• Early cardiac response is possible in patients with advanced cardiac AL amyloidosis.

  • Graded cardiac response at 90 days predicts survival in stage IIIb AL amyloidosis.

Patients with immunoglobulin light chain (AL) amyloidosis and stage IIIb cardiac involvement have a dismal outcome despite the introduction of novel treatments. However, a rapid hematologic response translates in better survival. We evaluated the impact of early cardiac response and its depth on outcome in 249 patients with newly diagnosed stage IIIb cardiac AL amyloidosis. Hematologic and cardiac responses were evaluated by intent to treat. After a median follow-up of 52 months, 219 (84%) patients died, and median survival was 4.2 months. The 30- and 90-day hematologic response rates were 22% (at least very good partial response [VGPR] in 9%) and 24% (at least VGPR in 15%), respectively. Early hematologic response resulted in better survival. At 90 days, 21 (8%) patients achieved a cardiac response (cardiac very good partial response [cardiac VGPR] in 12 cases and cardiac partial response [cardiac PR] in 9). At the 90-day landmark analysis, cardiac response resulted in longer survival (median, 54 months), also in those patients who have achieved at least VGPR (median, 62 vs 26 months, P = .011). Patients with cardiac VGPR had a longer survival than those with cardiac PR (median, 92 vs 24 months; P = .027), whereas patients without cardiac response had a poor survival (median, 6 months). A baseline difference of involved/uninvolved free light chains > 50 mg/L (odds ratio [OR], 0.21, P = .024) and a bone marrow plasma cell infiltrate > 10% (OR, 0.23, P = .040) were negative predictors of 90-day cardiac response. Early cardiac responses are rare but possible in stage IIIb AL amyloidosis and translate to longer survival.

Immunoglobulin light chain (AL) amyloidosis is caused by a usually small B-cell clone that synthesizes misfolded light chains that deposit in tissues and organs as insoluble amyloid fibrils. Cardiac involvement is present in almost 80% of cases, and it is the most prominent driver of prognosis.1 Severity of heart involvement is accurately evaluated by staging systems based on N-terminal natriuretic propeptide type-B (NT-proBNP) and cardiac troponins.2-4 The European staging system identifies stage IIIb patients as those with the most severe cardiac amyloidosis and poor survival.4,5 Recent data indicates that the outcome of these patients did not improve in recent years, despite the introduction of novel and effective treatments.6 There is the hope that daratumumab combinations, recently approved as standard of care in stage I-IIIa patients, may favorably impact the outcome of subjects with very advanced cardiac involvement, and a phase II study of daratumumab single agent (#NCT04131309; clinicaltrials.gov) is underway. Preliminary results were encouraging.7 

The International Society of Amyloidosis defines hematologic and cardiac response based on changes in circulating free light chains, on immunofixation of serum and urine,8,9 and on changes in NT-proBNP, respectively.8 Hematologic and cardiac response can be combined to assess whether the treatment induced a satisfactory response.10 Although hematologic response is graded, the possibility of grading cardiac response has been proposed11 and is being validated in a large international study.12 Achieving an early hematologic response (within 30 days from treatment initiation) is important in AL amyloidosis, and it is associated with longer survival and higher rates of organ responses.13 This is even more crucial in stage IIIb patients. The National Amyloid Center group of London, United Kingdom, suggested to evaluate hematologic response 30 days after starting chemotherapy and base the decision on the subsequent therapeutic strategy on this assessment.14 However, the possibility to achieve an early cardiac response in stage IIIb patients and its impact on outcome has not been evaluated yet.

In this study, we present a large series of 249 patients with newly diagnosed stage IIIb AL amyloidosis from a prospectively maintained database, we explore factors able to further stratify outcomes in this subgroup, and we evaluate the impact of early cardiac response and progression on overall survival (OS). We also assessed the applicability of the recently proposed graded cardiac response criteria and combined hematologic and organ response criteria in this setting.10-12 

The prospectively maintained database of 1378 patients with AL amyloidosis newly diagnosed at the Amyloid Research and Treatment Center in Pavia between 2004 and 2018 was searched for stage IIIb cases according to the European staging system. Two hundred forty-nine consecutive patients were included in the study. The study was approved by the ethics committee, and all subjects gave written informed consent for their clinical data to be used for research purposes, in accordance with the Declaration of Helsinki.

Cardiac stage IIIb was defined as NT-proBNP > 8500 ng/L and troponin I > 0.100 ng/mL at diagnosis according to the European staging system.4 The method for measurement of troponin I was changed in November 2018 to an ultrasensitive assay. Troponin I was then remeasured with the new assay on the frozen serum samples stored in our biobank in all patients who were evaluated at our center before November 2018 for consistency.

