fetuses at risk of aneuploidy and other disorders. [7] Unfortunately, neither maternal serum screening nor ultrasound provide information on the genetic constitution of a fetus or allow a definitive diagnosis to be made. [8] For this, fetal cells must be invasively sampled from the placenta (chorionic villus tissue), amniotic fluid or fetal blood – all of which increase the risk of miscarriage. [9,10] This increased risk makes the decision to use invasive prenatal diagnosis difficult, particularly as there are still only very limited treatment options. [11] As a result, the medical community has sought to develop reliable and safe methods for achieving non-invasive prenatal diagnosis (NIPD), in addition to future treatment options. [12] Through NIPD, researchers hope to improve screening uptake, and reduce morbidity and mortality associated with genetic disorders. [1] Ethicists argue that NIPD transects existing distinctions between screening and diagnostic tests, and has implications for informed consent or choice. [12]

Methods

MEDLINE, PubMed and Cochrane Library were searched weekly between September 2012 and April 2013 for original research articles, review articles and meta-analyses focussed on screening and diagnosis of fetal genetic disorders. MeSH headings used were: Genetics, Medical, Genetics Testing and Fetus. Search terms used were: non-invasive, whole-genome and sequencing. Results were limited to human studies written in English between 1995 and 2013.

Results

The search resulted in 422 articles being identified; these were subsequently examined. The majority of publications were original research and review articles, although there was one meta-analysis by Alfirevic et al. (2003). [6] Many publications (217) were excluded for their limited scope or irrelevance.

Maternal serum screening and ultrasound are current methods of choice for screening pregnancies at risk of genetic disorders. [8,13] However, both methods rely on measuring epiphenomena rather than core pathology. Consequently, both tests have limited sensitivity and specificity and can only be used within a relatively narrow gestational period. [14] To achieve a definitive diagnosis chorionic villi sampling (CVS), amniocentesis or cordocentesis must be used. [6,8]

CVS is an invasive diagnostic procedure performed after 10 weeks gestation that is used for karyotyping when first trimester screening suggests a high risk of aneuploidy. [8] It is also used for fetal DNA analysis if the parents are known to be carriers of an identifiable gene mutation, such as cystic fibrosis or thalassaemia. [9] The procedure involves ultrasound-guided aspiration of trophoblastic tissue using either the trans-cervical or trans-abdominal routes. The tissue is then analysed with fluorescence in situ hybridisation polymerase chain reaction (FISH PCR). Like CVS, amniocentesis involves ultrasound-guided aspiration of amniotic fluid but is performed after 15 weeks gestation. [6] Cordocentesis involves direct sampling of fetal blood from the umbilical cord but is rarely performed and will not be discussed further in this article.

The benefit of CVS is that it can be performed at an earlier gestation, facilitating earlier diagnosis and providing the opportunity to terminate the pregnancy by suction curettage of the uterus. The benefits of amniocentesis include the lower background rate of miscarriage and the avoidance of isolated placental mosaicism occurring in 1% of samples. [8] The primary risk with CVS and amniocentesis is miscarriage. The level of risk is similar for the two tests (reported risk ranges from 1% to 1 in 1600) and is operator dependent. [6,7] Researchers have attempted to reduce this risk by developing a NIPD that allows the direct analysis of fetal genetic materials. [2,12,14-22] Better screening tests will achieve a higher detection rate combined with a lower false positive rate, resulting in less invasive testing and fewer procedure-related miscarriages.

Much of the early work on NIPD focussed on the isolation of fetal nucleated cells that had entered into the maternal blood. [14] However, the concentrations of these cells were low, meaning the tests had low sensitivity and specificity. [15,22] Later methods were inspired by the presence of tumour-derived DNA in the plasma of cancer patients. [23,24] In 1997, Lo et al. (1997) observed an analogous phenomenon was present in pregnancy by identifying Y chromosomal DNA sequences in plasma of women carrying male foetuses. [25] Replication of this study has concluded that 10% of cell-free DNA (cffDNA) in a pregnant woman’s plasma originates from the fetus she carries. [14,17,18,20] Since then, several groups have developed NIPD tests but most were only capable of detecting gross abnormalities such as aneuploidies, and were limited by small sample size and substandard accuracy. [17,18,22,26,27] In June 2012, Kitzman et al. (2012) reconstructed the whole-genome sequence of a human fetus using samples obtained relatively noninvasively during the second trimester, including paternal buccal DNA and maternal and cffDNA from the pregnant mother’s plasma. [2] Predicting which genetic variants were passed from mother to fetus was achieved by resolving the mother’s haplotypes – groups of genetic variants residing on the same chromosomes – and combining this result with shotgun genome sequencing of the father’s DNA and deep sequencing of maternal plasma DNA. [19] Comparing the results of this method with cord blood taken at delivery found inheritance was predicted with 98.1% accuracy. The study sequenced only two fetuses at a cost of $50,000 each, and is yet to be reproduced. Researchers from Stanford University were able to sequence the fetal genome without a paternal saliva sample although this was less accurate than the method used by Kitzman et al. (2012). [18] This latter method forms the basis of commercially available NIPD tests being offered by laboratories. [28] In Australia, NIPD testing is currently limited to Trisomy 21, 18, 13 and abnormalities of sex chromosomes, is not eligible for a Medicare rebate and costs upwards of $1,250. [29] It is anticipated that analysing samples for NIPD locally will reduce the cost and drive demand. [30,31]

Discussion

Clinicians report that non-invasively diagnosing genetic disorders will reduce infant mortality and morbidity. [31] Ethicists argue the technology raises concerns for informed consent, rates of termination, reliability of future applications, inadvertent findings in clinical settings, commercial exploitation and inconsistent use of the technology internationally [12,32-36].

Informed Consent and Informed Choice

Ethicists believe NIPD testing transects existing distinctions between screening and diagnostic tests and has implications for informed consent and choice. [12] An example is screening for Down’s syndrome, a common genetic disorder. Although a significant number of women may not already achieve informed choice for screening, at least a subsequent invasive diagnosis provides another opportunity for reflection as they consent to the procedure (CVS or amniocentesis). [34,35] Replacing this multi-step screening process with highly-predictive cffDNA testing may reduce opportunities for exercising informed choice. [12] In addition, despite the belief that introducing cffDNA testing will promote parental reproductive choice, it may indeed make proceeding with an affected pregnancy more difficult for two reasons: First, the decreased risks associated with cffDNA might lead women to feel ‘pressured’ into agreeing to the tests, or undergoing testing without informed consent, even if they potentially lead to outcomes with which they disagree. [33,36] Second, the lower risks might cause a shift in the extent to which society is supportive of those who chose to have disabled children. [10] In turn, worries over social disapprobation could prompt a loopback effect, where women feel more pressured to test and to terminate their pregnancies.

Termination of pregnancy (TOP)

In Australia, there is broad agreement that TOP is ethically and legally permissible in some circumstances. [11,33,37] However, the laws are notoriously unclear, outdated and inconsistent between states and territories. [38,39] In many jurisdictions it is legally defensible for a clinician to perform a TOP at any gestation if they can justify the harms of continuing with the pregnancy outweigh the risks of termination. [40] For this reason, access to TOP is very much dependent on the clinician, which may be problematic if cffDNA testing becomes more widespread and moves outside the existing setting of medical genetics, where high standards of relevant ethical practice and the professional duty of non-directive counselling are firmly entrenched. [12]

Accuracy and reliability of NIPD

Despite improved accuracy by utilising fetal nucleic acids, the sensitivity and specificity of even the most accurate method is still less than 100%. [2] Maintaining an acceptably high sensitivity and specificity will also be a challenge, as researchers discover an ever-increasing number of sequences associated with pre-existing diseases. [12] To do this will require careful monitoring within different applications. [33] Without it, the personal, sociocultural, legal and ethical ramifications of false positives and negatives may be devastating. For example, additional invasive testing may be undertaken, healthy fetuses may be terminated, and children may suffer psychologically should they discover their parents would have terminated them if they had known of their diagnosis. [34]

Inadvertent findings in clinical settings

The Kitzman et al. method requires paternal buccal DNA to sequence the fetal genome and may therefore inadvertently disclose misattributed paternity. [41] However, so too may the Stanford University method that forms the basis of commercially available NIPD but that does not require paternal buccal DNA. [18] In a trial of 18 subjects, researchers using the latter method were able to predict 70% of the paternally inherited haplotypes in the fetus with 94–97% accuracy. [18] Of course, the correlation of these findings to the clinical setting would likely still require paternal buccal DNA to confirm paternity. The potential for inadvertent disclosure of misattributed paternity would be a particular concern if cffDNA testing were ever incorporated into routine antenatal screening as a greater number of women who may not have been adequately forewarned would be exposed to the risks such information may bring.

Commercial and international uses

The likely increase in the accessibility of NIPD using cffDNA tests made available via the internet has major implications, particularly for fetal sex selection. [12] In China [42] and India, [43] population skewing has already been observed as a result of unlawful sex selection practices favouring male children. Some ethicists believe cffDNA could significantly aggravate or extend this problem. [44] The development of cffDNA technology within the commercial sector is also a concern as some companies choose only to sell the service rather than invest in research and development eg: babygendermentor.com. The provision of testing direct-to-consumers raises a complex set of issues relating to the role of ‘gatekeepers’ in prenatal testing and access to non-clinical applications of the technology. [33] In addition, it may even impact upon the provision made through Medicare for ongoing care, including diagnostic confirmation, interventional procedures (such as TOP) and medical advice. [5] Having commercial players involved may result in elements of professional practice, including informed consent and counselling, being difficult to enforce considering international legislative and regulatory boundaries. [12] The cultural context is also highly relevant to how consumers access cffDNA testing. For example, its use in countries where access to safe TOP is limited or absent is ethically questionable and could cause significant social and medical problems. [45]

Conclusion

The utilisation of cffDNA for safe and reliable NIPD has opened the way for accurate sequencing of the fetal genome and the ability to diagnose an ever-increasing number of genetic anomalies and their clinical disorders. Once methods such as those by Kitzman et al. and researchers at Stanford University are refined and costs reduced it is likely the implementation of cffDNA testing will affect both specialist genetic and routine antenatal services, improve screening uptake, and reduce morbidity and mortality associated with genetic disorders. As a result of the pace of development, there is concern that cffDNA testing transects existing distinctions between screening and diagnostic tests, having implications for informed consent, termination rates and commercial. Given the complex set of ethical, legal and sociocultural issues raised by NIPD, both professional education and public engagement are urgently needed. Formal evaluation of each test should be required to determine its clinical accuracy, and laboratory standards should be developed alongside national best practice guidelines to ensure that cffDNA testing is only offered within agreed and well-supported pathways that take account of the aforementioned issues. This development has the potential to deliver tangible improvements in antenatal care within the next 5-10 years, and health systems and policy makers around the globe must now prepare to respond to further developments in cffDNA technology in a responsible, effective and timely manner.

