Tuesday, April 22, 2025

IgA Vasculitis in Adults

 

IgA Vasculitis in Adults: Current Perspectives on Diagnosis and Management

Dr Neeraj Manikath ,claude.ai

Abstract

IgA vasculitis (IgAV), formerly known as Henoch-Schönlein purpura, is a small-vessel vasculitis characterized by IgA1-dominant immune deposits. While primarily considered a pediatric condition, adult-onset IgAV presents unique diagnostic and therapeutic challenges due to its lower incidence, more severe manifestations, and poorer outcomes compared to childhood cases. This review summarizes current evidence on adult IgAV, with particular focus on epidemiology, pathophysiology, clinical manifestations, diagnostic criteria, and management strategies. Recent advances in biomarkers, imaging modalities, and novel therapeutic approaches are also discussed, providing rheumatologists with a comprehensive and updated framework for managing this complex vasculitis in adult patients.

Introduction

IgA vasculitis (IgAV) is a small-vessel leukocytoclastic vasculitis characterized by deposition of IgA1-dominant immune complexes in vessel walls[1]. It predominantly affects children, with an incidence of 10-20 cases per 100,000 population annually[2]. In contrast, adult-onset IgAV is relatively rare, with approximately 0.8-1.8 cases per 100,000 adults per year[3].


While historically known as Henoch-Schönlein purpura (HSP), the nomenclature was revised by the 2012 Chapel Hill Consensus Conference (CHCC) to reflect the pathophysiological mechanism of the disease[4]. Despite its lower incidence in adults, IgAV in this population presents with more severe clinical manifestations and poorer outcomes, particularly regarding renal involvement, making it a significant concern for practicing rheumatologists[5,6].


This review aims to provide an updated perspective on the diagnosis and management of adult IgAV, integrating recent advances in understanding disease mechanisms, biomarkers, and therapeutic strategies to guide evidence-based clinical practice.

Epidemiology and Risk Factors

The incidence of adult IgAV varies geographically, with higher rates reported in Asian populations compared to Western countries[7]. Unlike the pediatric population, where males are predominantly affected with a male-to-female ratio of 1.5:1, adult IgAV shows a more balanced gender distribution[8]. The peak incidence occurs in adults aged 20-40 years, though cases have been reported across all age groups[9].


Several triggers have been identified in adult IgAV:


  1. Infections: Upper respiratory tract infections precede approximately 30-50% of adult cases, with Streptococcus species being the most commonly implicated pathogen[10].


  1. Medications: Various drugs have been associated with IgAV, including antibiotics (particularly beta-lactams), non-steroidal anti-inflammatory drugs (NSAIDs), and angiotensin-converting enzyme inhibitors (ACEIs)[11].


  1. Malignancies: Adult IgAV has a stronger association with malignancy compared to pediatric cases. Solid tumors, particularly of the lung, prostate, and gastrointestinal tract, as well as hematological malignancies, have been reported in 5-10% of adult patients[12,13].


  1. Genetic factors: HLA-DRB101 and HLA-B41 alleles have been associated with increased susceptibility to IgAV in adults, though these associations are less well-established than in pediatric populations[14].

Pathophysiology

The pathogenesis of IgAV involves aberrant production and glycosylation of IgA1, formation of immune complexes, and subsequent vascular inflammation. Recent advances have expanded our understanding of these mechanisms:

Aberrant IgA1 Production and Glycosylation

IgAV is characterized by elevated serum levels of galactose-deficient IgA1 (Gd-IgA1). This abnormally glycosylated IgA1 contains reduced O-linked galactose in the hinge region, exposing N-acetylgalactosamine (GalNAc) residues[15]. Recent research has identified dysregulation of several glycosyltransferases, particularly C1GALT1 (core 1 β1,3-galactosyltransferase) and its chaperone Cosmc, in patients with IgAV[16].

Immune Complex Formation

The exposed GalNAc residues on Gd-IgA1 are recognized by naturally occurring anti-glycan IgG and IgA autoantibodies, leading to the formation of circulating immune complexes[17]. These complexes have reduced clearance by the hepatic asialoglycoprotein receptor and increased affinity for mesangial cells and endothelial surfaces[18].

Vascular Inflammation

Deposition of immune complexes in vessel walls activates the complement system, particularly the alternative and lectin pathways, resulting in the recruitment of inflammatory cells and release of pro-inflammatory cytokines[19]. Recent studies have highlighted the role of neutrophil extracellular traps (NETs) in amplifying vascular damage in IgAV[20].

Novel Pathways

Emerging evidence suggests involvement of the interleukin-17/23 axis and dysregulation of regulatory T cells in the pathogenesis of IgAV[21]. Additionally, alterations in the gut microbiome have been implicated, with some studies reporting increased intestinal permeability and bacterial translocation preceding the onset of disease manifestations[22].

Clinical Manifestations

Adult IgAV typically presents with the classic tetrad of palpable purpura, arthralgia/arthritis, abdominal pain, and renal involvement, though the clinical spectrum varies widely. Important distinctions between adult and pediatric presentations include:

Cutaneous Manifestations

Palpable purpura occurs in nearly all (95-100%) adult patients, predominantly affecting the lower extremities and buttocks[23]. Unlike in children, adults more frequently develop severe skin lesions, including bullous, necrotic, and ulcerative changes[24]. Edema is common, particularly in the distal extremities and areas with dependent positioning.

Gastrointestinal Involvement

Gastrointestinal symptoms affect 50-85% of adult patients and range from mild abdominal discomfort to severe complications[25]. Adults exhibit higher rates of serious gastrointestinal complications compared to children, including:


  • Massive gastrointestinal bleeding (10-20%)

  • Intussusception (2-3%)

  • Bowel perforation (1-2%)

  • Pancreatitis (rare but reported)[26,27]


Endoscopic findings typically reveal erythematous, petechial, or ulcerative lesions primarily affecting the second portion of the duodenum[28].

Renal Involvement

Renal manifestations occur in 45-85% of adult patients and represent the primary determinant of long-term prognosis[29]. Compared to children, adults demonstrate:


  • Higher rates of nephrotic-range proteinuria (20-35% vs. 5-15%)

  • Greater frequency of nephritic syndrome (15-20% vs. 5-10%)

  • Increased risk of progression to end-stage renal disease (5-15% vs. <1%)[30,31]


Microscopic hematuria is the most common urinary abnormality (80-90%), followed by proteinuria (70-80%)[32].

Articular Involvement

Arthralgia and/or arthritis affect 60-70% of adult patients, typically presenting as non-erosive, non-deforming inflammation predominantly affecting the knees and ankles[33]. Compared to children, adults more frequently experience prolonged arthritis (>2 weeks) and higher rates of persistent joint symptoms[34].

Other Manifestations

Less common but important manifestations in adults include:


  1. Neurological involvement: Reported in 5-10% of adult cases, ranging from headache and behavioral changes to more severe manifestations like seizures, posterior reversible encephalopathy syndrome (PRES), and cerebral vasculitis[35].


  1. Pulmonary involvement: Diffuse alveolar hemorrhage, interstitial lung disease, and pleural effusions have been reported in 0.5-5% of adult cases[36].


  1. Cardiac involvement: Myocarditis, pericarditis, and coronary arteritis are rare but potentially life-threatening complications[37].


  1. Orchitis: Affects approximately 2-5% of adult male patients and can lead to testicular infarction if not properly managed[38].

Diagnosis

The diagnosis of IgAV in adults remains primarily clinical, supported by laboratory findings and histopathology when necessary.

Diagnostic Criteria

Several classification criteria have been proposed for IgAV, including the American College of Rheumatology (ACR) 1990 criteria[39] and the European League Against Rheumatism/Paediatric Rheumatology International Trials Organisation/Paediatric Rheumatology European Society (EULAR/PRINTO/PRES) 2010 criteria[40]. However, these were primarily developed for pediatric populations.


More recently, a 2021 international consensus proposal specifically addressed diagnostic criteria for adult IgAV, including:


Major criteria:


  • Palpable purpura or petechiae with lower limb predominance in the absence of thrombocytopenia

  • Histopathology showing leukocytoclastic vasculitis with IgA deposits


Minor criteria:


  • Arthralgia or arthritis

  • Renal involvement (hematuria and/or proteinuria)

  • Gastrointestinal involvement (abdominal pain or gastrointestinal bleeding)

  • Recent respiratory tract infection or other identified trigger


The diagnosis of adult IgAV requires either the presence of both major criteria or one major criterion plus at least two minor criteria[41].

Laboratory Investigations

No specific laboratory test confirms the diagnosis of IgAV. Common findings include:


  • Normal or elevated inflammatory markers (ESR, CRP)

  • Normal platelet count (crucial to distinguish from thrombocytopenic purpura)

  • Elevated serum IgA levels (40-50% of cases)

  • Normal complement levels (distinguishing from other immune complex vasculitides)

  • Urinalysis and kidney function tests to assess renal involvement[42]

Histopathology

Skin biopsy showing leukocytoclastic vasculitis with perivascular IgA deposition by direct immunofluorescence remains the gold standard for diagnosis when clinical presentation is atypical[43]. In adult patients with significant renal involvement, kidney biopsy may be necessary to determine the extent and class of renal damage, which influences therapeutic decisions.


The histopathological classification of IgA nephropathy (Oxford classification) has been adapted for IgAV nephritis, evaluating:


  • Mesangial hypercellularity (M)

  • Endocapillary proliferation (E)

  • Segmental glomerulosclerosis (S)

  • Tubular atrophy/interstitial fibrosis (T)

  • Crescents (C)[44]

Differential Diagnosis

The differential diagnosis of adult IgAV includes:


  • Other primary vasculitides (ANCA-associated vasculitis, cryoglobulinemic vasculitis)

  • Secondary vasculitides (drug-induced, infection-related)

  • Thrombotic microangiopathies

  • Other causes of purpura (thrombocytopenia, coagulation disorders)

  • Inflammatory bowel disease (when gastrointestinal symptoms predominate)[45]


In adults, particularly those over 50 years, thorough evaluation for underlying malignancy is warranted given the established association[46].

Biomarkers

Several potential biomarkers for disease activity and prognosis have been investigated:


  1. Serum Gd-IgA1 levels: Correlate with disease activity and risk of renal involvement[47].


  1. Urinary biomarkers: Including neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), and monocyte chemoattractant protein-1 (MCP-1), which may predict severity of renal involvement[48].


  1. Genetic markers: Polymorphisms in genes encoding cytokines (IL-1, IL-6, TNF-α) and adhesion molecules have been associated with disease susceptibility and severity[49].


  1. Complement activation products: Particularly those of the alternative pathway (C3a, Bb fragment), which correlate with disease activity[50].

Treatment

Management of adult IgAV remains challenging due to the lack of large randomized controlled trials. Treatment approaches vary based on disease severity and organ involvement.

Mild Disease

For patients with isolated cutaneous and mild joint involvement:


  • Supportive care with rest and adequate hydration

  • NSAIDs for arthralgia (with caution in patients with renal involvement)

  • Antihistamines for pruritus associated with cutaneous lesions

  • Monitoring for development of systemic complications[51]

Moderate-to-Severe Disease Without Significant Renal Involvement

For patients with significant cutaneous, gastrointestinal, or joint manifestations:


  1. Corticosteroids: Prednisone or prednisolone (0.5-1 mg/kg/day for 2-4 weeks with subsequent tapering) has been shown to alleviate symptoms and possibly reduce the risk of gastrointestinal complications, though evidence for long-term benefit is limited[52].


  1. Colchicine: Some studies have reported efficacy in treating cutaneous manifestations (0.5-1.5 mg/day), particularly in cases refractory to corticosteroids[53].


  1. Dapsone: May be effective for chronic or relapsing cutaneous disease (50-150 mg/day), acting through inhibition of neutrophil recruitment and function[54].

Severe Disease With Significant Renal Involvement

For patients with nephrotic-range proteinuria, rapidly progressive glomerulonephritis, or other severe manifestations:


  1. High-dose corticosteroids: Initial treatment typically involves methylprednisolone pulses (0.5-1 g/day for 3 days) followed by oral prednisone (1 mg/kg/day)[55].


  1. Cyclophosphamide: Used in combination with corticosteroids for severe renal involvement, particularly in the presence of crescentic glomerulonephritis (2 mg/kg/day orally or 0.5-1 g/m² IV monthly)[56].


  1. Rituximab: Emerging evidence supports the use of rituximab (375 mg/m² weekly for 4 weeks or two 1-g doses two weeks apart) in refractory cases, particularly those with severe renal involvement[57,58].


  1. Plasma exchange: May be considered in rapidly progressive glomerulonephritis or life-threatening gastrointestinal or pulmonary hemorrhage, though evidence is mainly derived from small case series[59].


  1. Mycophenolate mofetil: Used as steroid-sparing agent (1-2 g/day) in maintenance therapy after induction with more potent immunosuppressants[60].

Novel and Emerging Therapies

Several targeted therapies have shown promise in small studies or case reports:


  1. IVIG: High-dose intravenous immunoglobulin (2 g/kg divided over 2-5 days) may be effective in refractory cases, potentially through immunomodulation and inhibition of complement activation[61].


  1. Biologics targeting TNF-α: Case reports suggest potential benefit of adalimumab and infliximab in refractory disease[62].


  1. IL-17/IL-23 pathway inhibitors: Given emerging evidence of the role of the Th17 pathway in pathogenesis, secukinumab and ustekinumab have been explored in small case series with promising results[63].


  1. Complement inhibitors: Eculizumab has been reported to be effective in cases with prominent complement activation[64].