Diagnosis of AL amyloidosis was established after histological identification and accurate typing of amyloid deposits on tissue biopsies with immunoelectron microscopy or mass spectrometry.15-17 Organ involvement was defined as per consensus criteria.18 The severity of renal involvement was assessed by the validated renal staging system with 24-hour proteinuria and estimated glomerular filtration rate (eGFR).19 

All the patients were evaluated by an expert cardiologist at the first evaluation at our center. During the cardiologic evaluation, heart failure treatment with diuretics was reviewed in all cases and optimized when needed before starting chemotherapy for AL amyloidosis. The New York Heart Association (NYHA) class was assessed both by the amyloid center cardiologist and the physician coordinating the patient’s evaluation. Assessment of NYHA class was concordant in all patients.

The treatment strategy was protocolized. Briefly, upfront bortezomib was not available before 2010, and patients were treated with oral melphalan dexamethasone (MDex) or immune modulatory drugs according to age (above or below 65 years) and eGFR (above or below 30 mL/min) to avoid oral melphalan in younger subjects and in those with renal failure. After 2010, bortezomib combinations (with melphalan or cyclophosphamide according to age and eGFR)5,20 were preferred, but MDex was still offered to subjects with contraindications to bortezomib (peripheral neuropathy or lung disease) and to those who preferred an oral treatment. Patients with immunoglobulin M expressing clones received rituximab-based therapy. All regimens were attenuated. Daily dexamethasone dose did not exceed 20 mg, and bortezomib was started at 0.7 or 1.0 mg/m2 and escalated to 1.3 according to tolerability.21-23 

Hematologic response was evaluated by intent to treat at 30 and 90 days from the day of starting chemotherapy and was assessed according to current validated criteria.8,9 Patients with a baseline difference between involved and uninvolved free light chains (dFLC) between 50 mg/L and 20 mg/L were evaluable with the low-dFLC criteria.24-28 Cardiac response, defined as a decrease of NT-proBNP > 300 ng/L and >30% from baseline,8 was evaluated by intent to treat after 90 days from starting treatment. Cardiac response was graded according to the recently proposed criteria for graded organ response: complete cardiac response (cardiac CR) if NT-proBNP was < 400 ng/L; very good partial cardiac response (cardiac VGPR) in the presence of a reduction of NT-proBNP > 60% and not meeting the cardiac CR definition; and partial cardiac response (cardiac PR) if NT-proBNP decrease was between 60% and 30%.11 A combined hematologic and cardiac and hematologic response (CHCR) model was tested for the identification of subjects with better outcome after treatment. CHCR was built referring to the proposed and validated combined hematologic and organ response model.10 More precisely, it was designed using combining scores of 0 to 3 for hematologic response (0, complete response [CR]; 1, very good partial response [VGPR]; 2, partial response [PR]; 3, no response [NR]) and 0 to 1 for cardiac response (0, cardiac response; 1, no cardiac response).

Fisher’s exact test and Mann-Whitney U test were used to assess differences in nominal and continuous variables between groups as appropriate. Median and interquartile range were reported for continuous variables. Survival was calculated from diagnosis to death or last contact. Survival curves were plotted according to Kaplan-Meier, and differences in OS were tested for significance with the log-rank test. A 30- and 90-day landmark analysis was performed to evaluate the impact of hematologic and cardiac response at specific time points. Cox hazard regression models and logistic regression analysis was performed to identify factors predicting survival and cardiac response, respectively. Receiver-operating characteristic analyses based on death at 3 months were used to identify dFLC and troponin I cutoffs best discriminating survival. MedCalc Statistical Software version 14.12.0 (MedCalc Software bvba, Ostend, Belgium; http://www.medcalc.org; 2014) was used for computation.

Patient characteristics are reported in Table 1. Of the 249 patients with stage IIIb AL amyloidosis, 74 (30%) had isolated heart involvement. Clone-related prognostic factors, namely dFLC > 180 mg/L and bone marrow plasma cellular infiltrate > 20%, were present in 67% and 16% of patients, respectively. Remarkably, a small but not negligible number were in NYHA class I (13% of cases). Patients received a bortezomib-based regimen in 118 (47%) cases. Of the 165 patients diagnosed between 2010 and 2018, 111 (67%) received bortezomib. Oral MDex was the most frequent treatment in patients who were not exposed to bortezomib.

Table 1.