Conflict of interest

None declared.

Correspondence

M Irwin: matt@irwinmd.com.au

References

[1] Bell, C.J., et al., Carrier testing for severe childhood recessive diseases by next-generation sequencing. Sci Transl Med, 2011. 3(65): p. 65ra4.

[2] Kitzman, J.O., et al., Noninvasive whole-genome sequencing of a human fetus. Sci Transl Med, 2012. 4(137): p. 137ra76.

[3] Allyse, M., et al., Cell-free fetal DNA testing for fetal aneuploidy and beyond: clinical integration challenges in the US context. Hum Reprod, 2012.

[4] Collaboration, T.C., in Cochrane Handbook for Systematic Reviews of Interventions, J. Higgins and J. Deeks, Editors. 2011.

[5] Tischler, R., et al., Noninvasive prenatal diagnosis: pregnant women’s interest and expected uptake. Prenat Diagn, 2011. 31(13): p. 1292-9.

[6] Alfirevic, Z., K. Sundberg, and S. Brigham, Amniocentesis and chorionic villus sampling for prenatal diagnosis. Cochrane Database Syst Rev, 2003(3): p. CD003252.

[7] Dugoff, L. and J.C. Hobbins, Invasive procedures to evaluate the fetus. Clin Obstet Gynecol, 2002. 45(4): p. 1039-53.

[8] Collins, S.L. and L. Impey, Prenatal diagnosis: types and techniques. Early Hum Dev, 2012. 88(1): p. 3-8.

[9] Mujezinovic, F. and Z. Alfirevic, Procedure-related complications of amniocentesis and chorionic villous sampling: a systematic review. Obstet Gynecol, 2007. 110(3): p. 687-94.

[10] Geaghan, S.M., Fetal laboratory medicine: on the frontier of maternal-fetal medicine. Clin Chem, 2012. 58(2): p. 337-52.

[11] van den Berg, M., et al., Accepting or declining the offer of prenatal screening for congenital defects: test uptake and women’s reasons. Prenat Diagn, 2005. 25(1): p. 84-90.

[12] Hall, A.B., A; Wright, C F, Non-Invasive Prenatal Diagnosis Using Cell-Free Fetal DNA Technology: Applications and Implications. Public Health Genomics, 2010. 13(4): p. 246-255.

[13] Wieacker, P. and J. Steinhard, The prenatal diagnosis of genetic diseases. Dtsch Arztebl Int, 2010. 107(48): p. 857-62.

[14] Lo, Y.M., Non-invasive prenatal diagnosis by massively parallel sequencing of maternal plasma DNA. Open Biol, 2012. 2(6): p. 120086.

[15] Bianchi, D.W., et al., PCR quantitation of fetal cells in maternal blood in normal and aneuploid pregnancies. Am J Hum Genet, 1997. 61(4): p. 822-9.

[16] Dan, S., et al., Prenatal detection of aneuploidy and imbalanced chromosomal arrangements by massively parallel sequencing. PLoS One, 2012. 7(2): p. e27835.

[17] Faas, B.H., et al., Non-invasive prenatal diagnosis of fetal aneuploidies using massively parallel sequencing-by-ligation and evidence that cell-free fetal DNA in the maternal plasma originates from cytotrophoblastic cells. Expert Opin Biol Ther, 2012. 12 Suppl 1: p. S19-26.

[18] Fan, H.C., et al., Non-invasive prenatal measurement of the fetal genome. Nature, 2012. 487(7407): p. 320-4.

[19] Kitzman, J.O., et al., Haplotype-resolved genome sequencing of a Gujarati Indian individual. Nat Biotechnol, 2011. 29(1): p. 59-63.

[20] Lo, Y.M., et al., Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med, 2010. 2(61): p. 61ra91.

[21] Muller, S.P., et al., Cell-free fetal DNA in specimen from pregnant women is stable up to 5 days. Prenat Diagn, 2011. 31(13): p. 1300-4.

[22] Bianchi, D.W., et al., Fetal gender and aneuploidy detection using fetal cells in maternal blood: analysis of NIFTY I data. Prenatal Diagnosis, 2002. 22(7): p. 609-615.

[23] Chen, X.Q., et al., Microsatellite alterations in plasma DNA of small cell lung cancer patients. Nat Med, 1996. 2(9): p. 1033-5.

[24] Nawroz, H., et al., Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med, 1996. 2(9): p. 1035-7.

[25] Lo, Y.M., et al., Presence of fetal DNA in maternal plasma and serum. Lancet, 1997. 350(9076): p. 485-7.

[26] Chiu, R.W. and Y.M. Lo, Non-invasive prenatal diagnosis by fetal nucleic acid analysis in maternal plasma: the coming of age. Semin Fetal Neonatal Med, 2011. 16(2): p. 88-93.

[27] Chu, T., et al., Statistical model for whole genome sequencing and its application to minimally invasive diagnosis of fetal genetic disease. Bioinformatics, 2009. 25(10): p. 1244-50.

[28] Skirton, H. and C. Patch, Factors affecting the clinical use of non-invasive prenatal testing: a mixed methods systematic review. Prenat Diagn, 2013. 33(6): p. 532-41.

[29] Douglass Hanly Moir Pathology, Verifi Prenatal Test, in http://dhm.com.au/media/21890505/verify_6pp_dhm_dl_fa_web.pdf2013, Douglass Hanly Moir Pathology: Sydney. p. 1-2.

[30] Kelly, S.E. and H.R. Farrimond, Non-invasive prenatal genetic testing: a study of public attitudes. Public Health Genomics, 2012. 15(2): p. 73-81.

[31] Sayres, L.C., et al., Cell-free fetal DNA testing: a pilot study of obstetric healthcare provider attitudes toward clinical implementation. Prenat Diagn, 2011. 31(11): p. 1070-6.

[32] Dormandy, E., et al., Low uptake of prenatal screening for Down syndrome in minority ethnic groups and socially deprived groups: a reflection of women’s attitudes or a failure to facilitate informed choices? International Journal of Epidemiology, 2005. 34(2): p. 346-352.

[33] Newson, A.J., Ethical aspects arising from non-invasive fetal diagnosis. Semin Fetal Neonatal Med, 2008. 13(2): p. 103-8.

[34] Schmitz, D., C. Netzer, and W. Henn, An offer you can’t refuse? Ethical implications of non-invasive prenatal diagnosis. Nat Rev Genet, 2009. 10(8): p. 515.

[35] Seavilleklein, V., Challenging the rhetoric of choice in prenatal screening. Bioethics, 2009. 23(1): p. 68-77.

[36] van den Berg, M., et al., Are counsellors’ attitudes influencing pregnant women’s attitudes and decisions on prenatal screening? Prenat Diagn, 2007. 27(6): p. 518-24.

[37] Drabsch, T., Abortion and the law in New South Wales, N.P.L.R. Service, Editor 2005, NSW Parliamentary Library Research Service: Sydney. p. 1-33.

[38] de Costa, C.M., et al., Introducing early medical abortion in Australia: there is a need to update abortion laws. Sex Health, 2007. 4(4): p. 223-6.

[39] De Crespigny, L.J. and J. Savulescu, Abortion: time to clarify Australia’s confusing laws. Med J Aust, 2004. 181(4): p. 201-3.

[40] Cica, N., Abortion Law in Australia, D.o.t.P. Library, Editor 1998, Department of Parliamentary Library: Canberra.

[41] Lucast, E.K., Informed consent and the misattributed paternity problem in genetic counseling. Bioethics, 2007. 21(1): p. 41-50.

[42] Cecilia Lai-wan, C., B. Eric, and C. Celia Hoi-yan, Attitudes to and practices regarding sex selection in China. Prenatal Diagnosis, 2006. 26(7): p. 610-613.

[43] George, S.M., Millions of missing girls: from fetal sexing to high technology sex selection in India. Prenat Diagn, 2006. 26(7): p. 604-9.

[44] Van Balen, F., Attitudes towards sex selection in the Western world. Prenat Diagn, 2006. 26(7): p. 614-8.

[45] Ballantyne, A., et al., Prenatal Diagnosis and Abortion for Congenital Abnormalities: Is It Ethical to Provide One Without the Other? The American Journal of Bioethics, 2009. 9(8): p. 48-56.

Categories
Review Articles Articles

The role of viruses in carcinogenesis

It is accepted that populations in the so-called developed world have gone through an ‘epidemiological transition’ where chronic disease has replaced infection as the primary cause of death. However, there is mounting evidence that infections play a key role in certain chronic diseases such as cancer. Cancers of infectious origin provide the perfect opportunity for harnessing the advances that have been made in the control of communicable diseases to attempt the control of noncommunicable diseases. Worldwide, one in every five malignancies can be attributed to infections: this figure is considered conservative and expected to rise. About two-thirds of these cancers occur in less developed countries. The majority of these malignancies are recognised to be caused by viruses via mechanisms of chronic inflammation, immunosuppression or the expression of oncogenic proteins. An understanding of virally mediated carcinogenesis may provide new targets for the development of specified viral therapy that not only impacts on viral infections but human cancer as well. From a public health perspective, viral carcinogenesis is important because it shows potential for preventative and therapeutic programmes to reduce the burden of cancer, particularly in less developed countries.

Introduction

The process of carcinogenesis involves multiple contributing factors. These include environmental, lifestyle, host factors, genetically inherited traits and infectious agents. Infectious agents are important from a public health aspect as they represent a significant and preventable cause of cancer. The infection-attributable cancer burden has been estimated at 1.9 million cases, or 17.8% of the total global cancer burden. [1] The percentage of infection-attributable cancer is higher in developing countries (26.3%) than in developed countries (7.7%), reflecting the higher prevalence of infectious diseases. Of these infection-associated cancers, viruses are the most common causative agents with 12.1% of cancers worldwide attributed to viral infections. [1]

This article aims to outline current knowledge of the role of viruses in mediating cancer, explore the main mechanisms involved and propose exciting preventative and therapeutic approach for virus-associated cancers in the 21st century.