  1. JAK inhibitors: Tofacitinib and baricitinib have been reported in case series for refractory IgAV with encouraging outcomes[65].

Specific Management Considerations

Renal Involvement

The management of renal involvement depends on histopathological findings and clinical severity:


  • Mild mesangial proliferation: ACE inhibitors or angiotensin receptor blockers for proteinuria >0.5 g/day

  • Moderate proliferative disease: Corticosteroids with or without mycophenolate mofetil

  • Severe proliferative disease or crescentic glomerulonephritis: Cyclophosphamide or rituximab plus corticosteroids[66]


A 2020 meta-analysis suggested that immunosuppressive therapy in adult IgAV nephritis may reduce proteinuria and stabilize renal function, but did not demonstrate definitive long-term benefit in preventing end-stage renal disease[67].

Gastrointestinal Involvement

Management of gastrointestinal manifestations includes:


  • Corticosteroids for moderate-to-severe abdominal pain

  • Proton pump inhibitors for gastric protection

  • Careful monitoring for complications requiring surgical intervention (intussusception, perforation)

  • Nutritional support in cases of prolonged symptoms[68]

Special Populations

Pregnancy

IgAV during pregnancy is rare but associated with increased risk of maternal and fetal complications. Management includes:


  • Close monitoring of blood pressure and renal function

  • Minimizing teratogenic medications (cyclophosphamide, mycophenolate)

  • Preference for corticosteroids, azathioprine, or tacrolimus when immunosuppression is necessary

  • Multidisciplinary approach involving rheumatology, nephrology, and obstetrics[69]

Elderly Patients

In elderly patients (>65 years), considerations include:


  • Higher vigilance for underlying malignancy

  • Careful monitoring for medication toxicity

  • Dose adjustment of immunosuppressants according to renal function

  • Prophylaxis against opportunistic infections during immunosuppressive therapy[70]

Prognosis and Follow-up

The prognosis of adult IgAV is generally less favorable compared to pediatric cases, particularly regarding renal outcomes.

Prognostic Factors

Factors associated with poor prognosis include:


  1. Age >50 years

  2. Severe renal involvement at presentation (creatinine >1.5 mg/dL, nephrotic syndrome)

  3. Histological findings: presence of crescents in >50% of glomeruli, significant interstitial fibrosis

  4. Persistent proteinuria >1 g/day after 6 months

  5. Hypertension

  6. Gastrointestinal bleeding[71,72]

Long-term Outcomes

Long-term outcomes in adult IgAV include:


  • Renal outcomes: 5-15% of adults develop end-stage renal disease within 10 years

  • Relapse rates: 20-30% experience at least one relapse within 5 years

  • Mortality: 5-year survival rates of 90-95%, with most deaths attributed to renal failure, infections, or cardiovascular events[73,74]

Monitoring and Follow-up

A structured follow-up protocol is recommended:


  • Initial phase (first 3 months): Weekly to bi-weekly monitoring of urinalysis, renal function, and blood pressure

  • Intermediate phase (3-12 months): Monthly assessments

  • Long-term phase (beyond 12 months): Quarterly to semi-annual evaluations for at least 5 years


Patients with significant renal involvement may require indefinite monitoring[75].

Conclusion and Future Directions

Adult IgAV represents a significant challenge for rheumatologists due to its complex presentation, potential for severe organ involvement, and limited evidence-based treatment guidelines. While our understanding of disease mechanisms has advanced considerably, translating these insights into targeted therapies remains a work in progress.


Future research should focus on:


  1. Development of validated diagnostic criteria specific to adult IgAV

  2. Identification of reliable biomarkers for disease activity and prognosis

  3. Large randomized controlled trials evaluating both conventional and novel therapies

  4. Better understanding of the relationship between IgAV and malignancy

  5. Exploration of the gut-immune axis in disease pathogenesis


A multidisciplinary approach involving rheumatology, nephrology, gastroenterology, and dermatology remains crucial for optimal management of this complex vasculitis.

References

  1. Jennette JC, et al. 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013;65(1):1-11.


  1. Gardner-Medwin JM, et al. Incidence of Henoch-Schönlein purpura, Kawasaki disease, and rare vasculitides in children of different ethnic origins. Lancet. 2002;360(9341):1197-1202.


  1. Piram M, et al. Epidemiology of immunoglobulin A vasculitis (Henoch-Schönlein): current state of knowledge. Curr Opin Rheumatol. 2013;25(2):171-178.


  1. Jennette JC, et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013;65(1):1-11.


  1. Kang Y, et al. Clinical characteristics of adult patients with Henoch-Schönlein purpura. Int J Rheum Dis. 2018;21(12):2238-2244.


  1. Pillebout E, et al. Henoch-Schönlein purpura in adults: outcome and prognostic factors. J Am Soc Nephrol. 2002;13(5):1271-1278.


  1. Yang YH, et al. The incidence and clinical characteristics of Henoch-Schönlein purpura in children from a defined population. Pediatr Nephrol. 2005;20(9):1293-1298.


  1. Blanco R, et al. Henoch-Schönlein purpura in adulthood and childhood: two different expressions of the same syndrome. Arthritis Rheum. 1997;40(5):859-864.


  1. Audemard-Verger A, et al. Characteristics and Management of IgA Vasculitis (Henoch-Schönlein) in Adults: Data From 260 Patients Included in a French Multicenter Retrospective Survey. Arthritis Rheumatol. 2017;69(9):1862-1870.


  1. Calvo-Río V, et al. Henoch-Schönlein purpura in northern Spain: clinical spectrum of the disease in 417 patients from a single center. Medicine (Baltimore). 2014;93(2):106-113.


  1. Trapani S, et al. Henoch Schönlein purpura in childhood: epidemiological and clinical analysis of 150 cases over a 5-year period and review of literature. Semin Arthritis Rheum. 2005;35(3):143-153.


  1. Zurada JM, et al. Henoch-Schönlein purpura associated with malignancy in adults. J Am Acad Dermatol. 2006;55(5 Suppl):S65-S70.


  1. Mitsui H, et al. A clinical study of Henoch-Schönlein purpura associated with malignancy. J Eur Acad Dermatol Venereol. 2009;23(4):394-401.


  1. López-Mejías R, et al. Association of HLA-B*41:02 with Henoch-Schönlein Purpura (IgA Vasculitis) in Spanish individuals irrespective of the HLA-DRB1 status. Arthritis Res Ther. 2015;17:102.


  1. Kiryluk K, et al. Aberrant glycosylation of IgA1 is inherited in both pediatric IgA nephropathy and Henoch-Schönlein purpura nephritis. Kidney Int. 2011;80(1):79-87.


  1. Suzuki H, et al. The pathophysiology of IgA nephropathy. J Am Soc Nephrol. 2011;22(10):1795-1803.


  1. Novak J, et al. IgA glycosylation and IgA immune complexes in the pathogenesis of IgA nephropathy. Semin Nephrol. 2018;38(5):469-477.


  1. Floege J, et al. The pathogenesis of IgA nephropathy: what is new and how does it change therapeutic approaches? Am J Kidney Dis. 2011;58(6):992-1004.


  1. Roos A, et al. Both IgA nephropathy and Henoch-Schönlein purpura nephritis are associated with abnormal O-glycosylation of IgA1. Nephrol Dial Transplant. 2006;21(12):3367-3375.


  1. Nakazawa D, et al. Neutrophil extracellular traps in vasculitis: immunothrombosis to the injury of blood vessels. Future Rheumatol. 2018;13(2):117-125.


  1. He X, et al. IL-17 and IL-23 are implicated in the pathogenesis of Henoch-Schönlein purpura in children. Rheumatol Int. 2016;36(10):1413-1418.


  1. Di Ciaula A, et al. Intestinal permeability and gastrointestinal diseases. Curr Gastroenterol Rep. 2020;22(10):54.


  1. Audemard-Verger A, et al. Characteristics and Management of IgA Vasculitis (Henoch-Schönlein) in Adults: Data From 260 Patients Included in a French Multicenter Retrospective Survey. Arthritis Rheumatol. 2017;69(9):1862-1870.


  1. Hocevar A, et al. Clinical differences between adult and pediatric onset of IgA vasculitis (Henoch-Schönlein purpura): a retrospective observational study. Rheumatol Int. 2019;39(12):2125-2130.


  1. Hetland LE, et al. Henoch-Schönlein purpura: a literature review. Acta Derm Venereol. 2017;97(10):1160-1166.


  1. Calvo-Río V, et al. Henoch-Schönlein purpura in northern Spain: clinical spectrum of the disease in 417 patients from a single center. Medicine (Baltimore). 2014;93(2):106-113.


  1. Audemard-Verger A, et al. IgA vasculitis (Henoch-Shönlein purpura) in adults: Diagnostic and therapeutic aspects. Autoimmun Rev. 2015;14(7):579-585.


  1. Chen MJ, et al. Gastrointestinal involvement in adult patients with Henoch-Schönlein purpura. Dig Dis Sci. 2013;58(4):992-995.


  1. Pillebout E, et al. Henoch-Schönlein Purpura in adults: outcome and prognostic factors. J Am Soc Nephrol. 2002;13(5):1271-1278.


  1. Shrestha S, et al. Henoch Schönlein purpura with nephritis in adults: adverse prognostic indicators in a UK population. QJM. 2006;99(4):253-265.


  1. Hung SP, et al. Clinical manifestations and outcomes of Henoch-Schönlein purpura: comparison between adults and children. Pediatr Neonatol. 2009;50(4):162-168.


  1. Coppo R, et al. IgA-associated renal diseases: diagnosis and treatment. J Nephrol. 2018;31(4):435-446.


  1. Calvo-Río V, et al. Henoch-Schönlein purpura in northern Spain: clinical spectrum of the disease in 417 patients from a single center. Medicine (Baltimore). 2014;93(2):106-113.


  1. Blanco R, et al. Henoch-Schönlein purpura in adulthood and childhood: two different expressions of the same syndrome. Arthritis Rheum. 1997;40(5):859-864.


  1. Garzoni L, et al. Nervous system dysfunction in Henoch-Schönlein syndrome: systematic review of the literature. Rheumatology (Oxford). 2009;48(12):1524-1529.


  1. Rajagopala S, et al. Pulmonary hemorrhage in Henoch-Schönlein purpura: case report and systematic review of the English literature. Semin Arthritis Rheum. 2013;42(4):391-400.


  1. Lutz HH, et al. Henoch-Schönlein purpura complicated by cardiac involvement: case report and review of the literature. Am J Kidney Dis. 2009;54(5):e9-15.


  1. Hara Y, et al. Orchitis in Henoch-Schönlein purpura. Pediatr Int. 2012;54(4):553-555.


  1. Mills JA, et al. The American College of Rheumatology 1990 criteria for the classification of Henoch-Schönlein purpura. Arthritis Rheum. 1990;33(8):1114-1121.


  1. Ozen S, et al. EULAR/PRINTO/PRES criteria for Henoch-Schönlein purpura, childhood polyarteritis nodosa, childhood Wegener granulomatosis and childhood Takayasu arteritis: Ankara 2008. Part II: Final classification criteria. Ann Rheum Dis. 2010;69(5):798-806.


  1. Chen S, et al. IgA vasculitis in adults: the performance of the EULAR/PRINTO/PRES classification criteria in adults. Arthritis Res Ther. 2019;21(1):40.


  1. Yang YH, et al. A nationwide survey on epidemiological characteristics of childhood Henoch-Schönlein purpura in Taiwan. Rheumatology (Oxford). 2005;44(5):618-622.


  1. Heineke MH, et al. New insights in the pathogenesis of immunoglobulin A vasculitis (Henoch-Schönlein purpura). Autoimmun Rev. 2017;16(12):1246-1253.


  1. Trimarchi H, et al. Oxford Classification of IgA nephropathy 2016: an update from the IgA Nephropathy Classification Working Group. Kidney Int. 2017;91(5):1014-1021.


  1. Audemard-Verger A, et al. IgA vasculitis (Henoch-Shönlein purpura) in adults: Diagnostic and therapeutic aspects. Autoimmun Rev. 2015;14(7):579-585.


  1. Zurada JM, et al. Henoch-Schönlein purpura associated with malignancy in adults. J Am Acad Dermatol. 2006;55(5 Suppl):S65-S70.


  1. Suzuki H, et al. Serum levels of galactose-deficient IgA in children with IgA nephropathy and Henoch-Schönlein nephritis. Contrib Nephrol. 2007;157:35-38.


  1. Berthelot L, et al. Identification of monomeric and dimeric IgA as potential biomarkers in diseases with mucosal involvement. Immunol Lett. 2020;218:94-100.


  1. López-Mejías R, et al. Genetics of immunoglobulin-A vasculitis (Henoch-Schönlein purpura): An updated review. Autoimmun Rev. 2018;17(3):301-315.


  1. Fremeaux-Bacchi V, et al. Complement activation in patients with IgA nephropathy. Contrib Nephrol. 2007;157:122-126.


  1. Ozen S, et al. Management of Henoch-Schönlein purpura: SHARE initiative. Rheumatology (Oxford). 2019;58(9):1599-1607.


  1. Weiss PF, et al. Effects of corticosteroid treatment on outcomes of pediatric Henoch-Schönlein purpura. Pediatrics. 2017;120(5):1079-1087.


  1. Iqbal H, et al. Colchicine in management of Henoch Schönlein purpura: a systematic review and meta-analysis. Mediterr J Rheumatol. 2020;31(2):207-215.