Characteristics of 249 patients with stage IIIb AL amyloidosis

VariablesOverall population
249 patients N (%), mean (IQR)
Age, years 68 (60-74) 
Sex, male 145 (58) 
Organ involvement (other than heart)  
Kidney 129 (52) 
Liver 43 (17) 
ST 51 (20) 
GI 6 (2) 
ANS 21 (8) 
PNS 23 (9) 
Number of involved organs 2 (1-3) 
Isolated heart involvement 74 (30) 
NYHA class  
33 (13) 
II 64 (26) 
III 139 (56) 
IV 13 (5) 
NT-proBNP, ng/L 17089 (12179-25014) 
Troponin I, ng/mL 0.265 (0.169-0.500) 
eGFR, mL/min × 1.73 m2 43 (27->60) 
eGFR < 30 mL/min × 1.73 m2 76 (31) 
Renal stage  
108 (43) 
II 111 (45) 
III 26 (10) 
Patients in dialysis at diagnosis 2 (1) 
dFLC, mg/L 259 (142-543) 
dFLC < 50 mg/L 13 (5) 
dFLC > 180 mg/L 167 (67) 
Involved/uninvolved FLC rate > 100 9 (4) 
BMPC, % 12 (8-20) 
BMPC > 20% 41 (16) 
Treatment  
MDex 98 (39) 
CyBorD 75 (30) 
BMDex 43 (18) 
IMiDs 28 (11) 
rituximab 5 (1) 
VariablesOverall population
249 patients N (%), mean (IQR)
Age, years 68 (60-74) 
Sex, male 145 (58) 
Organ involvement (other than heart)  
Kidney 129 (52) 
Liver 43 (17) 
ST 51 (20) 
GI 6 (2) 
ANS 21 (8) 
PNS 23 (9) 
Number of involved organs 2 (1-3) 
Isolated heart involvement 74 (30) 
NYHA class  
33 (13) 
II 64 (26) 
III 139 (56) 
IV 13 (5) 
NT-proBNP, ng/L 17089 (12179-25014) 
Troponin I, ng/mL 0.265 (0.169-0.500) 
eGFR, mL/min × 1.73 m2 43 (27->60) 
eGFR < 30 mL/min × 1.73 m2 76 (31) 
Renal stage  
108 (43) 
II 111 (45) 
III 26 (10) 
Patients in dialysis at diagnosis 2 (1) 
dFLC, mg/L 259 (142-543) 
dFLC < 50 mg/L 13 (5) 
dFLC > 180 mg/L 167 (67) 
Involved/uninvolved FLC rate > 100 9 (4) 
BMPC, % 12 (8-20) 
BMPC > 20% 41 (16) 
Treatment  
MDex 98 (39) 
CyBorD 75 (30) 
BMDex 43 (18) 
IMiDs 28 (11) 
rituximab 5 (1) 

ANS, autonomic nervous system involvement; BMDex, bortezomib, melphalan, and dexamethasone; BMPC, bone marrow plasma cells; CyBorD, cyclophosphamide, bortezomib, and dexamethasone; FLC, free light chain; GI, gastrointestinal; IMiDs, immunomodulatory drugs; IQR, interquartile range; PNS, peripheral nervous system; ST, soft tissues.

We compared baseline variables in NYHA class I patients and in the other stage IIIb patients (supplemental Table 1, available on the Blood Web site). The only significant difference was the concentration of troponin I (median, 0.196 vs 0.289 ng/mL, P = .017).

After a median follow-up of living patients of 52 months, 219 (84%) patients died, and median OS was 4.2 months. The cumulative proportion surviving was 91% at 1 month, 61% at 3 months, 41% at 6 months, and 26% at 12 months (Figure 1).

Figure 1.

OS in 249 patients with stage IIIb AL amyloidosis.

Figure 1.

OS in 249 patients with stage IIIb AL amyloidosis.

Close modal

Results of the univariable analysis for factors predicting OS are reported in Table 2. This analysis revealed that dFLC > 500 mg/L (hazard ratio [HR], 1.94; 95% CI, 1.43-2.63; P < .001) and troponin > 0.500 ng/mL (HR, 1.50; 95% CI, 1.10-2.50; P = .010) were associated with worse survival, whereas having a NYHA class I at diagnosis resulted in better outcome (HR, 0.57; 95% CI, 0.37-0.89; P < .014). Treatment regimens (considered as single regimens and as bortezomib-based vs non–bortezomib-based) were not associated with survival. We observed that reduced eGFR (<30 mL/min × 1.73 m2) was a protective factor (HR, 0.71; 95% CI, 0.52-0.96; P = .028). Due to the possible interference of reduced glomerular filtration on NT-proBNP concentration and staging, we elected to exclude from the multivariable analysis patients whose eGFR was <30 mL/min × 1.73 m2 (Table 2). On multivariable analysis, dFLC > 500 mg/L emerged as the only independent prognostic factor for survival at diagnosis (HR, 1.83; 95% CI, 1.28-2.62; P = .001). Overall, 69 (28%) patients had a dFLC level > 500 mg/L, and their median OS was 2.7 months. In 7 cases (10%), the involved/uninvolved free light chain ratio was >100.

Table 2.