Mechanisms by which viruses mediate cancer

The International Agency for Research on Cancer (IARC) recognises seven viral agents that have been linked with cancer: Hepatitis B Virus (HBV) and Hepatitis C Virus (HCV), Human Papilloma Virus (HPV), Epstein-Barr Virus (EBV), Karposi-Sarcoma Herpes Virus (Human Herpes Virus 8), Human T-cell leukemia virus type I (HTLV-1) and Human Immunodeficiency Virus type 1 (HIV). [2] These seven viruses classified as ‘carcinogenic to humans’, and the recently discovered Merkel Cell Virus which has not yet been included by the IARC, are summarised in Table 1.

The induction of cancer development by viruses requires persistent infection of the host.  It is hypothesised that long-term infection initiates cellular changes that predispose to cancer progression. [3]

In addition to persistent infection, the specific actions of these viruses are discussed below and can be broadly grouped into viruses that induce cancer by (i) chronic inflammation (eg. HCV), (ii) immunosuppression (eg. HIV) and by (iii) direct actions of viral oncogenic proteins (eg. EBV, HPV). [3]

(i) Cancer associated with chronic inflammation: Hepatitis B and C Viruses

Once a viral infection is initiated, recovery requires the activation of the innate and adaptive arms of the immune system. Acute inflammation is usually a short process that eliminates the pathogen. However, chronic inflammation may result if acute inflammation continues unresolved and fails to eradicate the pathogen. Chronic inflammation itself may promote carcinogenesis via the release of many factors including nitric oxide, cytokines and chemokines thus mediating DNA damage and effecting cell proliferation and neoangiogenesis. [3]

HBV and HCV infections are examples of chronic infections associated with ongoing inflammation. HBV and HCV are responsible for 54% and 31% of human hepatocellular carcinoma (HCC) cases worldwide. [4,5] These hepatotropic viruses can induce cirrhotic livers from which HCC can arise. This review will focus on HCV.

In those infected with HCV, 80% will develop chronic infection, and in 30 years 10-30% of these chronic HCV infections will develop cirrhosis. The subsequent rate of cirrhotic HCV liver disease developing HCC is 1-3% per year. [6] Since current WHO estimates suggest that 3% of the world’s population, or 150 million people, are HCV infected, this represents a significant virus-associated cancer burden.

HCV is a RNA virus of the hepacivirus family of the genus Flaviviridae. HCV does not integrate itself into the host genome and several viral proteins (core protein and the NS3, NS4B and NS5A) have been suggested as potential oncogenic candidates in-vitro. For example, the HCV NS5A protein has been shown to bind and sequester the cellular p53 protein to the perinuclear membrane, and it may be the be key to HCC development. [7] However, it is thought that HCC primarily occurs due to repeated rounds of hepatocyte destruction and regeneration from chronic inflammation, producing a procarcinogenic cirrhotic microenvironment, [3,8] rather than through the action of viral oncogenes. Cirrhosis appears to be the main risk factor for HCC, but exogenous factors could also play a role, such as chronic alcohol consumption, viral co-infection (such as HIV modulating immunosuppression), diabetes and obesity [4] highlighting the multifactorial triggers for the induction of cancer. [9]

HCV is also a well-established cause of essential mixed cryoglobulinemia, a lymphoproliferative disease that can evolve into B-cell non-Hodgkin lymphoma (NHL). [10] HCV has been suggested to be lymphotropic, but this is not well defined. [11] Again, since HCV has not been demonstrated to encode direct oncogenic proteins, the mechanisms of HCV-induced NHL are likely to be via chronic inflammation.

(ii) Cancer associated with immunosuppression and insertional mutagenesis: HIV

It is estimated that there are approximately 34.2 million individuals worldwide living with HIV infection, two-thirds of these being in sub-Saharan Africa. [12] People with HIV have a substantially higher risk of certain cancers compared with uninfected people of the same age. These cancers are termed AIDS-defining malignancies and include: Kaposi sarcoma, a mesenchymal tumour originating from lymphatic endothelial cells, cervical cancer and NHL. [13] Additionally, other types of cancer, such as Hodgkin’s disease (HD), anal cancer, lung cancer and testicular germ cell tumours appear to be more common among HIV-infected subjects compared to the general population and are termed AIDS-associated cancers. [14]

HIV is an RNA lentivirus of the Retroviridae family. The members of this family all integrate into the host chromosome and thus have the potential to cause direct insertional mutations or activation of cellular oncogenes.  Other members of the Retroviridae family, such as Mouse mammary tumour virus (MMTV) have a well-defined link with tumours in mice, which are likely mediated by insertional activation of cellular genes in breast tissue through hormone responsive elements in the MMTV promoter. [15] Similarly, insertional mutagenesis and the induction of lymphoma has been identified in humans treated with gammaretrovirus [16] and lentivirus vectors used in gene therapy. [17] In contrast, there is little evidence for an HIV oncogenic protein, although studies have suggested that the transactivator protein of viral gene expression, Tat, which has oncogenic potential, is secreted by HIV. It has also been suggested that Tat can re-enter non-infected cells blocking apoptosis and accelerating tumour formation. [18]

The above described AIDS-associated cancers are linked with low CD4+ T-cell counts, and this may lead to co-infections with other oncogenic viruses such as HPV (cervical cancer) and Kaposi’s sarcoma-associated herpesvirus (Kaposi’s sarcoma), or the reactivation of existing infections with opportunistic oncogenic viruses such as EBV (Burkitt’s lymphoma). [18] However, the specific mechanisms by which depressed immunity may increase the risk for cancer are unclear, except for KS and most subtypes of NHL that are strictly associated with a low CD4 count. [19] Supporting the link between cancer and immunosuppression, the pattern of cancers in immunosuppressed organ transplant recipients is similar to people with HIV/AIDS. [20]

Thus, the evidence suggests that HIV can be associated with carcinogenesis through insertional mutagenesis. Moreover, HIV may indirectly cause cancers by inducing a chronic state of immunosuppression, reducing immunosurveillance for neoplastic cells, and increasing the risk of reactivation of latent oncogenic viruses as well as the risk of acquiring new oncogenic viral infections.

(iii) Cancer associated with Oncogenic viruses

Of the identified and accepted carcinogenic viruses, EBV, HHV-8, HTLV-1and HPV are tumour viruses that express viral oncogenic proteins to exert carcinogenesis. HBV also produces the HBx protein that disrupts signal transduction and deregulates cell growth: however, HBV-associated carcinogenesis is believed to be mainly mediated through chronic inflammation as described for HCV. [3] Oncogenic viruses can transform cells by carrying viral oncogenes into a cell or by activating cellular proto-oncogenes. [5] The virally derived oncogenes produce transforming growth factors that deregulate growth control and proliferation, leading to malignant transformation. Specific examples are discussed below, with the oncogenic viruses divided into DNA and RNA tumour viruses.

DNA Tumour Viruses

EBV best illustrates DNA tumour viruses. EBV is a double-stranded DNA virus of the herpesviridae family, and causes infectious mononucleosis. Like all herpesviruses, EBV causes a life-long latent infection, and EBV is the primary cause of B-cell transformation in Burkitt’s lymphoma. [14] This was the first human tumour associated with an infectious agent. Since then, EBV has been implicated in a number of other cancers (see Table 1).

In the case of EBV-lymphoma, expression of the viral oncogene, latent membrane protein-1 (LMP1), transforms cells into lymphoblasts by the disruption of cellular signal transduction. [3] In contrast, in most NPCs, the viral BamHI-A reading frame-1 (BARF1) gene is expressed. BARF1 has been identified as an important oncogene in NPC pathology. [21] Thus, EBV has a number of different oncogene expression profiles associated with different cancers. EBV is extremely widespread in prevalence affecting more than 90% of the world’s population, [22] yet only a small fraction of the infected populations have a cancer attributable to EBV.  Therefore, beside viral factors, host responses also play a role in the neoplastic transformation of EBV-infected cells.

HHV-8 is a DNA virus of the herpesviridae family, and HHV-8 infection is strongly associated with Kaposi’s sarcoma. The mechanism, however, of HHV-8-induced carcinogenesis is very different to that of the related virus, EBV. HHV-8 infects endothelial cells and encodes a viral G protein-coupled receptor (vGPCR). This vGPCR has dysregulated signalling function and acts as an oncogene, inducing angioproliferative tumours. [23]

HPV is a DNA virus of the papillomavirus family, and there are 30-40 types. Approximately fifteen types of HPV are oncogenic viruses, causing 5.2% of total human cancers. [24] These cancers include those of the ano-genital mucosae (cervix, vagina, vulva, anus and penis), and the mouth and the pharynx. [24,25] The predominant transmission of these HPV infections is sexual. [26] While HPV is an accepted aetiological factor for oral and pharyngeal cancers, the major risk factors are tobacco and alcohol, with the effects of these exposures being multiplicative. [25] Oncogenic HPV can be detected by PCR in virtually all cases of cervical cancer, with specific genotypes HPV16 and 18 identified as the primary causes of cervical cancer. These viral genotypes have also been associated with 86-95% of HPV-associated non-cervical cancers. [26,27] These viruses infect the basal layer of the stratified epithelium and express two important viral oncoproteins, E6 and E7. [23] These proteins destabilise the cellular tumour suppressor genes, p53 and the retinoblastoma protein (RB). [28] This dysregulation of cellular growth directly leads to cell transformation and cancer.

RNA Tumour Viruses

HTLV-I is a retrovirus related to HIV, which is associated with adult T-cell leukaemia. Only 1% of HTLV-I infected individuals will develop leukaemia, and only after a long latency period of 20-30 years. [29] HTLV-I infection rates are elevated in certain Indigenous populations of Central and Northern Australia, as well as the southern islands of Japan, the Caribbean basin and South Africa. [14] Unlike HIV, HTLV-I infections are not associated with immunosuppression, but HTLV-I encodes an oncogenic protein; the viral Tax protein. [30,31] Tax is a transcription factor and is known to bind to a number of cellular genes involved in cell cycle progression and growth regulation, such as NFkB and p53. [32] Via promotion of transcription and cell cycle progression, Tax is proposed to set up a self-stimulating loop that causes continuous proliferation of infected T-cells, and ultimately leukaemia.