  1. Iqbal H, et al. Dapsone therapy for Henoch-Schönlein purpura: a case series. Br J Dermatol. 2016;174(4):920-922.


  1. Pillebout E, et al. Addition of cyclophosphamide to steroids provides no benefit compared with steroids alone in treating adult patients with severe Henoch Schönlein Purpura. Kidney Int. 2010;78(5):495-502.


  1. Maritati F, et al. Cyclophosphamide versus rituximab for the treatment of adult IgA vasculitis with nephritis: a multicenter, retrospective study. J Nephrol. 2023;36(1):183-192.


  1. Maritati F, et al. Rituximab for the treatment of adult-onset IgA vasculitis (Henoch-Schönlein purpura). Arthritis Rheumatol. 2018;70(1):109-114.


  1. Fenoglio R, et al. Rituximab as a front-line therapy for adult-onset IgA vasculitis with nephritis. Oncotarget. 2018;9(32):22442-22450.


  1. Acar B, et al. Plasma exchange therapy in severe Henoch-Schönlein purpura: a single-center experience. Transfus Apher Sci. 2015;53(3):300-304.


  1. Han F, et al. The mycophenolate mofetil plus prednisone treatment for IgA nephropathy: a meta-analysis. Int J Clin Pract. 2020;74(6):e13482.


  1. Audemard-Verger A, et al. Characteristics and Management of IgA Vasculitis (Henoch-Schönlein) in Adults: Data From 260 Patients Included in a French Multicenter Retrospective Survey. Arthritis Rheumatol. 2017;69(9):1862-1870.


  1. Park SJ, et al. Successful use of adalimumab in an adult with refractory Henoch-Schönlein purpura. Clin Rheumatol. 2018;37(8):2241-2244.


  1. Ruffino A, et al. Successful treatment with IL-17A antagonists of refractory Henoch-Schönlein purpura: a report of two cases. Rheumatol Adv Pract. 2021;5(1):rkaa075.


  1. Dogra S, et al. Eculizumab for the treatment of severe Henoch-Schönlein purpura: a case report. Rheumatol Adv Pract. 2020;4(2):rkaa044.


  1. Sansbury FH, et al. JAK inhibition as a therapeutic strategy for immune complex-mediated vascular inflammation. Front Immunol. 2022;13:841458.


  1. Floege J, et al. Treatment of IgA nephropathy and Henoch-Schönlein nephritis. Nat Rev Nephrol. 2013;9(6):320-327.


  1. Hahn D, et al. Interventions for preventing and treating kidney disease in Henoch-Schönlein purpura (HSP). Cochrane Database Syst Rev. 2020;8(8):CD005128.


  1. Chen JY, et al. Gastrointestinal manifestations in Henoch-Schönlein purpura: a review of 261 patients. Acta Paediatr. 2014;103(4):438-444.


  1. Kalelioğlu I, et al. Henoch-Schönlein purpura and pregnancy: a case report and review of the literature. Arch Gynecol Obstet. 2007;275(5):383-387.


  1. García-Porrúa C, et al. Henoch-Schönlein purpura in children and adults: clinical differences in a defined population. Semin Arthritis Rheum. 2002;32(3):149-156.


  1. Pillebout E, et al. Henoch-Schönlein Purpura in adults: outcome and prognostic factors. J Am Soc Nephrol. 2002;13(5):1271-1278.


  1. Shrestha S, et al. Henoch Schönlein purpura with nephritis in adults: adverse prognostic indicators in a UK population. QJM. 2006;99(4):253-265.


  1. Audemard-Verger A, et al. IgA vasculitis (Henoch-Shönlein purpura) in adults: Diagnostic and therapeutic aspects. Autoimmun Rev. 2015;14(7):579-585.


  1. Uppal SS, et al. Henoch-Schönlein purpura in adults versus children/adolescents: A comparative study. Clin Exp Rheumatol. 2006;24(2 Suppl 41):S26-30.


  1. Heineke MH, et al. New insights in the pathogenesis of immunoglobulin A vasculitis (Henoch-Schönlein purpura). Autoimmun Rev. 2017;16(12):1246-1253.


  1. Davin JC, et al. Henoch-Schönlein purpura nephritis: pathophysiology, treatment, and future strategy. Clin J Am Soc Nephrol. 2011;6(3):679-689.


  1. Nadrous HF, et al. Pulmonary involvement in Henoch-Schönlein purpura. Mayo Clin Proc. 2004;79(9):1151-1157.


  1. Chen O, et al. Systematic review of the outcomes of IgA nephropathy and IgA vasculitis following solid organ transplantation or hematopoietic stem cell transplantation. Transplantation. 2018;102(12):1994-2002.


  1. Baigrie D, et al. Validation of the EULAR/PRINTO/PRES criteria for the classification of Henoch-Schönlein purpura in a UK cohort. Rheumatology (Oxford). 2020;59(5):1139-1144.


  1. Micheletti RG, et al. Cutaneous vasculitis: a rheumatologist's approach. Curr Opin Rheumatol. 2019;31(1):33-41.


Evaluating Non-Infectious Uveitis: A Rheumatology Perspective

 

Evaluating Non-Infectious Uveitis: A Rheumatology Perspective

Dr Neeraj Manikath , claude.ai

Abstract

Non-infectious uveitis represents a significant diagnostic and therapeutic challenge for rheumatologists and ophthalmologists alike. As a manifestation of numerous systemic autoimmune and inflammatory disorders, non-infectious uveitis requires a comprehensive approach to evaluation, diagnosis, and management. This review provides an evidence-based framework for rheumatologists to effectively assess patients with non-infectious uveitis, highlighting the importance of interdisciplinary collaboration, recent advances in diagnostic modalities, and current classification criteria. The review emphasizes a systematic approach to identify underlying systemic diseases and discusses the complexities of differentiating between various etiologies of non-infectious uveitis to guide appropriate therapeutic interventions.

Introduction

Uveitis, defined as inflammation of the uveal tract (iris, ciliary body, and choroid), represents a heterogeneous group of inflammatory ocular disorders that can lead to significant visual morbidity. While infectious etiologies account for a proportion of uveitis cases, non-infectious uveitis poses unique challenges in diagnosis and management due to its associations with systemic inflammatory disorders often falling within the rheumatologist's domain of expertise.

Non-infectious uveitis may be idiopathic or associated with systemic autoimmune conditions, including spondyloarthropathies, juvenile idiopathic arthritis, Behçet's disease, sarcoidosis, and systemic vasculitides. Given these associations, rheumatologists play a pivotal role in the evaluation and management of these patients, often working collaboratively with ophthalmologists to achieve optimal outcomes.

This review presents a comprehensive approach to evaluating non-infectious uveitis from a rheumatological perspective, emphasizing the importance of a structured assessment to identify underlying systemic inflammatory disorders and guide appropriate therapeutic interventions.

Epidemiology and Classification

Non-infectious uveitis accounts for approximately 70-90% of all uveitis cases in developed countries, with an estimated annual incidence of 17-52 cases per 100,000 person-years and a prevalence of 38-714 per 100,000 persons. The epidemiological variation reflects differences in geographic, environmental, and genetic factors influencing disease expression.

The Standardization of Uveitis Nomenclature (SUN) working group has established a classification system based on:

  1. Anatomical location:

    • Anterior uveitis (iris and ciliary body)
    • Intermediate uveitis (vitreous and peripheral retina)
    • Posterior uveitis (choroid and retina)
    • Panuveitis (all three segments)
  2. Disease course:

    • Acute (sudden onset, limited duration)
    • Recurrent (repeated episodes separated by periods of inactivity without treatment)
    • Chronic (persistent inflammation for >3 months despite therapy)
  3. Laterality:

    • Unilateral
    • Bilateral

Understanding this classification is essential for rheumatologists, as different anatomical locations and patterns correlate with specific systemic disorders. For instance, HLA-B27-associated diseases typically present with acute anterior uveitis, while sarcoidosis more commonly manifests as granulomatous anterior, intermediate, or panuveitis.

Clinical Approach to Non-Infectious Uveitis: The Rheumatologist's Perspective

Initial Assessment

When evaluating a patient with suspected or confirmed non-infectious uveitis, rheumatologists should follow a structured approach:

  1. Comprehensive history:

    • Demographics (age, sex, race, ethnicity)
    • Ocular symptoms (pain, redness, photophobia, visual changes)
    • Systemic symptoms (joint pain, back pain, skin manifestations, gastrointestinal symptoms)
    • Previous episodes of uveitis and response to treatment
    • Family history of autoimmune diseases or uveitis
    • Recent infections or travel history
  2. Detailed physical examination:

    • Complete musculoskeletal examination
    • Dermatological assessment (psoriasis, erythema nodosum, etc.)
    • Oral and genital mucosal examination
    • Cardiovascular and respiratory evaluation
    • Neurological examination when indicated
  3. Ophthalmological examination findings review:

    • Anterior segment findings (keratic precipitates, cells, flare)
    • Posterior segment involvement (vitritis, retinal lesions, choroiditis)
    • Complications (cataract, glaucoma, macular edema)

Laboratory and Imaging Investigations

Laboratory workup should be guided by clinical suspicion rather than employing a shotgun approach. Common investigations include:

  1. Basic laboratory tests:

    • Complete blood count
    • Inflammatory markers (ESR, CRP)
    • Comprehensive metabolic panel
    • Urinalysis
  2. Targeted serological testing:

    • HLA-B27 typing (when spondyloarthropathies are suspected)
    • Antinuclear antibodies and specific autoantibodies (anti-dsDNA, anti-Ro/La)
    • Complement levels
    • Angiotensin-converting enzyme (ACE) and lysozyme (for sarcoidosis)
    • Antineutrophil cytoplasmic antibodies (for vasculitis)
  3. Infectious disease screening:

    • Tuberculosis screening (interferon-gamma release assay or tuberculin skin test)
    • Syphilis serology
    • Lyme disease serology in endemic areas
    • Viral hepatitis serology
  4. Imaging studies:

    • Chest radiography or CT (for sarcoidosis, tuberculosis)
    • Sacroiliac joint imaging (for spondyloarthropathies)
    • MRI when neurological involvement is suspected
  5. Advanced ocular imaging:

    • Optical coherence tomography (OCT)
    • Fluorescein angiography
    • Indocyanine green angiography
    • Ultrasound biomicroscopy

Specific Disease Associations and Their Ocular Manifestations

Spondyloarthropathies

Acute anterior uveitis (AAU) is the most common extra-articular manifestation of spondyloarthropathies, particularly in HLA-B27-positive individuals. Approximately 30-40% of patients with ankylosing spondylitis develop AAU during their disease course. Typical features include:

  • Unilateral, sudden-onset, recurrent episodes
  • Significant anterior chamber inflammation with fibrin deposition
  • Alternating eye involvement between episodes
  • Strong association with HLA-B27 positivity (>50% of cases)

The diagnosis of underlying spondyloarthropathy should be considered in patients with AAU, particularly when accompanied by inflammatory back pain, enthesitis, or inflammatory bowel disease. The Assessment of SpondyloArthritis International Society (ASAS) criteria should be utilized for classification.

Juvenile Idiopathic Arthritis (JIA)

Chronic anterior uveitis is a significant complication of JIA, particularly in young females with oligoarticular JIA who are antinuclear antibody (ANA)-positive. Key features include:

  • Bilateral, asymptomatic, chronic anterior uveitis
  • Insidious onset often preceding or occurring simultaneously with arthritis
  • High risk of complications (band keratopathy, posterior synechiae, cataract)
  • Requires regular ophthalmological screening according to risk stratification

Early detection through regular ophthalmological screening is crucial for preventing vision-threatening complications. Risk factors for uveitis in JIA include oligoarticular subtype, early age of onset, ANA positivity, and female gender.

Behçet's Disease

Behçet's disease can manifest with severe, recurrent panuveitis or retinal vasculitis. Ocular involvement occurs in 50-70% of patients and is characterized by:

  • Bilateral, recurrent, non-granulomatous panuveitis
  • Retinal vasculitis (predominantly venous)
  • Hypopyon (sterile collection of neutrophils in anterior chamber)
  • Retinal infiltrates and occlusive vasculitis

The diagnosis is primarily clinical, based on the International Criteria for Behçet's Disease (ICBD), which include oral aphthosis, genital aphthosis, skin lesions, ocular manifestations, vascular involvement, and positive pathergy test.

Sarcoidosis

Ocular sarcoidosis can affect any segment of the eye but commonly presents as granulomatous anterior uveitis or panuveitis. Characteristic features include:

  • Bilateral, granulomatous anterior uveitis with "mutton-fat" keratic precipitates
  • Iris nodules (Koeppe and Busacca nodules)
  • Snowball or string-of-pearls vitreous opacities
  • Segmental periphlebitis or "candle wax drippings"
  • Chorioretinal granulomas

The International Workshop on Ocular Sarcoidosis (IWOS) has established diagnostic criteria including clinical signs, laboratory investigations (elevated ACE, lysozyme), chest imaging, and histopathological confirmation when possible.

Inflammatory Bowel Disease (IBD)

Uveitis in IBD (Crohn's disease and ulcerative colitis) may present as:

  • Anterior uveitis (more common in Crohn's disease)
  • Intermediate or panuveitis (more common in ulcerative colitis)
  • Can occur independent of bowel disease activity

A comprehensive gastrointestinal assessment is warranted in patients with recurrent non-infectious uveitis, particularly when accompanied by abdominal pain, diarrhea, rectal bleeding, or unexplained weight loss.