Univariable and multivariable analysis for survival in patients with stage IIIb amyloidosis

Univariable analysis (249 patients)
VariablesHazard ratio95% CIP
Sex, male 1.22 0.96-1.67 0.158 
NYHA class I 0.57 0.37-0.89 0.014 
Troponin I > 0.500 ng/mL 1.50 1.10-2.50 0.010 
NT-proBNP(ln), ng/L 1.53 0.81-2.87 0.186 
eGFR < 30 mL/min × 1.73 m2 0.71 0.52-0.96 0.028 
LC isotype κ vs λ 1.18 0.84-1.66 0.326 
dFLC > 50 mg/L 0.53 0.26-1.09 0.083 
dFLC > 180 mg/L 1.17 0.87-1.56 0.296 
dFLC > 500 mg/L 1.94 1.43-2.63 <0.001 
BMPC > 20% 1.03 0.65-1.63 0.888 
Bortezomib-based treatment, yes 0.85 0.65-1.12 0.254 
Treatment with CyBorD, yes 0.81 0.59-1.11 0.192 
Treatment with BMDex, yes 1.22 0.85-1.74 0.274 
Treatment with MDex, yes 1.02 0.77-1.35 0.906 
Univariable analysis (249 patients)
VariablesHazard ratio95% CIP
Sex, male 1.22 0.96-1.67 0.158 
NYHA class I 0.57 0.37-0.89 0.014 
Troponin I > 0.500 ng/mL 1.50 1.10-2.50 0.010 
NT-proBNP(ln), ng/L 1.53 0.81-2.87 0.186 
eGFR < 30 mL/min × 1.73 m2 0.71 0.52-0.96 0.028 
LC isotype κ vs λ 1.18 0.84-1.66 0.326 
dFLC > 50 mg/L 0.53 0.26-1.09 0.083 
dFLC > 180 mg/L 1.17 0.87-1.56 0.296 
dFLC > 500 mg/L 1.94 1.43-2.63 <0.001 
BMPC > 20% 1.03 0.65-1.63 0.888 
Bortezomib-based treatment, yes 0.85 0.65-1.12 0.254 
Treatment with CyBorD, yes 0.81 0.59-1.11 0.192 
Treatment with BMDex, yes 1.22 0.85-1.74 0.274 
Treatment with MDex, yes 1.02 0.77-1.35 0.906 
Multivariable analysis (173 patients with eGFR > 30 mL/min × 1.73 m2)
NYHA class I 0.80 0.48-1.33 0.392 
Troponin I > 0.500 ng/mL 1.22 0.81-1.83 0.340 
dFLC > 500 mg/L 1.83 1.28-2.62 0.001 
Multivariable analysis (173 patients with eGFR > 30 mL/min × 1.73 m2)
NYHA class I 0.80 0.48-1.33 0.392 
Troponin I > 0.500 ng/mL 1.22 0.81-1.83 0.340 
dFLC > 500 mg/L 1.83 1.28-2.62 0.001 

BMDex, bortezomib, melphalan, and dexamethasone; CI, confidence interval; CyBorD, cyclophosphamide, bortezomib, and dexamethasone; LC, light chains.

A hematologic response was observed in 55 (22%) patients (CR, 3%; VGPR, 6%; PR, 13%) at 30 days from treatment initiation and in 59 (24%; CR, 5%; VGPR, 10%; PR, 9%) at 90 days. The presence and depth of hematologic response were significantly associated with OS at both day 30 and day 90 (Figure 2A-B). The benefit of a hematologic response on survival was observed both in patients treated with bortezomib and in those exposed to MDex (supplemental Figure 1).

Figure 2.

Hematologic response at 30 and 90 days in stage IIIb AL amyloidosis. Survival according to the depth and quality of hematologic response at 30 days from treatment initiation: CR/VGPR median survival, 52 months; PR median survival, 16 months; and NR median survival, 3 months. (A) Survival according to the depth and quality of hematologic response at 90 days from treatment initiation: CR/VGPR median survival, 52 months; PR median survival, 16 months; and NR median survival, 6 months. (B) A 30- and 90-day landmark analysis was performed.

Figure 2.

Hematologic response at 30 and 90 days in stage IIIb AL amyloidosis. Survival according to the depth and quality of hematologic response at 30 days from treatment initiation: CR/VGPR median survival, 52 months; PR median survival, 16 months; and NR median survival, 3 months. (A) Survival according to the depth and quality of hematologic response at 90 days from treatment initiation: CR/VGPR median survival, 52 months; PR median survival, 16 months; and NR median survival, 6 months. (B) A 30- and 90-day landmark analysis was performed.

Close modal

At 90 days, cardiac response was achieved in 21 (8%) cases, of which 12 obtained a cardiac VGPR and 9 a cardiac PR. None achieved a cardiac CR. Increase of diuretic usage within 90 days from diagnosis was observed in none of the patients who achieved a cardiac response. On the contrary, 20% of cardiac responders and 5% of those with no cardiac response had a reduction in diuretic usage, although this difference was not statistically significant (P = .097). In 204 (82%) patients, a cardiac progression was observed, whereas 24 (10%) were classified as stable. The 90-day landmark analysis showed that patients with a cardiac response had a longer OS (median 54 months) than those with stable cardiac disease (median 20 months) or cardiac progression (median 6 months; Figure 3A). Moreover, the deeper the cardiac response, the better was the benefit in survival (cardiac VGPR median survival, 92 months; cardiac PR median survival, 24 months; cardiac NR median survival, 6 months; Figure 3B). The presence of cardiac response resulted in better survival also in the subgroup of patients with dFLC > 500 mg/L at diagnosis (median survival, 41 vs 4 months, P = .015; supplemental Figure 2) and in those who achieved a CR/VGPR at 90 days (median survival, 62 vs 26 months, P = .011; Figure 3C).

