The Amyloidoses


Amyloid type

Protein

Abbreviation

Organ involvement

Comment

Immunoglobulin light chain

Monoclonal Ig light chain

AL

Kidney, liver, heart, GI tract, peripheral and autonomic nerves, soft tissues

Acquired plasma cell dyscrasia with synthesis of protein from bone marrow

Fibrinogen

Fibrinogen Aα

AFibα

Kidney, liver, spleen

Hereditary; hypertension common

Reactive

SAA

AA

Kidney, GI tract, liver, autonomic nervous system

Secondary to chronic inflammation, infection or neoplasm; protein synthesized from liver

Senile systemic

TTR wild type

ATTR-wt

Cardiac

Usually older males; protein synthesized from liver

TTR

TTR mutant

ATTR

Peripheral and autonomic nerves, heart, eye, occasional kidney

Hereditary; protein synthesized from liver

Apolipoprotein AI

Apolipoprotein AI

AApoI

Kidney (glomerular), liver heart, skin, larynx

Hereditary

Apolipoprotein AII

Apolipoprotein AII

AApoII

Kidney

Hereditary

Dialysis related

β2-microglobulin

Aβ 2M

Osteoarticular tissue, GI tract, blood vessels, heart

Inadequate renal clearance of protein

Lysozyme

Lysozyme

ALys

Kidney, liver, GI tract, spleen, lymph nodes, lung, thyroid, salivary glands

Hereditary


SAA serum amyloid A, TTR transthyretin, GI gastrointestinal, Ig immunoglobulin , AA serum amyloid A protein amyloidosis



It is now known that some amyloid fibrils may serve biological function(s). These protease-resistant β-pleated sheet assemblies are widely used in nature and comprise the so-called functional amyloids. In fact, amyloid formation seems to be an intrinsic propensity of polypeptides in general and the amyloid β-pleated sheet is a highly conserved structure through evolution. Functional amyloids have been found in a wide range of organisms, from bacteria to mammals, with functions as diverse as biofilm formation, development of aerial structures, scaffolding, regulation of melanin synthesis, epigenetic control of polyamines, and information transfer [11]. Presently, one of the methods under development for the treatment of amyloid disease involves directly inhibiting the formation of pathological amyloid . Given that pathological and functional amyloid share a common structure, some amyloidogenesis inhibitor drugs intended to prevent disease could disrupt functional amyloid formation. This could lead to undesirable side effects because functional amyloid seems to have a role in vital physiological processes in humans, including hemostasis and melanin synthesis [7]. Thus, amyloidogenesis inhibitor drugs must be designed with sufficient specificity to avoid interfering with these functions.


Case #1

Mr. P is a 69-year-old man with an 8-year history of type II diabetes who was followed by a nephrologist for renal insufficiency with a creatinine clearance of 45 ml/min and proteinuria of 5.3 g/day. His diabetes was well controlled with oral medications and he never required insulin. In addition, he had no evidence of diabetic retinopathy. For this reason, his nephrologist suspected that there was likely another etiology of his renal insufficiency and proteinuria and he was referred for a kidney biopsy. On H&E staining, Mr. P’s kidney biopsy demonstrated mild amorphous, eosinophilic deposition in the glomerulus, mesangium, and capillary membranes (see Fig. 14.1). A completely hyalinized glomerulus stained pink-red with Congo red (See Fig. 14.2). The same area showed apple-green birefringence with polarization (see Fig. 14.3).

What is the most likely diagnosis after the above biopsy result findings?



a.

AL Amyloidosis

 

b.

AA Amyloidosis

 

c.

Minimal change disease

 

d.

Diabetic nephropathy

 

e.

More information is required.

