In immunoglobulin light chain amyloidosis a small, indolent plasma cell clone synthesizes light chains that cause devastating organ damage. Early diagnosis, based on prompt recognition of “red-flags” before advanced cardiomyopathy ensues, is essential for improving outcomes. Differentiation from other systemic amyloidoses may require advanced technologies. Prognosis depends on the extent of cardiac involvement, and cardiac biomarkers guide the choice of therapy. The protean clinical presentation requires individualized treatment. Close monitoring of clonal and organ response guides therapy changes and duration. Conventional or high-dose alkylator-based chemotherapy is effective in almost two-thirds of patients. Combinations of proteasome inhibitors, dexamethasone, and alkylators achieve high response rates, although controlled studies are needed. Risk-adapted stem cell transplant and consolidation with novel agents may be considered in selected patients. Immune-modulatory drugs are good options for refractory/relapsed patients. Novel agents and therapeutic targets are expected to be exploited, in an integrated, more effective and less toxic treatment strategy.

The systemic amyloidoses comprise an increasing number of diseases characterized by multiorgan deposition of misfolded and aggregated autologous proteins as β-pleated sheet fibrils.1  Immunoglobulin light chain (AL) amyloidosis is the most common (incidence ∼10 patients per million per year)2  and the most severe because it often targets the heart.3  A small, usually indolent plasma cell (PC) clone synthesizes an unstable, misfolded light chain (LC), which is prone to aggregate and form amyloid fibrils. This process causes systemic toxicity and devastating organ dysfunction.4  Genetic characteristics of amyloid LCs have been associated with kidney5  and heart6  predilection, but mechanisms of tissue specificity and organ dysfunction are poorly understood. Over the past decade, effective regimens have been developed markedly improving survival,7,8  but the 25% to 30% early death rate has not changed, with patients dying within a few weeks of cardiac failure due to late diagnosis. Early diagnosis remains the, as yet, elusive key for improving the care of this dreadful but treatable disease.

The protean clinical features of AL amyloidosis reflect its systemic nature and are detailed in Figure 1. Combinations such as nephrotic syndrome and heart failure, simultaneous peripheral and autonomic neuropathy in nondiabetic patients, “left ventricular hypertrophy” on echocardiography without consistent electrocardiographic evidence or low limb lead voltages, hepatomegaly with normal imaging, or albuminuria in patients with MGUS or myeloma should raise suspicion of amyloidosis.

Figure 1

Pathway for a patient with suspected AL amyloidosis. Signs and symptoms derive from organ involvement: heart and kidney in 70% of patients each, liver in 17% of patients, soft tissues in 17% of patients, peripheral and autonomous nervous system in 15% of patients each, and gastrointestinal tract in 10% of patients. Clinical manifestations with head and neck purpura, unexplained submandibular swelling, signs of heart diastolic dysfunction such as jugular venous distention (white arrowhead), or stiff indented macroglossia, although found in no more than 15% of patients, are prototypic, but late manifestations of the disease. Symptoms and signs derive from advanced organ failure caused by the amyloid-forming process, whereas biomarkers of heart and kidney function (NT-proBNP and albuminuria) may allow identifying presymptomatic patients with initial, possibly reversible, amyloidotic organ damage. The diagnostic process involves the detection of the amyloid deposits and of the underlying PC clone. Considering the wide clinical heterogeneity of this disease, the assessment of the organ dysfunction is necessary in order to define the risk of chemotherapy. The flowchart represents the authors’ view on the current approach for management of a patient with AL amyloidosis. NT-proBNP cutoffs are not applicable in patients with end-stage renal failure. MDex represents the standard of care in most countries, and risk-adapted CTD is an alternative. Although increasingly used in clinical practice, the clinical benefit of adding bortezomib to dexamethasone and alkylating agents frontline remains unproven and is still being tested in controlled trials. ASCT with HMD, autologous stem cell transplantation with high-dose melphalan; BMDex, bortezomib, melphalan, dexamethasone; CTD, cyclophosphamide, thalidomide, dexamethasone; CyBorD, cyclophosphamide, bortezomib, dexamethasone; eGFR, estimated glomerular filtration rate; FLC, serum-free LC measurement; IF, immunofixation; iFISH, interphase fluorescence in situ hybridization; IHC, immunohistochemistry; LC-MS, laser capture microdissection and mass spectrometry; MDex, melphalan dexamethasone; MGUS/MM, monoclonal gammopathy of undetermined significance/multiple myeloma; NT-proBNP, amino-terminal pro–natriuretic peptide type-B; PS, performance status.

Figure 1

Pathway for a patient with suspected AL amyloidosis. Signs and symptoms derive from organ involvement: heart and kidney in 70% of patients each, liver in 17% of patients, soft tissues in 17% of patients, peripheral and autonomous nervous system in 15% of patients each, and gastrointestinal tract in 10% of patients. Clinical manifestations with head and neck purpura, unexplained submandibular swelling, signs of heart diastolic dysfunction such as jugular venous distention (white arrowhead), or stiff indented macroglossia, although found in no more than 15% of patients, are prototypic, but late manifestations of the disease. Symptoms and signs derive from advanced organ failure caused by the amyloid-forming process, whereas biomarkers of heart and kidney function (NT-proBNP and albuminuria) may allow identifying presymptomatic patients with initial, possibly reversible, amyloidotic organ damage. The diagnostic process involves the detection of the amyloid deposits and of the underlying PC clone. Considering the wide clinical heterogeneity of this disease, the assessment of the organ dysfunction is necessary in order to define the risk of chemotherapy. The flowchart represents the authors’ view on the current approach for management of a patient with AL amyloidosis. NT-proBNP cutoffs are not applicable in patients with end-stage renal failure. MDex represents the standard of care in most countries, and risk-adapted CTD is an alternative. Although increasingly used in clinical practice, the clinical benefit of adding bortezomib to dexamethasone and alkylating agents frontline remains unproven and is still being tested in controlled trials. ASCT with HMD, autologous stem cell transplantation with high-dose melphalan; BMDex, bortezomib, melphalan, dexamethasone; CTD, cyclophosphamide, thalidomide, dexamethasone; CyBorD, cyclophosphamide, bortezomib, dexamethasone; eGFR, estimated glomerular filtration rate; FLC, serum-free LC measurement; IF, immunofixation; iFISH, interphase fluorescence in situ hybridization; IHC, immunohistochemistry; LC-MS, laser capture microdissection and mass spectrometry; MDex, melphalan dexamethasone; MGUS/MM, monoclonal gammopathy of undetermined significance/multiple myeloma; NT-proBNP, amino-terminal pro–natriuretic peptide type-B; PS, performance status.

