ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Treatment of AA (secondary) amyloidosis

Treatment of AA (secondary) amyloidosis
Literature review current through: Jan 2024.
This topic last updated: Feb 18, 2022.

INTRODUCTION — AA (secondary) amyloidosis is a disorder characterized by the extracellular tissue deposition of fibrils composed of fragments of serum amyloid A protein (SAA), an acute phase reactant. AA amyloidosis may complicate a number of chronic inflammatory conditions, including rheumatoid arthritis (RA), juvenile idiopathic arthritis, ankylosing spondylitis (AS), inflammatory bowel disease, familial periodic fever syndromes, chronic infections, and certain neoplasms (table 1) [1,2].

The treatment of AA amyloidosis is presented here. The pathogenesis, clinical manifestations, major causes, and approach to diagnosis of AA amyloidosis are presented separately, as are the clinical manifestations and treatment of the musculoskeletal and renal manifestations of amyloid diseases. (See "Pathogenesis of AA amyloidosis" and "Overview of amyloidosis", section on 'Clinical manifestations' and "Causes and diagnosis of AA amyloidosis and relation to rheumatic diseases" and "Musculoskeletal manifestations of amyloidosis" and "Renal amyloidosis".)

PROGNOSIS — If untreated, AA amyloidosis is a serious disease with a significant mortality due to end-stage kidney disease, infection, heart failure, bowel perforation, or gastrointestinal bleeding [3-5]. Patients with persistently high circulating levels of serum amyloid A protein (SAA) are at particular risk of these complications [6,7]. A progressive improvement in survival rates has been reported among patients with AA amyloidosis, likely reflecting, in part, improved treatment strategies for associated inflammatory disorders, as well as earlier detection [2,3,8,9].

In a review of 374 cases of AA amyloidosis, for example, median survival from diagnosis was 133 months, with older age, reduced serum albumin concentration, end-stage renal failure at baseline, and the degree by which the SAA concentration was elevated during follow-up being of poorer prognostic significance [3].

In a review of 48 cases of AA amyloidosis undergoing kidney transplantation, recurrence rate in those with persistent SAA elevations was 3 times those with the lowest mean SAA levels achieved by suppression post-transplant [10].

Importance of control of the underlying disease — The fibril precursor in AA amyloid, SAA, is a normal plasma protein that is produced by hepatocytes as part of the physiologic acute phase response. A chronic inflammatory state leads to sustained high levels of the acute phase proteins [11]. Successful treatment of the underlying inflammatory process by, for example, surgical resection of the focus of infection or the tumor, cytotoxic agents and/or biologic agents in rheumatoid arthritis (RA), or antibiotics with chronic infection, results in reduced hepatic production of the acute phase response and a fall in circulating SAA down to normal healthy levels. Over time, this can lead to stabilization of or improvement in renal function, to reduction in urinary protein excretion, and to partial resolution of amyloid deposits (as assessed in studies by scintigraphy with radiolabeled serum amyloid P component [SAP]) [12-18]. This was illustrated in a report of 80 patients with AA amyloid, mostly due to juvenile idiopathic arthritis or to RA, who were prospectively followed for a median of four years; in this series, the systemic amyloid load was assessed by yearly SAP scintigraphy [19]. The following findings were noted:

Forty-two patients had median serum SAA concentration within the reference range (<10 mg/L); amyloid deposits regressed in 25 and stabilized in 14. Among patients with renal disease at baseline, proteinuria typically fell, while the serum creatinine concentration was either stable or improved. Two patients who required dialysis remained on dialysis.

The outcomes were variable in the other patients. However, among those in whom the serum SAA was persistently above 50 mg/L, the amyloid load usually increased, and organ function deteriorated. In one patient who underwent kidney transplantation, proteinuria and renal amyloid deposits recurred within 36 months.

There was a variable relation between changes in amyloid load and changes in organ function. In some patients, for example, renal function was stable despite a reduction in amyloid load, while others had improved renal function despite a stable amyloid load.

The estimated 10-year survival was much higher in the patients with median serum SAA below 10 mg/L (90 versus 40 percent in those with higher median values), with 60 percent regression as assessed by I-123-labeled SAP scintigraphy, if this level was maintained [2,3,19].

Similar findings have been noted in other uncontrolled series [12,13,16-18,20-22]. As examples:

In a 10- to 20-year follow-up of 16 adults given alkylating agents because of AA amyloid due to rheumatic disease (RA, ankylosing spondylitis, or juvenile chronic polyarthritis), treated patients had substantial preservation of renal function compared with 48 untreated controls [12].

In three patients with Crohn disease who underwent kidney transplantation because of renal amyloidosis, immunosuppressive therapy led to remission of the inflammatory bowel disease [13]. Serial SAP scintigraphy demonstrated regression of amyloid deposits in two patients and stabilization of amyloid in the third.

In four patients with systemic AA amyloidosis complicating Castleman disease (angiofollicular lymph node hyperplasia), resection of the tumors may have led to clinical regression of the amyloid, which was also corroborated by SAP scintigraphy [15,23].