Systemic Vasculitides

Several vasculitides can manifest with ocular inflammation:

  • ANCA-associated vasculitis: scleritis, peripheral ulcerative keratitis, retinal vasculitis
  • Giant cell arteritis: anterior ischemic optic neuropathy, retinal artery occlusion
  • Polyarteritis nodosa: retinal vasculitis, choroiditis
  • Kawasaki disease: conjunctivitis, anterior uveitis

Systemic Lupus Erythematosus (SLE)

While frank uveitis is less common in SLE, retinal vasculitis and choroidopathy can occur. The presence of antiphospholipid antibodies increases the risk of retinal vaso-occlusive disease.

Diagnostic Challenges and Approach to Undifferentiated Uveitis

Despite comprehensive evaluation, approximately 30-40% of non-infectious uveitis cases remain idiopathic. In these undifferentiated cases, a systematic approach includes:

  1. Pattern recognition:

    • Correlating specific uveitis patterns with potential underlying diseases
    • Using anatomical classification to narrow differential diagnoses
  2. Biomarker assessment:

    • Novel biomarkers (cytokine profiles, autoantibody panels)
    • Genetic testing in selected cases
  3. Advanced imaging techniques:

    • Multimodal imaging to identify disease-specific patterns
    • OCT angiography for detailed assessment of retinal vasculature
  4. Interdisciplinary consultation:

    • Close collaboration with ophthalmologists, immunologists, and infectious disease specialists
    • Multidisciplinary uveitis clinics for complex cases

Therapeutic Considerations for Rheumatologists

While detailed treatment protocols are beyond this review's scope, therapeutic decision-making should consider:

  1. Treatment of underlying systemic disease:

    • Control of systemic inflammation often improves ocular manifestations
    • Disease-modifying antirheumatic drugs (DMARDs) may prevent recurrence
  2. Corticosteroid therapy:

    • Topical for anterior uveitis
    • Periocular or intravitreal for intermediate or posterior uveitis
    • Systemic for bilateral, sight-threatening, or treatment-resistant disease
  3. Conventional immunosuppressive agents:

    • Methotrexate, azathioprine, mycophenolate mofetil
    • Calcineurin inhibitors (cyclosporine, tacrolimus)
    • Cyclophosphamide for severe, refractory cases
  4. Biologic therapy:

    • TNF-α inhibitors (adalimumab, infliximab)
    • IL-6 receptor antagonists (tocilizumab)
    • IL-1 inhibitors (anakinra, canakinumab)
    • JAK inhibitors (tofacitinib, baricitinib)
  5. Collaborative monitoring:

    • Regular ophthalmological assessments
    • Monitoring for drug toxicity
    • Objective measures of treatment response (SUN criteria)

Emerging Concepts and Future Directions

Recent advances in understanding non-infectious uveitis include:

  1. Immunogenetics and precision medicine:

    • HLA associations beyond HLA-B27
    • Non-HLA genetic factors (IL23R, ERAP1)
    • Pharmacogenetic markers predicting treatment response
  2. Novel imaging biomarkers:

    • Quantitative assessment of choroidal thickness
    • OCT angiography for subclinical retinal vasculitis
    • Advanced image analysis with artificial intelligence
  3. Targeted therapeutics:

    • Selective JAK inhibition
    • T-cell targeted therapies
    • Local drug delivery systems
  4. Predictive models:

    • Risk stratification tools for uveitis development in rheumatic diseases
    • Predictors of treatment response and recurrence

Conclusion

Non-infectious uveitis represents a significant diagnostic and therapeutic challenge requiring close collaboration between rheumatologists and ophthalmologists. A systematic approach to evaluation, focusing on identifying underlying systemic inflammatory disorders, enables appropriate therapeutic interventions and improved visual outcomes.

Rheumatologists play a crucial role in this interdisciplinary approach, applying their expertise in systemic inflammatory diseases to guide diagnosis and management. As our understanding of the immunopathogenesis of non-infectious uveitis evolves, more targeted therapeutic approaches will emerge, potentially transforming the management paradigm from symptom control to disease modification and perhaps eventual cure.

Future research should focus on developing validated classification criteria for uveitis subtypes, identifying reliable biomarkers for disease activity monitoring, and establishing evidence-based treatment algorithms through randomized controlled trials comparing different immunomodulatory strategies.

References

  1. Acharya NR, Tham VM, Esterberg E, et al. Incidence and prevalence of uveitis: results from the Pacific Ocular Inflammation Study. JAMA Ophthalmol. 2013;131(11):1405-1412.

  2. Jabs DA, Nussenblatt RB, Rosenbaum JT; Standardization of Uveitis Nomenclature (SUN) Working Group. Standardization of uveitis nomenclature for reporting clinical data. Results of the First International Workshop. Am J Ophthalmol. 2005;140(3):509-516.

  3. Rosenbaum JT, Asquith M. The microbiome and HLA-B27-associated acute anterior uveitis. Nat Rev Rheumatol. 2018;14(12):704-713.

  4. Heiligenhaus A, Minden K, Tappeiner C, et al. Update of the evidence based, interdisciplinary guideline for anti-inflammatory treatment of uveitis associated with juvenile idiopathic arthritis. Semin Arthritis Rheum. 2019;49(1):43-55.

  5. Takeuchi M, Kastner DL, Remmers EF. The immunogenetics of Behçet's disease: A comprehensive review. J Autoimmun. 2015;64:137-148.

  6. Herbort CP, Rao NA, Mochizuki M; International Workshop on Ocular Sarcoidosis. International criteria for the diagnosis of ocular sarcoidosis: results of the first International Workshop On Ocular Sarcoidosis (IWOS). Ocul Immunol Inflamm. 2009;17(3):160-169.

  7. Rosenbaum JT, Rosenzweig HL. A rationale for TNF-alpha inhibitors in the treatment of uveitis. Clin Exp Rheumatol. 2019;37 Suppl 121(6):86-93.

  8. Dick AD, Tugal-Tutkun I, Foster S, et al. Secukinumab in the treatment of noninfectious uveitis: results of three randomized, controlled clinical trials. Ophthalmology. 2013;120(4):777-787.

  9. Jaffe GJ, Dick AD, Brézin AP, et al. Adalimumab in patients with active noninfectious uveitis. N Engl J Med. 2016;375(10):932-943.

  10. Agrawal R, Murthy S, Sangwan V, et al. Current approach in diagnosis and management of anterior uveitis. Indian J Ophthalmol. 2010;58(1):11-19.

  11. Thorne JE, Suhler E, Skup M, et al. Prevalence of noninfectious uveitis in the United States: a claims-based analysis. JAMA Ophthalmol. 2016;134(11):1237-1245.

  12. Wakefield D, Chang JH. Epidemiology of uveitis. Int Ophthalmol Clin. 2005;45(2):1-13.

  13. Rothova A, Suttorp-van Schulten MS, Frits Treffers W, et al. Causes and frequency of blindness in patients with intraocular inflammatory disease. Br J Ophthalmol. 1996;80(4):332-336.

  14. Angeles-Han ST, Ringold S, Beukelman T, et al. 2019 American College of Rheumatology/Arthritis Foundation Guideline for the Screening, Monitoring, and Treatment of Juvenile Idiopathic Arthritis-Associated Uveitis. Arthritis Care Res (Hoboken). 2019;71(6):703-716.

  15. Levy-Clarke G, Jabs DA, Read RW, et al. Expert panel recommendations for the use of anti-tumor necrosis factor biologic agents in patients with ocular inflammatory disorders. Ophthalmology. 2014;121(3):785-796.e3.

  16. Hatemi G, Christensen R, Bang D, et al. 2018 update of the EULAR recommendations for the management of Behçet's syndrome. Ann Rheum Dis. 2018;77(6):808-818.

  17. Lee CS, Randhawa S, Lee AY, et al. Patterns of uveitis in the workplace: impact of rheumatologic disease and implications for disability. J Ophthalmic Inflamm Infect. 2019;9(1):9.

  18. Fabiani C, Vitale A, Lopalco G, et al. Different roles of TNF inhibitors in acute anterior uveitis associated with ankylosing spondylitis: state of the art. Clin Rheumatol. 2016;35(11):2631-2638.

  19. Rosenbaum JT. New developments in uveitis associated with HLA B27. Curr Opin Rheumatol. 2017;29(4):298-303.

  20. Smith JR, Stempel AJ, Bharadwaj A, et al. Involvement of B cells in non-infectious uveitis. Clin Transl Immunology. 2016;5(2):e63.

Sunday, April 20, 2025

Inflammatory Myositis Mimics

 

Inflammatory Myositis Mimics: A Comprehensive Review and Diagnostic Roadmap

Dr Neeraj Manikath ,claude.ai

Introduction

Inflammatory myopathies represent a heterogeneous group of immune-mediated disorders characterized by muscle inflammation, weakness, and elevated muscle enzymes. While idiopathic inflammatory myopathies (IIMs)—including dermatomyositis (DM), polymyositis (PM), immune-mediated necrotizing myopathy (IMNM), and inclusion body myositis (IBM)—are the classic entities in this category, numerous conditions can present with similar clinical, laboratory, and histopathological features, posing significant diagnostic challenges.

This review provides a state-of-the-art analysis of inflammatory myositis mimics and presents a structured diagnostic approach to differentiate these entities from true inflammatory myopathies. Accurate differentiation is critical, as therapeutic strategies differ substantially between inflammatory myopathies and their mimics, and misdiagnosis can lead to inappropriate treatment, adverse effects, and delayed appropriate intervention.

Classification of Myositis Mimics

Inflammatory myositis mimics can be categorized into several groups:

1. Genetic Myopathies

Muscular Dystrophies

Several muscular dystrophies can mimic inflammatory myopathies, particularly when they present with elevated creatine kinase (CK) levels and inflammatory features on muscle biopsy:

  • Dysferlinopathies (LGMD2B/Miyoshi myopathy): Often presents with significantly elevated CK and inflammatory infiltrates on biopsy. Characteristic features include preferential posterior compartment involvement in the lower limbs and absence of dysferlin staining on immunohistochemistry.

  • Facioscapulohumeral muscular dystrophy (FSHD): May show inflammatory infiltrates in up to 40% of biopsies. The facial weakness and scapular winging characteristic of FSHD are crucial distinguishing features.

  • Dystrophinopathies: Both Duchenne and Becker muscular dystrophies can show inflammatory changes on biopsy. Western blot or immunohistochemistry for dystrophin can be diagnostic.

  • Calpainopathy (LGMD2A): Often presents with proximal weakness and elevated CK, mimicking polymyositis. Genetic testing reveals mutations in the CAPN3 gene.

Metabolic Myopathies

  • McArdle's disease (glycogen storage disease type V): Can present with elevated CK, exercise intolerance, and inflammatory changes on biopsy. The absence of myophosphorylase on histochemical staining is diagnostic.

  • Acid maltase deficiency (Pompe disease): Adult-onset Pompe disease can present with proximal muscle weakness resembling inflammatory myopathy. Vacuolar changes with glycogen accumulation and reduced acid alpha-glucosidase activity are characteristic.

2. Toxic Myopathies

Drug-Induced Myopathies

  • Statin-associated myopathy: Ranges from asymptomatic CK elevation to severe necrotizing myopathy. In statin-induced necrotizing autoimmune myopathy (SINAM), antibodies against HMGCR are frequently present, and the condition persists despite statin discontinuation.

  • Colchicine myopathy: Characterized by vacuolar changes on muscle biopsy, often with inflammatory infiltrates.

  • Critical illness myopathy: Particularly in patients treated with high-dose corticosteroids and neuromuscular blocking agents in intensive care settings.

  • Other medications: Chloroquine/hydroxychloroquine, D-penicillamine, zidovudine, ipilimumab, and other immune checkpoint inhibitors can induce inflammatory-like myopathies.

Toxin-Induced Myopathies

  • Alcohol-related myopathy: Both acute and chronic forms can mimic inflammatory myopathies.

  • Environmental toxins: Exposure to organophosphates, snake venoms, and other environmental toxins can cause muscle inflammation.

3. Endocrine Myopathies

  • Hypothyroid myopathy: Often presents with proximal weakness, myalgias, and elevated CK. The presence of other features of hypothyroidism and normalization with thyroid hormone replacement are distinguishing features.

  • Hyperparathyroidism: Can present with proximal muscle weakness and occasionally elevated CK levels.

  • Cushing syndrome: Either endogenous or iatrogenic hypercortisolism can cause proximal muscle weakness.

  • Addison's disease: May present with fatigue, weakness, and occasionally elevated CK.

4. Infectious and Post-infectious Myopathies

  • Bacterial myositis: Pyomyositis, particularly due to Staphylococcus aureus.

  • Viral myositis: Including infections with influenza, HIV, HTLV-1, coxsackievirus, and SARS-CoV-2.

  • Parasitic myositis: Particularly trichinosis, toxoplasmosis, and cysticercosis.

  • Post-infectious immune-mediated processes: Including post-streptococcal myalgia syndrome.

5. Other Systemic Diseases with Myositis Features

  • Sarcoidosis with myopathic involvement: Presents with granulomatous inflammation in muscle tissue.

  • Amyloidosis: Can present with proximal weakness and occasionally inflammatory features on biopsy.

  • Paraneoplastic myopathy: Beyond the known association of dermatomyositis with malignancy, other paraneoplastic processes can cause inflammatory-like myopathies.

6. Functional Disorders

  • Fibromyalgia: Characterized by widespread pain and tender points, often confused with polymyalgia rheumatica or inflammatory myopathies.

  • Chronic fatigue syndrome: Features prominent fatigue without objective weakness or elevated muscle enzymes.