The growing cancer burden attributable to viruses

While there are only seven viruses clearly recognised as carcinogenic to humans, this is conservative, with the discovery of new associations between infections, particularly viruses, and cancer anticipated.

MCV is a recently discovered DNA virus that is found to be associated with approximately 80% of Merkel Cell Carcinomas, an aggressive form of skin cancer. [33] MCV is a relatively common virus, yet only leads to cancer in rare circumstances. It is thought that this is because for MCV to become carcinogenic, two rare mutagenic steps must occur:  viral integration and T antigen mutation. Integration of MCV is not a regulated event, unlike for HIV and HTLV-I, and occurs rarely. The integration, probably of only parts of the MCV genome into cells, renders the virus replication-incompetent, but allows parts of the virus, such as the T-antigens, to be maintained in these cells. [34] MCV T antigens can be oncogenic, and target cellular tumour suppressors and cell cycle regulatory proteins. Thus, the whole replicative virus may not be present, but the residual oncogenic T-antigen is, and can promote transformation of the cell leading to cancer.

Cancerous cells themselves are generally not transmissible. In humans, during the two known physiological routes for tumour cell transmission (pregnancy and organ transplantation), the immune system is altered.  Transplacental transmission of lymphoma, acute leukaemia, melanoma and carcinoma have been observed, as well as acute leukaemia cells transmitted to the foetuses in multiple case pregnancies with the subsequent disease development in the newborn. [35] Similarly, in organ transplantation, donor derived tumour cells have been observed, with the immunosuppressive therapy following transplantation potentially facilitating the engraftment and growth of donor derived tumour cells. [35] Fortunately, these transmissible tumours are rare, with the development of donor-derived tumours in solid organ transplant recipients at 0.04%. [15] Additionally there have been rare case reports of human contagious cancers documented via needle stick (colonic adenocarcinoma), [36] and a surgeon contracting a malignant fibrous histiocytoma from a patient following an intraoperative cut to his left palm. [37]

Cancer prevention and public health strategies

In theory, the cancers resulting from viral infections represent an exciting potential for public health intervention strategies and therapeutics to prevent these cancers.  In particular, the high number of cancers attributable to viral infections in developing countries presents a real need and opportunity for public health programs to reduce both infectious disease and cancer burden. [38]

The mode of transmission of the seven IARC-recognised carcinogenic viruses is provided in Table 2. The implementation of public health education, awareness, treatment and prevention programs to reduce the horizontal spread of these viruses and manage these viral infections in patients is a public health priority, but has the additional benefit of reduction in the associated cancer risks.

Public health programs should be prioritised to target vertical transmission of viral infections such as HBV and HIV. The WHO outlined targets and recommendations in 2010 with the prevention of mother to child transmission (PMTCT) strategy, targeting anti-retroviral therapy (ART) in pregnant women and providing guidelines for HIV in relation to infant breastfeeding. [39] Similar guidelines may be applicable to our Indigenous population afflicted by HTLV-I, which has a well described increased mother to child transmission rate associated with breastfeeding. [40] However, breastfeeding recommendations in resource poor settings need careful consideration. [41]

More outstanding, are the successful programs for screening and management of viral infections associated with cancer. For example, the National Cervical Screening Program (NCSP) in Australia has had a huge benefit in reducing the mortality rates from cervical cancer from 3.9/100,000 in 1991 to 1.9/100,000 in 2007, [42], demonstrating that cancer prevention via monitoring oncogenic viral infections is a real possibility. [43] Additionally, such programs as the NSW Cancer Council ‘B positive’ program, implemented in 2008 aims to increase HBV awareness and the treatment and management of chronic HBV infection to reduce the risk of HCC. [44]

Vaccination and treatments to prevent cancer-associated viral infections

Historically, the world has experienced, with polio and smallpox, elimination or virtual elimination of viral diseases through vaccination. There are now vaccines available for both HBV and HPV, two major infectious causes of HCC and cervical cancer, respectively. The HBV surface antigen is the basis for the vaccine against the HBV, which was first available in the 1980s, and is the first vaccine for prevention of a human cancer. [45] Vaccination programmes of children with the HBV vaccine have already proved successful in protecting against chronic carriage and HCC, [46,47] and HBV vaccination has now been introduced into the Australian childhood immunisation schedule. Long-term and full coverage of newborns against HBV has the potential of reducing HCC by approximately 85%. [14]

The two currently marketed vaccines for HPV utilise the L1 coat protein in the form of virus-like particles to prevent persistent infection with HPV16 and HPV18. [48-50] These viral subtypes are estimated to cause 71.8% of all HPV-related cancers, cervical and non-cervical. [25] These vaccines need to be administered prior to exposure to HPV16 and 18, which makes delivery in a public health setting more difficult than an infant setting. In Australia in 2007, the National HPV vaccination Program was made available to teenage women, and is now part of the school age vaccination program. From 2013 will also be made available to 12-13 year old males. [51] However, the current cost is not practical for all groups, especially those in developing countries, [14] and although the HPV vaccination program in developing countries is supported by the WHO, the applicability and benefits of HPV vaccination have been queried and recently suspended in India. [52] The efficacy of these HPV vaccines in preventing infections at sites other then the cervix, vagina and vulva should be assessed. [27] Specifically, research is required on the administration to high-risk groups (e.g. men who have sex with men and HIV positive people) for anal cancer. [24]

Unfortunately, the described RNA viruses associated with cancer, HIV and HCV, are highly genetically variable and therefore prove to be difficult candidates for prophylactic vaccines. For these viruses, anti-viral therapy appears to be more successful. For example, the risk of infection-associated cancers in HIV positive individuals is related to ongoing HIV replication. The use of suppressive highly active antiretroviral therapy (HAART) has dramatically reduced the risk for opportunistic infections and improved overall life expectancy in patients with HIV-infection and AIDS. [53] A significant decrease in the incidence of KS has been observed in patients treated with HAART. [19] Moreover, HAART and preserved CD4 count preferentially reduces the risk of malignancies associated with oncogenic infections. [54] Similarly, patients with HCV who were prescribed the anti-viral agent, interferon, showed regression of their splenic lymphoma. [55]

Recently approved HCV NS3-4A protease inhibitors are proving effective in clearing and curing HCV infection. In the future, this may significantly impact on HCV infection rates and subsequent incidence of HCC.

Exciting Therapeutic Targets

Our understanding of mechanisms of viral initiation of carcinogenesis has provided the opportunity to design innovative, targeted cancer therapies based on the pathways disrupted by the transforming viral genes.

For example, recent studies reveal that the cellular survivin oncoprotein is activated by MCV large T antigen protein via targeting the cellular Rb (p53) protein, and that survivin inhibitors can delay MCV-induced tumour progression in animal models. [56] Clinical trials are now in progress to determine whether these survivin inhibitors have any therapeutic benefits. Additionally, MCV is a target for cell-mediated immune responses, and so important research efforts are being focused on immunologic therapies that may benefit MCC patients. [56] These findings provide a proof of principle for specifically treating virus-associated cancers by targeting the mechanisms by which they induce oncogenesis. In the case of MCV, a promising rational drug target has been uncovered within only four years of the initial discovery of MCV as a causative cancer agent. Similarly, other new treatments for cancer might be rapidly developed should we identify new viral associations with malignancies.

Conclusion

Viruses are an important aetiological cause of human cancers, especially in the developing world where they lead to a significant burden of disease.  Although viruses make an important contribution to human cancer development, it is often difficult to prove the association of viral infections with cancer, due to latency in tumour development and the multifaceted interaction with the host.  It is reasonable to think that the calculations of cancers attributable to viruses are underestimates and that cancers other than the ones described may also be associated with viral infections. The viruses in this review exemplify the best-established human tumour viruses, but there are many other potential candidates.  Undoubtedly, as our knowledge of carcinogenesis and viruses expand, further cancer-associated viruses will be discovered. From a public health point of view, infectious diseases are often preventable or treatable; therefore, cancers associated with infections are, or may become, preventable.  Prevention may be in the form of vaccination, novel therapies to target the immune system or oncogenic proteins, or education and public health interventions.

Conflict of interest

None declared.

Correspondence

V Boon: boon0035@flinders.edu.au

References

[1] de Martel C, Ferlay J, Franceschi S, Vignat J, Bray F, Forman D, et al. Global burden of cancers attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol. 2012 Jun;13(6):607-15. PubMed PMID: 22575588. Epub 2012/05/12. eng.

[2] Bouvard V, Baan R, Straif K, Grosse Y, Secretan B, El Ghissassi F, et al. A review of human carcinogens–Part B: biological agents. Lancet Oncol. 2009 Apr;10(4):321-2. PubMed PMID: 19350698. Epub 2009/04/08. eng.

[3] Carrillo-Infante C, Abbadessa G, Bagella L, Giordano A. Viral infections as a cause of cancer (review). Int J Oncol. 2007 Jun;30(6):1521-8. PubMed PMID: 17487374. Epub 2007/05/10. eng.

[4] El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012 May;142(6):1264-73 e1. PubMed PMID: 22537432. Pubmed Central PMCID: 3338949. Epub 2012/04/28. eng.

[5] Parkin DM. The global health burden of infection-associated cancers in the year 2002. Int J Cancer. 2006 Jun 15;118(12):3030-44. PubMed PMID: 16404738. Epub 2006/01/13. eng.

[6] Rosen HR. Clinical practice. Chronic hepatitis C infection. N Engl J Med. 2011 Jun 23;364(25):2429-38. PubMed PMID: 21696309. Epub 2011/06/24. eng.

[7] Farazi PA, DePinho RA. Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer. 2006 Sep;6(9):674-87. PubMed PMID: 16929323. Epub 2006/08/25. eng.

[8] Farazi PA, Glickman J, Horner J, Depinho RA. Cooperative interactions of p53 mutation, telomere dysfunction, and chronic liver damage in hepatocellular carcinoma progression. Cancer Res. 2006 May 1;66(9):4766-73. PubMed PMID: 16651430. Epub 2006/05/03. eng.

[9] Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology. 2004 Nov;127(5 Suppl 1):S35-50. PubMed PMID: 15508101. Epub 2004/10/28. eng.

[10] de Martel C, Franceschi S. Infections and cancer: established associations and new hypotheses. Crit Rev Oncol Hematol. 2009 Jun;70(3):183-94. PubMed PMID: 18805702. Epub 2008/09/23. eng.