Close modal

Clinical manifestations of AL amyloidosis reflect advanced organ damage. Early diagnosis requires switching from traditional symptoms- and signs-bound diagnostics to sensitive biomarkers signaling presymptomatic organ damage. The progressive, clinically silent involvement of heart and kidneys can be detected early by simple tests. The NT-proBNP is the most sensitive, although not specific, marker for amyloid cardiomyopathy. A concentration >332 ng/L has 100% sensitivity9  and even in asymptomatic patients with normal echocardiogram predicts the development of cardiac amyloidosis.10  Treatment at the asymptomatic stage may prevent irreversible organ damage, and better organ function will allow adequate treatment delivery.

Undetected advanced organ failure can ensue even during careful follow-up of individuals with MGUS, unless looked for specifically. We would recommend that all patients with MGUS and abnormal FLC ratio (who are at higher risk of developing amyloidosis and should undergo lifelong monitoring for symptomatic myeloma11 ) have, additionally, NT-proBNP and urine albumin assessed at each visit. Unexplained positive results should promptly trigger procedures to diagnose amyloidosis.

Diagnosing AL amyloidosis involves 4 steps: proving systemic amyloid deposition, typing the deposits, assessing the monoclonal disease, and defining the extent of systemic damage including risk stratification/staging. Localized AL amyloidosis, resulting from in situ (eg, skin, airways, and urinary tract) production of LC, usually does not necessitate systemic therapy and should be differentiated from systemic amyloidosis, characterized by visceral involvement.12 

Demonstration of amyloid deposition in a tissue biopsy by Congo red staining remains the gold standard, although novel methods have been proposed.13,14  The most accessible site is periumbilical fat that can be easily and innocuously aspirated. Labial salivary gland biopsy is positive in >50% of patients with negative fat biopsy.15  If needed, target organs can be biopsied after careful assessment of hemostasis. Tissue deposits should be typed using mass spectrometry (the current gold standard),16,17  immuno–electron microscopy,18  or immunohistochemistry in specialized laboratories.19  Confirmation of fibril type is critical because a dozen proteins can cause systemic amyloidosis,13  each requiring distinct therapy. Even in patients with MGUS, cardiomyopathy caused by V122I mutant transthyretin (carrier rate of 4% in Afro-Caribbeans) or by wild-type transthyretin in elderly men with senile systemic amyloidosis, and amyloidosis reactive to chronic inflammation (AA) should be carefully considered in differential diagnosis.20,21  Gene sequencing is needed when familial amyloidosis is possible: such as in those with isolated neuropathic or cardiac disease (transthyretin amyloidosis), isolated renal involvement (fibrinogen amyloidosis), corneal lattice dystrophy, progressive bilateral facial paralysis and cutis laxa (gelsolin amyloidosis), dry mouth/gastrointestinal/kidney/liver involvement (lysozyme amyloidosis), and renal/liver/cardiac involvement in relatively asymptomatic patients (apolipoprotein-A1 amyloidosis). Amyloid typing and gene sequencing are available at all major referral centers.

The identification of amyloidogenic monoclonal proteins requires serum and urine immunofixation combined with FLC quantification.22  Half of all amyloidogenic PC clones produce LC only, with typically modest bone marrow infiltrate (median 5% to 10%). The λ clones dominate κ ones by 4:1, unlike the 2:3 ratio in myeloma. Fluorescence in situ hybridization of bone marrow PC and investigations to rule out symptomatic myeloma, including skeletal survey, should be done at baseline.11 

Assessment of amyloid-related organ damage is the next step. Echocardiography, including tissue Doppler and strain imaging, defines baseline cardiac function, in addition to widely available and well-standardized cardiac biomarkers.23,24  Cardiac magnetic resonance imaging is useful in diagnosing and possibly monitoring amyloid deposits.25  Renal involvement is best evaluated by eGFR and albuminuria. Liver function tests and size document involvement. Whole body imaging techniques like serum amyloid P component scintigraphy are useful, where available, for diagnosis and monitoring.26 99mTc-3,3-diphosphono-1,2-propanodicarboxylic acid or pyrophosphate scans help in differentiating cardiac AL from transthyretin amyloidosis (both mutated and wild type) and may spare cardiac biopsy.27  Simple parameters like poor performance status, New York Heart Association class ≥3, and low systolic blood pressure (<100 mm Hg) are useful bedside indicators of poor outcome.28  Because cardiac damage determines survival and treatment tolerability, the Mayo staging system29  using NT-proBNP (>332 ng/L) and cardiac troponin-T (cTnT)/troponin-I (cTnT, >0.035 ng/mL; cardiac troponin-I, >0.1 ng/mL), or more recently high-sensitivity cTnT (>0.077 ng/mL),30  is the most robust method for risk stratification. Stage III patients have a median survival of 3.5 to 8 months.28-31  High FLC levels predict poor outcome and early mortality32,33  and have been incorporated in the Mayo staging system.34  Renal function (especially when eGFR <30 mL/min) affects cardiac biomarkers concentration, and the Mayo staging is not directly applicable for patients in renal failure. BNP is more useful than NT-proBNP in these patients.35 

Preliminary cytogenetic data suggest that the amyloidogenic clone may represent an early stage in PC dyscrasias progression.36  The recent observation that after ASCT patients with AL amyloidosis survive longer than subjects with multiple myeloma, especially those achieving complete response (CR),37  may indicate that the AL clone is more susceptible to chemotherapy than the myeloma clone or, alternatively, that it takes longer to resurface due to its slower proliferative rate.

Treating AL amyloidosis needs to be adapted to the heterogeneous manifestations and seriousness of the disease, resulting from different patterns and severity of organ involvement. Low-risk patients may be candidates for aggressive therapy, and high-risk subjects need gentle yet rapidly acting regimens (Figure 1). Clinical benefit depends on profound hematologic response translating into organ, especially cardiac or renal, response. Treatment is aimed at rapidly eliminating the toxic LC, ideally achieving CR (negative serum and urine immunofixation and normal FLC ratio) or at least very good partial response (VGPR; difference between involved and uninvolved FLC <40 mg/L). Partial response (decrease in difference between involved and uninvolved FLC >50%) is now considered an unsatisfactory end point.38  Therapy can be continued for 1 to 2 cycles beyond best response for consolidation. Hematologic and cardiac response should be assessed frequently, every 2 cycles (or 3 months after ASCT), promptly switching therapy, if ineffective, to prevent progression of organ damage. An NT-proBNP decrease of >30% and >300 ng/L (if baseline is ≥650 ng/L)38  guides treatment adequacy, mindful of increases in renal failure35  and immune-modulatory therapy.39  The best renal outcomes are seen in patients who achieve VGPR or better.40  Renal responses can be delayed and, hence, difficult to use as a guide for therapy adequacy. Factors influencing progression to dialysis are lower GFR (<60 mL/min), lower serum albumin, and less than VGPR.40 

Most available evidence derives from small uncontrolled trials and retrospective series (Table 1)41  with only 1 randomized trial completed in the past 15 years. It failed to demonstrate an advantage for ASCT over MDex, even accounting for the high (24%) transplant-related mortality,42  but was before the era of cardiac biomarkers. Therefore, it is of paramount importance that patients are entered into controlled trials whenever possible.