TREATMENT OF AA AMYLOIDOSIS — As illustrated by the preceding observations, the preferred therapy of AA amyloidosis is control of the underlying inflammatory disease and thus near complete suppression of serum amyloid A protein (SAA) production. Treatment needs to be specific for the underlying disease; for example, colchicine is the accepted therapy for AA amyloidosis complicating familial Mediterranean fever (FMF), and agents active against proinflammatory cytokines (interleukin [IL] 1 beta, tumor necrosis factor [TNF] alpha, IL-6) have been shown to have efficacy in some cases of AA amyloidosis due to rheumatic disorders and to hereditary autoinflammatory disease. Eprodisate, a glycosaminoglycan mimetic that was developed to interfere with fibril formation in AA amyloidosis, was withdrawn from clinical development after phase 3 studies failed to show benefit. Agents that enhance clearance of amyloid from tissue are undergoing clinical studies. The efficacy of dialysis and kidney transplantation in those patients who progress to end-stage kidney disease is discussed separately. (See "Renal amyloidosis".)

Colchicine — Colchicine has become accepted prophylactic therapy to prevent AA amyloidosis in FMF, where doses of 0.6 to 1.2 mg/day in adults markedly reduced the frequency of attacks of abdominal pain, diminished the incidence of clinical renal disease (including prevention of recurrence in the kidney transplant), and stabilized the glomerular filtration rate in patients with mild proteinuria. In the large series reported from Tel Hashomer Hospital in Israel, the rate of development of proteinuria at 9 to 11 years was 1.7 in 960 compliers versus 49 percent in 54 non-compliers [24]. In 86 patients who already had nonnephrotic proteinuria prior to therapy, colchicine led to resolution of proteinuria in five and to stabilization in 68. (See "Management of familial Mediterranean fever".)

Among FMF patients with nephrotic syndrome due to AA amyloid, prevention of disease progression and a reduction in protein excretion can be achieved. However, a higher colchicine dose of 1.5 to 2.0 mg/day appears to be required, and therapy should be instituted before the plasma creatinine concentration reaches 1.5 mg/dL (132 micromol/L) [25,26]. Protein excretion in responders can be reduced to below 1 to 2 grams/day with this regimen; the benefit is gradual, occurring over a one- to two-year period [25]. A small number of patients do, however, appear to be resistant to the effects of colchicine, possibly reflecting genetic subsets of underlying disease or variable pharmacokinetics of the drug and anti-IL-1 therapies are the second-line treatment of choice [27,28].

Colchicine is not likely to be effective in salvaging renal function in FMF patients who already have chronic renal failure, since irreversible glomerular injury is probably present. However, it can prevent recurrent disease in the transplant and prevent progressive amyloid deposition in other sites [26]. The optimal colchicine dose in this setting is 1.5 to 2.0 mg/day; lower doses are less predictably effective. (See "Renal amyloidosis".)

Anecdotal successes with chronic colchicine therapy (0.6 mg twice daily) have also been reported in patients with AA amyloid due to inflammatory bowel disease, in patients with Behçet syndrome, and in intravenous drug users with suppurative skin lesions. Proteinuria due to renal amyloidosis may be markedly reduced, and renal function may remain stable over long periods of time [29-32]. However, repeat renal biopsy in one such patient did not demonstrate a reduction in total amyloid deposition despite almost complete resolution of proteinuria [31].

Dimethylsulfoxide — Dimethylsulfoxide (DMSO) may have anti-amyloid activity in several forms of the disorder; it has been shown to be active in murine models of AA amyloidosis and in case reports of patients with AA amyloid due to rheumatoid arthritis (RA) or to Crohn disease [33-35]. Use of DMSO is limited, however, by its acrid odor and, because of its potent solvent activity, by the difficulty attaining purity for pharmaceutical usage [36].

Cytotoxic and immunosuppressive agents — Azathioprine, chlorambucil, methotrexate, and cyclophosphamide have been shown to be helpful in AA amyloidosis complicating treatment-responsive inflammatory diseases in individual case reports and small series. As examples:

A small retrospective study reported that cyclophosphamide may provide a significant survival benefit in patients with RA and renal AA amyloid [22]. In this study of 15 French patients, of whom six received monthly cyclophosphamide following confirmation of renal involvement, the survival of patients treated with pulse cyclophosphamide was longer than that of those administered non-alkylating drug regimens (mean survival of 165 and 46 months, respectively) [22]. Trends toward decreased proteinuria and maintenance of renal function were also noted with cyclophosphamide.

Similar results were confirmed in a cohort study reported from Japan [17]. Prospective studies are required to properly assess the role and toxicity of this agent in this setting.

Anticytokine therapy — Increasing use of biologics with activities against proinflammatory cytokines (TNF-alpha, IL-1, and IL-6) for diseases such as RA, psoriatic arthritis, and ankylosing spondylitis (AS) may reduce the risk of development of AA amyloid as well as treating existing amyloidosis.

The majority of published experience that has shown efficacy has involved patients with RA and has reported on the first-generation TNF-alpha antagonists (ie, etanercept, infliximab, and adalimumab) [37-40]. In one large study involving 86 patients with RA complicated by AA amyloidosis, etanercept was superior to cyclophosphamide independent of the SAA 1.3 allele [41]. Less information is available regarding the newer TNF-alpha antagonists (certolizumab pegol and golimumab).

The effectiveness of the TNF antagonists appears to be directly related to their ability to control the underlying inflammatory disorder and may be enhanced by pulse therapy with glucocorticoids for induction [42]. This experience has been extended to AA amyloidosis complicating AS and Crohn disease, with regression of tissue amyloid occurring as early as three months, documented by gastrointestinal biopsies in some instances [43-45]. Efficacy of biologic response modifiers (BRMs) may be responsible in part for the marked decline in the incidence of renal replacement therapy for amyloidosis associated with inflammatory rheumatic diseases reported to nationwide registries in Finland during the period 1999 to 2008 [46].