[11] Zignego AL, Ferri C, Monti M, LaCivita L, Giannini C, Careccia G, et al. Hepatitis C virus as a lymphotropic agent: evidence and pathogenetic implications. Clin Exp Rheumatol. 1995 Nov-Dec;13 Suppl 13:S33-7. PubMed PMID: 8730474. Epub 1995/11/01. eng.

[12] Organisation WH. HIV/AIDS Fact Sheet No 360. Geneva WHO Media centre; 2011.

[13] Control CfD. 1993 revised classification system for HIV infection and expanded surveillance case definition for AIDS among adolescents and adults. Morb Mortal Wkly Rep. 1992;41:1-19.

[14] Harford JB. Viral infections and human cancers: the legacy of Denis Burkitt. Br J Haematol. 2012 Mar;156(6):709-18. PubMed PMID: 22233526. Epub 2012/01/12. eng.

[15] Braoudaki M, Tzortzatou-Stathopoulou F. Tumorigenesis related to retroviral infections. J Infect Dev Ctries. 2011 Nov;5(11):751-8. PubMed PMID: 22112727. Epub 2011/11/25. eng.

[16] Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008 Sep;118(9):3132-42. PubMed PMID: 18688285. Pubmed Central PMCID: 2496963. Epub 2008/08/09. eng.

[17] Cesana D, Sgualdino J, Rudilosso L, Merella S, Naldini L, Montini E. Whole transcriptome characterization of aberrant splicing events induced by lentiviral vector integrations. J Clin Invest. 2012 May 1;122(5):1667-76. PubMed PMID: 22523064. Pubmed Central PMCID: 3336994. Epub 2012/04/24. eng.

[18] Huynh D, Vincan E, Mantamadiotis T, Purcell D, Chan CK, Ramsay R. Oncogenic properties of HIV-Tat in colorectal cancer cells. Curr HIV Res. 2007 Jul;5(4):403-9. PubMed PMID: 17627503. Epub 2007/07/14. eng.

[19] Barbaro G, Barbarini G. HIV infection and cancer in the era of highly active antiretroviral therapy (Review). Oncol Rep. 2007 May;17(5):1121-6. PubMed PMID: 17390054. Epub 2007/03/29. eng.

[20] Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM. Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet. 2007 Jul 7;370(9581):59-67. PubMed PMID: 17617273. Epub 2007/07/10. eng.

[21] Decaussin G, Sbih-Lammali F, de Turenne-Tessier M, Bouguermouh A, Ooka T. Expression of BARF1 gene encoded by Epstein-Barr virus in nasopharyngeal carcinoma biopsies. Cancer Res. 2000 Oct 1;60(19):5584-8. PubMed PMID: 11034107. Epub 2000/10/18. eng.

[22] Liebowitz D. Nasopharyngeal carcinoma: the Epstein-Barr virus association. Semin Oncol. 1994 Jun;21(3):376-81. PubMed PMID: 8209269. Epub 1994/06/01. eng.

[23] Zheng ZM. Viral oncogenes, noncoding RNAs, and RNA splicing in human tumor viruses. Int J Biol Sci. 2010;6(7):730-55. PubMed PMID: 21152115. Pubmed Central PMCID: 2999850. Epub 2010/12/15. eng.

[24] Stanley MA, Winder DM, Sterling JC, Goon PK. HPV infection, anal intra-epithelial neoplasia (AIN) and anal cancer: current issues. BMC cancer. 2012;12:398. PubMed PMID: 22958276. Pubmed Central PMCID: 3488563.

[25] Parkin DM, Bray F. Chapter 2: The burden of HPV-related cancers. Vaccine. 2006 Aug 31;24 Suppl 3:S3/11-25. PubMed PMID: 16949997.

[26] Gillison ML, Castellsague X, Chaturvedi A, Goodman MT, Snijders P, Tommasino M, et al. Comparative epidemiology of HPV infection and associated cancers of the head and neck and cervix. Int J Cancer. 2013 Apr 9. PubMed PMID: 23568556.

[27] Gillison ML, Chaturvedi AK, Lowy DR. HPV prophylactic vaccines and the potential prevention of noncervical cancers in both men and women. Cancer. 2008 Nov 15;113(10 Suppl):3036-46. PubMed PMID: 18980286.

[28] Baak JP, Kruse AJ, Garland SM, Skaland I, Janssen EA, Tabrizi S, et al. Combined p53 and retinoblastoma protein detection identifies persistent and regressive cervical high-grade squamous intraepithelial lesions. Am J Surg Pathol. 2005 Aug;29(8):1062-6. PubMed PMID: 16006801. Epub 2005/07/12. eng.

[29] Popovic M, Reitz MS, Jr., Sarngadharan MG, Robert-Guroff M, Kalyanaraman VS, Nakao Y, et al. The virus of Japanese adult T-cell leukaemia is a member of the human T-cell leukaemia virus group. Nature. 1982 Nov 4;300(5887):63-6. PubMed PMID: 6982418. Epub 1982/11/04. eng.

[30] Ding W, Albrecht B, Kelley RE, Muthusamy N, Kim SJ, Altschuld RA, et al. Human T-cell lymphotropic virus type 1 p12(I) expression increases cytoplasmic calcium to enhance the activation of nuclear factor of activated T cells. J Virol. 2002 Oct;76(20):10374-82. PubMed PMID: 12239314. Pubmed Central PMCID: 136546. Epub 2002/09/20. eng.

[31] Grassmann R, Aboud M, Jeang KT. Molecular mechanisms of cellular transformation by HTLV-1 Tax. Oncogene. 2005 Sep 5;24(39):5976-85. PubMed PMID: 16155604. Epub 2005/09/13. eng.

[32] Cheng H, Ren T, Sun SC. New insight into the oncogenic mechanism of the retroviral oncoprotein Tax. Protein Cell. 2012 Aug;3(8):581-9. PubMed PMID: 22865346. Pubmed Central PMCID: 3553852. Epub 2012/08/07. eng.

[33] Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science. 2008 Feb 22;319(5866):1096-100. PubMed PMID: 18202256. Pubmed Central PMCID: 2740911. Epub 2008/01/19. eng.

[34] Shuda M, Feng H, Kwun HJ, Rosen ST, Gjoerup O, Moore PS, et al. T antigen mutations are a human tumor-specific signature for Merkel cell polyomavirus. Proc Natl Acad Sci U S A. 2008 Oct 21;105(42):16272-7. PubMed PMID: 18812503. Pubmed Central PMCID: 2551627. Epub 2008/09/25. eng.

[35] Welsh JS. Contagious cancer. Oncologist. 2011;16(1):1-4. PubMed PMID: 21212437. Pubmed Central PMCID: 3228048. Epub 2011/01/08. eng.

[36] Gugel EA, Sanders ME. Needle-stick transmission of human colonic adenocarcinoma. N Engl J Med. 1986 Dec 4;315(23):1487. PubMed PMID: 3785302. Epub 1986/12/04. eng.

[37] Gartner HV, Seidl C, Luckenbach C, Schumm G, Seifried E, Ritter H, et al. Genetic analysis of a sarcoma accidentally transplanted from a patient to a surgeon. N Engl J Med. 1996 Nov 14;335(20):1494-6. PubMed PMID: 8890100. Epub 1996/11/14. eng.

[38] Danaei G. Global burden of infection-related cancer revisited. Lancet Oncol. 2012 Jun;13(6):564-5. PubMed PMID: 22575586. Epub 2012/05/12. eng.

[39] Organisation WH. Mother-to-child transmission of HIV 2013. Available from: http://www.who.int/hiv/topics/mtct/en/.

[40] Moriuchi H, Masuzaki H, Doi H, Katamine S. Mother-to-child transmission of human T-cell lymphotropic virus type 1. Pediatr Infect Dis J. 2013 Feb;32(2):175-7. PubMed PMID: 23328821. Epub 2013/01/19. eng.

[41] van Tienen C, Jakobsen M, Schim van der Loeff M. Stopping breastfeeding to prevent vertical transmission of HTLV-1 in resource-poor settings: beneficial or harmful? Arch Gynecol Obstet. 2012 Jul;286(1):255-6. PubMed PMID: 22228317. Pubmed Central PMCID: 3374111. Epub 2012/01/10. eng.

[42] Welfare AIoHa. National Cervical Screening Program Key Statistics 2012. Available from: http://www.cancerscreening.gov.au/internet/screening/publishing.nsf/Content/facts.

[43] Frazer IH. Cervical cancer vaccine development. Sex Health. 2010 Sep;7(3):230-4. PubMed PMID: 20719210. Epub 2010/08/20. eng.

[44] NSW CC. About the B Positive Program Woolloomooloo NSW: Cancer Council NSW; 2013 [cited 2012 September 2012]. Available from: http://www.cancercouncil.com.au/52127/reduce-risks/cancer-screening-reduce-risks/b-positive-hepatitis-b-program/about-the-b-positive-program/.

[45] Blumberg BS. The discovery of the hepatitis B virus and the invention of the vaccine: a scientific memoir. J Gastroenterol Hepatol. 2002 Dec;17 Suppl:S502-3. PubMed PMID: 12534787. Epub 2003/01/22. eng.

[46] Whittle H, Jaffar S, Wansbrough M, Mendy M, Dumpis U, Collinson A, et al. Observational study of vaccine efficacy 14 years after trial of hepatitis B vaccination in Gambian children. BMJ. 2002 Sep 14;325(7364):569. PubMed PMID: 12228132. Pubmed Central PMCID: 124550. Epub 2002/09/14. eng.

[47] Ni YH, Huang LM, Chang MH, Yen CJ, Lu CY, You SL, et al. Two decades of universal hepatitis B vaccination in taiwan: impact and implication for future strategies. Gastroenterology. 2007 Apr;132(4):1287-93. PubMed PMID: 17433322. Epub 2007/04/17. eng.

[48] Harper DM. Why am I scared of HPV? CA Cancer J Clin. 2004 Sep-Oct;54(5):245-7. PubMed PMID: 15371282. Epub 2004/09/17. eng.

[49] Villa LL. Prophylactic HPV vaccines: reducing the burden of HPV-related diseases. Vaccine. 2006 Mar 30;24 Suppl 1:S23-8. PubMed PMID: 16194583. Epub 2005/10/01. eng.