Table 1

Selected therapy regimens in AL amyloidosis

Therapy regimensDosing scheduleN (F)H (stage III)HR/CR (OR)Most common grade ≥3 SAE100-d mortalityMedian PFS/OS (y)
Controlled phase 3 trial        
 MDex M 10 mg/m2 + Dex 40 mg days 1-4 q28 43 (100%) 46% 68%/32% (39%) Overall 16% 2% TTP 2.7/OS 4.7 
 vs ASCT42  M 140-200 mg/m2 37 (100%) 48% 67%/41% (45%) Hemodialysis 22% 24% TTP 2.7/OS 1.8 
Autologous stem cell transplantation        
 ASCT45  M 100-200 mg/m2 421 (76%) 45% M200 -/43% — M200 9% M200 3.4/8.4 
M100-140 -/24% (53%) M100-140 14% M100-140 1.8/3.8 
 ASCT41  M 100-200 mg/m2 434 49% (25%) 76%/39% (47%) — 10% CR -/not reached 
PR -/8.9 
NR -/2.7 
 Risk-adapted ASCT48  M 100/140/200 mg/m2 40 (100%) 65% (28%) 79%/58% (70%) During BDex, Tp 43%, cardiac 20%, anemia 13% ASCT 10% At 2 y: 69%/82% 
B 1.3 mg/m2 (OW first 2 cycles, then TW) BDex 4% 
Dex 20 mg day of B and following day 
Conventional chemotherapy        
 MDex55,56  M 0.22 mg/kg + Dex 40 mg days 1-4 q28 46 (100%) 70% 67%/33% (48%) Overall 11%, infection 6% 4% 3.8/5.1 
Immune-modulatory–based therapy        
 CTD57  C 500 mg on days 1, 8, 15 q21/28 75 (41%) 59% 74%/21% (27%) Grade ≥2: sedation 40%, fluid retention 21% 4% 1.7/3.4 
T 50-200 mg/d 
Dex 40 mg on days 1-4, 9-12 q21 
or 20 mg on days 1-4 and 15-18q28 
 LDex61  L up to 25 mg on days 1-21 q28 22 (41%) 64% (23%) 41%/- (23%) Overall 86%, neutropenia 45%, Tp 27%, rash 18%, infection 18%, fatigue 18% 18% 1.6/- 
Dex 40 mg on days 1-4, 15-18 q28 
added in nonresponders to L alone 
 CLD66  C 300 mg/m2 on days 1, 8, 15 q 28 35 (11%) 63% (43%) 60%/11% (31%) Overall 74%, Np 40%, Tp 40%, rash 10%, thrombosis 10% 9% 2.4/3.1 
L 15 mg on days 1-21 q 28 
Dex 40 mg on days 1, 8, 15, 22 q28 
 MLD69  M 0.18 mg/kg on days 1-4 q28 26 (100%) 58% 58%/23%, 42% with full-dose L (50%) Overall 81%, Np 11%, heart failure 11% — At 2 y: 54%/81% 
L 5-15 mg on days 1-21 q28 
Dex 40 mg on days 1-4 q28 
 PDex63  Pomalidomide 2 mg/d 33 (0) 82% (25%) 48%/3% (15%) Np 30%, infection 27%, arrhythmia 21%, fatigue 18% 3% 1.2/2.3 
Dex 40 mg OW 
Proteasome inhibitor–based therapy        
 Bortezomib60  1.6 mg/m2 OW 70 (0) 56% OW: 69%/38% Overall OW 50%, TW 79% 3% At 1 y: OW: 72%/93% 
1.3 mg/m2 TW TW: 67%/24% (29%) Tp 18% (TW), vomiting 12% (TW) TW: 75%/84% 
 CyBorD50  C 350 mg/m2 on days 1, 8, 15 B 1.0-1.3 mg/m2 TW 43 (47%) 74% (46%) 81%/65% frontline, 22% pretreated (46%) 19% discontinued (due to neuropathy in 14%) At 2 y: 53%/98% 
Dex 20 mg TW  
 Ixazomib71  4.0 mg on days 1, 8, 15 q28 (MTD) 16 (0) 69% (6%) 42%/8%, (Any grade) nausea 31%, diarrhea 25%, thrombocytopenia 25% — 
Dex 40 mg on days 1-4 (added in nonresponders) VGPR 25% (-) 
Therapy regimensDosing scheduleN (F)H (stage III)HR/CR (OR)Most common grade ≥3 SAE100-d mortalityMedian PFS/OS (y)
Controlled phase 3 trial        
 MDex M 10 mg/m2 + Dex 40 mg days 1-4 q28 43 (100%) 46% 68%/32% (39%) Overall 16% 2% TTP 2.7/OS 4.7 
 vs ASCT42  M 140-200 mg/m2 37 (100%) 48% 67%/41% (45%) Hemodialysis 22% 24% TTP 2.7/OS 1.8 
Autologous stem cell transplantation        
 ASCT45  M 100-200 mg/m2 421 (76%) 45% M200 -/43% — M200 9% M200 3.4/8.4 
M100-140 -/24% (53%) M100-140 14% M100-140 1.8/3.8 
 ASCT41  M 100-200 mg/m2 434 49% (25%) 76%/39% (47%) — 10% CR -/not reached 
PR -/8.9 
NR -/2.7 
 Risk-adapted ASCT48  M 100/140/200 mg/m2 40 (100%) 65% (28%) 79%/58% (70%) During BDex, Tp 43%, cardiac 20%, anemia 13% ASCT 10% At 2 y: 69%/82% 
B 1.3 mg/m2 (OW first 2 cycles, then TW) BDex 4% 
Dex 20 mg day of B and following day 
Conventional chemotherapy        
 MDex55,56  M 0.22 mg/kg + Dex 40 mg days 1-4 q28 46 (100%) 70% 67%/33% (48%) Overall 11%, infection 6% 4% 3.8/5.1 
Immune-modulatory–based therapy        
 CTD57  C 500 mg on days 1, 8, 15 q21/28 75 (41%) 59% 74%/21% (27%) Grade ≥2: sedation 40%, fluid retention 21% 4% 1.7/3.4 
T 50-200 mg/d 
Dex 40 mg on days 1-4, 9-12 q21 
or 20 mg on days 1-4 and 15-18q28 
 LDex61  L up to 25 mg on days 1-21 q28 22 (41%) 64% (23%) 41%/- (23%) Overall 86%, neutropenia 45%, Tp 27%, rash 18%, infection 18%, fatigue 18% 18% 1.6/- 
Dex 40 mg on days 1-4, 15-18 q28 
added in nonresponders to L alone 
 CLD66  C 300 mg/m2 on days 1, 8, 15 q 28 35 (11%) 63% (43%) 60%/11% (31%) Overall 74%, Np 40%, Tp 40%, rash 10%, thrombosis 10% 9% 2.4/3.1 
L 15 mg on days 1-21 q 28 
Dex 40 mg on days 1, 8, 15, 22 q28 
 MLD69  M 0.18 mg/kg on days 1-4 q28 26 (100%) 58% 58%/23%, 42% with full-dose L (50%) Overall 81%, Np 11%, heart failure 11% — At 2 y: 54%/81% 
L 5-15 mg on days 1-21 q28 
Dex 40 mg on days 1-4 q28 
 PDex63  Pomalidomide 2 mg/d 33 (0) 82% (25%) 48%/3% (15%) Np 30%, infection 27%, arrhythmia 21%, fatigue 18% 3% 1.2/2.3 
Dex 40 mg OW 
Proteasome inhibitor–based therapy        
 Bortezomib60  1.6 mg/m2 OW 70 (0) 56% OW: 69%/38% Overall OW 50%, TW 79% 3% At 1 y: OW: 72%/93% 
1.3 mg/m2 TW TW: 67%/24% (29%) Tp 18% (TW), vomiting 12% (TW) TW: 75%/84% 
 CyBorD50  C 350 mg/m2 on days 1, 8, 15 B 1.0-1.3 mg/m2 TW 43 (47%) 74% (46%) 81%/65% frontline, 22% pretreated (46%) 19% discontinued (due to neuropathy in 14%) At 2 y: 53%/98% 
Dex 20 mg TW  
 Ixazomib71  4.0 mg on days 1, 8, 15 q28 (MTD) 16 (0) 69% (6%) 42%/8%, (Any grade) nausea 31%, diarrhea 25%, thrombocytopenia 25% — 
Dex 40 mg on days 1-4 (added in nonresponders) VGPR 25% (-) 

