New treatment options are required for primary systemic amyloid light chain (AL) amyloidosis. This phase 1/2 dose-escalation study aimed to determine the maximum tolerated dose (MTD) of lenalidomide in combination with melphalan and dexamethasone (M-dex), and assess the efficacy and tolerability of this therapy for patients with de novo AL amyloidosis. Twenty-six patients were enrolled across 4 cohorts: M-dex + lenalidomide 5, 10, 15, and 20 mg once daily on days 1 to 21 in a 28-day cycle. No dose limiting toxicity (DLT) was observed in cohorts 1, 2, and 3. 4. Seven patients in cohort 4, M-dex + lenalidomide 20 mg/day, experienced DLT. MTD was defined as 15 mg of lenalidomide. A complete hematologic response was achieved in 42% at the dose of 15 mg of lenalidomide per day. After a median follow-up of 19 months, estimated 2-year overall survival (OS) and event-free survival (EFS) were 80.8% and 53.8%, respectively. Hematologic and organ responses were both associated with superior EFS rates (P = .0001). A higher EFS was also observed in patients whose free light chains decreased by more than 50% during therapy (P = .019). Lenalidomide 15 mg/d + M-dex is a new effective combination therapy in patients with newly diagnosed AL amyloidosis. This study is registered at www.clinicaltrials.gov as NCT00621400.

Systemic amyloid light chain (AL) amyloidosis is a protein conformational disease consecutive to a marrow clonal plasma cell proliferation. AL amyloidosis results from tissue deposition of amyloid protein, mainly consisting of monoclonal light chain fragments produced by the clone.1  In most patients, the amyloidogenic monoclonal immunoglobulin can be detected in serum and/or urine, either by immunofixation or by the use of free light chain assay (FLC). Multiple organ dysfunction comes from extracellular amyloid deposits and the natural history of AL amyloidosis is that of a progressive and fatal disease. AL amyloidosis therapy aims at rapidly reducing the supply of amyloid-forming monoclonal free light chains by suppression of underlying plasma cell dyscrasia. The regimens used to treat subjects with systemic AL amyloidosis are based on those used in multiple myeloma therapy. Hematologic response usually translates into clinically improved organ function and combines with a substantial survival advantage and improved quality of life. Strategies for the effective management of subjects with systemic AL amyloidosis have been reported.2,3  First-line therapy includes treatments such as high-dose melphalan followed by autologous stem cell transplantation (ASCT) in patients eligible for high-dose therapy, or oral melphalan + dexamethasone (M-dex). In a recent prospective randomized trial performed in patients with newly diagnosed AL amyloidosis comparing oral M-dex given monthly with a high-dose therapeutic approach, the median overall survival (OS) for patients receiving M-Dex was 56 months, indicating that this combination could be considered as an interesting therapeutic option in patients with newly diagnosed AL amyloidosis.4  Lenalidomide has shown significant activity in patients with multiple myeloma alone and in combination with dexamethasone, in relapse or in newly diagnosed patients.5,6  Lenalidomide has also been associated in patients with de novo multiple myeloma with melphalan and prednisone. This oral melphalan and prednisone regimen was well tolerated with a significant response rate of approximately 80%.7  The recommended dose in the melphalan and prednisone regimen was 0.18 mg/kg melphalan given for 4 consecutive days every 4 to 6 weeks, and 10 mg/d lenalidomide for 21 consecutive days every 4 weeks. Based on these encouraging results in myeloma patients, lenalidomide + dexamethasone was tested in patients with relapsing AL amyloidosis. Sanchorawala et al reported that the initial dose of lenalidomide used (25 mg/d) was poorly tolerated in such patients, but a reduced dose of 15 mg/d was well tolerated and effective, with an overall hematologic response rate of 67% in combination with dexamethasone.8  Dispenzieri et al confirmed that the combination of lenalidomide + dexamethasone achieved a 75% hematologic response rate, with a 42% organ response rate, with a median follow-up of 17 months in a series mainly including relapsed patients.9  These authors also recommended a lower dose of 15 mg/d. Both groups concluded that lenalidomide + dexamethasone was an interesting option in patients with AL amyloidosis.