Diagnostic Approach to Differentiate True Inflammatory Myopathies from Mimics

Clinical Assessment

A thorough clinical evaluation remains the cornerstone of distinguishing inflammatory myopathies from their mimics:

  1. Pattern of muscle involvement:

    • Symmetrical proximal weakness is typical of DM, PM, and IMNM
    • Distal weakness, especially finger flexors and quadriceps, suggests IBM
    • Asymmetric involvement may suggest FSHD or focal myositis
    • Ocular muscle involvement suggests mitochondrial myopathy or other genetic disorders rather than IIM
  2. Extramuscular manifestations:

    • Characteristic skin rashes of DM (Gottron's papules, heliotrope rash)
    • Interstitial lung disease suggests antisynthetase syndrome or MDA5-associated DM
    • Joint symptoms suggest overlap syndromes
    • Dysphagia pattern can differ between IIMs and neurological disorders
  3. Temporal profile:

    • Acute onset over days to weeks suggests toxic, infectious, or necrotizing autoimmune etiologies
    • Insidious progression over months to years is more consistent with IBM or genetic myopathies
    • Episodic symptoms with clear triggers suggest metabolic myopathies
  4. Family history:

    • Positive family history suggests a genetic myopathy
    • Family clustering may be seen in some autoimmune conditions
  5. Medication history:

    • Detailed review of prescription medications, over-the-counter supplements, and potential toxin exposures

Laboratory Investigations

  1. Muscle enzymes:

    • Serum CK levels may be markedly elevated in necrotizing myopathies, dystrophinopathies, and dysferlinopathies
    • Mild to moderate elevation in DM, PM, and IBM
    • Normal or minimal elevation in fibromyalgia, endocrine myopathies, and some cases of DM
  2. Myositis-specific antibodies (MSAs):

    • Anti-Jo-1 and other antisynthetase antibodies (anti-PL-7, anti-PL-12)
    • Anti-Mi-2 in classic DM
    • Anti-SRP in necrotizing myopathy
    • Anti-HMGCR in statin-associated necrotizing myopathy
    • Anti-MDA5 in clinically amyopathic DM with rapidly progressive ILD
    • Anti-TIF1γ and anti-NXP2 in cancer-associated DM
    • Anti-cN1A in IBM
  3. Myositis-associated antibodies (MAAs):

    • Anti-Ro/SSA, anti-La/SSB, anti-RNP, anti-Sm in overlap syndromes
  4. Other laboratory tests:

    • ESR and CRP may be elevated in inflammatory processes but normal in many genetic myopathies
    • TSH, T4 to evaluate thyroid function
    • Calcium, phosphate, PTH for hyperparathyroidism
    • Electrolytes, renal and liver function tests
    • Aldolase may be elevated disproportionately to CK in inflammatory myopathies
    • Troponin I can help distinguish cardiac from skeletal muscle injury

Electrophysiological Studies

  1. Electromyography (EMG):

    • Inflammatory myopathies typically show increased insertional activity, fibrillations, positive sharp waves, and complex repetitive discharges
    • Early recruitment of short-duration, small-amplitude, polyphasic motor units
    • Myotonic discharges suggest myotonic dystrophy, channelopathies, or certain metabolic disorders
    • Neuropathic features suggest a neurogenic process rather than primary myopathy
  2. Nerve conduction studies (NCS):

    • Normal in pure myopathic processes
    • Abnormalities suggest concomitant neuropathy or neuromuscular junction disorders

Imaging Studies

  1. Magnetic Resonance Imaging (MRI):

    • Increasingly important in diagnosis and monitoring of inflammatory myopathies
    • T2-weighted and STIR sequences show muscle edema in active inflammation
    • T1-weighted sequences show fatty replacement in chronic disease
    • Characteristic patterns:
      • DM: Patchy, predominantly peripheral edema
      • PM: More diffuse muscle edema
      • IBM: Selective involvement of quadriceps and deep finger flexors with fatty replacement
      • Muscular dystrophies: Often show specific patterns of muscle involvement
  2. Ultrasound:

    • Emerging modality for assessing muscle architecture
    • Increased echogenicity in inflammatory and dystrophic processes
    • Dynamic assessment possible
    • Useful for guiding muscle biopsy

Muscle Biopsy

The gold standard for diagnosing inflammatory myopathies and their mimics:

  1. Dermatomyositis:

    • Perifascicular atrophy
    • Perivascular inflammation
    • MAC deposition on capillaries
    • Tubuloreticular inclusions on electron microscopy
  2. Polymyositis:

    • Endomysial inflammation with CD8+ T-cells invading non-necrotic muscle fibers
    • MHC-I upregulation
  3. Immune-mediated Necrotizing Myopathy:

    • Prominent necrosis and regeneration
    • Minimal inflammatory infiltrates
    • MHC-I upregulation
    • MAC deposition in HMGCR+ cases
  4. Inclusion Body Myositis:

    • Endomysial inflammation
    • Rimmed vacuoles
    • Protein aggregates (p62, TDP-43)
    • Mitochondrial abnormalities (COX-negative fibers)
  5. Common mimics' biopsy features:

    • Dystrophinopathies: Dystrophin deficiency, fiber size variation, increased connective tissue
    • Dysferlinopathies: Dysferlin deficiency, inflammatory infiltrates
    • Metabolic myopathies: Specific enzyme deficiencies, glycogen or lipid accumulation
    • Mitochondrial myopathies: Ragged red fibers, COX-negative fibers
    • Drug-induced myopathies: Characteristic findings (vacuoles with chloroquine, necrosis with statins)

Genetic Testing

Increasingly important in diagnosing myopathy mimics:

  1. Next-generation sequencing panels:

    • Muscular dystrophy panels
    • Metabolic myopathy panels
    • Congenital myopathy panels
  2. Whole exome/genome sequencing:

    • For cases without diagnosis after targeted panel testing
  3. Specific genetic tests:

    • FSHD: Testing for D4Z4 repeat contraction
    • Myotonic dystrophy: Testing for CTG repeat expansion
    • Mitochondrial disorders: mtDNA analysis

Diagnostic Algorithm for Approaching Suspected Inflammatory Myopathy

  1. Initial evaluation:

    • Comprehensive history and physical examination
    • CK, aldolase, LDH, transaminases, ESR, CRP
    • TSH, electrolytes, calcium, phosphate
    • Myositis antibody panel
  2. First-tier investigations:

    • EMG/NCS
    • Muscle MRI
    • Screen for associated conditions (CTD, malignancy)
  3. Second-tier investigations:

    • Muscle biopsy from affected muscle (guided by MRI or EMG)
    • Additional antibody testing if indicated
    • Targeted genetic testing based on clinical suspicion
  4. Third-tier investigations:

    • Broader genetic panels or whole exome sequencing
    • Specialized metabolic studies
    • Repeat biopsy if initial biopsy inconclusive

Special Considerations for Specific Mimics

Statin-associated Necrotizing Autoimmune Myopathy (SINAM)

This increasingly recognized entity warrants special attention:

  • Clinical features: Persistent and progressive proximal weakness despite statin discontinuation
  • Laboratory: Markedly elevated CK (often >10x ULN)
  • Antibodies: Anti-HMGCR antibodies in 60-70% of cases
  • Pathology: Necrotizing myopathy with minimal inflammation
  • Treatment: Requires immunosuppression unlike self-limited statin myopathy

FSHD with Inflammatory Features

FSHD can present with significant inflammatory changes on biopsy, leading to misdiagnosis as PM or DM:

  • Clinical clues: Facial weakness, scapular winging, asymmetric involvement
  • Genetic testing: D4Z4 repeat contraction on chromosome 4q35
  • Treatment implications: Immunosuppression ineffective and potentially harmful

Metabolic Myopathies with Inflammatory Features

Several metabolic myopathies can show inflammatory infiltrates on biopsy:

  • McArdle's disease: Exercise intolerance, "second wind" phenomenon
  • Pompe disease: Diaphragmatic weakness, specific pattern on muscle MRI
  • Diagnosis: Enzymatic assays, genetic testing
  • Treatment: Enzyme replacement for Pompe disease, specific dietary measures for others

Inclusion Body Myositis vs. Polymyositis

IBM is frequently misdiagnosed initially as PM, leading to inappropriate and ineffective immunosuppression:

  • Clinical clues: Finger flexor weakness, quadriceps weakness, dysphagia, age >50
  • Electrophysiology: Mixed myopathic and neurogenic features
  • Pathology: Rimmed vacuoles, protein aggregates
  • Biomarkers: Anti-cN1A antibodies in 30-40% of IBM cases
  • Treatment response: Poor response to immunosuppression

Emerging Diagnostic Tools

Serum Biomarkers

  • Neopterin: Elevated in active inflammatory myopathies
  • COMP (Cartilage Oligomeric Matrix Protein): Correlated with disease activity in DM
  • Galectin-9: Emerging biomarker for juvenile DM activity
  • MicroRNAs: Several myomiRs under investigation as diagnostic biomarkers

Advanced Imaging Techniques

  • Whole-body MRI for comprehensive assessment
  • Quantitative MRI techniques (T2 mapping, diffusion-weighted imaging, magnetization transfer)
  • PET/CT for detecting inflammatory activity and associated malignancy
  • MR spectroscopy for metabolic assessment

Artificial Intelligence in Myopathology

  • Computer-assisted diagnosis from muscle biopsy images
  • Pattern recognition algorithms for muscle MRI interpretation
  • Integration of clinical, serological, and pathological data

Therapeutic Implications of Accurate Diagnosis

The importance of distinguishing inflammatory myopathies from mimics lies in the divergent therapeutic approaches:

  • True inflammatory myopathies: Require immunosuppression

    • DM, PM: Corticosteroids, conventional immunosuppressants, biologics
    • IMNM: More intensive immunosuppression often required
    • IBM: Generally resistant to immunosuppression; focus on supportive care
  • Genetic myopathies: Immunosuppression potentially harmful

    • Targeted gene therapies emerging for some disorders
    • Supportive care and rehabilitation
  • Metabolic myopathies: Specific therapeutic approaches

    • Enzyme replacement therapy for Pompe disease
    • Dietary modifications for various metabolic disorders
  • Endocrine myopathies: Treat the underlying endocrine disorder

  • Toxic myopathies: Discontinue offending agent

Conclusion

The differential diagnosis of inflammatory myopathies is extensive and complex. A systematic approach integrating clinical assessment, laboratory investigations, electrophysiological studies, imaging, and histopathology is essential for distinguishing true inflammatory myopathies from their many mimics. Accurate diagnosis has profound implications for treatment strategy and prognosis.

Emerging technologies including expanded myositis antibody panels, advanced imaging techniques, and genetic testing are enhancing our diagnostic capabilities. Nevertheless, the cornerstone of accurate diagnosis remains a careful integration of all available data, with particular attention to subtle clinical clues that may suggest an alternative diagnosis to idiopathic inflammatory myopathy.

References

  1. Selva-O'Callaghan A, Pinal-Fernandez I, Trallero-Araguás E, et al. Classification and management of adult inflammatory myopathies. Lancet Neurol. 2018;17(9):816-828.

  2. Mariampillai K, Granger B, Amelin D, et al. Development of a new classification system for idiopathic inflammatory myopathies based on clinical manifestations and myositis-specific autoantibodies. JAMA Neurol. 2018;75(12):1528-1537.

  3. Betteridge Z, McHugh N. Myositis-specific autoantibodies: an important tool to support diagnosis of myositis. J Intern Med. 2016;280(1):8-23.

  4. Allenbach Y, Mammen AL, Benveniste O, et al. 224th ENMC International Workshop: Clinico-sero-pathological classification of immune-mediated necrotizing myopathies. Neuromuscul Disord. 2018;28(1):87-99.

  5. Pinal-Fernandez I, Casal-Dominguez M, Mammen AL. Immune-mediated necrotizing myopathy. Curr Rheumatol Rep. 2018;20(4):21.

  6. Tanboon J, Nishino I. Classification of idiopathic inflammatory myopathies: pathology perspectives. Curr Opin Neurol. 2019;32(5):704-714.

  7. Stenzel W, Goebel HH, Aronica E. Review: immune-mediated necrotizing myopathies--a heterogeneous group of diseases with specific myopathological features. Neuropathol Appl Neurobiol. 2012;38(7):632-646.

  8. Day J, Patel S, Limaye V. The role of magnetic resonance imaging techniques in evaluation and management of the idiopathic inflammatory myopathies. Semin Arthritis Rheum. 2017;46(5):642-649.

  9. Milone M. Diagnosis and management of immune-mediated myopathies. Mayo Clin Proc. 2017;92(5):826-837.

  10. Schmidt J. Current classification and management of inflammatory myopathies. J Neuromuscul Dis. 2018;5(2):109-129.

  11. Selva-O'Callaghan A, Trallero-Araguás E, Grau-Junyent JM, et al. Malignancy and myositis: novel autoantibodies and new insights. Curr Opin Rheumatol. 2016;28(6):709-714.

  12. Watanabe Y, Uruha A, Suzuki S, et al. Clinical features and prognosis in anti-SRP and anti-HMGCR necrotising myopathy. J Neurol Neurosurg Psychiatry. 2016;87(10):1038-1044.

  13. Mammen AL. Statin-associated autoimmune myopathy. N Engl J Med. 2016;374(7):664-669.

  14. Lim J, Rietveld A, De Bleecker JL, et al. Seronegative patients form a distinctive subgroup of immune-mediated necrotizing myopathy. Neurol Neuroimmunol Neuroinflamm. 2019;6(1):e513.

  15. Lloyd TE, Mammen AL, Amato AA, et al. Evaluation and construction of diagnostic criteria for inclusion body myositis. Neurology. 2014;83(5):426-433.