[50] Lu B, Kumar A, Castellsague X, Giuliano AR. Efficacy and safety of prophylactic vaccines against cervical HPV infection and diseases among women: a systematic review & meta-analysis. BMC Infect Dis. 2011;11:13. PubMed PMID: 21226933. Pubmed Central PMCID: 3034689. Epub 2011/01/14. eng.

[51] Ageing AGDoHa. HPV School Vaccination Program Canberra2013 [cited 2012 September 2012]. Available from: http://hpv.health.gov.au/.

[52] Mattheij I, Pollock AM, Brhlikova P. Do cervical cancer data justify HPV vaccination in India? Epidemiological data sources and comprehensiveness. J R Soc Med. 2012 Jun;105(6):250-62. PubMed PMID: 22722970. Epub 2012/06/23. eng.

[53] Mocroft A, Devereux H, Kinloch-de-Loes S, Wilson D, Madge S, Youle M, et al. Immunological, virological and clinical response to highly active antiretroviral therapy treatment regimens in a complete clinic population. Royal Free Centre for HIV Medicine. AIDS. 2000 Jul 28;14(11):1545-52. PubMed PMID: 10983641. Epub 2000/09/13. eng.

[54] Vogel M, Friedrich O, Luchters G, Holleczek B, Wasmuth JC, Anadol E, et al. Cancer risk in HIV-infected individuals on HAART is largely attributed to oncogenic infections and state of immunocompetence. Eur J Med Res. 2011 Mar 28;16(3):101-7. PubMed PMID: 21486722. Pubmed Central PMCID: 3352206. Epub 2011/04/14. eng.

[55] Hermine O, Lefrere F, Bronowicki JP, Mariette X, Jondeau K, Eclache-Saudreau V, et al. Regression of splenic lymphoma with villous lymphocytes after treatment of hepatitis C virus infection. N Engl J Med. 2002 Jul 11;347(2):89-94. PubMed PMID: 12110736. Epub 2002/07/12. eng.

[56] Arora R, Shuda M, Guastafierro A, Feng H, Toptan T, Tolstov Y, et al. Survivin is a therapeutic target in Merkel cell carcinoma. Sci Transl Med. 2012 May 9;4(133):133ra56. PubMed PMID: 22572880. Epub 2012/05/11. eng.

 

Categories
Review Articles Articles

Oncolytic Virotherapy: The avant-garde approach to oncological treatment via infectious agents

Over the past twenty years, advances in translational medicine have resulted in new and exciting treatments in the area of oncology. New modalities have arisen out of the need to address existing limitations in conventional treatments such as chemotherapy and radiotherapy. What started out as an outrageous idea in the 20th century to use potentially dangerous infectious agents such as viruses to kill cancer cells has gradually evolved into a maturing field, which has the promising potential to incorporate conventional and immunological aspects of treatment within a microbial-based system. Finally, in 2006, the introduction of the world’s first approved oncolytic virus by China heralded a milestone in the clinical application of this approach. This article will examine use of oncolytic viruses in cancer treatment with emphasis on its current status and strategies, possible immune mechanism and future considerations.

Introduction

Oncolytic viruses are self-replicating viruses which can target and lyse cancer cells specifically. [1] Since the early 1900s, it was recognised that natural viral infections in cancer patients are sometimes associated with tumour regression. Indeed, case reports noted instances where influenza or measles infections in leukemia patients resulted in remissions. [2] Interest in utilising these ‘cancer-killing’ viruses peaked in the 1950-60s but the rise of chemotherapy and radiotherapy meant that progress in this field stagnated until the 1990s, when genetic engineering and better understanding of viruses and tumours revived the development of oncolytic viruses. [3] A breakthrough in the clinical translation of oncolytic viruses finally came in 2006 with the world’s first approved oncolytic virus- H101 (a genetically modified adenovirus) for head and neck cancers. [4]

Why oncolytic virotherapy?

Conventional treatments such as chemotherapy and radiotherapy have been the cornerstone of oncological management for many years. While we have achieved a considerable amount of success in many cancers, there are often criticisms against conventional treatments in terms of their limitations (e.g. transient effects against metastasis) and flaws (e.g. poor toxicity profile). [5] In recent years, gene therapy and immunotherapy have emerged as alternatives but results have been mixed. In 2002, the development of leukaemia in x-linked severe combined immunodeficiency (X-SCID) patients due to insertional mutagenesishas severely affected public confidence in gene therapy. [6] While immunotherapy remains promising, the current emphasis on specific targeting neglects the ability of tumour cells to mutate and change antigen profiles, resulting in variable clinical outcomes. [7]

In view of these insufficiencies, there has been renewed interest in oncolytic virotherapy, an interesting cross-disciplinary approach to treatment based virology, genetic engineering and immunology. The initial thinking behind this approach was simple- certain viruses exhibit tropism for cancer cells, which either express specific receptors for viral entry or lack anti-viral mechanisms that are normally intact in normal cells. [8] Once viral entry is achieved, replication of viruses continues until cell lysis occurs; allowing their progeny to infect other cancer cells.  If viral spread is homogenous, the oncolytic effect can be amplified many times and this effectively destroys the whole tumour. [8] As therapeutic genes encoding pro-apoptotic and immune effectors can be incorporated into the viral genome, an effective anti-tumour response may be initiated and magnified with each replication. [9] The ingenuity of this idea is that it exploits the infectious nature of viruses and uses it as a carrier and amplifier of other therapeutic agents. The latter may be crucial in exploiting synergistic anti-tumour effects between distinct treatment modalities.

Current status of oncolytic virotherapy

A variety of natural occurring and genetically modified viruses have been tested in clinical trials (Table 1).

Natural occurring oncolytic viruses are chosen for their low pathogenicity and inherent specificity for tumour cells. [10] Conversely, genetically modified viruses are those that are modified to promote tumour specificity, for example through use of tumour-specific promoters and gene deletions, or reduce pathogenicity by serial passage through cell culture. [8] Based on clinical data, it has been shown that virotherapy has a favourable toxicity and safety profile as compared to conventional treatment; the most common side effects being fever,flu-like symptoms and safety issues mainly concerning viral shedding and mutation-induced pathogenesis. [1,11] For the latter, dosing limitations and use of pro-drug activating suicide genes have addressed many of these issues. [1,6]

A straightforward dose-response relationship is not often observed as viral replication occurs in a heterogeneous tumour microenvironment and depends on factors such as availability of cell surface receptors and anti-viral responses. [12] Efficacy varies between different viruses but is reasonable at this early stage of development. The clinical trial for H101 reported complete remissions and partial responses in three and eleven out of forty-six patients respectively while another modified adenovirus- ONYX-15 was also used in head and neck cancer trials and achieved tumour growth stabilisation in eight out of twenty-two patients and tumour necrosis in five out of twenty-two patients.  [11,13]

It appears that limitations in efficacy were due to certain barriers. Firstly, viruses are not adept at surviving in the circulation. They are subjected to neutralising antibodies, complement and sequestration by the reticuloendothelial system. [8] In some cases, previous viral exposure may result in pre-existing anti-viral antibodies. For example, almost all individuals have antibodies to measles while reovirus infections are prevalent in about half the population. [14,15] Potent anti-viral responses such as type 1 interferons (IFNs) may also inhibit viral replication within the tumour. [8] Secondly, viruses have to endure acidotic and hypoxic conditions,transverse necrotic tumour regions and areas of poor vasculature in order to survive and infect tumour cells. [1] Thereafter, the availability of cell receptors may become a limiting factor in viral entry. [8] These obstacles are expected as viruses are foreign but this does not mean they are unsuitable therapeutic agents. On the contrary, viruses have the advantage of alerting the immune system to attack their infected target(s).

Enhancing oncolytic virotherapy via protective strategies

Protective strategies are aimed at improving delivery of viruses and avoiding viral clearance. The systemic delivery of viruses can be improved by preventing uptake of viruses by liver Kupffer cells (specialised macrophage cells). In mouse studies, viral delivery can be enhanced by clodronate-containing liposomes. [1] Clodronate is a selective macrophage-depleting agent that can temporarily inhibit viral uptake by Kuppfer cells, thereby allowing more virus particles to reach the tumour site. [1] Recent interest is focused on cell-carrier based delivery of oncolytic viruses, which aims to protect viruses from systemic and intra-tumoural barriers by packaging within a cell carrier that supports viral replication and targets tumour cells, its microenvironment or the tissue/organ in which the tumour resides. [16]

Cell-carriers targeting tumour cells include tumour-infiltrating lymphocytes (TILs) and cytokine-induced killer (CIK) cells. [16] TILs are T cells which accumulate in tumours and possess T cell receptors (TCRs) that recognise tumour-associated antigens (TAAs) in the context of major histocompatibility complex (MHC). [17] Since TILs are inherently cytotoxic to T cells, using such a carrier synergistically enhances the anti-tumour effects of oncolytic viruses. Highly-specific TAAs are rare and use of less-specific TAAs may lead to non-specific targeting of normal cells. [18] Production of TILs against TAAs is also an expensive and tedious process, which argues against its widespread clinical application. [16] Conversely, although cytotoxic lymphocytes like CIKs have a lower tumour-specificity, these cells are non-MHC dependent and can proliferate ex vivo without antigen stimulation. [18] Thorne et al. injected vaccinia virus-containing CIKs into nude mice and found that the VV/CIK combination was able to accurately target tumour cells and also improved the survival rate of mice as compared to VV administration alone. [19] To improve specificity, Yoon et al. engineered Her-2/neu expressing CIKs which can target ovarian cancer cells in nude mice with high affinity. Results suggest that this approach was more effective in killing cancer cells than administering Herceptin alone. [20] Nonetheless, mechanisms underlying the tumour-specificity of CIKs remain unclear and should be studied further.