To improve the hematologic and organ response rates, it might be interesting to combine M-dex, which many investigators consider as the standard of care at the time of diagnosis, and lenalidomide. Until now, no trial of this specific combination has been performed in patients with newly diagnosed AL-amyloidosis patients, and the toxicity of this regimen is unknown. We therefore have initiated a multicenter single-arm, open-label phase 1/2 dose-escalation study of lenalidomide administered in combination with M-dex.

Patients

Patients from 18 to 70 years with a diagnosis of de novo AL amyloidosis, including measurable hematologic and amyloid-related end-organ involvement, were eligible. The diagnosis of AL-amyloidosis required a histologic tissue diagnosis of amyloid deposition proven by Congo red staining of a tissue biopsy, plus proof of plasma cell dyscrasia (measurable M-protein in serum or urine by immunofixation or electrophoresis, and/or elevated free light chain [FLC] higher than 100 mg/L and an abnormal FLC ratio). Adequate organ function defined as absolute neutrophil count > 1.0 × 109/L, platelet count > 100 × 109/L, aspartate transaminase (AST) and alanine transaminase (ALT) < 2× upper normal limit, total bilirubin < 1.5 mg/dL, creatinin serum level < 150μM, and ECOG performance status of < 2 at study entry were required. For females of childbearing potential a negative serum or urine pregnancy test at screening visit, as well as within 24 hours of starting lenalidomide, was mandatory. All patients were counseled at a minimum of every 28 days about pregnancy precautions and risks of fetal exposure due to lenalidomide therapy. Patients had to be disease free from any prior malignancies for > 5 years with exception of currently treated basal cell, squamous cell carcinoma of the skin, or carcinoma in situ of the cervix or breast.

Patients were excluded if they had symptomatic multiple myeloma, if they had received prior treatment with lenalidomide or any other experimental drug within 28 days of baseline, or any prior treatment for amyloidosis, if they were known positive for HIV or infectious hepatitis, type A, B, or C. Pregnant or breast feeding females were excluded. There was no eligibility restriction for any degree of cardiac involvement.

All patients provided written, informed consent in accordance with the Declaration of Helsinki.

Study design

Patients were enrolled in this phase 1/2, multicenter, single-arm, open-label study of dose escalation of lenalidomide administered in combination with M-dex (www.clinicaltrials.gov NCT00621400, Eudract 2007-004739-43) on 11 sites in France, between March 2008 and February 2009. The primary end point of this study was to determine the incidence of dose-limiting toxicities (DLT) during the first cycle of lenalidomide at a given dose level in combination with M-dex to define the maximal tolerated dose (MTD) in a dose-escalation study design. Secondary objectives were to determine hematologic response rate of the combination, organ response rate, and safety (type, frequency, severity, and relationship of adverse events [AEs] to study treatment), and to determine the prognostic value of FLC reduction during therapy. The study was carried out in accordance with Good Clinical Practice guidelines and the Declaration of Helsinki, and was approved by a single Institutional Review Board/Independent Ethics Committee (Tours, France) for all the centers involved according to French law.

A standard 3 + 3 dose-escalation design was used to determine the MTD. For each dose level, 3 subjects were exposed with up to 6 subjects depending on DLT. If no DLT was observed among the first cohort of 3 subjects at a given dose level, the next set of 3 subjects was entered on the following higher dose. If 2 or more DLTs were observed, the previous dose level was identified as MTD. If 1 DLT was observed on the initial 3 subjects, an additional group of 3 subjects was entered. If none of the additional 3 subjects experienced any DLT, the dose was escalated; otherwise the lower dose was identified as MTD. After the MTD had been defined, 9 additional subjects were enrolled at MTD to define the safety profile further and to estimate the hematologic and organ response rate.

Using National Cancer Institute common toxicity criteria during the first 4 weeks of treatment (1 cycle), DLT was defined as at least grade 2 cardiac arrhythmia, at least grade 3 nonhematologic toxicity, grade 4 neutropenia lasting > 7 days, or any grade 4 other hematologic toxicity or treatment delay due to toxicity occurring during the first cycle.