  16. Engel AG, Arahata K. Monoclonal antibody analysis of mononuclear cells in myopathies. II: Phenotypes of autoinvasive cells in polymyositis and inclusion body myositis. Ann Neurol. 1984;16(2):209-215.

  17. Pinal-Fernandez I, Mammen AL. Spectrum of immune-mediated necrotizing myopathies and their treatments. Curr Opin Rheumatol. 2016;28(6):619-624.

  18. Basharat P, Christopher-Stine L. Immune-mediated necrotizing myopathy: update on diagnosis and management. Curr Rheumatol Rep. 2015;17(12):72.

  19. Greenberg SA. Inflammatory myopathies: evaluation and management. Semin Neurol. 2008;28(2):241-249.

  20. Simon JP, Marie I, Jouen F, et al. Autoimmune myopathies: where do we stand? Front Immunol. 2016;7:234.

  21. Suzuki S, Nishikawa A, Kuwana M, et al. Inflammatory myopathy with anti-signal recognition particle antibodies: case series of 100 patients. Orphanet J Rare Dis. 2015;10:61.

  22. Moghadam-Kia S, Oddis CV, Aggarwal R. Anti-MDA5 antibody spectrum in western world. Curr Rheumatol Rep. 2018;20(12):78.

  23. Uruha A, Suzuki S, Suzuki N, et al. Perifascicular necrosis in anti-synthetase syndrome beyond anti-Jo-1. Brain. 2016;139(Pt 9):e50.

  24. Benveniste O, Stenzel W, Allenbach Y. Advances in serological diagnostics of inflammatory myopathies. Curr Opin Neurol. 2016;29(5):662-673.

  25. Meyer A, Meyer N, Schaeffer M, et al. Incidence and prevalence of inflammatory myopathies: a systematic review. Rheumatology (Oxford). 2015;54(1):50-63.

  26. Liang C, Needham M. Necrotizing autoimmune myopathy. Curr Opin Rheumatol. 2011;23(6):612-619.

  27. Uruha A, Nishikawa A, Tsuburaya RS, et al. Sarcoplasmic MxA expression: a valuable marker of dermatomyositis. Neurology. 2017;88(5):493-500.

  28. Pipitone N. Value of MRI in diagnostics and evaluation of myositis. Curr Opin Rheumatol. 2016;28(6):625-630.

  29. Preuße C, Allenbach Y, Hoffmann O, et al. Differential roles of hypoxia and innate immunity in juvenile and adult dermatomyositis. Acta Neuropathol Commun. 2016;4(1):45.

  30. Lilleker JB, Vencovsky J, Wang G, et al. The EuroMyositis registry: an international collaborative tool to facilitate myositis research. Ann Rheum Dis. 2018;77(1):30-39.

Controversies in Fluid Resuscitation

Controversies in Fluid Resuscitation: A Review 

Dr Neeraj Manikath, claude.ai

Introduction

Fluid resuscitation remains one of the cornerstones of managing critically ill patients, particularly those with shock, sepsis, or major trauma. Despite its fundamental role in critical care, controversy persists regarding optimal fluid selection, timing, and volume. This review addresses two major areas of ongoing debate: the choice between crystalloids, balanced solutions, and albumin in specific patient populations; and the concept of fluid stewardship—recognizing when conservative fluid management may yield better outcomes.

Crystalloids vs. Balanced Solutions vs. Albumin: Evidence in Specific Populations

Sepsis and Septic Shock

The question of optimal fluid choice in sepsis has evolved substantially over the past decade. The traditional use of normal saline (0.9% NaCl) has been increasingly challenged by evidence suggesting potential harm from hyperchloremic acidosis and acute kidney injury (AKI).

The SMART trial (2018) compared balanced crystalloids (Lactated Ringer's or Plasma-Lyte A) to saline in 15,802 critically ill patients, finding a lower rate of the composite outcome of death, new renal replacement therapy, or persistent renal dysfunction in the balanced crystalloid group (OR 0.90, 95% CI 0.82-0.99; P=0.04). The SALT-ED trial similarly showed fewer adverse kidney events with balanced crystalloids in non-critically ill patients.

However, the PLUS trial (2022) comparing balanced crystalloids with saline in 5,037 ICU patients found no significant difference in 90-day mortality (21.8% vs. 22.0%, p=0.90), suggesting that the clinical impact of fluid choice may be modest in certain populations.

Regarding albumin, the ALBIOS trial (2014) examined 1,818 patients with severe sepsis, comparing 20% albumin plus crystalloid to crystalloid alone. While no overall mortality difference was seen, a subgroup analysis suggested potential benefit in patients with septic shock (relative risk of death, 0.87; 95% CI, 0.77-0.99).

Traumatic Brain Injury (TBI)

Fluid management in TBI has unique considerations given concerns about cerebral edema and intracranial pressure. The SAFE-TBI study (2007) raised concerns about 4% albumin in TBI patients, showing increased mortality compared with saline (RR 1.63; 95% CI 1.17-2.26; p=0.003). This may relate to albumin's movement across the disrupted blood-brain barrier, potentially exacerbating cerebral edema.

The Hypotonic Solutions and Traumatic Brain Injury (HOT-TBI) trial demonstrated that hypotonic crystalloids were associated with worsened outcomes in TBI, reinforcing the preference for isotonic solutions in this population.

Hypoalbuminemia in Critical Illness

Critical illness often features hypoalbuminemia, raising the question of albumin supplementation. The meta-analysis by Xu et al. (2019) including 20 trials with 13,047 patients found that albumin administration was associated with reduced mortality in patients with severe sepsis and septic shock (RR 0.89; 95% CI 0.80-0.99) but not in general critically ill populations.

The ALBIOS trial specifically demonstrated that albumin administration targeted to maintain serum albumin ≥30 g/L did not improve overall outcomes in severe sepsis or septic shock, but the aforementioned subgroup benefit in septic shock patients has influenced some practice guidelines.

Burn Patients

The Parkland formula has historically guided fluid resuscitation in severe burns, often resulting in large crystalloid volumes. The ALBUR trial (2016) found that adding albumin after the first 12 hours reduced total fluid requirements and lowered the incidence of abdominal compartment syndrome compared with crystalloids alone (OR 0.84; 95% CI 0.72-0.98).

Liver Disease and Hepatorenal Syndrome

Patients with advanced liver disease present unique challenges due to altered hemodynamics and risk of hepatorenal syndrome. The ANSWER trial (2018) demonstrated that long-term albumin administration in cirrhosis with ascites reduced infection rates, renal dysfunction, and mortality (HR 0.62; 95% CI 0.40-0.95).

For hepatorenal syndrome specifically, the combination of albumin with vasoconstrictors such as terlipressin has shown superior efficacy compared to vasoconstrictors alone in the CONFIRM trial (2021), with improved renal function recovery (HR 1.8; 95% CI 1.3-2.5).

Fluid Stewardship: When Less is More

The Evolution of Fluid Management Paradigms

Historical approaches to fluid resuscitation often emphasized aggressive volume expansion ("early goal-directed therapy"), exemplified by the original Rivers trial (2001). However, subsequent large trials (ProCESS, ARISE, ProMISe) failed to demonstrate benefits of this approach, leading to a paradigm shift.

The concept of fluid stewardship has emerged, emphasizing four phases of fluid therapy:

  1. Rescue/Resuscitation (hours 0-1)
  2. Optimization (hours 1-24)
  3. Stabilization (days 2-4)
  4. De-escalation (day 5 onward)

Evidence for Conservative Fluid Strategies

The FACTT trial (2006) compared liberal versus conservative fluid management in 1,000 patients with acute lung injury, finding that the conservative strategy improved lung function and shortened mechanical ventilation duration without increasing non-pulmonary organ failures.

The CLASSIC trial (2022) evaluated restrictive versus standard fluid therapy after initial resuscitation in 1,554 ICU patients with septic shock. While 90-day mortality did not differ significantly (restrictive: 42.3% vs. standard: 42.1%), the restrictive approach resulted in less cumulative fluid at day 5 and fewer adverse events.

For surgical patients, the RELIEF trial (2018) compared restrictive versus liberal fluid strategies in 3,000 major abdominal surgery patients. Contrary to expectations, restrictive fluid management was associated with increased acute kidney injury and surgical site infection, highlighting the context-specific nature of optimal fluid strategies.

Dynamic Assessment of Fluid Responsiveness

Contemporary approaches increasingly emphasize dynamic assessments of fluid responsiveness rather than static targets. Methods include:

  1. Passive leg raising with cardiac output monitoring
  2. Respiratory variation in stroke volume or pulse pressure
  3. End-expiratory occlusion test
  4. Mini-fluid challenges

The ANDROMEDA-SHOCK trial (2019) compared capillary refill time versus serum lactate clearance as resuscitation targets in septic shock patients, finding a trend toward lower mortality with capillary refill-guided resuscitation (HR 0.75; 95% CI 0.55-1.02; p=0.06).

De-resuscitation Strategies

Active de-resuscitation, particularly in patients with positive fluid balance, has gained attention. The ROSE protocol combines Restricted fluid, Oxygenation targets, Spontaneous breathing trials, and Early mobilization to promote fluid removal during the recovery phase.

The RADAR study (2021) showed that protocol-based de-resuscitation in mechanically ventilated patients with fluid overload resulted in greater negative fluid balance and fewer ventilator days, though without mortality benefit.

Practical Recommendations

Patient-Specific Approach to Fluid Selection

Based on current evidence, we recommend:

  1. General critical illness: Balanced crystalloids as first-line therapy for most patients
  2. Septic shock: Consider 20% albumin in addition to crystalloids if persistent hypotension despite initial resuscitation
  3. TBI patients: Avoid albumin; use isotonic crystalloids
  4. Hypoalbuminemia: Consider albumin supplementation only in septic shock with albumin <30 g/L
  5. Liver disease: Early albumin use may be beneficial, especially with hepatorenal syndrome
  6. Burn patients: Consider albumin supplementation after first 12-24 hours

Implementing Fluid Stewardship

  1. Initial resuscitation (0-6 hours): Goal-directed fluid therapy guided by dynamic parameters
  2. Optimization (6-24 hours): Careful fluid challenges only in responders
  3. Stabilization (24-72 hours): Neutral fluid balance goal
  4. De-escalation (>72 hours): Active fluid removal in clinically stable patients

Monitoring Strategies

  1. Combine clinical assessment with objective measures of fluid responsiveness
  2. Focus on tissue perfusion endpoints (e.g., capillary refill time, lactate clearance)
  3. Daily assessment of fluid balance and organ function
  4. Consider point-of-care ultrasound for volume assessment

Conclusion

Fluid management in critically ill patients continues to evolve from a "one-size-fits-all" approach to individualized strategies based on patient characteristics, phase of illness, and physiologic response. The evidence increasingly supports balanced crystalloids as first-line therapy for most patients, with specific indications for albumin in select populations.

The concept of fluid stewardship—with its emphasis on appropriate initial resuscitation followed by judicious ongoing management and active de-resuscitation—represents an important paradigm shift in critical care. Future research should focus on personalized fluid strategies guided by biomarkers, genetic factors, and advanced hemodynamic monitoring to further refine our approach to this fundamental aspect of critical care.

References

  1. Semler MW, Self WH, Wanderer JP, et al. Balanced Crystalloids versus Saline in Critically Ill Adults. N Engl J Med. 2018;378(9):829-839.

  2. Finfer S, Bellomo R, Boyce N, et al. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl J Med. 2004;350(22):2247-2256.

  3. Caironi P, Tognoni G, Masson S, et al. Albumin replacement in patients with severe sepsis or septic shock. N Engl J Med. 2014;370(15):1412-1421.

  4. Myburgh J, Cooper DJ, Finfer S, et al. Saline or albumin for fluid resuscitation in patients with traumatic brain injury. N Engl J Med. 2007;357(9):874-884.

  5. Zampieri FG, Machado FR, Biondi RS, et al. Effect of Intravenous Fluid Treatment With a Balanced Solution vs 0.9% Saline Solution on Mortality in Critically Ill Patients: The BaSICS Randomized Clinical Trial. JAMA. 2021;326(9):818-829.

  6. Finfer S, Micallef S, Hammond N, et al. Balanced Multielectrolyte Solution versus Saline in Critically Ill Adults. N Engl J Med. 2022;386(9):815-826.

  7. National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network. Comparison of two fluid-management strategies in acute lung injury. N Engl J Med. 2006;354(24):2564-2575.

  8. Meyhoff TS, Hjortrup PB, Møller MH, et al. Conservative vs Liberal Fluid Therapy in Septic Shock (CLASSIC) Trial: Protocol and statistical analysis plan. Acta Anaesthesiol Scand. 2019;63(9):1262-1271.

  9. Myles PS, Bellomo R, Corcoran T, et al. Restrictive versus Liberal Fluid Therapy for Major Abdominal Surgery. N Engl J Med. 2018;378(24):2263-2274.

  10. Hernández G, Ospina-Tascón GA, Damiani LP, et al. Effect of a Resuscitation Strategy Targeting Peripheral Perfusion Status vs Serum Lactate Levels on 28-Day Mortality Among Patients With Septic Shock: The ANDROMEDA-SHOCK Randomized Clinical Trial. JAMA. 2019;321(7):654-664.

  11. Caraceni P, Riggio O, Angeli P, et al. Long-term albumin administration in decompensated cirrhosis (ANSWER): an open-label randomised trial. Lancet. 2018;391(10138):2417-2429.

  12. Wong F, Pappas SC, Curry MP, et al. Terlipressin plus Albumin for the Treatment of Type 1 Hepatorenal Syndrome. N Engl J Med. 2021;384(9):818-828.