B, bortezomib; BDex, bortezomib dexamethasone; C, cyclophosphamide; CLD, cyclophosphamide, lenalidomide, dexamethasone; Dex, dexamethasone; F, frontline therapy; H, heart involvement; HR, overall hematologic response; L, lenalidomide; LDex, lenalidomide plus dexamethasone; M, melphalan; MLD, melphalan, lenalidomide, dexamethasone; MTD, maximum tolerated dose; N, number of patients; Np, neutropenia; NR, nonresponders; OR, organ response; OS, overall survival; OW, once weekly; PDex, pomalidomide plus dexamethasone; PFS, progression-free survival; PR, partial response; q, every; SAE, severe adverse events; T, thalidomide; Tp, thrombocytopenia; TTP, time to hematologic progression; TW, twice weekly.

Intention-to-treat response rates are reported.

Frontline therapy

ASCT should be considered in low-risk patients (∼15% to 20%) given the long-term survival in responders. Improvements in patient selection, and particularly the exclusion of subjects with elevated cardiac biomarkers, can reduce transplant-related mortality to <5%.43-45  Current eligibility criteria include the following: cTnT <0.06 ng/mL, NT-proBNP <5000 ng/L,43  age <65 years, performance status 0 to 2, ejection fraction >45%, systolic blood pressure >90 mm Hg (standing), and CO diffusion capacity >50%.46  Melphalan dose may be risk adapted (100, 140, or 200 mg/m2) based on age and renal and cardiac function. After risk-adapted melphalan, consolidation with bortezomib (or thalidomide) increases CR rate to ∼50%, with median survival approaching 8 years.47-49  Alternatively, patients not willing to undergo ASCT can receive stem cell–sparing regimens, such as CyBorD,50,51  as “induction therapy,” transplanting only subjects not attaining CR. However, the definition of the durability of responses with CyBorD requires longer follow-up.

Stage III patients with NT-proBNP >8500 ng/L are at high-risk of death within a few weeks28  and need rapidly active treatment. They are extremely fragile and sensitive to treatment toxicity, particularly high-dose dexamethasone-induced fluid retention and arrhythmias. Standard regimens cannot overcome their dismal outcome.52  Bortezomib combinations, particularly CyBorD, given the rapid action and the vulnerability of amyloidogenic PC to bortezomib,53,54  seem ideal (long-term survivors reported), but prospective upfront studies are still lacking.50  Reduced doses of dexamethasone (10-20 mg) and bortezomib (0.7-1.0 mg/m2) are recommended in high-risk patients with cautious week-by-week dose increase.

Most patients (∼60%) are intermediate risk and are best treated with combination chemotherapy, MDex, although some may still be suitable for ASCT in centers of experience. There is a trend to treat these patients with bortezomib-containing regimens, but evidence of superiority, tolerability (particularly in the elderly), and comparable documented long-lasting responses of standard regimen MDex remain lacking.42,55,56  An ongoing randomized trial comparing MDex with MDex plus bortezomib aims to establish the standard of care in these subjects. If contraindications to ASCT are potentially reversible, a stem cell–sparing regimen is preferable, and CyBorD or CTD57  may be considered.

Patients with overt myeloma and symptomatic AL amyloidosis have a poor prognosis58  and require risk-adapted chemotherapy. Asymptomatic amyloid deposits will not alter the prognosis in myeloma. AL amyloidosis caused by an underlying (usually lymphoplasmacytic) lymphoma poses a particular therapeutic challenge especially due to the low achievable CR/VGPR rates. Suitable patients should be considered upfront for ASCT; otherwise, a rapidly acting combination regimen including rituximab and bortezomib should be used.