Treatment schedule was as follows: in addition to melphalan 0.18 mg/kg/d on days 1-4 in each 28-day cycle and dexamethasone 40 mg/d on days 1-4 in each 28-day cycle, subjects received lenalidomide. Three successive cohorts of subjects were exposed to escalating doses of lenalidomide (5, 10, and 15 mg once daily on days 1-21 in a 28-day cycle). The dosage of lenalidomide in cohort 3 (ie, 15 mg/d on days 1-21) had been anticipated as the highest tolerable, according to the reports of previous publications showing that this dosage was the optimal dosage in the relapse setting, using lenalidomide as single-agent or with dexamethasone, and without combining with melphalan. However, in November 2008, no DLT had been observed at the lenalidomide dose of 15 mg/d in combination with M-dex, so the protocol was amended to open a fourth cohort at 20 mg/d. Patients could receive up to 9 cycles of treatment according to the protocol. A prophylactic anti-thrombotic treatment consisting of aspirin, low-molecular-weight heparin (LMWH) or warfarin at the physician's discretion was recommended. In spite of aspirin prophylaxis, patient 103 experienced one thrombosis event during cycle 2. The protocol was amended and daily LMWH administration was mandatory during the first 4 cycles of the protocol. In absence of thrombosis during these first 4 months of therapy, aspirin could be introduced, replacing LMWH infusion, for the further treatment cycles. Granulocyte colony-stimulating factor was not permitted during the first cycle of therapy.

Assessment

Baseline assessments and procedures included a complete physical examination, assessment of performans status, amyloid-related organ involvement, and laboratory assessments for hematology and clinical chemistry, serum/urine M-protein analysis, FLC analysis, bone marrow aspirate, and biopsy. Patients were regularly seen during therapy for a follow-up visit including physical examination and laboratory assessments, every 4 weeks or more often, if clinically required.

Organ involvement and response to therapy were assessed according to the international consensus guidelines defined during the 10th International Symposium on Amyloidosis,10  except that bone marrow studies were not required to define a complete response.4  A clinical response was defined as improvement of an organ known to be involved with amyloid. A complete hematologic response was defined as the complete disappearance of the monoclonal immunoglobulin or light chain in a serum or urine specimen; a partial hematologic response was defined as more than a 50% reduction in these proteins. Responses were assessed during the rest period of each treatment cycle, at the end-of-treatment visit, and then every 4 to 6 weeks until progression. Safety was assessed by the incidence of AE or any clinically laboratory changes. Adverse events were recorded throughout the study and until 30 days after the last dose of lenalidomide. Any patient receiving one dose of treatment with lenalidomide was assessable for safety.

Statistical analysis

Kaplan-Meier curves for event-free survival (EFS, defined by the time between diagnosis and the occurrence of progression, relapse, or death) and overall survival (OS) were plotted and compared by the use of the log-rank test.

Patient characteristics

A total of 27 patients with newly diagnosed AL amyloidosis were enrolled in the 4 cohorts of this study. One patient (313) withdrew consent during the first month of the trial and was lost to follow-up. Twenty-six patients were thus assessable for efficacy and toxicity. Baseline demographics and disease characteristics are shown in Table 1. Overall median age was 57 years (range, 27-70 years); 10 patients (38%) had 1 organ involved and 16 patients (62%) 2 organs or more. A total of 10 patients (38%) were considered at high risk.4,11  The plasma cell dyscrasia was λ in 20, κ in 5, and biclonal in 1.