 



A314216_1_En_14_Fig1_HTML.jpg


Fig. 14.1
Section shows obliteration of the glomerulus by amyloid deposition, (a). Glomerulus with slight thickening of the capillary membranes and mild expansion of the mesangium by amyloid deposition, (b). H&E, 200X



A314216_1_En_14_Fig2_HTML.jpg


Fig. 14.2
Congo red stained section shows pink-red amyloid in the glomerulus (arrow). Congo red, 400X



A314216_1_En_14_Fig3_HTML.jpg


Fig. 14.3
Yellow-green birefringence with polarization (arrow). Congo red, 400X

The kidney is most frequently affected in AL , AA, and several of the hereditary amyloidoses. Kidney biopsy is often required to identify the underlying disease. Amyloid deposits can be seen throughout the kidney but predominate in the glomerulus [12]. By light microscopy, amyloid appears as an amorphous, eosinophilic material in the mesangium and capillary loops. When amyloid is suspected, the tissue for Congo red staining should be cut at 10-micron thickness, rather than the 2-micron sections that are normally prepared from renal biopsies. Amyloid deposition in the tubulointerstitium can also lead to tubular atrophy and interstitial fibrosis even in the absence of significant glomerular deposition. Regardless of the location of the amyloid deposits, birefringent Congo red staining is seen. Because the amyloid fibrils are composed primarily of the amyloid protein and not extracellular matrix polysaccharides such as collagen, periodic acid-Schiff (PAS) staining is only weakly positive. Glomerular deposition often leads to significant proteinuria in the nephrotic range with rates up to 20 g/day. Since the protein is primary albumin, the associated edema can be severe and refractory to diuretics, as well as difficult to manage, especially in the setting of cardiac dysfunction and autonomic neuropathy seen in some amyloid subtypes. Conversely, when glomerular involvement is minimal and amyloid deposition is primarily in the tubulointerstitium, the resulting proteinuria is minimal, glomerular filtration is reduced, and creatinine is increased. Immunofluorescence (IF) and immunohistochemistry (IHC) are negative for intact immunoglobulin, complement and fibrin, but in AL amyloid are frequently positive for immunoglobulin light chain .

Although amyloid fibril deposition is often evident on kidney biopsy by electron microscopy, it can be overlooked if the index of suspicion is not sufficiently high. The fibrils are nonbranching, randomly arrayed, and have a diameter of 8–10 nm. The diagnosis of amyloid may be missed if there is secondary effacement of epithelial foot processes affected by amyloid deposition in the setting of only mild amyloid deposition in the mesangium. Such pathologic findings can give the false impression of minimal change glomerulonephritis [13]. Misdiagnosis may also occur due to confusion with the morphologic appearance of diabetic nephropathy. In a study of 26 cases of both AA and AL renal amyloidosis, there were many morphologic changes seen that mimic diabetic nephropathy; such as diffuse and nodular patterns, capsular drop-like deposits along Bowman’s capsule, deposition in the afferent and efferent arterioles, early stage glomerular microaneurysm, and accumulation of amyloid along the tubular basement membrane [14] .


Case #1 Follow-up and Discussion

The correct answer is e. Congo red staining can define presence of amyloidosis but the type of amyloidosis is still very important to determine for treatment purposes. As stated above, IF and IHC are negative for intact immunoglobulin, complement, and fibrin in AA amyloidosis, but in AL amyloid are frequently positive for immunoglobulin light chain. For confirmation of diagnosis, Mr. P’s biopsy specimen was also sent to the Mayo Clinic for laser capture microdissection and mass spectrometry (LCM-MS)-based proteomic analysis and was determined to be AL λ-type amyloidosis.

Serum amyloid P component is a normal plasma protein that constitutes approximately 5 % of all amyloid deposits. It is a member of the pentraxin family of proteins that are involved in the acute inflammatory process, another example of such is C-reactive protein. Of note, radiolabeled serum amyloid P component scintigraphy is a noninvasive and quantitative method for imaging amyloid deposits, which produces diagnostic images in most patients with systemic amyloidosis, and can be used repeatedly to monitor the course of the disease. However, it has not been produced commercially and has very limited availability [15] .

Renal disease is common in many forms of amyloid and a major source of morbidity. Without treatment, end stage kidney disease (ESKD) will usually occur at variable rates over time. However, some forms of amyloid such as senile systemic (TTR protein) and dialysis-related amyloid (β2-microglobulin) do not typically involve the kidney.