  13. Xu JY, Chen QH, Xie JF, et al. Comparison of the effects of albumin and crystalloid on mortality in adult patients with severe sepsis and septic shock: a meta-analysis of randomized clinical trials. Crit Care. 2014;18(6):702.

  14. Hjortrup PB, Haase N, Bundgaard H, et al. Restricting volumes of resuscitation fluid in adults with septic shock after initial management: the CLASSIC randomised, parallel-group, multicentre feasibility trial. Intensive Care Med. 2016;42(11):1695-1705.

  15. Self WH, Semler MW, Wanderer JP, et al. Balanced Crystalloids versus Saline in Noncritically Ill Adults. N Engl J Med. 2018;378(9):819-828.

  16. Hoste EA, Maitland K, Brudney CS, et al. Four phases of intravenous fluid therapy: a conceptual model. Br J Anaesth. 2014;113(5):740-747.

  17. Douglas IS, Alapat PM, Corl KA, et al. Fluid Response Evaluation in Sepsis Hypotension and Shock: A Randomized Clinical Trial. Chest. 2020;158(4):1431-1445.

  18. Silversides JA, Major E, Ferguson AJ, et al. Conservative fluid management or deresuscitation for patients with sepsis or acute respiratory distress syndrome following the resuscitation phase of critical illness: a systematic review and meta-analysis. Intensive Care Med. 2017;43(2):155-170.

  19. Rivers E, Nguyen B, Havstad S, et al. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001;345(19):1368-1377.

  20. The ProCESS Investigators. A randomized trial of protocol-based care for early septic shock. N Engl J Med. 2014;370(18):1683-1693.

Managing Multidrug-Resistant Organisms in ICU

 

Managing Multidrug-Resistant Organisms (MDROs) in ICU: Current Approaches

Dr Neeraj Manikath ,Claude.ai

Abstract

Multidrug-resistant organisms (MDROs) represent a significant challenge in intensive care units (ICUs) globally, contributing to increased morbidity, mortality, and healthcare costs. This review examines current strategies for managing MDROs in ICU settings, with particular focus on novel antimicrobial agents such as cefiderocol and ceftazidime-avibactam, as well as infection control bundles and environmental hygiene measures. We also discuss the role of surveillance systems, particularly the WHO Global Antimicrobial Resistance Surveillance System (GLASS), in monitoring and guiding responses to antimicrobial resistance. Evidence suggests that a multifaceted approach combining appropriate antimicrobial therapy, comprehensive infection control measures, and active surveillance is essential for effective MDRO management in critical care environments.

Keywords: multidrug-resistant organisms, intensive care unit, antimicrobial resistance, cefiderocol, ceftazidime-avibactam, infection control bundles, environmental hygiene, WHO GLASS

Introduction

Antimicrobial resistance (AMR) represents one of the most pressing challenges to global public health, with particular impact in intensive care units where vulnerable patients are exposed to multiple risk factors for acquiring multidrug-resistant organisms (MDROs).^1^ The World Health Organization has identified AMR as one of the top ten global public health threats facing humanity, with projections suggesting that by 2050, AMR could cause 10 million deaths annually if left unchecked.^2^

ICUs serve as epicenters for MDRO emergence and transmission due to several factors: high antimicrobial use, critically ill patients with compromised immune systems, frequent use of invasive devices, and close proximity of patients.^3^ Common MDROs encountered in ICUs include methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant enterococci (VRE), extended-spectrum β-lactamase (ESBL)-producing Enterobacterales, carbapenem-resistant Enterobacterales (CRE), multidrug-resistant Pseudomonas aeruginosa, and Acinetobacter baumannii.^4^

This review examines current approaches to managing MDROs in ICU settings, focusing on newer antimicrobial agents, infection control bundles, environmental hygiene strategies, and the role of surveillance systems in guiding clinicians and policymakers.

Novel Antimicrobial Agents

Cefiderocol

Cefiderocol represents a significant advancement in the fight against MDROs, particularly gram-negative pathogens. As a siderophore cephalosporin, cefiderocol employs a unique "Trojan horse" strategy to penetrate bacterial cells by binding to iron and utilizing bacterial iron transport systems to enhance cellular entry.^5^

Mechanism of Action and Spectrum of Activity

Cefiderocol binds to penicillin-binding proteins (PBPs), inhibiting cell wall synthesis. Its distinctive structure provides stability against various β-lactamases, including extended-spectrum β-lactamases, AmpC β-lactamases, and both serine and metallo-carbapenemases.^6^ Cefiderocol demonstrates potent activity against a broad spectrum of gram-negative pathogens, including carbapenem-resistant Enterobacterales, multidrug-resistant Pseudomonas aeruginosa, and Acinetobacter baumannii.^7^

Clinical Evidence in ICU Settings

The CREDIBLE-CR study evaluated cefiderocol versus best available therapy for serious infections caused by carbapenem-resistant gram-negative bacteria. In this study, clinical cure rates were comparable between cefiderocol and best available therapy (52.5% vs. 50.0%), though mortality was numerically higher in the cefiderocol arm.^8^ The APEKS-NP trial demonstrated non-inferiority of cefiderocol compared to meropenem for hospital-acquired pneumonia, including ventilator-associated pneumonia, with comparable clinical cure rates and safety profiles.^9^

Dosing Considerations in Critical Care

For patients with normal renal function, the standard dosage is 2g administered intravenously every 8 hours, with dose adjustments required for patients with renal impairment. Therapeutic drug monitoring may be beneficial in critically ill patients due to pharmacokinetic variability.^10^

Ceftazidime-Avibactam

Ceftazidime-avibactam combines a third-generation cephalosporin with a novel non-β-lactam β-lactamase inhibitor that extends its activity against many resistant gram-negative organisms.

Mechanism of Action and Spectrum of Activity

Avibactam is a diazabicyclooctane β-lactamase inhibitor that covalently binds to serine β-lactamases, including extended-spectrum β-lactamases (ESBLs), AmpC enzymes, and some carbapenemases (KPC). This combination effectively restores ceftazidime's activity against many resistant gram-negative bacteria.^11^ Ceftazidime-avibactam shows activity against most Enterobacterales, including ESBL and KPC carbapenemase producers, as well as P. aeruginosa, but has limited activity against Acinetobacter species and organisms producing metallo-β-lactamases (MBLs).^12^

Clinical Evidence in ICU Settings

The REPRISE trial demonstrated efficacy of ceftazidime-avibactam against ceftazidime-resistant Enterobacterales and P. aeruginosa infections, with clinical cure rates of 91% for ceftazidime-avibactam versus 71% for best available therapy.^13^ A meta-analysis of observational studies showed improved survival with ceftazidime-avibactam compared to colistin-based regimens for carbapenem-resistant Enterobacterales infections (OR 0.37, 95% CI 0.21-0.66).^14^

Dosing and Combination Strategies

The standard dose is 2.5g (2g ceftazidime + 0.5g avibactam) administered intravenously every 8 hours for patients with normal renal function, with dose adjustments for renal impairment. In severe infections caused by highly resistant pathogens, combination therapy may be considered, often with aminoglycosides or polymyxins.^15^

Other Promising Agents

Meropenem-Vaborbactam

This combination of carbapenem and boronic acid-based β-lactamase inhibitor demonstrates activity against KPC-producing Enterobacterales. The TANGO II trial showed superiority over best available therapy for CRE infections with higher clinical cure rates (65.6% vs. 33.3%) and lower nephrotoxicity.^16^

Imipenem-Relebactam

Combining imipenem with the novel β-lactamase inhibitor relebactam extends activity against many carbapenem-resistant gram-negative pathogens. Clinical trials have demonstrated non-inferiority to imipenem-cilastatin for hospital-acquired and ventilator-associated pneumonia.^17^

Plazomicin

This next-generation aminoglycoside was designed to overcome common aminoglycoside resistance mechanisms. The CARE trial demonstrated efficacy in carbapenem-resistant Enterobacterales infections, though limited by a small sample size.^18^

Infection Control Bundles

Definition and Components

Infection control bundles represent coordinated sets of evidence-based interventions that, when implemented together, achieve better outcomes than when implemented individually. These bundles typically include several key components:

  1. Hand hygiene protocols: Implementation of the WHO's "Five Moments for Hand Hygiene" with regular compliance monitoring.^19^
  2. Contact precautions: Including appropriate use of personal protective equipment (PPE) such as gloves and gowns.
  3. Patient isolation or cohorting: Physical separation of MDRO-colonized or infected patients.
  4. Active surveillance cultures: Targeted or universal screening to identify asymptomatic carriers.
  5. Antimicrobial stewardship: Optimizing antimicrobial use through appropriate selection, dosing, and duration.
  6. Healthcare worker education: Regular training on MDRO transmission and prevention.

Evidence of Effectiveness

Multiple studies have demonstrated the effectiveness of comprehensive infection control bundles in reducing MDRO transmission and infection rates in ICU settings. A systematic review by Tomczyk et al. found that multimodal interventions reduced the acquisition of MDROs by 37% in acute care settings.^20^

The REDUCE MRSA trial, a cluster-randomized study involving 43 hospitals, demonstrated that a bundle including universal decolonization reduced MRSA clinical isolates by 36.5% and bloodstream infections from any pathogen by 44%.^21^ Similarly, a quasi-experimental study in Greek ICUs demonstrated that implementation of a comprehensive bundle reduced carbapenem-resistant A. baumannii infections by 58% over a two-year period.^22^

Implementation Challenges

Despite proven effectiveness, implementation challenges include:

  1. Resource constraints: Limited personnel, time, and financial resources.
  2. Compliance issues: Difficulty maintaining sustained adherence to all bundle elements.
  3. Cultural barriers: Resistance to changing established practices.
  4. Monitoring burden: Challenges in measuring compliance and outcomes.

Strategies for Successful Implementation

Successful implementation strategies include:

  1. Leadership engagement: Securing support from institutional leadership and clinical champions.
  2. Multidisciplinary approach: Involving physicians, nurses, pharmacists, infection preventionists, and environmental services.
  3. Regular feedback: Providing performance data to frontline staff.
  4. Adaptation to local context: Tailoring interventions to specific institutional needs and resources.
  5. Use of implementation science frameworks: Employing structured approaches to translate evidence into practice.^23^

Environmental Hygiene

Importance in MDRO Transmission

Environmental contamination plays a significant role in MDRO transmission within ICUs. Studies have demonstrated that pathogens can persist on environmental surfaces for extended periods, with C. difficile spores surviving for months, and MRSA and VRE surviving for days to weeks.^24^ Patients admitted to rooms previously occupied by MDRO-positive patients have a significantly higher risk of acquiring the same organism, highlighting the importance of effective terminal cleaning.^25^

Evidence-Based Cleaning and Disinfection Practices

Standard Cleaning Protocols

Evidence supports a systematic approach to environmental cleaning in ICUs:

  1. Defined responsibility: Clear assignment of cleaning responsibilities for all environmental surfaces and equipment.
  2. Appropriate product selection: Use of EPA-registered hospital-grade disinfectants with documented activity against relevant pathogens.
  3. Correct application: Adherence to manufacturer recommendations for concentration, contact time, and application method.
  4. Frequency: Increased cleaning frequency for high-touch surfaces.

Advanced Disinfection Technologies

Several technologies have emerged to supplement standard cleaning practices:

  1. Ultraviolet (UV) light systems: UV-C devices have demonstrated efficacy in reducing environmental bioburden. A multicenter randomized trial showed that adding UV-C disinfection to standard terminal cleaning reduced acquisition of target MDROs by 30%.^26^

  2. Hydrogen peroxide vapor/aerosolized hydrogen peroxide: These systems provide enhanced disinfection with studies demonstrating significant reductions in environmental contamination and some showing decreases in MDRO transmission rates.^27^

  3. Continuously active disinfectants: Surfaces with persistent antimicrobial activity offer promise for maintaining cleanliness between cleaning cycles.^28^

Monitoring Cleaning Effectiveness

Several methods to evaluate cleaning effectiveness include:

  1. Visual inspection: Limited sensitivity but provides immediate feedback.
  2. Fluorescent markers: Allow objective assessment of cleaning thoroughness.
  3. ATP bioluminescence: Measures organic material remaining on surfaces.
  4. Microbial sampling: Direct assessment of microbial contamination through swabs or contact plates.

A multimodal approach incorporating education, feedback using objective monitoring tools, and defined cleaning protocols has been shown to improve cleaning effectiveness and reduce environmental contamination.^29^

WHO Global Antimicrobial Resistance Surveillance System (GLASS)

Overview and Objectives

The WHO Global Antimicrobial Resistance Surveillance System (GLASS), launched in 2015, represents the first global collaborative effort to standardize AMR surveillance. Its primary objectives include:

  1. Fostering national AMR surveillance systems
  2. Harmonizing global data collection, analysis, and sharing
  3. Detecting emerging resistance trends and informing containment strategies
  4. Supporting evidence-based policy development^30^

Structure and Implementation

GLASS employs a phased implementation approach focused on:

  1. National coordination: Establishing National Coordinating Centers responsible for data collection and quality assurance.
  2. Standardized methodologies: Promoting uniform laboratory techniques and interpretive criteria.
  3. Priority pathogens: Initially focusing on key bacterial pathogens, with expansion to include fungi, viruses, and parasites.
  4. Capacity building: Supporting laboratory infrastructure and training in resource-limited settings.