Relapsing-refractory patients

The choice of drugs depends on previous treatment and clinical presentation. An attempt to restore response with the regimen used frontline can be made if first remission was prolonged. A bortezomib combination is the first choice in subjects, without severe neuropathy, unexposed to this drug.59,60  Lenalidomide and pomalidomide are good options for overcoming resistance to alkylators, bortezomib, and thalidomide.61-64  Lenalidomide requires careful monitoring of renal function.65  Combination of lenalidomide and alkylators may improve the response rate, but at the cost of significant myelosuppression.66-70  New drugs (carfilzomib, ixazomib,71  and bendamustine72 ) are under evaluation and may widen the therapeutic options. At present, there are no data to support maintenance treatment.

Supportive therapy

Supportive therapy, recently reviewed in Merlini et al3  and Palladini et al,73  is vital. Patient education with daily weights, judicious diuretic use, salt-poor albumin, cautious angiotensin converting enzyme-inhibitors, and close multidisciplinary monitoring makes lifesaving differences. Renal and cardiac transplantation may prolong survival and improve quality of life in subjects with irreversible organ damage. Young patients with isolated advanced cardiac involvement should be considered for heart transplant followed by ASCT.74-77  The use of left ventricular assist devices needs further study.78 

Biomarker-based staging and frequent response assessment has helped to improve the survival of patients with AL amyloidosis. When the diagnosis is made early, before advanced organ dysfunction ensues, recovery of organ function and prolonged survival are possible. Changing the approach to monitoring MGUS may reduce late diagnoses, preventing early deaths, but this remains a major challenge. A new framework for randomized clinical trials with novel agents will establish contemporary standards of care.79  Novel agents and therapeutic approaches, such as those targeting amyloid deposits,80  are now under development and, hopefully in the near future, will be used synergistically to give a concrete hope of curing AL amyloidosis.

The authors thank Dr Helen J. Lachmann and Dr Laura Obici for critical reading of the manuscript.

This work was supported by grants from “Associazione Italiana per la Ricerca sul Cancro” Special Program Molecular Clinical Oncology 5 per mille n. 9965 “Harnessing tumor cell/microenvironment cross talk to treat mature B cell tumors.”

Contribution: G.M., A.D.W., and G.P. designed the review and wrote the manuscript.

Conflict-of-interest disclosure: G.M. received honoraria from Neotope, Pfizer, and Millennium. A.D.W. received honoraria from Janssen-Cilag. The remaining author declares no competing financial interests.

Correspondence: Giampaolo Merlini, Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Viale Golgi, 19 – 27100 Pavia, Italy; e-mail: [email protected].

1
Merlini
 
G
Bellotti
 
V
Molecular mechanisms of amyloidosis.
N Engl J Med
2003
, vol. 
349
 
6
(pg. 
583
-
596
)
2
Kyle
 
RA
Linos
 
A
Beard
 
CM
et al. 
Incidence and natural history of primary systemic amyloidosis in Olmsted County, Minnesota, 1950 through 1989.
Blood
1992
, vol. 
79
 
7
(pg. 
1817
-
1822
)
3
Merlini
 
G
Seldin
 
DC
Gertz
 
MA
Amyloidosis: pathogenesis and new therapeutic options.
J Clin Oncol
2011
, vol. 
29
 
14
(pg. 
1924
-
1933
)
4
Merlini
 
G
Stone
 
MJ
Dangerous small B-cell clones.
Blood
2006
, vol. 
108
 
8
(pg. 
2520
-
2530
)
5
Comenzo
 
RL
Wally
 
J
Kica
 
G
et al. 
Clonal immunoglobulin light chain variable region germline gene use in AL amyloidosis: association with dominant amyloid-related organ involvement and survival after stem cell transplantation.
Br J Haematol
1999
, vol. 
106
 
3
(pg. 
744
-
751
)
6
Perfetti
 
V
Palladini
 
G
Casarini
 
S
et al. 
The repertoire of λ light chains causing predominant amyloid heart involvement and identification of a preferentially involved germline gene, IGLV1-44.
Blood
2012
, vol. 
119
 
1
(pg. 
144
-
150
)
7
Kumar
 
SK
Gertz
 
MA
Lacy
 
MQ
et al. 
Recent improvements in survival in primary systemic amyloidosis and the importance of an early mortality risk score.
Mayo Clin Proc
2011
, vol. 
86
 
1
(pg. 
12
-
18
)
8
Merlini
 
G
CyBorD: stellar response rates in AL amyloidosis.
Blood
2012
, vol. 
119
 
19
(pg. 
4343
-
4345
)
9
Palladini
 
G
Campana
 
C
Klersy
 
C
et al. 
Serum N-terminal pro-brain natriuretic peptide is a sensitive marker of myocardial dysfunction in AL amyloidosis.
Circulation
2003
, vol. 
107
 
19
(pg. 
2440
-
2445
)
10
Wechalekar
 
AD
Gillmore
 
JD
Wassef
 
N
Lachmann
 
HJ
Whelan
 
C
Hawkins
 
PN
Abnormal N-terminal fragment of brain natriuretic peptide in patients with light chain amyloidosis without cardiac involvement at presentation is a risk factor for development of cardiac amyloidosis.
Haematologica
2011
, vol. 
96
 
7
(pg. 
1079
-
1080
)
11
Kyle
 
RA
Durie
 
BG
Rajkumar
 
SV
et al. 
International Myeloma Working Group
Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management.
Leukemia
2010
, vol. 
24
 
6
(pg. 
1121
-
1127
)
12
Gertz
 
MA
Comenzo
 
R
Falk
 
RH
et al. 
Definition of organ involvement and treatment response in immunoglobulin light chain amyloidosis (AL): a consensus opinion from the 10th International Symposium on Amyloid and Amyloidosis, Tours, France, 18-22 April 2004.
Am J Hematol
2005
, vol. 
79
 
4
(pg. 
319
-
328
)
13
Sipe
 
JD
Benson
 
MD
Buxbaum
 
JN
et al. 
Nomenclature Committee of the International Society of Amyloidosis
Amyloid fibril protein nomenclature: 2012 recommendations from the Nomenclature Committee of the International Society of Amyloidosis.
Amyloid
2012
, vol. 
19
 
4
(pg. 
167
-
170
)
14
Nilsson
 
KP
Ikenberg
 
K
Aslund
 
A
et al. 
Structural typing of systemic amyloidoses by luminescent-conjugated polymer spectroscopy.
Am J Pathol
2010
, vol. 
176
 