The most common form of systemic amyloidosis is AL amyloidosis, with a reported incidence of 8.9 per million person years [10]. AL amyloidosis is of interest to the hematologist because it is caused by a neoplastic plasma cell or B-cell clone which synthesizes abnormal amounts of a specific immunoglobulin (Ig) which results in the dysfunction of one or more involved organs. Systemic amyloid may also be seen in 5–15 % of individuals with multiple myeloma . AL amyloidosis should be suspected in any patient with nondiabetic nephrotic syndrome, nonischemic cardiomyopathy with an echocardiogram showing concentric hypertrophy, increase of NTproBNP in the absence of primary heart disease, presence of hepatomegaly or increase of alkaline phosphatase without an imaging abnormality, peripheral and/or autonomic neuropathy, unexplained facial or neck purpura or macroglossia. Any patient who presents with any one of these signs should undergo a biopsy to detect amyloid deposits and blood screening for monoclonal immunoglobulin light chains . If a monoclonal protein is present, a bone marrow examination should be performed to evaluate for the presence of multiple myeloma . In a retrospective review of 100 known AL amyloid patients, bone marrow core biopsy revealed a plasma cell dyscrasia in 83 % (λ, 65; κ, 18) of cases [16]. Amyloid deposits were observed in 60 % of the bone marrow core biopsy specimens and, when present, were detected most often in blood vessel walls only (39 out of 60). Congo red staining of subcutaneous fat obtained by aspiration is a reliable and noninvasive test that positively identified amyloid deposits in 78 % of patients. If negative, a biopsy of the labial salivary glands may detect amyloid deposits in 50 % of patients. If this is also negative, then an involved organ should be biopsied when the clinical index of suspicion is high (see Fig. 14.4). Both IF and IHC staining are negative for intact immunoglobulin (Ig), but often positive for Ig light chain, which should be restricted to either of the two light chains (κ or λ). A limitation of IHC is reduced specificity due to background staining by normal light chain deposition. Another is the failure of commercial agents to detect the amyloid light chain because of conformational changes in the amyloid fibril that masks the relevant epitope. In one study, 12 of 34 patients (35.3 %) with known AL amyloidosis had negative IF staining for both κ and λ chains [17]. In contrast, AA amyloid is usually more accurately detected with standard antibodies against the AA protein .

A314216_1_En_14_Fig4_HTML.gif


Fig. 14.4
Proposed diagnostic algorithm for AL amyloid. (Source: Reprinted with permission. © 2011 American Society of Clinical Oncology. All rights reserved. Merlini G: J Clin Oncol Vol. (29), 2011: 1924–1933)

The differential diagnosis of AL amyloid includes light chain deposition disease (LCDD). The amyloid deposits of LCDD are distributed in a uniform, granular pattern throughout the glomerulus and the tubular basement membranes. The PAS staining is much more intense than AL amyloid due to the inflammatory response stimulated by the light chain deposition. In AL amyloid the λ light chain isotype predominates, whereas in LCDD the κ isotype is more common. Given the lack of β-pleated fibril formation, LCDD does not emit apple-green birefringence.

Other considerations in the pathologic differential diagnosis of amyloidosis include fibrillary glomerulonephritis and immunotactoid glomerulopathy. In fibrillary glomerulonephritis, the morphologic appearance is indistinguishable from amyloid in that there is glomerular accumulation of nonbranching, randomly arranged fibrils. Like amyloid, there is often a lack of inflammatory cell infiltrate in the glomerulus. It differs from amyloid in that the fibrils are larger (usually 18–20 nm) and lack reactivity with Congo red and light chain IHC stains [18]. Immunotactoid glomerulopathy may be considered a subtype of fibrillary with similar morphologic and histochemical characteristics. However, this is a process where much larger fibrils (ranging from 34 to 49 nm) are deposited in an ordered and parallel orientation. An association with lymphoproliferative disorders has been identified .