Figure 3.

Early cardiac response (90 days) in stage IIIb AL amyloidosis. Survival in patients with a cardiac response (median survival, 54 months), cardiac stable disease (median survival, 20 months), or cardiac progression (median survival, 6 months). (A) Survival according to depth of cardiac response: cardiac VGPR (median survival, 92 months), cardiac PR (median survival, 24 months), and cardiac NR (median survival, 6 months). (B) Survival according cardiac response in patients who attained a profound hematologic response (ie, at least VGPR). (C) Ninety-day landmark analysis was performed.

Figure 3.

Early cardiac response (90 days) in stage IIIb AL amyloidosis. Survival in patients with a cardiac response (median survival, 54 months), cardiac stable disease (median survival, 20 months), or cardiac progression (median survival, 6 months). (A) Survival according to depth of cardiac response: cardiac VGPR (median survival, 92 months), cardiac PR (median survival, 24 months), and cardiac NR (median survival, 6 months). (B) Survival according cardiac response in patients who attained a profound hematologic response (ie, at least VGPR). (C) Ninety-day landmark analysis was performed.

Close modal

A CHCR model was built to evaluate the impact of the combination of hematologic response and cardiac response on survival after 90 days from treatment initiation. At this timepoint, CHCR score was 0 to 2 in 39 (16%) and 3 to 4 in 210 (84%) cases. At the 90-day landmark analysis, patients with a CHCR score of 0 to 2 had a significantly longer OS (median, 51 vs 6 months; P < .001; Figure 4). None of the patients who achieved a cardiac response had less than a hematologic PR, and only 3 of them achieved less than a VGPR.

Figure 4.

A composite hematologic and cardiac response at 90 days in stage IIIb AL amyloidosis. Differences in survival among patients with a CHCR score of 0 to 2 and 3 to 4 after 90 days from treatment initiation. A 90-day landmark analysis was performed.

Figure 4.

A composite hematologic and cardiac response at 90 days in stage IIIb AL amyloidosis. Differences in survival among patients with a CHCR score of 0 to 2 and 3 to 4 after 90 days from treatment initiation. A 90-day landmark analysis was performed.

Close modal

Finally, we searched for predictors of cardiac response at 90 days (Table 3). At univariable logistic regression analysis, having a dFLC > 50 mg/L (OR, 0.21; 95% CI, 0.05-0.82; P = .024) and BMPC > 10% (OR, 0.23; 95% CI, 0.06-0.94; P = .040) at diagnosis was associated with a lower probability of reaching cardiac response. Type of treatment did not emerge as a predictor of cardiac response.

Table 3.

Univariable logistic regression for predictors of cardiac response at 90 days

VariablesOdds ratio95% CIP
Sex, male 0.64 0.25-1.62 0.349 
NYHA class I 1.13 0.35-3.70 0.835 
Troponin I > 0.500 ng/mL 1.02 0.31-3.30 0.974 
eGFR < 30 mL/min × 1.73 m2 0.90 0.32-2.54 0.845 
LC isotype κ vs λ 0.53 0.20-1.46 0.221 
dFLC > 50 mg/L 0.21 0.05-0.82 0.024 
dFLC > 180 mg/L 0.56 0.22-1.41 0.217 
dFLC > 500 mg/L 0.97 0.30-3.13 0.959 
BMPC > 10% 0.23 0.06-0.94 0.040 
Bortezomib-based treatment, yes 1.13 0.45-2.86 0.788 
Treatment with CyBorD, yes 1.18 0.44-3.16 0.736 
Treatment with BMDex, yes 0.90 0.24-3.34 0.875 
Treatment with MDex, yes 0.96 0.37-2.48 0.933 
VariablesOdds ratio95% CIP
Sex, male 0.64 0.25-1.62 0.349 
NYHA class I 1.13 0.35-3.70 0.835 
Troponin I > 0.500 ng/mL 1.02 0.31-3.30 0.974 
eGFR < 30 mL/min × 1.73 m2 0.90 0.32-2.54 0.845 
LC isotype κ vs λ 0.53 0.20-1.46 0.221 
dFLC > 50 mg/L 0.21 0.05-0.82 0.024 
dFLC > 180 mg/L 0.56 0.22-1.41 0.217 
dFLC > 500 mg/L 0.97 0.30-3.13 0.959 
BMPC > 10% 0.23 0.06-0.94 0.040 
Bortezomib-based treatment, yes 1.13 0.45-2.86 0.788 
Treatment with CyBorD, yes 1.18 0.44-3.16 0.736 
Treatment with BMDex, yes 0.90 0.24-3.34 0.875 
Treatment with MDex, yes 0.96 0.37-2.48 0.933 

BMDex, bortezomib, melphalan, and dexamethasone; CyBorD, cyclophosphamide, bortezomib, and dexamethasone; LC, light chains.