Efficacy of the rapidly acting IL-1 receptor antagonist (IL-1ra) anakinra has been established for the 20 to 30 percent of cases of "idiopathic" AA amyloidosis reported in large series [47] and is effective in retarding recurrence of AA amyloid in combination with colchicine after kidney transplantation [48].

Since 2006, with the first report of the effectiveness of a humanized anti-IL-6 receptor antibody (tocilizumab) for the treatment of AA amyloidosis complicating juvenile chronic polyarthritis, there has been a large experience with the use of this biologic agent to improve AA amyloid complicating adult RA, as well as isolated case reports in patients with ankylosing spondylitis, Behçet syndrome, polyarteritis nodosa, Castleman disease, Sweet syndrome, and Crohn disease [49-54]. Tocilizumab has been used effectively in patients with RA and renal insufficiency due to AA amyloid [55], with biologic activity manifesting as a dramatic and rapid drop in C-reactive protein (CRP) and SAA levels to normal compared with more traditional treatments, such as methotrexate or TNF-alpha antagonists, with a significantly lower incidence of progression to hemodialysis over time [56]. In a retrospective review of 42 patients with RA and AA amyloidosis treated with tocilizumab or TNF antagonists, the former was more efficient in normalizing SAA levels, and was a more effective strategy for treating AA amyloidosis [57]. By contrast, although TNF inhibitors may reduce SAA levels in clinical practice, complete normalization is rare [52].

Only limited information is available regarding the efficacy of other biologics, such as abatacept and rituximab, which are effective in reducing disease activity but do not have direct anticytokine activity, in reversing the development of AA amyloid [58,59]. A possible therapeutic effect for IL-17 inhibition with regard to extrahepatic SAA synthesis in inflammatory rheumatic diseases has yet to be explored [60].

The impact of anticytokine therapy in preventing AA amyloid in hereditary autoinflammatory diseases, such as FMF, TNF receptor-associated periodic syndrome (TRAPS), or Muckle-Wells disease, is increasingly clear. AA amyloidosis may complicate conditions related to pyrin mutations (in particular, homozygosity for the M694V mutation in MEFV), with, and, very rarely, without the phenotype of FMF, TRAPS, the full spectrum of cryopyrinopathies (familial cold autoinflammatory syndrome, which is also termed familial cold urticaria; Muckle-Wells syndrome; and neonatal onset multisystem inflammatory disorder), and mevalonate kinase deficiency (also known as hyperimmunoglobulinemia D syndrome), with varying incidences depending upon disease and ethnic background [61].

The bulk of early experience has focused on the central role of the NALP3 inflammasome, with mutations resulting in constitutive overproduction of IL-1 beta in the cryopyrinopathies, and on the efficacy of anakinra in attenuating both disease and resulting amyloid [62-64]. Similar experience has accrued with the use of the long-acting anti-IL-1 monoclonal antibody canakinumab before and after kidney transplantation [65,66]. As yet, less information is available regarding the potential efficacy for AA amyloidosis of other synthetic agents (eg, caspase 1 inhibitors) or alternative anticytokines (eg, IL-1 TRAP or IL-18 inhibitors) that may be effective in reducing cryopyrinopathy disease manifestations [67]. With regard to TRAPS and FMF, both etanercept and anakinra have been reported as effective treatments for disease manifestations and for lowering of SAA levels, although anti-IL-1 treatments are increasingly favored [68,69]. However, similar efficacy to that seen with etanercept has not been reported with use of other TNF inhibitors, infliximab and adalimumab, in patients with TRAPS [61], and whether these agents are effective for preventing or reversing AA amyloidosis has not been described. IL-6 receptor blockade with tocilizumab may act downstream of inflammasome activation and has been reported to be an effective therapy for AA amyloidosis complicating both FMF and TRAPS; in a series of 20 patients with autoinflammatory disease and AA amyloidosis, including four patients with kidney transplants, a rapid drop in SAA levels to normal occurred within 10 days and was sustained through 23 months, with stabilization or regression of amyloid on SAP scintigraphy [70].

Anticytokine therapy remains an option for patients presenting with renal insufficiency who undergo kidney transplantation as prophylaxis to prevent recurrence of AA amyloid in the allograft; in particular, IL-1 inhibitors may be especially beneficial in this setting [69,71]. In addition, the calcineurin inhibitor FK506 (tacrolimus), which is commonly used to prevent allograft rejection, may also slow progression of AA amyloid, based on animal studies [72]. Tocilizumab was also shown to be beneficial for AA amyloidosis in a family with the SAA1.1 haplotype linked to a polymorphism in the SAA promoter [73].

Demonstration of the efficacy of available anticytokines holds the promise that novel monoclonal antibodies and low molecular weight (mw) inhibitors in phase 2/3 trials may also prove useful for the inhibition of the deposition phase of SAA amyloid [74]. In particular, tofacitinib, which is approved for the treatment of RA, has potential for the inhibition of both IL-6 and SAA in AA amyloidosis [75]. Identification of the central role of activation of the inflammasome in the generation of proinflammatory cytokines (eg IL-1beta, IL-6, IL-18) as a key pathogenic event in some autoinflammatory diseases that may be associated with the development of AA amyloid is also an active area of drug development [76,77]. The role of SAA as a signal for activation of the NLRP3 inflammasome appears to be complicated. Delipidated SAA stimulates IL-1 secretion through a mechanism dependent on NLRP3 expression and caspase-1 activity, an effect abrogated by binding to high-density lipoprotein [78]; a direct effect of SAA oligomers on the inflammasome analogous to that shown for other forms of amyloid has been postulated, but not yet shown [79].