It is now clear that an individual patient may have both a monoclonal gammopathy and a hereditary variant which creates the confounding scenario of two possible sources of the amyloid forming protein. AL amyloidosis often responds to chemotherapy that suppresses the underlying clonal plasma-cell disorder, but chemotherapy has no role in the treatment of hereditary amyloidosis and may be harmful. This was initially described in theUK where 350 patients with systemic amyloidosis in whom a diagnosis of sporadic AL amyloid was suggested by clinical findings (negative family history and the presence of a monoclonal gammopathy). However, DNA genotyping of whole blood for the most common causes of hereditary amyloidosis yielded mutations in 10 % of patients, most often in the genes encoding fibrinogen Aα and TTR [19]. Thus, in these cases the monoclonal gammopathy was considered incidental and these patients were not treated with chemotherapy. Since hereditary amyloid has variable penetrance, the family history proved to be an ineffective screening test. This series also revealed that in those patients with true AL amyloidosis, the Ig light chain fibrils were identified by IHC staining in only 38 % of the cases . This low value reflects the failure of anti-light chain antibodies to bind to light chain fragments once they have formed into an amyloid fibril. Thus, although IHC staining for amyloid forming proteins can be useful, protein- or DNA-based screening is recommended due to the limitations of IHC. The Memorial Sloan Kettering group took a more targeted approach to hereditary screening for all patients referred for an evaluation of systemic amyloidosis. In their study of 178 patients, screening took place for those who met the following criteria: (1) asymptomatic African Americans were screened for the presence of a mutant TTR (the Val122Ile variant of TTR occurs in 4 % of African Americans); (2) patients with dominant peripheral nervous system involvement were screened for variants of TTR, apolipoprotein AI and AII, fibrinogen Aα, and lysozyme (peripheral neuropathy is a common presentation of AL amyloidosis and also several of the hereditary variants); (3) and patients with isolated renal amyloidosis and no amyloid in the bone marrow were screened for the fibrinogen Aα variant [20]. Of those who were screened, 6 % had both an incidental monoclonal gammopathy and a true hereditary amyloid protein identified in the same patient .

Because of sample size limitations of the tissue biopsy and the aforementioned limitations of IHC, a newer technique of LCM-MS-based proteomic analysis has been developed at Mayo Clinic to identify amyloid protein in both a specific and sensitive fashion. Laser capture microdissection (LCM) is used to specifically study those areas in the biopsy sample that are positive for Congo red staining, thus increasing the yield of the result. In brief, the process involves the microdissection of amyloid from the tissue biopsy. This is then digested into tryptic peptides and analyzed by liquid chromatography electrospray in tandem mass spectrometry (MS). The MS raw data files are queried by different computer algorithms to search protein databases for the compatible protein. A training set of 50 patients with cardiac amyloid was compared with the current gold standard approach (includes an extensive clinical investigation for plasma cell disorders, serum and genetic testing for amyloidogenic TTR variants, and IHC for TTR, SAA, Ig κ, Ig λ, and serum amyloid P component). This was later validated in 41 additional cases yielding a specificity of 100 % and sensitivity of 98 %, whereas IHC was comparatively informative in only 42 % [21, 22]. In a later study on amyloid diseases associated with neuropathy (includes AL, ATTR, AGel, and AApoAI), the specific amyloid subtype was identified in 21 different nerve biopsies by LCM-MS without assistance from clinical information [22]. In future, LCM-MS will likely become the new gold standard for identifying the protein forming the amyloid deposits when it becomes more generally available. However, there are limitations in LCM-MS, including that the mutations must be both known and available in protein databases and that the amino acid changes must lead to alterations significant enough to be detected by mass spectroscopy .



Non-AL Amyloid


Mutated genes that are associated with hereditary amyloid include apolipoprotein AI (Apo AI) , TTR, fibrinogen Aα chain, lysozyme, cystatin C, gelsolin, and apolipoprotein AII. Each of them is associated with clinical amyloidosis syndromes and has distinct clinical manifestations such as age of onset, presenting signs, site of organ involvement, and rate of progression and prognosis. The most common clinical manifestation of AApoAI is a slowly progressive, non-proteinuric renal failure due to tubular deposits of amyloid fibril. It is also associated with extensive deposits in the liver and spleen and no cardiomyopathy. The Gly26Arg mutation is most common among patients with Irish ancestry. Three Irish families were studied to assess the natural history of the disease and the usage of renal transplantation. As opposed to the generally rapid progression of renal failure seen in AL amyloid, the progression seen in AApoAI amyloid is slow. Renal failure usually presents as hypertension and mild proteinuria between age 18 and 55 years [23]. Histology demonstrated tubulointerstitial fibrosis with the unusual finding of amyloid deposition in the medulla. The UK study also supports the durable nature of renal transplantation in such patients. Over a median of 9 years from transplantation, eight of ten patients were alive, and seven with a functioning graft [24]. Renal transplantation may be used successfully to treat this disorder with uncommon failure of the graft due to amyloid recurrence. Patients who present with familial tubulointerstitial nephritis pattern and associated liver disease require a high index of suspicion for AApoAI amyloidosis .