2
(pg. 
563
-
574
)
15
Foli
 
A
Palladini
 
G
Caporali
 
R
et al. 
The role of minor salivary gland biopsy in the diagnosis of systemic amyloidosis: results of a prospective study in 62 patients.
Amyloid
2011
, vol. 
18
 
suppl 1
(pg. 
80
-
82
)
16
Vrana
 
JA
Gamez
 
JD
Madden
 
BJ
Theis
 
JD
Bergen
 
HR
Dogan
 
A
Classification of amyloidosis by laser microdissection and mass spectrometry-based proteomic analysis in clinical biopsy specimens.
Blood
2009
, vol. 
114
 
24
(pg. 
4957
-
4959
)
17
Brambilla
 
F
Lavatelli
 
F
Di Silvestre
 
D
et al. 
Reliable typing of systemic amyloidoses through proteomic analysis of subcutaneous adipose tissue.
Blood
2012
, vol. 
119
 
8
(pg. 
1844
-
1847
)
18
Arbustini
 
E
Morbini
 
P
Verga
 
L
et al. 
Light and electron microscopy immunohistochemical characterization of amyloid deposits.
Amyloid
1997
, vol. 
4
 
3
(pg. 
157
-
170
)
19
Schönland
 
SO
Hegenbart
 
U
Bochtler
 
T
et al. 
Immunohistochemistry in the classification of systemic forms of amyloidosis: a systematic investigation of 117 patients.
Blood
2012
, vol. 
119
 
2
(pg. 
488
-
493
)
20
Lachmann
 
HJ
Booth
 
DR
Booth
 
SE
et al. 
Misdiagnosis of hereditary amyloidosis as AL (primary) amyloidosis.
N Engl J Med
2002
, vol. 
346
 
23
(pg. 
1786
-
1791
)
21
Comenzo
 
RL
Zhou
 
P
Fleisher
 
M
Clark
 
B
Teruya-Feldstein
 
J
Seeking confidence in the diagnosis of systemic AL (Ig light-chain) amyloidosis: patients can have both monoclonal gammopathies and hereditary amyloid proteins.
Blood
2006
, vol. 
107
 
9
(pg. 
3489
-
3491
)
22
Palladini
 
G
Russo
 
P
Bosoni
 
T
et al. 
Identification of amyloidogenic light chains requires the combination of serum-free light chain assay with immunofixation of serum and urine.
Clin Chem
2009
, vol. 
55
 
3
(pg. 
499
-
504
)
23
Falk
 
RH
Diagnosis and management of the cardiac amyloidoses.
Circulation
2005
, vol. 
112
 
13
(pg. 
2047
-
2060
)
24
Buss
 
SJ
Emami
 
M
Mereles
 
D
et al. 
Longitudinal left ventricular function for prediction of survival in systemic light-chain amyloidosis: incremental value compared with clinical and biochemical markers.
J Am Coll Cardiol
2012
, vol. 
60
 
12
(pg. 
1067
-
1076
)
25
Banypersad
 
SM
Sado
 
DM
Flett
 
AS
et al. 
Quantification of myocardial extracellular volume fraction in systemic AL amyloidosis: an equilibrium contrast cardiovascular magnetic resonance study.
Circ Cardiovasc Imaging
2013
, vol. 
6
 
1
(pg. 
34
-
39
)
26
Hawkins
 
PN
Lavender
 
JP
Pepys
 
MB
Evaluation of systemic amyloidosis by scintigraphy with 123I-labeled serum amyloid P component.
N Engl J Med
1990
, vol. 
323
 
8
(pg. 
508
-
513
)
27
Rapezzi
 
C
Quarta
 
CC
Guidalotti
 
PL
et al. 
Usefulness and limitations of 99mTc-3,3-diphosphono-1,2-propanodicarboxylic acid scintigraphy in the aetiological diagnosis of amyloidotic cardiomyopathy.
Eur J Nucl Med Mol Imaging
2011
, vol. 
38
 
3
(pg. 
470
-
478
)
28
Wechalekar
 
AD
Schonland
 
SO
Kastritis
 
E
et al. 
A European collaborative study of treatment outcomes in 346 patients with cardiac stage III AL amyloidosis.
Blood
2013
, vol. 
121
 
17
(pg. 
3420
-
3427
)
29
Dispenzieri
 
A
Gertz
 
MA
Kyle
 
RA
et al. 
Serum cardiac troponins and N-terminal pro-brain natriuretic peptide: a staging system for primary systemic amyloidosis.
J Clin Oncol
2004
, vol. 
22
 
18
(pg. 
3751
-
3757
)
30
Palladini
 
G
Barassi
 
A
Klersy
 
C
et al. 
The combination of high-sensitivity cardiac troponin T (hs-cTnT) at presentation and changes in N-terminal natriuretic peptide type B (NT-proBNP) after chemotherapy best predicts survival in AL amyloidosis.
Blood
2010
, vol. 
116
 
18
(pg. 
3426
-
3430
)
31
Kristen
 
AV
Giannitsis
 
E
Lehrke
 
S
et al. 
Assessment of disease severity and outcome in patients with systemic light-chain amyloidosis by the high-sensitivity troponin T assay.
Blood
2010
, vol. 
116
 
14
(pg. 
2455
-
2461
)
32
Dispenzieri
 
A
Lacy
 
MQ
Katzmann
 
JA
et al. 
Absolute values of immunoglobulin free light chains are prognostic in patients with primary systemic amyloidosis undergoing peripheral blood stem cell transplantation.
Blood
2006
, vol. 
107
 
8
(pg. 
3378
-
3383
)
33
Wechalekar
 
AD
Wassef
 
N
Lachmann
 
H
et al. 
High early mortality and poor outcomes for patients with AL amyloidosis presenting with high serum free light chains - a new risk stratification model [abstract].
Haematologica
2009
, vol. 
94
 
suppl 2
pg. 
222
  
Abstract 0544
34
Kumar
 
S
Dispenzieri
 
A
Lacy
 
MQ
et al. 
Revised prognostic staging system for light chain amyloidosis incorporating cardiac biomarkers and serum free light chain measurements.
J Clin Oncol
2012
, vol. 
30
 
9
(pg. 
989
-
995
)
35
Palladini
 
G
Foli
 
A
Milani
 
P
et al. 
Best use of cardiac biomarkers in patients with AL amyloidosis and renal failure.
Am J Hematol
2012
, vol. 
87
 
5
(pg. 
465
-
471
)
36
Bochtler
 
T
Hegenbart
 
U
Heiss
 
C
et al. 
Hyperdiploidy is less frequent in AL amyloidosis compared with monoclonal gammopathy of undetermined significance and inversely associated with translocation t(11;14).
Blood
2011
, vol. 
117
 