Binding to cofactors and peptidic inhibitors — New classes of agents may eventually prove efficacious by interfering with fibril formation [80,81]. Low mw anionic sulfonates or sulfates, as well as low mw heparins, impede AA fibril formation in vitro and attenuate the development of murine AA amyloid [82,83]. This hypothesis was tested in a randomized, multicenter international phase II/III trial of the first of these compounds (eprodisate) for the treatment of 150 patients with AA amyloid [84]. In this study, eprodisate showed a clinical benefit in delaying the decline in renal function for AA amyloidosis patients, defined as a reduction in risk and delayed time to doubling of serum creatinine, to 50 percent decrease in creatinine clearance, or to progression to dialysis, end-stage kidney disease, or death (from all causes). Unfortunately, a second international phase 3 randomized trial (ClinicalTrials.gov identifier: NCT012157470) failed to confirm benefit, and drug development has been terminated. An alternative strategy is the design of low mw peptides complementary to SAA domains critical for self-aggregation in order to inhibit fibrillogenesis [85].

Clearance of amyloid deposits from tissue — Two approaches to the use of immunotherapy as a mechanism to promote clearance of existing amyloid and thereby treat patients with significant organ-system compromise have reached the stage of clinical trials.

A novel bis (proline) compound (CPHPC) has been developed that binds with high affinity to serum amyloid P component (SAP), binding to amyloid fibrils, resulting in rapid clearance of SAP by the liver and depleting its serum level by approximately 90 percent. In a preliminary report, this resulted in disappearance of deposits in three types of systemic amyloidosis (AL, ATTR, and AA), as assessed by SAP scintigraphy [86]. This effect was sustained in follow-up studies [87]; however, approximately 20 percent of P-component remained associated with the amyloid deposits, which were only slowly resorbed. This limitation was addressed initially in a murine model for AA amyloid, in which SAP depletion following CPHPC was then followed with treatment using polyclonal anti-SAP, resulting in a rapid complement-dependent, macrophage-derived giant cell reaction that appeared to clear visceral amyloid deposits [88]. Administration of a humanized monoclonal anti-SAP antibody following CPHPC treatment has been shown to trigger clearance of amyloid deposits from the liver, kidney, and other tissues in an early phase trial in patients with systemic amyloidosis, including a patient with AA amyloidosis, as well as patients with AL and hereditary forms of amyloidosis [89]. This effect has subsequently been updated to include patients with cardiac amyloidosis [90] preparatory to a phase 3 multicenter trial. This approach may provide a new and generic therapeutic approach to the treatment of systemic and possibly localized forms of amyloidosis.

An alternative approach is the use of monoclonal anti-AA antibodies as agents for the in vivo imaging of AA amyloidosis and potential mobilization of amyloid deposits from tissue [91]. One of these antibodies directed to an epitope derived from a cleavage site of SAA was found to recognize a cryptic epitope shared with human light chain (AL) amyloid [92]. Extended efficacy for AA amyloidosis remains to be established.

Investigational approaches — Accumulating knowledge regarding the structural basis of fibrillogenesis has stimulated alternative approaches to generic therapy for the amyloidoses. Sometimes referred to as akin to putting "sand in the (beta) sheets," they include the following:

Design of or search for ligands that stabilize the native conformation of subunit proteins

Capping and stabilization of prefibrillar aggregates

Ligands or monoclonal antibodies specific for lipid-free SAA oligomers

Development of drugs that inhibit the adoption of the beta-pleated sheet configuration (either generically or specifically to the type of amyloid)

Use of chaperones to reverse aggregation

Alterations of the milieu that is responsible for off-pathway aggregation

Development of other methods to stimulate resorption of amyloid from tissue

Animal models — In addition to the use of low mw sulfates such as eprodisate, other studies have examined the potential efficacy of novel compounds that affect glycosaminoglycan biosynthesis and that retard the development of AA amyloid in experimental models [93]. Colchicine, dimethylsulfoxide, inhibition of interactions between SAA and the receptor for advanced glycation end-products (RAGE), fenofibrate, FK506, and lovastatin have also been shown to have efficacy in retarding the deposition phase of murine AA amyloid [72,94,95]. In a mouse model of accelerated AA amyloidogenesis, alkylating agents (chlorambucil and cyclophosphamide) were found to be more active than methotrexate and azathioprine in suppressing amyloidogenesis [96]. Proton pump inhibitors had an antiinflammatory effect and caused regression of AA amyloid in a murine knock-in model for cryopyrinopathy [97].

A novel transgenic murine model for AA amyloid may provide an in vivo system in which the kinetics of amyloid regression and the efficacy of novel therapeutics might be tested [98,99].

Murine studies have validated treatment approaches that knock down SAA using a low mw inhibitor of IL-6 [100], induction of macrophage apoptosis using clodronate-containing liposomes [101] for the attenuation of the deposition phase, and the triggering of a complement-dependent, macrophage-derived giant cell reaction by anti-human SAP antibodies for the rapid removal of massive established visceral amyloid without adverse effects [88].

SUMMARY AND RECOMMENDATIONS

Complications and mortality risk – In untreated patients, AA (secondary) amyloidosis carries a significant risk of mortality due to end-stage kidney disease, infection, heart failure, bowel perforation, or gastrointestinal bleeding. Patients with persistently high circulating levels of SAA are at particular risk of these complications of disease. Other adverse prognostic signs included older age, reduced serum albumin concentration, and end-stage kidney failure at baseline. (See 'Prognosis' above.)