Reactive systemic amyloidosis is comprised of the AA protein (SAA) secondary to an associated inflammatory condition. Examples of such disorders include rheumatoid arthritis, tuberculosis, chronic osteomyelitis, inflammatory bowel disease, and familial Mediterranean fever (FMF) [25]. The underlying disease causes chronically active inflammation. Of note, secondary amyloidosis can also occasionally occur in patients with neoplasms such as hepatocellular carcinoma, renal cell carcinoma, Castleman’s disease, Hodgkin’s disease, and hairy cell leukemia.

A process called “localized amyloid” may occur in individual organs, in the absence of systemic involvement. The reason for localized deposition is unknown, but it is hypothesized that the deposits result from local synthesis of the amyloid protein, rather than the deposition of light chains produced elsewhere. In a series of 20 cases of localized amyloidosis diagnosed between 1993 and 2003 in solitary organs involving skin, soft tissues, oropharynx, larynx, lung, bladder, colon, conjunctiva, and lymph nodes, no patient progressed to systemic disease over an average of 7 years [26] .


Treatment



Case #2

Mr. P (from case #1) had an extensive pretreatment evaluation. He had a normal troponin but an elevation in N-terminal pro-brain natriuretic peptide (NT-pro-BNP) 467 ng/L. An echocardiogram showed a left ventricular ejection fraction (LVEF) of 54 % and mild concentric left ventricular hypertrophy with normal global left ventricular systolic function. He had a cardiac MRI which confirmed a normal left ventricular systolic function with an LVEF of 57 %. There was no evidence of myocardial scar or infiltration suggesting that he had no amyloid involvement of his heart. He had an abdominal ultrasound which was negative for hepatosplenomegaly and a normal alkaline phosphatase suggesting no hepatic involvement. He had an excellent performance status. Thus, it was determined that Mr. P would be an appropriate candidate for treatment with high-dose melphalan followed by autologous stem cell transplantation hematopoietic stem cell transplantation (HSCT). Overall, Mr. P tolerated the treatment well over time and his proteinuria was reduced from > 5 to 1.5 g/day. His creatinine clearance remained stable. What clinical features in a patient with AL amyloidosis can predict outcomes?



a.

Number of organs involved

 

b.

Degree of cardiac involvement

 

c.

Degree of renal involvement

 

d.

Serum free light chains (FLCs)

 

e.

Serum FLCs and degree of cardiac involvement

 

Early treatment intervention is critical in AL amyloid . Upon confirmation of the diagnosis, treatment should commence without delay given the progressive course of the disease. Delays in treatment can increase the number of organs involved or the severity of individual organ impairment. In turn, both factors will limit the available treatments, as patients with advanced single- or multiorgan involvement are less likely to tolerate aggressive regimens, such as high-dose chemotherapy followed by hematopoietic stem cell transplantation (HSCT) .

Treatment is divided into two broad categories, namely high-dose chemotherapy followed by autologous HSCT versus chemotherapy alone. To date, the literature remains mixed in definitively deeming one modality superior over the other. However, for reasons to be discussed below, HSCT remains the favored approach for most clinicians whenever feasible .

Stem cell transplant was first explored as a treatment option in the 1990s. The initial case series published in 1998 raised concern about high treatment-related mortality (TRM). Comenzo reported on a series of 25 patients with primary amyloidosis who were treated with dose-intensive intravenous melphalan followed by an autologous stem cell transplant. [27]. On an intention-to-treat basis, the 3-month mortality associated with therapy was 20 % (5 of 25). Two of the five patients had significant cardiac amyloid involvement and suffered sudden cardiac death [27]. Those with less than two organs involved, and those without cardiac involvement, faired significantly better than their counterparts with more extensive involvement. Moreau et al. reported similar findings in a series of 21 patients with systemic AL amyloidosis [28] . The conditioning regimen consisted of high-dose melphalan either alone or in combination with 12 Gy of total body irradiation [28]. Forty-three percent (9 of 21) of patients died within 1 month of transplantation. Patients with less than two organs involved had improved survival compared to those with more extensive organ involvement. Organ involvement was defined as creatinine clearance < 30 mL/min, protein excretion > 3 g/24 h, congestive heart failure, neuropathy, or hepatomegaly associated with alkaline phosphatase level of > 200 IU/L.