14
(pg. 
3809
-
3815
)
37
Dispenzieri
 
A
Seenithamby
 
K
Lacy
 
MQ
et al. 
Patients with immunoglobulin light chain amyloidosis undergoing autologous stem cell transplantation have superior outcomes compared with patients with multiple myeloma: a retrospective review from a tertiary referral center.
Bone Marrow Transplant
2013
 
[Epub ahead of print]
38
Palladini
 
G
Dispenzieri
 
A
Gertz
 
MA
et al. 
New criteria for response to treatment in immunoglobulin light chain amyloidosis based on free light chain measurement and cardiac biomarkers: impact on survival outcomes.
J Clin Oncol
2012
, vol. 
30
 
36
(pg. 
4541
-
4549
)
39
Dispenzieri
 
A
Dingli
 
D
Kumar
 
SK
et al. 
Discordance between serum cardiac biomarker and immunoglobulin-free light-chain response in patients with immunoglobulin light-chain amyloidosis treated with immune modulatory drugs.
Am J Hematol
2010
, vol. 
85
 
10
(pg. 
757
-
759
)
40
Pinney
 
JH
Lachmann
 
HJ
Bansi
 
L
et al. 
Outcome in renal Al amyloidosis after chemotherapy.
J Clin Oncol
2011
, vol. 
29
 
6
(pg. 
674
-
681
)
41
Gertz
 
MA
Lacy
 
MQ
Dispenzieri
 
A
et al. 
Autologous stem cell transplant for immunoglobulin light chain amyloidosis: a status report.
Leuk Lymphoma
2010
, vol. 
51
 
12
(pg. 
2181
-
2187
)
42
Jaccard
 
A
Moreau
 
P
Leblond
 
V
et al. 
Myélome Autogreffe (MAG) and Intergroupe Francophone du Myélome (IFM) Intergroup
High-dose melphalan versus melphalan plus dexamethasone for AL amyloidosis.
N Engl J Med
2007
, vol. 
357
 
11
(pg. 
1083
-
1093
)
43
Gertz
 
MA
Lacy
 
MQ
Dispenzieri
 
A
et al. 
Refinement in patient selection to reduce treatment-related mortality from autologous stem cell transplantation in amyloidosis.
Bone Marrow Transplant
2013
, vol. 
48
 
4
(pg. 
557
-
561
)
44
Mangatter
 
A
Schoenland
 
SO
Hansberg
 
M
et al. 
Improvement of long-term survival after high-dose melphalan in patients with light chain amyloidosis responding to induction chemotherapy [abstract].
Blood
2008
, vol. 
112
 
11
 
Abstract 3334
45
Cibeira
 
MT
Sanchorawala
 
V
Seldin
 
DC
et al. 
Outcome of AL amyloidosis after high-dose melphalan and autologous stem cell transplantation: long-term results in a series of 421 patients.
Blood
2011
, vol. 
118
 
16
(pg. 
4346
-
4352
)
46
Palladini
 
G
Merlini
 
G
Transplantation vs. conventional-dose therapy for amyloidosis.
Curr Opin Oncol
2011
, vol. 
23
 
2
(pg. 
214
-
220
)
47
Cohen
 
AD
Zhou
 
P
Chou
 
J
et al. 
Risk-adapted autologous stem cell transplantation with adjuvant dexamethasone +/- thalidomide for systemic light-chain amyloidosis: results of a phase II trial.
Br J Haematol
2007
, vol. 
139
 
2
(pg. 
224
-
233
)
48
Landau
 
H
Hassoun
 
H
Rosenzweig
 
MA
et al. 
Bortezomib and dexamethasone consolidation following risk-adapted melphalan and stem cell transplantation for patients with newly diagnosed light-chain amyloidosis.
Leukemia
2013
, vol. 
27
 
4
(pg. 
823
-
828
)
49
Comenzo
 
RL
Fein
 
DE
Hassoun
 
H
et al. 
Long-term outcomes of patients with systemic light chain amyloidosis (AL) treated at diagnosis with risk-adapted stem cell transplant and consolidation with novel agents [abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 3150
50
Venner
 
CP
Lane
 
T
Foard
 
D
et al. 
Cyclophosphamide, bortezomib, and dexamethasone therapy in AL amyloidosis is associated with high clonal response rates and prolonged progression-free survival.
Blood
2012
, vol. 
119
 
19
(pg. 
4387
-
4390
)
51
Mikhael
 
JR
Schuster
 
SR
Jimenez-Zepeda
 
VH
et al. 
Cyclophosphamide-bortezomib-dexamethasone (CyBorD) produces rapid and complete hematologic response in patients with AL amyloidosis.
Blood
2012
, vol. 
119
 
19
(pg. 
4391
-
4394
)
52
Dietrich
 
S
Schönland
 
SO
Benner
 
A
et al. 
Treatment with intravenous melphalan and dexamethasone is not able to overcome the poor prognosis of patients with newly diagnosed systemic light chain amyloidosis and severe cardiac involvement.
Blood
2010
, vol. 
116
 
4
(pg. 
522
-
528
)
53
Sitia
 
R
Palladini
 
G
Merlini
 
G
Bortezomib in the treatment of AL amyloidosis: targeted therapy?
Haematologica
2007
, vol. 
92
 
10
(pg. 
1302
-
1307
)
54
Oliva
 
L
Pengo
 
N
Palladini
 
G
et al. 
Proteasome activity and stress in light chain amyloidosis [abstract].
Amyloid
2010
, vol. 
17
 
suppl 1
 
99. Abstract OP-043
55
Palladini
 
G
Perfetti
 
V
Obici
 
L
et al. 
Association of melphalan and high-dose dexamethasone is effective and well tolerated in patients with AL (primary) amyloidosis who are ineligible for stem cell transplantation.
Blood
2004
, vol. 
103
 
8
(pg. 
2936
-
2938
)
56
Palladini
 
G
Russo
 
P
Nuvolone
 
M
et al. 
Treatment with oral melphalan plus dexamethasone produces long-term remissions in AL amyloidosis.
Blood
2007
, vol. 
110
 
2
(pg. 
787
-
788
)
57
Wechalekar
 
AD
Goodman
 
HJ
Lachmann
 
HJ
Offer
 
M
Hawkins
 
PN
Gillmore
 
JD
Safety and efficacy of risk-adapted cyclophosphamide, thalidomide, and dexamethasone in systemic AL amyloidosis.
Blood
2007
, vol. 
109
 