Potential benefits of treatment and prevention – Successful treatment of the underlying inflammatory process can lead to stabilization of or improvement in renal function, to reduction in protein excretion, and to partial resolution of amyloid deposits. (See 'Importance of control of the underlying disease' above.)

Therapeutic interventions – The preferred therapy of AA amyloid is control of the underlying inflammatory disease. Colchicine has become accepted therapy for prophylaxis and treatment of familial Mediterranean fever (FMF), and anti-proinflammatory cytokines (interleukin [IL] 1 beta, tumor necrosis factor [TNF] alpha, IL-6) have been shown to have efficacy in some cases of AA amyloidosis due to rheumatic disorders and to hereditary autoinflammatory disease. Agents under development interfere with fibril formation. (See 'Treatment of AA amyloidosis' above.)

  1. Westermark GT, Fändrich M, Westermark P. AA amyloidosis: pathogenesis and targeted therapy. Annu Rev Pathol 2015; 10:321.
  2. Papa R, Lachmann HJ. Secondary, AA, Amyloidosis. Rheum Dis Clin North Am 2018; 44:585.
  3. Lachmann HJ, Goodman HJ, Gilbertson JA, et al. Natural history and outcome in systemic AA amyloidosis. N Engl J Med 2007; 356:2361.
  4. Yilmaz M, Unsal A, Sokmen M, et al. Renal involvement in AA amyloidosis: clinical outcomes and survival. Kidney Blood Press Res 2013; 37:33.
  5. Bunker D, Gorevic P. AA amyloidosis: Mount Sinai experience, 1997-2012. Mt Sinai J Med 2012; 79:749.
  6. Tanaka F, Migita K, Honda S, et al. Clinical outcome and survival of secondary (AA) amyloidosis. Clin Exp Rheumatol 2003; 21:343.
  7. Falck HM, Maury CP, Teppo AM, Wegelius O. Correlation of persistently high serum amyloid A protein and C-reactive protein concentrations with rapid progression of secondary amyloidosis. Br Med J (Clin Res Ed) 1983; 286:1391.
  8. Lane T, Pinney JH, Gilbertson JA, et al. Changing epidemiology of AA amyloidosis: clinical observations over 25 years at a single national referral centre. Amyloid 2017; 24:162.
  9. Palladini G, Riva E, Basset M, et al. Prognostication of survival and progression to dialysis in AA amyloidosis. Amyloid 2017; 24:136.
  10. Law S, Cohen O, Lachmann HJ, et al. Renal transplant outcomes in amyloidosis. Nephrol Dial Transplant 2021; 36:355.
  11. Sack GH Jr. Serum amyloid A - a review. Mol Med 2018; 24:46.
  12. Berglund K, Thysell H, Keller C. Results, principles and pitfalls in the management of renal AA-amyloidosis; a 10-21 year followup of 16 patients with rheumatic disease treated with alkylating cytostatics. J Rheumatol 1993; 20:2051.
  13. Lovat LB, Madhoo S, Pepys MB, Hawkins PN. Long-term survival in systemic amyloid A amyloidosis complicating Crohn's disease. Gastroenterology 1997; 112:1362.
  14. Waldenström H. On the formation and disappearance of amyloid in man. Acta Chir Scand 1928; 63:479.
  15. Shimojima Y, Takei Y, Tazawa K, et al. Histopathological regression of systemic AA amyloidosis after surgical treatment of a localized Castleman's disease. Amyloid 2006; 13:184.
  16. Ortiz-Santamaría V, Olivé A, Valls-Roc M, Tena X. Treatment of AA amyloid with chlorambucil. Rheumatology (Oxford) 2002; 41:833.
  17. Nakamura T, Yamamura Y, Tomoda K, et al. Efficacy of cyclophosphamide combined with prednisolone in patients with AA amyloidosis secondary to rheumatoid arthritis. Clin Rheumatol 2003; 22:371.
  18. Mpofu S, Teh LS, Smith PJ, et al. Cytostatic therapy for AA amyloidosis complicating psoriatic spondyloarthropathy. Rheumatology (Oxford) 2003; 42:362.
  19. Gillmore JD, Lovat LB, Persey MR, et al. Amyloid load and clinical outcome in AA amyloidosis in relation to circulating concentration of serum amyloid A protein. Lancet 2001; 358:24.
  20. Hawkins PN, Richardson S, Vigushin DM, et al. Serum amyloid P component scintigraphy and turnover studies for diagnosis and quantitative monitoring of AA amyloidosis in juvenile rheumatoid arthritis. Arthritis Rheum 1993; 36:842.
  21. Deschênes G, Prieur AM, Hayem F, et al. Renal amyloidosis in juvenile chronic arthritis: evolution after chlorambucil treatment. Pediatr Nephrol 1990; 4:463.
  22. Chevrel G, Jenvrin C, McGregor B, Miossec P. Renal type AA amyloidosis associated with rheumatoid arthritis: a cohort study showing improved survival on treatment with pulse cyclophosphamide. Rheumatology (Oxford) 2001; 40:821.
  23. Lachmann HJ, Gilbertson JA, Gillmore JD, et al. Unicentric Castleman's disease complicated by systemic AA amyloidosis: a curable disease. QJM 2002; 95:211.
  24. Zemer D, Pras M, Sohar E, et al. Colchicine in the prevention and treatment of the amyloidosis of familial Mediterranean fever. N Engl J Med 1986; 314:1001.