Despite the high TRM of the earliest case series, a retrospective study suggested that there could be a benefit in overall survival and quality of life. One series included 126 patients, half of whom received HSCT and half of whom received chemotherapy alone. The groups were matched with respect to sex, age, left ventricular ejection fraction, interventricular septal wall thickness, peripheral nerve involvement, serum creatinine, and bone marrow plasmacytosis. The overall survival rates at 1 year were 89 % in the HSCT arm and 71 % in the chemotherapy arm, and 71 versus 41 % at 4 years [29] .

Jaccard et al. conducted the only prospective randomized controlled trial that compared chemotherapy to autologous stem cell transplantation. One hundred patients were randomized to receive either melphalan and dexamethasone or high-dose melphalan followed by HSCT. The median survival in the melphalan plus dexamethasone arm was statistically significant longer at 56.9 months compared to 22.2 months in the arm receiving high-dose melphalan followed by stem cell transplantation [30]. Thus, the study failed to show a survival benefit in the transplantation arm. The results of this study should be interpreted with caution. There are limitations to the conclusions that can be drawn based on small number of patients included, the way the data was analyzed, and controversy regarding the selection criteria. From a statistical point of view, the data was evaluated using intention-to-treat analysis (ITT). In ITT, the analysis of results is based on the initial treatment assignment and not on the treatment eventually received in an effort to minimize artifact, such as nonrandom attrition from one arm, when interpreting the results. Of the 50 patients assigned to the stem cell transplant arm, 10 died prior to receiving this treatment. The majority succumbed to sudden death or progressive heart failure. In ITT analysis, these deaths were counted as mortalities in the transplant arm even though the patients never underwent transplantation. In a relatively small sample size, results can be profoundly altered by statistics of this kind .

This study highlights many issues surrounding AL amyloid and transplantation. Multiple myeloma and AL amyloid are both clonal plasma cell disorders and respond to similar treatments. The treatment of AL amyloid has been extrapolated from well-established treatments of multiple myeloma. However, the treatment-related toxicity is very different in both because of the pattern of end-organ damage. In multiple myeloma, patients have a significant burden of disease in the bone marrow but generally have well-preserved organ function with the exception of renal impairment [31]. In contradistinction, AL amyloid patients often have very little disease in the bone marrow with an average tumor burden of 5 % plasma cells [32, 33]. However, the burden of end-organ impairment is significantly higher. This results in starkly different treatment-related toxicity during stem cell mobilization and administration of high-dose chemotherapy .

The high number of deaths prior to transplant may reflect a liberal inclusion criteria for the Jaccard study when compared to other large centers treating AL amyloid [34, 35]. For example, over 84 % of the patients had two or more organs involved and over 25 % of the patients had New York Heart Association (NYHA) Grade III or IV heart failure. The previously discussed retrospective studies and case series by Comenzo and Moreau demonstrate the importance of careful patient selection when considering transplant. These early studies strongly suggest that patients with extensive organ involvement and/or severe cardiac impairment fair significantly worse and should probably be excluded from transplant due to high TRM [27, 28]. The TRM in the transplant arm of the Jaccard study was 24 % which is double the rate of what had previously been reported in single-center studies [28, 34, 3638]. Gertz et al. reported that at the Mayo clinic there was a 40 % reduction in TRM after 2006, which is largely attributed to improved selection criteria [31]. Thus, in the prospective randomized trial by Jaccard, the benefit of transplant may have been masked by inappropriate patient selection .