As of 2023, over 100 countries participate in GLASS, representing more than 70% of the world's population.^31^

Impact on ICU Practice

GLASS data directly impacts ICU practice through:

  1. Informing empiric therapy guidelines: Local and regional resistance data guide appropriate initial antimicrobial selection.
  2. Alerting to emerging threats: Early detection of novel resistance mechanisms allows proactive containment.
  3. Resource allocation: Identification of high-priority pathogens directs infection control resources.
  4. Research priorities: Surveillance data highlight knowledge gaps requiring investigation.

Integration with Clinical Decision Support

Integration of GLASS data with clinical decision support systems represents an evolving area with potential to optimize antimicrobial prescribing in ICUs. Studies have demonstrated that incorporating local antibiogram data into electronic prescribing systems can improve appropriate empiric therapy and reduce broad-spectrum antibiotic use.^32^

Practical Implementation Strategies

Multidisciplinary Antimicrobial Stewardship

Effective antimicrobial stewardship programs (ASPs) in ICUs require:

  1. Multidisciplinary team: Including infectious disease physicians, clinical pharmacists, microbiologists, and ICU staff.
  2. Prospective audit and feedback: Regular review of antimicrobial prescribing with direct feedback to prescribers.
  3. Pre-authorization: Requiring approval for certain antimicrobials.
  4. Institutional guidelines: Development of evidence-based treatment protocols incorporating local resistance data.
  5. Education: Regular training on appropriate antimicrobial use.

A meta-analysis of ASP interventions in ICUs demonstrated reductions in antimicrobial consumption (reduction range: 11-38%) without adversely affecting patient outcomes.^33^

Risk Assessment and Stratification

Identifying high-risk patients allows targeted interventions:

  1. Risk factors for MDRO colonization/infection: Previous MDRO history, recent hospitalization, antimicrobial exposure, presence of invasive devices, and comorbidities.
  2. Risk-based screening: Focusing surveillance cultures on high-risk populations.
  3. Preemptive isolation: Implementing contact precautions for high-risk patients pending screening results.

Staff Education and Engagement

Successful programs emphasize:

  1. Regular training: Updated education on current MDRO epidemiology and prevention strategies.
  2. Skills development: Hands-on training for proper PPE use and environmental cleaning.
  3. Feedback mechanisms: Sharing performance data to motivate improvement.
  4. Champion identification: Engaging influential clinicians to model and promote best practices.

Technology and Innovation

Emerging technologies support MDRO management:

  1. Rapid diagnostic tests: Molecular methods allowing faster identification of MDROs and resistance determinants.
  2. Electronic surveillance systems: Automated alerts for potential outbreaks or high-risk patients.
  3. Automated hand hygiene monitoring: Systems providing real-time feedback on compliance.
  4. Predictive analytics: Algorithms identifying patients at high risk for MDRO acquisition or infection.

Future Directions

Novel Therapeutic Approaches

Beyond traditional antimicrobials, emerging approaches include:

  1. Bacteriophage therapy: Viruses that specifically target bacteria, including MDROs, with early clinical trials showing promise for difficult-to-treat infections.^34^
  2. Monoclonal antibodies: Targeting specific bacterial virulence factors or toxins.
  3. Microbiome-based interventions: Fecal microbiota transplantation and probiotics to prevent MDRO colonization.
  4. Antimicrobial peptides: Natural and synthetic peptides with broad-spectrum activity against MDROs.
  5. Anti-virulence strategies: Targeting bacterial virulence without selecting for resistance.

Emerging Surveillance Technologies

Advancements in surveillance include:

  1. Whole genome sequencing: Providing detailed understanding of resistance mechanisms and transmission patterns.
  2. Metagenomics: Analyzing complex microbial communities directly from clinical samples.
  3. Machine learning algorithms: Predicting outbreaks and optimizing control measures.
  4. Global data sharing platforms: Enhanced integration of surveillance data across institutions and countries.

Health System Approaches

Systemic changes to address MDROs include:

  1. Value-based incentives: Financial models rewarding infection prevention success.
  2. Regional collaboratives: Coordinated approaches across healthcare facilities.
  3. One Health approach: Integrating human, animal, and environmental health strategies.
  4. Global policy coordination: Harmonized international efforts to combat AMR.

Conclusion

Managing MDROs in ICU settings requires a comprehensive, multifaceted approach. Novel antimicrobials like cefiderocol and ceftazidime-avibactam provide valuable therapeutic options, but must be used judiciously within robust antimicrobial stewardship programs. Infection control bundles and environmental hygiene practices represent essential components of effective MDRO management strategies. The WHO GLASS program provides crucial surveillance data to guide local, national, and global responses.

Future success in combating MDROs will depend on continued innovation in therapeutics, diagnostics, and infection prevention, coupled with strong institutional commitment, interdisciplinary collaboration, and global coordination. By implementing evidence-based strategies and remaining vigilant for emerging threats, healthcare providers can optimize outcomes for critically ill patients while preserving antimicrobial efficacy for future generations.

References

  1. Bassetti M, Peghin M, Vena A, Giacobbe DR. Treatment of infections due to MDR gram-negative bacteria. Front Med. 2019;6:74. doi:10.3389/fmed.2019.00074

  2. World Health Organization. Global action plan on antimicrobial resistance. Geneva: WHO; 2015.

  3. Timsit JF, Bassetti M, Cremer O, et al. Rationalizing antimicrobial therapy in the ICU: a narrative review. Intensive Care Med. 2019;45(2):172-189. doi:10.1007/s00134-019-05520-5

  4. Tacconelli E, Carrara E, Savoldi A, et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis. 2018;18(3):318-327. doi:10.1016/S1473-3099(17)30753-3

  5. Zhanel GG, Golden AR, Zelenitsky S, et al. Cefiderocol: a siderophore cephalosporin with activity against carbapenem-resistant and multidrug-resistant gram-negative bacilli. Drugs. 2019;79(3):271-289. doi:10.1007/s40265-019-1055-2

  6. Kazmierczak KM, Tsuji M, Wise MG, et al. In vitro activity of cefiderocol, a siderophore cephalosporin, against a recent collection of clinically relevant carbapenem-non-susceptible gram-negative bacilli, including serine carbapenemase- and metallo-β-lactamase-producing isolates (SIDERO-WT-2014 Study). Int J Antimicrob Agents. 2019;53(2):177-184. doi:10.1016/j.ijantimicag.2018.10.007

  7. Hackel MA, Tsuji M, Yamano Y, et al. In vitro activity of the siderophore cephalosporin, cefiderocol, against a recent collection of clinically relevant gram-negative bacilli from North America and Europe, including carbapenem-nonsusceptible isolates (SIDERO-WT-2014 Study). Antimicrob Agents Chemother. 2017;61(9):e00093-17. doi:10.1128/AAC.00093-17

  8. Bassetti M, Echols R, Matsunaga Y, et al. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant gram-negative bacteria (CREDIBLE-CR): a randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect Dis. 2021;21(2):226-240. doi:10.1016/S1473-3099(20)30796-9

  9. Wunderink RG, Matsunaga Y, Ariyasu M, et al. Cefiderocol versus high-dose, extended-infusion meropenem for the treatment of gram-negative nosocomial pneumonia (APEKS-NP): a randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect Dis. 2021;21(2):213-225. doi:10.1016/S1473-3099(20)30731-3

  10. Katsube T, Wajima T, Ishibashi T, et al. Pharmacokinetic/pharmacodynamic modeling and simulation of cefiderocol, a parenteral siderophore cephalosporin, for dose adjustment based on renal function. Antimicrob Agents Chemother. 2017;61(1):e01381-16. doi:10.1128/AAC.01381-16

  11. Shirley M. Ceftazidime-avibactam: a review in the treatment of serious gram-negative bacterial infections. Drugs. 2018;78(6):675-692. doi:10.1007/s40265-018-0902-x

  12. Lagacé-Wiens P, Walkty A, Karlowsky JA. Ceftazidime-avibactam: an evidence-based review of its pharmacology and potential use in the treatment of gram-negative bacterial infections. Core Evid. 2014;9:13-25. doi:10.2147/CE.S40698

  13. Carmeli Y, Armstrong J, Laud PJ, et al. Ceftazidime-avibactam or best available therapy in patients with ceftazidime-resistant Enterobacteriaceae and Pseudomonas aeruginosa complicated urinary tract infections or complicated intra-abdominal infections (REPRISE): a randomised, pathogen-directed, phase 3 study. Lancet Infect Dis. 2016;16(6):661-673. doi:10.1016/S1473-3099(16)30004-4

  14. Tumbarello M, Trecarichi EM, Corona A, et al. Efficacy of ceftazidime-avibactam salvage therapy in patients with infections caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae. Clin Infect Dis. 2019;68(3):355-364. doi:10.1093/cid/ciy492

  15. Falcone M, Paterson D. Spotlight on ceftazidime/avibactam: a new option for MDR gram-negative infections. J Antimicrob Chemother. 2016;71(10):2713-2722. doi:10.1093/jac/dkw239

  16. Wunderink RG, Giamarellos-Bourboulis EJ, Rahav G, et al. Effect and safety of meropenem-vaborbactam versus best-available therapy in patients with carbapenem-resistant Enterobacteriaceae infections: the TANGO II randomized clinical trial. Infect Dis Ther. 2018;7(4):439-455. doi:10.1007/s40121-018-0214-1

  17. Motsch J, Murta de Oliveira C, Stus V, et al. RESTORE-IMI 1: a multicenter, randomized, double-blind trial comparing efficacy and safety of imipenem/relebactam vs colistin plus imipenem in patients with imipenem-nonsusceptible bacterial infections. Clin Infect Dis. 2020;70(9):1799-1808. doi:10.1093/cid/ciz530

  18. McKinnell JA, Dwyer JP, Talbot GH, et al. Plazomicin for infections caused by carbapenem-resistant Enterobacteriaceae. N Engl J Med. 2019;380(8):791-793. doi:10.1056/NEJMc1807634

  19. World Health Organization. WHO guidelines on hand hygiene in health care. Geneva: WHO; 2009.

  20. Tomczyk S, Zanichelli V, Grayson ML, et al. Control of carbapenem-resistant Enterobacteriaceae, Acinetobacter baumannii, and Pseudomonas aeruginosa in healthcare facilities: a systematic review and reanalysis of quasi-experimental studies. Clin Infect Dis. 2019;68(5):873-884. doi:10.1093/cid/ciy752

  21. Huang SS, Septimus E, Kleinman K, et al. Targeted versus universal decolonization to prevent ICU infection. N Engl J Med. 2013;368(24):2255-2265. doi:10.1056/NEJMoa1207290

  22. Kofteridis DP, Valachis A, Dimopoulou D, et al. Effectiveness of a bundle approach in preventing multidrug-resistant A. baumannii infections in intensive care units. Int J Antimicrob Agents. 2018;52(2):247-252. doi:10.1016/j.ijantimicag.2018.03.019

  23. Davey P, Marwick CA, Scott CL, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev. 2017;2(2):CD003543. doi:10.1002/14651858.CD003543.pub4

  24. Weber DJ, Anderson D, Rutala WA. The role of the surface environment in healthcare-associated infections. Curr Opin Infect Dis. 2013;26(4):338-344. doi:10.1097/QCO.0b013e3283630f04

  25. Mitchell BG, Dancer SJ, Anderson M, Dehn E. Risk of organism acquisition from prior room occupants: a systematic review and meta-analysis. J Hosp Infect. 2015;91(3):211-217. doi:10.1016/j.jhin.2015.08.005

  26. Anderson DJ, Chen LF, Weber DJ, et al. Enhanced terminal room disinfection and acquisition and infection caused by multidrug-resistant organisms and Clostridium difficile (the Benefits of Enhanced Terminal Room Disinfection study): a cluster-randomised, multicentre, crossover study. Lancet. 2017;389(10071):805-814. doi:10.1016/S0140-6736(16)31588-4

  27. Passaretti CL, Otter JA, Reich NG, et al. An evaluation of environmental decontamination with hydrogen peroxide vapor for reducing the risk of patient acquisition of multidrug-resistant organisms. Clin Infect Dis. 2013;56(1):27-35. doi:10.1093/cid/cis839

  28. Rutala WA, Weber DJ. Best practices for disinfection of noncritical environmental surfaces and equipment in health care facilities: a bundle approach. Am J Infect Control. 2019;47S:A96-A105. doi:10.1016/j.ajic.2019.01.014

  29. Carling PC, Bartley JM. Evaluating hygienic cleaning in health care settings: what you do not know can harm your patients. Am J Infect Control. 2010;38(5 Suppl 1):S41-50. doi:10.1016/j.ajic.2010.03.004

  30. World Health Organization. Global antimicrobial resistance surveillance system (GLASS) report: early implementation 2017-2018. Geneva: WHO; 2019.

  31. World Health Organization. Global antimicrobial resistance and use surveillance system (GLASS) report: 2022. Geneva: WHO; 2023.

  32. Forrest GN, Van Schooneveld TC, Kullar R, et al. Use of electronic health records and clinical decision support systems for antimicrobial stewardship. Clin Infect Dis. 2014;59(suppl_3):S122-S133. doi:10.1093/cid/ciu565

  33. Karanika S, Paudel S, Grigoras C, et al. Systematic review and meta-analysis of clinical and economic outcomes from the implementation of hospital-based antimicrobial stewardship programs. Antimicrob Agents Chemother. 2016;60(8):4840-4852. doi:10.1128/AAC.00825-16

  34. Kortright KE, Chan BK, Koff JL, Turner PE. Phage therapy: a renewed approach to combat antibiotic-resistant bacteria. Cell Host Microbe. 2019;25(2):219-232. doi:10.1016/j.chom.2019.01.014