  25. Livneh A, Zemer D, Langevitz P, et al. Colchicine treatment of AA amyloidosis of familial Mediterranean fever. An analysis of factors affecting outcome. Arthritis Rheum 1994; 37:1804.
  26. Livneh A, Zemer D, Siegal B, et al. Colchicine prevents kidney transplant amyloidosis in familial Mediterranean fever. Nephron 1992; 60:418.
  27. Ozen S, Kone-Paut I, Gül A. Colchicine resistance and intolerance in familial mediterranean fever: Definition, causes, and alternative treatments. Semin Arthritis Rheum 2017; 47:115.
  28. Druyan A, Giat E, Livneh A, et al. Effect of interleukin-1 inhibition in a cohort of patients with colchicine-resistant familial Mediterranean fever treated consecutively with anakinra and canakinumab. Clin Exp Rheumatol 2021; 39 Suppl 132:75.
  29. Greenstein AJ, Sachar DB, Panday AK, et al. Amyloidosis and inflammatory bowel disease. A 50-year experience with 25 patients. Medicine (Baltimore) 1992; 71:261.
  30. Tasdemir I, Sivri B, Turgan C, et al. The expanding spectrum of a disease. Behçet's disease associated with amyloidosis. Nephron 1989; 52:154.
  31. Tan AU Jr, Cohen AH, Levine BS. Renal amyloidosis in a drug abuser. J Am Soc Nephrol 1995; 5:1653.
  32. Hausmann MJ, Maor E, Kachko L. Colchicine-sensitive nephrotic syndrome due to AA amyloidosis. Amyloid 2011; 18:169.
  33. Iwakiri R, Sakemi T, Fujimoto K. Dimethylsulfoxide for renal dysfunction caused by systemic amyloidosis complicating Crohn's disease. Gastroenterology 1999; 117:1031.
  34. Amemori S, Iwakiri R, Endo H, et al. Oral dimethyl sulfoxide for systemic amyloid A amyloidosis complication in chronic inflammatory disease: a retrospective patient chart review. J Gastroenterol 2006; 41:444.
  35. Scheinberg MA, Pernambuco JC, Benson MD. DMSO and colchicine therapy in amyloid disease. Ann Rheum Dis 1984; 43:421.
  36. Jimenez RA, Willkens RF. Dimethyl sulfoxide: a perspective of its use in rheumatic diseases. J Lab Clin Med 1982; 100:489.
  37. Gottenberg JE, Merle-Vincent F, Bentaberry F, et al. Anti-tumor necrosis factor alpha therapy in fifteen patients with AA amyloidosis secondary to inflammatory arthritides: a followup report of tolerability and efficacy. Arthritis Rheum 2003; 48:2019.
  38. Fernández-Nebro A, Tomero E, Ortiz-Santamaría V, et al. Treatment of rheumatic inflammatory disease in 25 patients with secondary amyloidosis using tumor necrosis factor alpha antagonists. Am J Med 2005; 118:552.
  39. Nakamura T, Higashi S, Tomoda K, et al. Etanercept can induce resolution of renal deterioration in patients with amyloid A amyloidosis secondary to rheumatoid arthritis. Clin Rheumatol 2010; 29:1395.
  40. Keersmaekers T, Claes K, Kuypers DR, et al. Long-term efficacy of infliximab treatment for AA-amyloidosis secondary to chronic inflammatory arthritis. Ann Rheum Dis 2009; 68:759.
  41. Nakamura T, Higashi S, Tomoda K, et al. Effectiveness of etanercept vs cyclophosphamide as treatment for patients with amyloid A amyloidosis secondary to rheumatoid arthritis. Rheumatology (Oxford) 2012; 51:2064.
  42. Perry ME, Stirling A, Hunter JA. Effect of etanercept on serum amyloid A protein (SAA) levels in patients with AA amyloidosis complicating inflammatory arthritis. Clin Rheumatol 2008; 27:923.
  43. Park YK, Han DS, Eun CS. Systemic amyloidosis with Crohn's disease treated with infliximab. Inflamm Bowel Dis 2008; 14:431.
  44. Kobak S, Oksel F, Kabasakal Y, Doganavsargil E. Ankylosing spondylitis-related secondary amyloidosis responded well to etanercept: a report of three patients. Clin Rheumatol 2007; 26:2191.
  45. Kuroda T, Wada Y, Kobayashi D, et al. Effective anti-TNF-alpha therapy can induce rapid resolution and sustained decrease of gastroduodenal mucosal amyloid deposits in reactive amyloidosis associated with rheumatoid arthritis. J Rheumatol 2009; 36:2409.
  46. Immonen K, Finne P, Grönhagen-Riska C, et al. A marked decline in the incidence of renal replacement therapy for amyloidosis associated with inflammatory rheumatic diseases - data from nationwide registries in Finland. Amyloid 2011; 18:25.
  47. Lane T, Wechalekar AD, Gillmore JD, et al. Safety and efficacy of empirical interleukin-1 inhibition using anakinra in AA amyloidosis of uncertain aetiology. Amyloid 2017; 24:189.
  48. Simsek C, Karatas M, Tatar E, et al. The efficacy of interleukin-1 antagonist drugs in combination with colchicine in patients with FMF-AA with colchicine resistance after kidney transplantation: A study with histopathologic evidence. Clin Transplant 2021; 35:e14309.