The considerable heterogeneity in the prognosis of AL amyloid depends on the number and degree of organs involved. At present, the patients must satisfy the criteria listed in Table 14.2 to be eligible for transplant [3941]. Multiple studies have validated these criteria in the selection of patients [28, 4245]. The degree of cardiac impairment, as measured by troponin and NT-pro-BNP is the most potent predictor of outcome [46, 47]. Both values have been incorporated into the Mayo stage and the revised Mayo stage. The Mayo stage is obtained information from 242 patients newly diagnosed with AL amyloidosis [48]. Patients were stratified into three groups based on NT-pro-BNP and troponin T levels. Stage I is defined as Cardiac troponin < 0.035 mcg/L and NT-proBNP < 332 ng/L. Stage II is defined as elevation in one value, either the troponin or NT-proBNP being above the defined threshold values. Stage III is defined as both values being above the threshold values . The median survival is 26 months in stage I, 11 months in stage II, and 4 months in stage III. The staging was also applied to patients receiving stem cell transplant. Of note, patients with stage III disease had less than a 12-month survival even with transplantation [49]. The Revised Mayo staging adds serum FLC and uses different cutoff values of NT-pro-BNP and Troponin T levels to improve risk stratification [50]. As with the original Mayo stage, those with more advanced stages have a poorer prognosis both with and without transplantation.


Table 14.2
Criteria for autologous stem cell transplant

























Age ≤ 70 years

Troponin T < 0.06 ng/mL

NT-proBNP < 5000 ng/L

Creatinine clearance ≥ 30 mL/min (unless on chronic stable dialysis)

Eastern Cooperative Oncology Group (ECOG) performance status ≤ 2

NYHA functional status Class I or II

No more than two organs significantly involved (liver, heart, kidney, or autonomic nerve)

No large pleural effusions

No dependency on oxygen therapy

Adequate factor X levels


Case #2 Follow-up and Discussion

Based on the revised Mayo staging, serum FLCs and cardiac involvements are the most important predictors of outcomes. Choice e is correct.

In summary, many questions remain regarding the utility of stem cell transplantation in AL amyloid. More studies are needed using the now widely accepted eligibility criteria to best select AL amyloid patients for transplant. It also remains to be seen what role transplantation will play as newer immunomodulatory agents and proteasome inhibitors come into use. For example, Dispenzieri et al. reported that in a group of transplant eligible patients who opted for chemotherapy alone the median survival was 42 months, a number which rivals the results seen with transplant [51]. Future studies will need to examine the role of autologous stem cell transplant in the setting of newer chemotherapeutic agents .


Melphalan and Dexamethasone



Case #3

Mr. C is a 76-year-old man who presented to his primary care physician with the complaint of “foamy urine.” He had a normal creatinine. A 24-h urine collection revealed 4.9 g of proteinuria. Serum and urine immunofixation revealed λ type Bence Jones protein. FLCs λ were elevated at 23.6 mg/dL (reference range 0.57–2.63 mg/dL). The κ/λ FLC ratio was 0.08 (reference range 0.26–1.65). He had a kidney biopsy which showed AL amyloid, λ light chain with mild to moderate involvement of glomerular and arterial vessels. A bone marrow biopsy followed which showed a λ-restricted plasma cells and amyloid in the vessel walls. What is the best treatment option for this patient?



a.

Bortezomib

 

b.

Melphalan and dexamethasone

 

c.

High-dose chemotherapy followed by stem cell transplantation.

 

Patients who are ineligible for HSCT due to the severity of organ involvement are precisely the patients who require an effective treatment that provides a rapid response to stabilize or reverse the progression of their disease . Borrowing from the experience with multiple myeloma treatments, several studies examined melphalan and prednisone [52, 53]. Melphalan acts by alkylating DNA bases and cross linking DNA strands, which results in DNA fragmentation due to impaired repair, prevention of DNA synthesis or transcription, and the induction of mispairing of the nucleotide leading to mutations.

In two randomized controlled studies, melphalan and prednisone were superior to colchicine [53, 54]. Importantly, melphalan seemed to be effective and safe in patients with cardiac impairment [55]. This is significant because, as previously discussed, cardiac impairment is a major prognostic indicator as well as a major predictor for TRM and morbidity. The response rate for melphalan and prednisone was 28 % and the time to response was protracted with 70 % of patients experiencing a response in 1 year and an additional 20 % requiring 2 years to show a response to therapy. Based on the Mayo staging and the revised Mayo staging, previously discussed, only patients with stage I disease have a median survival of more than 2 years. Thus, many AL amyloid patients will not live long enough to see a benefit from treatment that requires up to 24 months to reach its maximum effect. For this reason, alternative regimens were explored in the hopes of achieving a brisker response.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 17, 2017 | Posted by in NEPHROLOGY | Comments Off on The Amyloidoses

Full access? Get Clinical Tree

Get Clinical Tree app for offline access