In comparison, cell carriers targeting the tumour microenvironment have well-studied. Examples include mesenchymal stem cells (MSCs) and tumour-associated macrophages (TAMs). [18] MSCs are often attracted by inflammatory chemokines expressed in the microenvironment while TAMs tend to accumulate in hypoxic areas and regions of chronic inflammation in the tumour. [16] Mader et al. reported that in mouse studies, intra-peritoneal injection of a measles virus-MSC combination can prolong the survival period of mice with ovarian cancer. [21] Similarly, a clinical study found that intravenous injection of autologous MSCs carrying the modified adenovirus-ICOVIR 5 into four children with metastatic neuroblastoma was found to induce a complete clinical response in one child who also achieved complete remission within 3 years. [22] However, as MSCs are potentially tumourigenic, there is a trade-off between exploiting its propensity for tumour accumulation and the risk of enhancing tumour growth. The main criticism against targeting the tumour microenvironment relates to the inability of these cell carriers to deliver viruses directly into tumour cells. However, modifying the microenvironment through the engineering of viruses containing genes encoding pro-apoptotic and pro-inflammatory cytokines may circumvent this limitation by disrupting tumour-promoting interactions between the microenvironment and cancer cells. [16] Furthermore, the administration of proteases such as relaxin to degrade the extracellular matrix or fusogenic membrane glycoproteins to promote cell-to-cell fusion before oncolytic therapy may facilitate intratumoural spread of viruses. [23,34]

Lastly, the targeting of tumour-associated tissues and organs is also achieved by carriers such as dendritic cells (DCs) and peripheral blood lymphocytes (PBLs). [18] These carriers are attractive because they circulate through lymphoid organs such as the lymph nodes and spleen, which are sites of micrometastases and T-cell priming. [16] DC or PBL mediated delivery of VSV and reoviruses have been shown to purge metastases in lymphoid organs. Qiao et al. found that a VSV/PBL combination partially purged B16 metastases in mice 2-3 days after administration. [25] The oncolysis of metastatic cells by VSV also primed anti-tumour T cell responses effectively and probably contributed to fast purging. [16] Targeting of tissues/organs is the least specific but this negates the requirement for highly-specific tumour markers. In addition, the circulatory paths of these cell carriers are well-characterised, allowing better prediction of their tumour trafficking patterns. [18]

The mechanisms of viral loading, amplification and transfer are equally important in enhancing cell-carrier based strategies. Willmon et al. suggested that the loading of viruses depends on the multiplicity of infection (MOI), which is the ratio of infectious agent to infection target (i.e. cell carrier). [16] In high MOI loading, a higher viral loading density may be achieved but many viral particles will be stuck to the cell’s external surface and become susceptible to neutralising antibodies. [26] Conversely, in low MOI loading, most viral particles will be internalised although the viral loading density may be lower. [26] This approach may help avoid neutralisation and is suitable for individuals who have pre-existing antibodies against the oncolytic virus (e.g. measles and reovirus).

The carrier’s ability to support viral replication determines the amount of virus delivered. As viral replication can be affected by intact IFN responses in normal cells and also requires synchronous timing with carrier bursting, tumour cells have been implicated as possible carriers. [27] A successful example has been shown in the use of VSV-infected carcinoma cells to target lung metastases in mice but safety issues concerning the tumourigenity of tumour cell-based carriers remain. [28]

The transfer of virus from cell carrier to tumour cells is crucial as exposed viral particles are susceptible to neutralisation. In some viruses such human immunodeficiency virus (HIV), viral spread is mediated by a virological synapse (a specialised form of immunological synapse) between TCRs on T cells and MHC on adjacent cells. [29] By identifying viruses which utilises a virological synapse, oncolytic viruses can transfer safely between cells.

Besides cell-carrier based strategies, immunosuppression has been considered as a means of inhibiting anti-viral immunity. In rat glioma models, cyclophosphamide and cyclosporin A (CPA) have been shown to enhance HSV-mediated oncolysis by inhibiting tumour-mediated phagocyte infiltration. [30] However, recent studies suggest that such agents can be immunostimulatory and there is increasing recognition that anti-viral immunity also contribute to effective anti-tumour responses; implying that immune mechanisms of oncolytic virotherapy may need to be examined further. [31]

Immune mechanisms of oncolytic virotherapy

The direct oncolytic effects of oncolytic virotherapy are well appreciated. Successful infection and efficient spread of oncolytic viruses determine the extent of tumour lysis; leading to emphasis on developing viruses that replicated robustly and extensively. [31] However, the lack of a straightforward dose-response relationship suggests that other oncolytic mechanisms are present. The immune system may play paradoxical roles in enhancing or impeding anti-tumour responses mediated by oncolytic viruses. [32]

Innate immune responses have been shown to inhibit viral replication in rat glioma models as indicated by rapid decrease in HSV/VV titers with concomitant increase in natural killer (NK) cell infiltration following oncolytic virotherapy. [30,33] However, viral-mediated recruitment of NK cells is advantageous as NK cells are cytotoxic and associated with tumour regression. NK cells and DCs are also involved in reciprocal interactions. [31] In vitro experiments involving Mel888 melanoma cell lines showed that reovirus-infected DCs induced IFN-β production, which in turn activated NK cells. [34] Activation of NK cells resulted in cytotoxic effects against Mel888 cells and reciprocal maturation of DCs. [34] As DCs are involved in antigen presentation to T cells, DC maturation may also promote adaptive anti-tumour responses. However, DC functions are virus-dependent as studies showed that wild-type measles and adenoviruses are inhibitory and neutral respectively. [31] It appears that the timing of viral clearance is crucial and prolonging this therapeutic window by immunosuppression may be beneficial. This is because some immunosuppressive agents may suppress anti-viral responses while stimulating anti-tumour responses. For example, similar to HSV, rat glioma studies indicate that CPA may promote VV replication while inducing a cytokine storm, which enhances activity of tumour-associated cytotoxic lymphocytes. [35]

Adaptive anti-tumour responses may be shaped by two models of immune activation: the infectious non-self (INS) and ‘danger’ models. [31] The former refers to the provision of pathogen-associated molecular patterns (PAMPs) such as viral nucleic acids to pattern-recognition receptors (e.g. toll-like receptors) on antigen-presenting cells (APCs) while the latter refers to the release of endogenous ‘danger’ signals such TAAs to APCs. [31] In the INS model, the presence of viral PAMPs induces activation and proliferation of antibodies and T cells upon antigen presentation. Infection of tumour cells is therefore not a pre-requisite for anti-tumour responses, which may instead be due to bystander effects of anti-viral responses. This was illustrated by Breitbach et al. in a murine colorectal cancer model whereby administration of HSV and VV infected only a small number of cancer cells but triggered massive destruction of non-infected cancer cells. [36] Conversely, the ‘danger’ model is more in line with oncolysis of tumour cells. Greiner et al. showed that an attenuated VV was capable of lysing human melanoma cells with subsequent development of an anti-TAA response. [37] These two models are not mutually exclusive, suggesting that the actual anti-tumour effect may be mediated by both, with their relative contributions dependent on the immunogenicity of the oncolytic virus or the tumour. [31] It is therefore apparent from an immunological perspective that effective oncolyic virotherapy may capitalise on the use of highly immunogenic viruses in a bystander effect or alternatively, promoting efficient anti-TAA responses via engineering of TAA-expressing viral vectors in poorly immunogenic viruses.  [31,38]

The mechanisms involved in oncolytic virotherapy are summarised in Figure 1.

Future work

The current development of oncolytic virotherapy is based on rational designing but this approach may not always lead to the most selective and potent viruses. Buazon and Hermiston suggested that directed evolution might help researchers identify viruses with these desired characteristics. This involves growing diverse viruses in conditions that enhance diversity and passaging them through conditions mimicking the tumour microenvironment. [39] Application of directed evolution to colon cancer cell lines resulted in the adenovirus ColoAd1, which was 2-3 logs more potent than the advanced ONYX-15 and also had a therapeutic window 3-4 logs greater than the standard Ad5. [4,40] Furthermore, ColoAd1 is more sensitive to the anti-viral cidofovir (CDV) than either of its parents (Ad11p and Ad3) and this was due to directed evolution. [41] Thus, such an approach has a promising safety profile.

Integrating oncolyic virotherapy with existing treatments must be considered. Current studies have indicated promising results with radiotherapy and chemotherapy. A combination of modified HSV and radiotherapy have shown additive cell-killing effects in colorectal cancer assays and increased tumour regression in human glioma xenograft models. [42,43] It was suggested that radiotherapy could have improved viral replication and spread through irradiation-induced cellular changes. Similarly, pre-clinical studies have indicated that combination of ONXY-15 with cisplatin and 5-flurouracil in oesophageal cancers had a 39% increase in response rate as compared to chemotherapy alone (79% versus 40%). [44] Although both radiotherapy and chemotherapy are immunosuppressive to some extent, synergistic effects can be achieved by selecting the right virus and radiation/drug dosage. Combination of immunotherapy and virotherapy is at an early stage but success has been seen with VSV therapy combined with IL-2 and regulatory T cell (Treg) depletion in terms of enhanced NK cell activity and increased viral delivery. [45]

In the short term, the conflict between intra-tumoural and intravenous/intra-peritoneal administration of oncolytic viruses needs to be resolved. Mastrangelo et al showed that intra-tumoural injection of a GM-CSF-modified VV was able to induce regression in distant non-injected metastatic sites in melanoma patients but systemic effects were absent in intra-tumoural injections of ONYX-15 or reovirus. [46] Intravenous administration of the latter viruses was efficacious but resulted in thrombocytopenia and transaminitis respectively. [47] Therefore, when deciding the route of administration, a clear clinical endpoint must be established (targeting primary tumour or metastatic sites) and this will guide the type of virus used and effects (beneficial and detrimental) observed, and eventually how the patient is managed.

To conclude, oncolytic virotherapy has its antecedent in early observations and experiments detailing viral-mediated tumour regressions. Despite being neglected for decades, its resurgence reflects a current trend towards exploring new oncological treatments and a genuine hope that it can deliver better clinical outcomes.  Encouraging early results and the increasing availability of solutions to its problems suggest that it is well-poised to be the avant-garde of next-generation cancer therapeutics.

Conflict of interest

None declared.

Correspondence

K Ho: koho2292@uni.sydney.edu.au

References

[1] Prestwich RJ, Errington F, Harrington KJ, Pandha HS, Selby P, Melcher A. Oncolytic Viruses:  Do they have a role in anti-cancer therapy? Clin Med Oncol 2008; 2:83-96.

[2] Pelner L, Fowler GA, Nauts HC. Effects of concurrent infections and their toxins on the course of leukemia. Acta Med Scand Suppl 1958; 338: 1–47.

[3] Kelly E, Russell SJ. History of Oncolytic Viruses: Genesis to Genetic Engineering. Mol Ther 2007; 15(4):651-9.