2
(pg. 
457
-
464
)
58
Girnius
 
S
Seldin
 
DC
Skinner
 
M
et al. 
Short and long-term outcome of treatment with high-dose melphalan and stem cell transplantation for multiple myeloma-associated AL amyloidosis.
Ann Hematol
2010
, vol. 
89
 
6
(pg. 
579
-
584
)
59
Kastritis
 
E
Wechalekar
 
AD
Dimopoulos
 
MA
et al. 
Bortezomib with or without dexamethasone in primary systemic (light chain) amyloidosis.
J Clin Oncol
2010
, vol. 
28
 
6
(pg. 
1031
-
1037
)
60
Reece
 
DE
Hegenbart
 
U
Sanchorawala
 
V
et al. 
Efficacy and safety of once-weekly and twice-weekly bortezomib in patients with relapsed systemic AL amyloidosis: results of a phase 1/2 study.
Blood
2011
, vol. 
118
 
4
(pg. 
865
-
873
)
61
Dispenzieri
 
A
Lacy
 
MQ
Zeldenrust
 
SR
et al. 
The activity of lenalidomide with or without dexamethasone in patients with primary systemic amyloidosis.
Blood
2007
, vol. 
109
 
2
(pg. 
465
-
470
)
62
Palladini
 
G
Russo
 
P
Foli
 
A
et al. 
Salvage therapy with lenalidomide and dexamethasone in patients with advanced AL amyloidosis refractory to melphalan, bortezomib, and thalidomide.
Ann Hematol
2012
, vol. 
91
 
1
(pg. 
89
-
92
)
63
Dispenzieri
 
A
Buadi
 
F
Laumann
 
K
et al. 
Activity of pomalidomide in patients with immunoglobulin light-chain amyloidosis.
Blood
2012
, vol. 
119
 
23
(pg. 
5397
-
5404
)
64
Sanchorawala
 
V
Wright
 
DG
Rosenzweig
 
M
et al. 
Lenalidomide and dexamethasone in the treatment of AL amyloidosis: results of a phase 2 trial.
Blood
2007
, vol. 
109
 
2
(pg. 
492
-
496
)
65
Specter
 
R
Sanchorawala
 
V
Seldin
 
DC
et al. 
Kidney dysfunction during lenalidomide treatment for AL amyloidosis.
Nephrol Dial Transplant
2011
, vol. 
26
 
3
(pg. 
881
-
886
)
66
Kumar
 
SK
Hayman
 
SR
Buadi
 
FK
et al. 
Lenalidomide, cyclophosphamide, and dexamethasone (CRd) for light-chain amyloidosis: long-term results from a phase 2 trial.
Blood
2012
, vol. 
119
 
21
(pg. 
4860
-
4867
)
67
Kastritis
 
E
Terpos
 
E
Roussou
 
M
et al. 
A phase 1/2 study of lenalidomide with low-dose oral cyclophosphamide and low-dose dexamethasone (RdC) in AL amyloidosis.
Blood
2012
, vol. 
119
 
23
(pg. 
5384
-
5390
)
68
Palladini
 
G
Russo
 
P
Milani
 
P
et al. 
A phase II trial of cyclophosphamide, lenalidomide and dexamethasone in previously treated patients with AL amyloidosis.
Haematologica
2013
 
98(3):433-436
69
Moreau
 
P
Jaccard
 
A
Benboubker
 
L
et al. 
Lenalidomide in combination with melphalan and dexamethasone in patients with newly diagnosed AL amyloidosis: a multicenter phase 1/2 dose-escalation study.
Blood
2010
, vol. 
116
 
23
(pg. 
4777
-
4782
)
70
Sanchorawala
 
V
Patel
 
JM
Sloan
 
JM
Shelton
 
AC
Zeldis
 
JB
Seldin
 
DC
Melphalan, lenalidomide and dexamethasone for the treatment of immunoglobulin light chain amyloidosis: results of a phase II trial.
Haematologica
2013
 
98(5):789-792
71
Merlini
 
G
Sanchorawala
 
V
Zonder
 
JA
et al. 
MLN9708, a novel, investigational oral proteasome inhibitor, in patients with relapsed or refractory light-chain amyloidosis (AL): results of a phase 1 study [abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 731
72
Palladini
 
G
Schonland
 
SO
Milani
 
P
et al. 
Treatment of AL amyloidosis with bendamustine[abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 4057
73
Palladini
 
G
Merlini
 
G
Systemic amyloidoses: what an internist should know.
Eur J Intern Med
 
In press
74
Gillmore
 
JD
Goodman
 
HJ
Lachmann
 
HJ
et al. 
Sequential heart and autologous stem cell transplantation for systemic AL amyloidosis.
Blood
2006
, vol. 
107
 
3
(pg. 
1227
-
1229
)
75
Sack
 
FU
Kristen
 
A
Goldschmidt
 
H
et al. 
Treatment options for severe cardiac amyloidosis: heart transplantation combined with chemotherapy and stem cell transplantation for patients with AL-amyloidosis and heart and liver transplantation for patients with ATTR-amyloidosis.
Eur J Cardiothorac Surg
2008
, vol. 
33
 
2
(pg. 
257
-
262
)
76
Lacy
 
MQ
Dispenzieri
 
A
Hayman
 
SR
et al. 
Autologous stem cell transplant after heart transplant for light chain (Al) amyloid cardiomyopathy.
J Heart Lung Transplant
2008
, vol. 
27
 
8
(pg. 
823
-
829
)
77
Dey
 
BR
Chung
 
SS
Spitzer
 
TR
et al. 
Cardiac transplantation followed by dose-intensive melphalan and autologous stem-cell transplantation for light chain amyloidosis and heart failure.
Transplantation
2010
, vol. 
90
 
8
(pg. 
905
-
911
)
78
Swiecicki
 
PL
Edwards
 
BS
Kushwaha
 
SS
Dispenzieri
 
A
Park
 
SJ
Gertz
 
MA
Left ventricular device implantation for advanced cardiac amyloidosis.
J Heart Lung Transplant
2013
, vol. 
32
 
5
(pg. 
563
-
568
)
79
Comenzo
 
RL
Reece
 
D
Palladini
 
G
et al. 
Consensus guidelines for the conduct and reporting of clinical trials in systemic light-chain amyloidosis.
Leukemia
2012
, vol. 
26
 
11
(pg. 
2317
-
2325
)
80
Bodin
 
K
Ellmerich
 
S
Kahan
 
MC
et al. 
Antibodies to human serum amyloid P component eliminate visceral amyloid deposits.
Nature
2010
, vol. 
468
 
7320
(pg. 
93
-
97
)
Sign in via your Institution