  49. Okuda Y, Takasugi K. Successful use of a humanized anti-interleukin-6 receptor antibody, tocilizumab, to treat amyloid A amyloidosis complicating juvenile idiopathic arthritis. Arthritis Rheum 2006; 54:2997.
  50. Nishida S, Hagihara K, Shima Y, et al. Rapid improvement of AA amyloidosis with humanised anti-interleukin 6 receptor antibody treatment. Ann Rheum Dis 2009; 68:1235.
  51. Inoue D, Arima H, Kawanami C, et al. Excellent therapeutic effect of tocilizumab on intestinal amyloid a deposition secondary to active rheumatoid arthritis. Clin Rheumatol 2010; 29:1195.
  52. Redondo-Pachón MD, Enríquez R, Sirvent AE, et al. Tocilizumab treatment for nephrotic syndrome due to amyloidosis in Behcet's disease. Ren Fail 2013; 35:547.
  53. Jung JY, Kim YB, Kim JW, et al. Biologic therapy for amyloid A amyloidosis secondary to rheumatoid arthritis treated with interleukin 6 therapy: Case report and review of literature. Medicine (Baltimore) 2021; 100:e26843.
  54. Eriksson P, Mölne J, Wirestam L, Sjöwall C. Successful Treatment of AA Amyloidosis in Ankylosing Spondylitis Using Tocilizumab: Report of Two Cases and Review of the Literature. Front Med (Lausanne) 2021; 8:661101.
  55. Fukuda M, Sawa N, Hoshino J, et al. Tocilizumab preserves renal function in rheumatoid arthritis with AA amyloidosis and end-stage kidney disease: Two case reports. Clin Nephrol 2021; 95:54.
  56. Okuda Y. AA amyloidosis - Benefits and prospects of IL-6 inhibitors. Mod Rheumatol 2019; 29:268.
  57. Okuda Y, Ohnishi M, Matoba K, et al. Comparison of the clinical utility of tocilizumab and anti-TNF therapy in AA amyloidosis complicating rheumatic diseases. Mod Rheumatol 2014; 24:137.
  58. Narváez J, Hernández MV, Ruiz JM, et al. Rituximab therapy for AA-amyloidosis secondary to rheumatoid arthritis. Joint Bone Spine 2011; 78:101.
  59. Sawamura M, Sawa N, Fujiwara H, et al. Abatacept Improves Intractable Protein-Losing Enteropathy Secondary to AA Amyloidosis in a Patient With Rheumatoid Arthritis. Mayo Clin Proc Innov Qual Outcomes 2020; 4:815.
  60. Sorić Hosman I, Kos I, Lamot L. Serum Amyloid A in Inflammatory Rheumatic Diseases: A Compendious Review of a Renowned Biomarker. Front Immunol 2020; 11:631299.
  61. Ombrello AK. Modern therapies in AA amyloidosis. In: Amyloid and Related Disorders: Surgical Pathology and Clinical Correlations, Picken MM, Dogan A, Herrera GA (Eds), Humana Press, New York 2012. p.399.
  62. Leslie KS, Lachmann HJ, Bruning E, et al. Phenotype, genotype, and sustained response to anakinra in 22 patients with autoinflammatory disease associated with CIAS-1/NALP3 mutations. Arch Dermatol 2006; 142:1591.
  63. Thornton BD, Hoffman HM, Bhat A, Don BR. Successful treatment of renal amyloidosis due to familial cold autoinflammatory syndrome using an interleukin 1 receptor antagonist. Am J Kidney Dis 2007; 49:477.
  64. Lane T, Loeffler JM, Rowczenio DM, et al. AA amyloidosis complicating the hereditary periodic fever syndromes. Arthritis Rheum 2013; 65:1116.
  65. Varan Ö, Kucuk H, Babaoglu H, et al. Efficacy and safety of interleukin-1 inhibitors in familial Mediterranean fever patients complicated with amyloidosis. Mod Rheumatol 2019; 29:363.
  66. Trabulus S, Korkmaz M, Kaya E, Seyahi N. Canakinumab treatment in kidney transplant recipients with AA amyloidosis due to familial Mediterranean fever. Clin Transplant 2018; 32:e13345.
  67. Portincasa P. Colchicine, Biologic Agents and More for the Treatment of Familial Mediterranean Fever. The Old, the New, and the Rare. Curr Med Chem 2016; 23:60.
  68. Gattorno M, Pelagatti MA, Meini A, et al. Persistent efficacy of anakinra in patients with tumor necrosis factor receptor-associated periodic syndrome. Arthritis Rheum 2008; 58:1516.
  69. Moser C, Pohl G, Haslinger I, et al. Successful treatment of familial Mediterranean fever with Anakinra and outcome after renal transplantation. Nephrol Dial Transplant 2009; 24:676.
  70. Lane T, Gillmore JD, Wechalekar AD, et al. Therapeutic blockade of interleukin-6 by tocilizumab in the management of AA amyloidosis and chronic inflammatory disorders: a case series and review of the literature. Clin Exp Rheumatol 2015; 33:S46.
  71. Stankovic Stojanovic K, Delmas Y, Torres PU, et al. Dramatic beneficial effect of interleukin-1 inhibitor treatment in patients with familial Mediterranean fever complicated with amyloidosis and renal failure. Nephrol Dial Transplant 2012; 27:1898.
  72. Ueda M, Ando Y, Nakamura M, et al. FK506 inhibits murine AA amyloidosis: possible involvement of T cells in amyloidogenesis. J Rheumatol 2006; 33:2260.
  73. Sikora J, Kmochová T, Mušálková D, et al. A mutation in the SAA1 promoter causes hereditary amyloid A amyloidosis. Kidney Int 2022; 101:349.