[4] Garber K. China approves world’s first oncolyic virus therapy for cancer treatment. J Natl Cancer Inst 2006; 98:298-300.

[5] Gavhane YN, Shete AS, Bhagat AK, Shinde VR, Bhong KK, Khairnar GA, et al. Solid Tumours: Facts, Challenges and Solutions. IJPSR 2011; 2(1):1-12.

[6] McCormick F. Future prospects for oncolytic therapy. Oncogene 2005; 24:7817-9.

[7] Waldmann TA. Immunotherapy: Past, Present and Future.  Nat Med 2003; 9(3):269-77.

[8] Wong HH, Lemonine NR, Wang Y. Oncolytic viruses: Overcoming the Obstacles. Viruses 2010; 2(1):78-106.

[9] Chen L, Chen D, Gong M, Na M, Li L, Wu H, et al. Concomitant use of Ad5/35 chimeric oncolytic adenovirus with TRAIL gene and taxol produces synergistic cytotoxicity in gastric cancer cells. Cancer Lett 2009; 284:141-8.

[10] Cervantes-Garcia D, Ortiz-Lopez R, Mayek-Perez N, Rojas-Martinez A. Oncolytic virotherapy. Ann Hepatol 2008; 7(1):34-45.

[11] Lu W, Zheng S, Li XF, Huang JJ, Zheng X, Li Z. Intra-tumour injection of H101, a recombinant adenovirus, in combination with chemotherapy in patients with advanced cancers: a pilot phase II clinical trial. World J Gastroenterol 2004; 10(24):3634-8.

[12] Aghi M, Martuza RL. Oncolytic viral therapies-the clinical experience. Oncogene 2005; 24:7802-16.

[13] Ganly I, Kirn D, Eckhardt G, Rodriguez GI, Soutar DS, Otto R, et al. A phase I study of Onyx-015, an E1B attenuated adenovirus, administered intratumourally to patients with recurrent head and neck cancer. Clin Cancer Res 2000; 6(3):798-806.

[14] Tyler KL. Mammalian reoviruses. In Lippencott Fields Virology, Williams and Wilkens: Philidelphia; 2001.

[15] Iankov ID, Blechacz B, Liu C, Schmeckpeper JD, Tarara JE, Federspiel MJ, et al. Infected cell carriers: a new strategy for systemic delivery of oncolytic measles in cancer virotherapy. Mol Ther 2007; 15:114-22.

[16] Wilmon C, Harrington K, Kottke T, Prestwich R, Melcher A,Vile R. Cell carriers for oncolytic viruses: Fed Ex for cancer therapy. Mol Ther 2009; 17(10):1667-76.

[17] Goff SL, Smith FO, Klapper JA, Sherry R, Wunderlich JR, Steinberg SM, et al. Tumour infiltrating lymphocyte therapy for metastatic melanoma: analysis of tumours resected for TIL. J Immunother 2010; 33(8):840-7.

[18] Su X, Zhang L. Advances in cell carriers for oncolytic viruses in cancer therapy. Tumour 2011; 31(1):85-8.

[19] Thorne SH, Negrin RS, Contag CH. Synergistic antitumour effects of immune cell-viral biotherapy. Science 2006; 331(5768):1780-84.

[20] Yoon SH, Lee JM, Woo SJ, Park MJ, Park SJ, Kim HS, et al. Transfer of Her-2/neu specificity into cytokine-induced killer (CIK) cells with RNA encoding chimeric immune receptor (CIR). J Clin Immunol 2009; 29(6):806-14.

[21] Mader EK, Maeyama Y, Lin Y, Butler GW, Russell HM, Galanis E, et al. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res 2009; 15(23):7246-55.

[22] Garcia-Castro J, Alemany R, Cascallo M, Martinez-Quintanilla J, Arriero-Mdel M, Lassaletta A, et al. Treatment of metastatic neuroblastoma with systemic oncolyic virotherapy delivered by autologous mesenchymal stem cells: an exploratory study. Cancer Gene Ther 2010; 17(7):476-83.

[23] Kim JH, Lee YS, Kim H, Huang JH, Yoon AR, Yun CO. Relaxin expression from tumour-targeting adenoviruses and its intra-tumoural spread, apoptosis induction, and efficacy. J Natl Cancer 2006; 98:1482-93.

[24] Bateman A, Bullough F, Murphy S, Emiliusen L, Lavillette D, Cosset FL, et al. Fusogenic membrane glycoproteins as a novel class of genes for the local and immune-mediated control of tumour growth. Cancer Res 2000; 60:1492-7.

[25] Qiao J, Kottke T, Willmon C, Galivo F, Wongthida P, Diaz RM, et al. Purging metastases in lymphoid organs using a combination of antigen-nonspecific adoptive T cell therapy,oncolytic virotherapy and immunotherapy. Nat Med 2008; 14(1):37-44.

[26] Kottke T, Diaz RM, Kaluza K, Pulido J, Galivo F, Wongthida P, et al. Use of biological therapy to enhance both virotherapy and adoptive T-cell therapy for cancer. Mol Ther 2008; 16:1910-8.

[27] Ottolino-Perry K, Diallo JS, Lichty BD, Bell JC, McCart JA. Intelligent design: combination therapy with oncolytic viruses. Mol Ther 2010; 18(2):251-63.

[28] Garcia-Castro J, Martinez-Palacio J, Lillo R, Garcia-Sanchez F, Alemany R, Madero L, et al. Tumour cells as cellular vehicles to deliver gene therapies to metastatic tumours. Cancer Gen Ther 2005; 12(4):341-9.

[29] Groot F, Welsch S, Sattentau QJ. Efficient HIV-1 transmission from macrophages to T cells across transient virological synapses. Blood 2008; 111:4660-3.

[30] Fulci G, Breymann L, Gianni D, Kurozomi K, Rhee SS, Yu J, et al. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc Natl Acad Sci 2006; 103:12873-8.

[31] Prestwich RJ, Errington F, Diaz RM, Pandha HS, Harrington KJ, Melcher AA, et al. The Case of Oncolytic Viruses Versus the Immune System: Waiting on the Judgment of Solomon. Hum Gene Ther 2009; 20(10):1119-32.

[32] Prestwich RJ, Harrington KJ, Pandha HS, Vile RG, Melcher AA, Errington F. Oncolytic viruses: a novel form of immunotherapy. Expert Rev Anticancer Ther 2008; 8(10):1581-8.

[33] Lun XQ, Jang JH, Tang N, Deng H, Head R, Bell JC, et al. Efficacy of systemically administered oncolytic vaccinia virotherapy for malignant gliomas is enhanced by combination therapy with rapamycin or cyclophosphamide. Clin Cancer Res 2009; 15:2777-88.

[34] Prestwich RJ, Errington F, Ilett EJ, Morgan RS, Scott KJ, Kottke T, et al. Tumour infection by oncolytic reovirus primes adaptive anti-tumour immunity. Clin Cancer Res 2008; 14:7358-66.

[35] Brentjens RJ, Riviere I, Hollyman D, Taylor C, Nikhamin Y, Stefanski J, et al. Unexpected Toxicity of Cyclophosphamide Followed by Adoptively Transferred CD19-Targeted T Cells in a Patient with Bulky CLL. Molecular Therapy. 2009; 17(Suppl 1):S157.

[36] Breitbach CJ, Paterson JM, Lemay CG, Falls TJ, Mcguire A, Parato KA, et al. Targeted inflammation during oncolytic virus therapy severely compromises tumour blood flow. Mol Ther 2007; 15:1686-93.

[37] Greiner S, Humrich JY, Thuman P, Sauter B, Schuler G, Jenne L. The highly attenuated vaccinia virus strain modified virus Ankara induces apoptosis in melanoma cells and allows bystander dendritic cells to generate a potent anti-tumoural immunity. Clin Exp Immunol 2006; 146:344-53.

[38] Diaz RM, Galivo F, Kottke T, Wongthida P, Qiao J, Thompson J, et al. Oncolytic immunovirotherapy for melanoma using vesicular stomatitis virus. Cancer Res 2007; 67:2840-8.

[39] Bauzon M, Hermiston TW. Oncolytic Viruses: The Power of Directed Evolution. Adv Viro 2012; doi:10.1155/2012/586389.

[40] Kuhn I, Harden P, Bauzon M, Chartier C, Nye J, Thorne S, et al. Directed evolution generates a novel oncolytic virus for the treatment of cancer treatment. PLoS ONE 2008; 3(6): e2409. doi:10.1371/journal.pone.0002409.

[41] Bauzon M, Jin F, Kretschmer P, Hermiston T. In vitro analysis of cidofovir and genetically engineered TK expression as potential approaches for the intervention of ColoAd1-based treatment of cancer. Gene Ther 2009; 16(9):1169-74.

[42] Stanziale SF, Petrowsky H, Joe JK, Roberts GD, Zager JS, Gusani NJ, et al. Ionizing radiation potentiates the antitumour efficacy of oncolytic herpes simplex virus G207 by upregulating ribonucleotide reductase. Surgery 2002; 132:353-9.

[43] Advani SJ, Sibley GS, Song PY, Hallahan DE, Kataoka Y, Roizman B. Enhancement of replication of genetically engineered herples simplex viruses by ionizing radiation: a new paradigm for destruction of therapeutically intractable tumours. Gene Ther 1998; 5:160-5.

[44] Heise C, Sampson-Johannes A, Williams A, McCormick F, Von Hoff DD, Kim DH. ONYX-15, an E1B gene-attenuated adenovirus, causes tumour-specific cytolysis and antitumoural efficacy that can be augmented by standard chemotherapeutic agents. Nat Med 1997; 3:639-45.

[45] Kottke T, Galivo F, Wongthida P, Diaz RM, Thompson J, Jevremovic D, et al. Treg depletion-enhanced IL-2 treatment facilitates therapy of established tumours using systemically delivered oncolytic virus. Mol Ther 2008; 16:1217-26.

[46] Mastrangelo MJ, Maguire HC Jr, Eisenlohr LC, Laughlin CE, Monken CE, Mccue PA, et al. Intratumoural recombinant GM-CSF-encoding virus as gene therapy in patients with cutaneous melanoma. Cancer Gene Ther 1999; 6:409-22.

[47] Liu TC, Kirn D. Systemic efficacy with oncolytic virus therapeutics: clinical proof-of-concept and future directions. Cancer Res 2007; 67(2):429-32.