This is the largest study on stage IIIb AL amyloidosis published so far and the first specifically addressing cardiac response in stage IIIb AL amyloidosis and showing that early NT-proBNP improvements are associated with improved outcomes in patients with very advanced AL amyloidosis, including those who attain deep hematologic response.

As previously reported, the outcome of stage IIIb patients was extremely poor also in our series, with a median survival of only 4.2 months and 74% of patients dying within 1 year from diagnosis.14,29 A nonnegligible proportion (13%) of patients in this series collected with a protocolized assessment of signs and symptoms at baseline were classified as NYHA class I. However, this did not have an independent impact on survival, and the only variable independently predicting the outcome of stage IIIb patients was a high (>500 mg/L) dFLC level.

Somewhat surprisingly, a severe reduction of eGFR (<30 mL/min) was associated with better survival. We interpreted this observation as an effect of severely reduced glomerular filtration on the clearance of NT-proBNP, which might have led to classifying as stage IIIb some patients with relatively less advanced heart involvement. Dittrich et al explored the impact of low eGFR (<50 mL/min) on the performance of the staging systems in AL amyloidosis and found that reduced glomerular filtration was an independent predictor of poorer outcome.30 We had previously shown that in patients with end-stage renal failure (eGFR < 15 mL/min), BNP was a better predictor of survival than NT-proBNP.31 Based on the results of Dittrich et al, we advocated further studies on the impact of severely reduced eGFR on staging.32 The present observation further emphasizes the need for larger studies addressing the impact of reduced glomerular filtration as a possible independent prognostic factor, as well as a variable interfering with cardiac staging in AL amyloidosis. This could allow refinements in staging and in eligibility criteria for clinical trials.

In the present study, early (90 days after treatment initiation) NT-proBNP response was unfortunately rare (8%). Nevertheless, cardiac response was able to significantly improve OS even in stage IIIb subjects. Not only was cardiac response possible at this early assessment after treatment initiation, but it could also be graded according to the recent proposal by the Mayo Clinic group,11,12 and deeper cardiac response was associated with a more pronounced improvement of survival.

As previously reported by Manwani et al,14 hematologic response at 30 and 90 days also predicted survival in this group. In the present series, the 30-day rate of CR/VGPR was lower than that reported by the London group (9% vs 21%), but OS seemed to be better (52 vs 26 months). The proportion of patients treated with a bortzomib-based regimen was similar (48% vs 46%), and it is possible that other factors explain the lower CR/VGPR rate in our study. For instance, a higher proportion of patients in our series were NYHA class III/IV (61% vs 38%).

Remarkably, cardiac response further prolonged OS also in subjects who attained a deep hematologic response (at least VGPR), indicating that both hematologic and cardiac response can be used in early assessment of treatment efficacy also in this population of patients with very advanced heart involvement. Indeed, the recently proposed criteria for combined hematologic and organ response assessment provided a sharp discrimination of outcome in our series. Interestingly, when we analyzed possible predictors of irreversible cardiac dysfunction, we found that a relatively low dFLC value (>50 mg/L) made cardiac response less likely. However, cardiac response improved survival also in patients with high dFLC. Overall, these observations corroborate the link between cardiac dysfunction and circulating free light chains in AL amyloidosis that was supported by previous in vitro and in vivo studies.33-35 

As previously reported,6 the type of treatment did not have an impact on OS. However, our study was not designed to assess a treatment effect on survival, because treatment allocation was influenced by variables with a prognostic impact (age and eGFR). Nevertheless, we confirmed the data reported by Vaxman et al29 that bortezomib-based treatment does not seem to have an impact on the proportion of patients dying within the first 12 months from diagnosis. Yet, it is clear that improvement of treatment of stage IIIb AL amyloidosis is urgently needed. Hopefully, the introduction of novel and rapidly acting drugs such as daratumumab may change the clinical history also of these patients.36 

We recognize that, despite the large number of patients included in the study, a possible limitation is the low number of hematologic and cardiac responses. Given the rarity of the disease and the peculiar frailty of patients with stage IIIb AL amyloidosis, only large international cooperative studies can further address the impact of cardiac response in these subjects with advanced cardiac disease.

In conclusion, our study shows that cardiac dysfunction can be rapidly reversible in patients with stage IIIb AL amyloidosis. These patients should be treated with delicate and rapidly acting regimens to induce profound reductions of the amyloid light chain. Hopefully, the proportion of patients in whom this result is obtained will improve with the recent introduction of more effective anti–plasma cell agents7 and by immunotherapy targeting the amyloid deposits.37,38 Early assessment and grading of cardiac response is a useful complement to assessment of hematologic response in stage IIIb patients and may be used to guide therapeutic decisions.