  74. Semerano L, Thiolat A, Minichiello E, et al. Targeting IL-6 for the treatment of rheumatoid arthritis: Phase II investigational drugs. Expert Opin Investig Drugs 2014; 23:979.
  75. Migita K, Izumi Y, Jiuchi Y, et al. Effects of Janus kinase inhibitor tofacitinib on circulating serum amyloid A and interleukin-6 during treatment for rheumatoid arthritis. Clin Exp Immunol 2014; 175:208.
  76. Zahid A, Li B, Kombe AJK, et al. Pharmacological Inhibitors of the NLRP3 Inflammasome. Front Immunol 2019; 10:2538.
  77. Vong CT, Tseng HHL, Yao P, et al. Specific NLRP3 inflammasome inhibitors: promising therapeutic agents for inflammatory diseases. Drug Discov Today 2021; 26:1394.
  78. Shridas P, De Beer MC, Webb NR. High-density lipoprotein inhibits serum amyloid A-mediated reactive oxygen species generation and NLRP3 inflammasome activation. J Biol Chem 2018; 293:13257.
  79. Masters SL, O'Neill LA. Disease-associated amyloid and misfolded protein aggregates activate the inflammasome. Trends Mol Med 2011; 17:276.
  80. Ke PC, Zhou R, Serpell LC, et al. Half a century of amyloids: past, present and future. Chem Soc Rev 2020; 49:5473.
  81. Nevone A, Merlini G, Nuvolone M. Treating Protein Misfolding Diseases: Therapeutic Successes Against Systemic Amyloidoses. Front Pharmacol 2020; 11:1024.
  82. Kisilevsky R, Lemieux LJ, Fraser PE, et al. Arresting amyloidosis in vivo using small-molecule anionic sulphonates or sulphates: implications for Alzheimer's disease. Nat Med 1995; 1:143.
  83. Zhu H, Yu J, Kindy MS. Inhibition of amyloidosis using low-molecular-weight heparins. Mol Med 2001; 7:517.
  84. Dember LM, Hawkins PN, Hazenberg BP, et al. Eprodisate for the treatment of renal disease in AA amyloidosis. N Engl J Med 2007; 356:2349.
  85. Sosnowska M, Skibiszewska S, Kamińska E, et al. Designing peptidic inhibitors of serum amyloid A aggregation process. Amino Acids 2016; 48:1069.
  86. Pepys MB, Herbert J, Hutchinson WL, et al. Targeted pharmacological depletion of serum amyloid P component for treatment of human amyloidosis. Nature 2002; 417:254.
  87. Gillmore JD, Tennent GA, Hutchinson WL, et al. Sustained pharmacological depletion of serum amyloid P component in patients with systemic amyloidosis. Br J Haematol 2010; 148:760.
  88. Bodin K, Ellmerich S, Kahan MC, et al. Antibodies to human serum amyloid P component eliminate visceral amyloid deposits. Nature 2010; 468:93.
  89. Richards DB, Cookson LM, Berges AC, et al. Therapeutic Clearance of Amyloid by Antibodies to Serum Amyloid P Component. N Engl J Med 2015; 373:1106.
  90. Richards DB, Cookson LM, Barton SV, et al. Repeat doses of antibody to serum amyloid P component clear amyloid deposits in patients with systemic amyloidosis. Sci Transl Med 2018; 10.
  91. Wall JS, Kennel SJ, Richey T, et al. Generation and characterization of anti-AA amyloid-specific monoclonal antibodies. Front Immunol 2011; 2:32.
  92. Wall JS, Kennel SJ, Williams A, et al. AL amyloid imaging and therapy with a monoclonal antibody to a cryptic epitope on amyloid fibrils. PLoS One 2012; 7:e52686.
  93. Kisilevsky R, Szarek WA, Ancsin J, et al. Novel glycosaminoglycan precursors as anti-amyloid agents, part III. J Mol Neurosci 2003; 20:291.
  94. Yan SD, Zhu H, Zhu A, et al. Receptor-dependent cell stress and amyloid accumulation in systemic amyloidosis. Nat Med 2000; 6:643.
  95. van der Hilst JC, Kluve-Beckerman B, Bodar EJ, et al. Lovastatin inhibits formation of AA amyloid. J Leukoc Biol 2008; 83:1295.
  96. Shtrasburg S, Pras M, Rabinovich E, et al. Attempts at suppression of amyloidogenesis in a mouse model by a variety of anti-inflammatory agents. Autoimmun Rev 2012; 12:18.
  97. Bertoni A, Carta S, Baldovini C, et al. A novel knock-in mouse model of cryopyrin-associated periodic syndromes with development of amyloidosis: Therapeutic efficacy of proton pump inhibitors. J Allergy Clin Immunol 2020; 145:368.
  98. Kluve-Beckerman B, Hardwick, Du L, Benson MD. AA amyloidosis: potential therapy with antisense oligonucleotides. Amyloid 2011; 18 Suppl 1:200.
  99. Gorevic PD. Amyloid and inflammation. Proc Natl Acad Sci U S A 2013; 110:16291.
  100. Ranganath S, Bhandari A, Avitahl-Curtis N, et al. Discovery and Characterization of a Potent Interleukin-6 Binding Peptide with Neutralizing Activity In Vivo. PLoS One 2015; 10:e0141330.
  101. Lundmark K, Vahdat Shariatpanahi A, Westermark GT. Depletion of spleen macrophages delays AA amyloid development: a study performed in the rapid mouse model of AA amyloidosis. PLoS One 2013; 8:e79104.
Topic 5588 Version 29.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