This study was supported by a grant from CARIPLO “Harnessing the plasma cell secretory capacity against systemic light chain amyloidosis” (n. 2018-0257), by a grant from the Italian Ministry of Health “Towards effective, patient-tailored anti-plasma cell therapies in AL amyloidosis: Predicting drug response and overcoming drug resistance” (GR-2018-12368387), and by a grant from Cancer Research UK, FCAECC, and AIRC under the Accelerator Award 2017 Program “Early detection and intervention: Understanding the mechanisms of transformation and hidden resistance of incurable haematological malignancies.”

Contribution: M.B. designed the study, wrote the manuscript, collected and analyzed data, and evaluated patients; P.M. designed the study, collected data, and evaluated patients; G.P. designed the study, wrote the manuscript, and evaluated patients; A.F., M. Nuvolone, P.B., M. Nanci, F.F., and C.B. evaluated patients; and G.M. evaluated patients, critically reviewed the manuscript, and gave final approval.

Conflict-of-interest disclosure: M.B. reports honoraria for lectures from Jannsen. P.M. reports honoraria for lectures from Pfizer and Jannsen-Cilag and travel support from Celgene. M. Nuvolone reports honoraria for lectures from Jannsen. A.F. reports honoraria for lectures from Jannsen. G.P. reports advisory board membership and honoraria from Janssen, Protego, and Zentalis; and honoraria from Pfizer, Sebia, Siemens, and The Binding Site. The remaining authors declare no competing financial interests.

Correspondence: Giovanni Palladini, Amyloidosis Research and Treatment Center, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Viale Golgi, 19, Pavia, Italy 27100; e-mail: giovanni.palladini@unipv.it.

1.
Merlini
G
,
Dispenzieri
A
,
Sanchorawala
V
, et al
.
Systemic immunoglobulin light chain amyloidosis
.
Nat Rev Dis Primers
.
2018
;
4
(
1
):
38
.
2.
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
;
22
(
18
):
3751
-
3757
.
3.
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
;
30
(
9
):
989
-
995
.
4.
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
;
121
(
17
):
3420
-
3427
.
5.
Palladini
G
,
Sachchithanantham
S
,
Milani
P
, et al
.
A European collaborative study of cyclophosphamide, bortezomib, and dexamethasone in upfront treatment of systemic AL amyloidosis
.
Blood
.
2015
;
126
(
5
):
612
-
615
.
6.
Palladini
G
,
Schonland
S
,
Merlini
G
, et al
.
Systemic Light Chain Amyloidosis across Europe: Key Outcomes from a Retrospective Study of 4500 Patients
.
ASH Annual Meeting & Exposition
.
11-14 December 2021
.
7.
Kastritis
E
,
Minnema
MC
,
Dimopoulos
MA
, et al
.
Efficacy and Safety of Daratumumab Monotherapy in Newly Diagnosed Patients with Stage 3B Light Chain Amyloidosis: A Phase 2 Study By the European Myeloma Network ASH Annual Meeting & Exposition
.
ASH Annual Meeting & Exposition
.
11-14 December 2021
.
8.
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
;
30
(
36
):
4541
-
4549
.
9.
Palladini
G
,
Schönland
SO
,
Sanchorawala
V
, et al
.
Clarification on the definition of complete haematologic response in light-chain (AL) amyloidosis
.
Amyloid
.
2021
;
28
(
1
):
1
-
2
.
10.
Sidana
S
,
Milani
P
,
Binder
M
, et al
.
A validated composite organ and hematologic response model for early assessment of treatment outcomes in light chain amyloidosis
.
Blood Cancer J
.
2020
;
10
(
4
):
41
.
11.
Muchtar
E
,
Dispenzieri
A
,
Leung
N
, et al
.
Depth of organ response in AL amyloidosis is associated with improved survival: grading the organ response criteria
.
Leukemia
.
2018
;
32
(
10
):
2240
-
2249
.
12.
Muchtar
E
,
Dispenzieri
A
,
Wisniowski
B
, et al
.
Graded Cardiac Response Criteria for AL Amyloidosis: The Impact of Depth of Cardiac Response on Survival
.
ASH Annual Meeting & Exposition
.
December 2021
;
11-14
.
13.
Kastritis
E
,
Fotiou
D
,
Theodorakakou
F
, et al
.
Timing and impact of a deep response in the outcome of patients with systemic light chain (AL) amyloidosis
.
Amyloid
.
2021
;
28
(
1
):
3
-
11
.
14.
Manwani
R
,
Foard
D
,
Mahmood
S
, et al
.
Rapid hematologic responses improve outcomes in patients with very advanced (stage IIIb) cardiac immunoglobulin light chain amyloidosis
.
Haematologica
.
2018
;
103
(
4
):
e165
-
e168
.
15.
Fernández de Larrea
C
,
Verga
L
,
Morbini
P
, et al
.
A practical approach to the diagnosis of systemic amyloidoses
.
Blood
.
2015
;
125
(
14
):
2239
-
2244
.
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
;
114
(
24
):
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
;
119
(
8
):
1844
-
1847
.
18.
Gertz
MA
,
Merlini
G
.
Definition of organ involvement and response to treatment in AL amyloidosis: an updated consensus opinion
.
Amyloid
.
2010
;
17
:
48
-
49
.
19.
Palladini
G
,
Hegenbart
U
,
Milani
P
, et al
.
A staging system for renal outcome and early markers of renal response to chemotherapy in AL amyloidosis
.
Blood
.
2014
;
124
(
15
):
2325
-
2332
.
20.
Kastritis
E
,
Leleu
X
,
Arnulf
B
, et al
.
Bortezomib, melphalan, and dexamethasone for light-chain amyloidosis
.
J Clin Oncol
.
2020
;
38
(
28
):
3252
-
3260
.
21.
Palladini
G
,
Milani
P
,
Merlini
G
.
Management of AL amyloidosis in 2020
.
Blood
.
2020
;
136
(
23
):
2620
-
2627
.
22.
Palladini
G
,
Merlini
G
.
How I treat AL amyloidosis
.
Blood
.
2022
;
139
(
19
):
2918
-
2930
.
23.
Palladini
G
,
Celant
S
,
Milani
P
, et al
.
AIFA’s Monitoring Registries Group. A nationwide prospective registry of bortezomib-based therapy in light chain (AL) amyloidosis
.
Leuk Lymphoma
.
2022
;
63
(
1
):
205
-
211
.
24.
Milani
P
,
Basset
M
,
Russo
F
,
Foli
A
,
Merlini
G
,
Palladini
G
.
Patients with light-chain amyloidosis and low free light-chain burden have distinct clinical features and outcome
.
Blood
.
2017
;
130
(
5
):
625
-
631
.
25.
Dittrich
T
,
Bochtler
T
,
Kimmich
C
, et al
.
AL amyloidosis patients with low amyloidogenic free light chain levels at first diagnosis have an excellent prognosis
.
Blood
.
2017
;
130
(
5
):
632
-
642
.
26.
Sidana
S
,
Tandon
N
,
Dispenzieri
A
, et al
.
Clinical presentation and outcomes in light chain amyloidosis patients with non-evaluable serum free light chains
.
Leukemia
.
2018
;
32
(
3
):
729
-
735
.
27.
Nguyen
VP
,
Rosenberg
A
,
Mendelson
LM
,
Comenzo
RL
,
Varga
C
,
Sanchorawala
V
.
Outcomes of patients with AL amyloidosis and low serum free light chain levels at diagnosis
.
Amyloid
.
2018
;
25
(
3
):
156
-
159
.
28.
Qiu
Y
,
Zhang
CL
,
Shen
KN
, et al
.
Clinical presentation and prognosis of light-chain amyloidosis patients with unmeasurable free light-chain levels
.
Ann Hematol
.
2018
;
97
(
12
):
2465
-
2470
.
29.
Vaxman
I
,
Kumar
SK
,
Buadi
F
, et al
.
Outcomes among newly diagnosed AL amyloidosis patients with a very high NT-proBNP: implications for trial design
.
Leukemia
.
2021
;
35
(
12
):
3604
-
3607
.
30.
Dittrich
T
,
Benner
A
,
Kimmich
C
, et al
.
Performance analysis of AL amyloidosis cardiac biomarker staging systems with special focus on renal failure and atrial arrhythmia
.
Haematologica
.
2019
;
104
(
7
):
1451
-
1459
.
31.
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
;
87
(
5
):
465
-
471
.
32.
Palladini
G
,
Milani
P
,
Merlini
G
.
Predicting survival in light chain amyloidosis
.
Haematologica
.
2019
;
104
(
7
):
1294
-
1296
.
33.
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
;
107
(
19
):
2440
-
2445
.
34.
Palladini
G
,
Lavatelli
F
,
Russo
P
, et al
.
Circulating amyloidogenic free light chains and serum N-terminal natriuretic peptide type B decrease simultaneously in association with improvement of survival in AL
.
Blood
.
2006
;
107
(
10
):
3854
-
3858
.
35.
Diomede
L
,
Rognoni
P
,
Lavatelli
F
, et al
.
Investigating heart-specific toxicity of amyloidogenic immunoglobulin light chains: A lesson from C. elegans
.
Worm
.
2014
;
3
(
3
):
e965590
.
36.
Kastritis
E
,
Palladini
G
,
Minnema
MC
, et al
.
ANDROMEDA Trial Investigators. Daratumumab-based treatment for immunoglobulin light-chain amyloidosis
.
N Engl J Med
.
2021
;
385
(
1
):
46
-
58
.
37.
Edwards
CV
,
Rao
N
,
Bhutani
D
, et al
.
Phase 1a/b study of monoclonal antibody CAEL-101 (11-1F4) in patients with AL amyloidosis
.
Blood
.
2021
;
138
(
25
):
2632
-
2641
.
38.
Merlini
G
,
Palladini
G
.
Two-hit strategy for treating AL amyloidosis?
.
Blood
.
2021
;
138
(
25
):
2596
-
2598
.

Author notes

For original data, please contact giovanni.palladini@unipv.it.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Supplemental data

Sign in via your Institution