Table 1

Patient characteristics

CharacteristicsAll patients (n = 26)
NumberPercentage
Age, y   
    Median 57 
    Range 27-70 
Sex   
    Male 16 62 
    Female 10 38 
No. of involved organs   
    Median 
    Range 1-6 
Heart 15 58 
Kidney 15 58 
Liver 27 
Peripheral nerve 12 
Patients with high-risk disease 10 38 
Involved serum FLC, mg/L   
    Median 187 
    Range 18-9650 
NT-pro BNP, ng/L   
    Median 1140 
    Range 194-21 091 
    Missing 11 
> 332 ng/L12 12/15 80 
CharacteristicsAll patients (n = 26)
NumberPercentage
Age, y   
    Median 57 
    Range 27-70 
Sex   
    Male 16 62 
    Female 10 38 
No. of involved organs   
    Median 
    Range 1-6 
Heart 15 58 
Kidney 15 58 
Liver 27 
Peripheral nerve 12 
Patients with high-risk disease 10 38 
Involved serum FLC, mg/L   
    Median 187 
    Range 18-9650 
NT-pro BNP, ng/L   
    Median 1140 
    Range 194-21 091 
    Missing 11 
> 332 ng/L12 12/15 80 

NT-pro BNP indicates N-terminal pro brain natriuretic peptide.

DLT and definition of MTD (Table 2)

Three patients were enrolled in cohort 1, M-dex + 5 mg of lenalidomide, and no DLT was observed. Four patients (1 patient, 204, died from progressive disease during the first month of therapy and was not assessable for DLT) were enrolled in cohort 2, M-dex + 10 mg of lenalidomide, and no DLT was observed. Three patients were enrolled in cohort 3, M-dex + 15 mg of lenalidomide, no DLT was observed. Nine additional patients were treated to further define safety at this dose level, which had been anticipated to be the highest. No DLT was observed on these 9 patients. The protocol was thus subsequently amended to open cohort 4, M-dex + 20 mg of lenalidomide. Among the first 3 patients treated in cohort 4, 1 experienced DLT: grade 4 anemia. Further 4 patients received M-dex + 20 mg of lenalidomide and 3 of them experienced DLT: 1 grade 4 neutropenia lasting 10 days, 1 grade 3 muscle pain, and 1 grade 3 cerebrovascular ischemia. Overall, on the 7 patients treated in cohort 4, 4 DLTs were reported, and 15 mg of lenalidomide was defined as MTD, in combination with M-dex.

Table 2

DLT, responses, and outcome

Patient no.Dose of Len, mg/dDLTHematologic responseOR*No. of cyclesRelapse progressionDeath
101 − PR + (H,L,K) − − 
102 − Stable − − − 
103 − PR − − 
204 10 − No − 
205 10 − PR + (K) − 
206 10 − CR + (K) − − 
207 10 − Stable − − 
308 15 − CR + (K) − − 
309 15 − PR + (H) − − 
310 15 − No − 
311 15 − Stable − − 
312 15 − PR − − 
314 15 − PR + (H,K) − − 
315 15 − No − 
316 15 − Stable − − 
317 15 − CR + (K) − − 
318 15 − CR + (K,L) − − 
319 15 − CR + (H,K) − − 
320 15 − CR + (L) − − 
421 20 − No − 
422 20 − PR + (H,K) − − 
423 20 Grade 4 anemia No − − 
424 20 Grade 4 neutropenia, 10 days PR + (K) − − 
425 20 Grade 3 cerebrovascular ischemia Stable − − − 
426 20 Grade 3 muscle pain No − − 
427 20 − PR + (H,K) − − 
Patient no.Dose of Len, mg/dDLTHematologic responseOR*No. of cyclesRelapse progressionDeath
101 − PR + (H,L,K) − − 
102 − Stable − − − 
103 − PR − − 
204 10 − No − 
205 10 − PR + (K) − 
206 10 − CR + (K) − − 
207 10 − Stable − − 
308 15 − CR + (K) − − 
309 15 − PR + (H) − − 
310 15 − No − 
311 15 − Stable − − 
312 15 − PR − − 
314 15 − PR + (H,K) − − 
315 15 − No − 
316 15 − Stable − − 
317 15 − CR + (K) − − 
318 15 − CR + (K,L) − − 
319 15 − CR + (H,K) − − 
320 15 − CR + (L) − − 
421 20 − No − 
422 20 − PR + (H,K) − − 
423 20 Grade 4 anemia No − − 
424 20 Grade 4 neutropenia, 10 days PR + (K) − − 
425 20 Grade 3 cerebrovascular ischemia Stable − − − 
426 20 Grade 3 muscle pain No − − 
427 20 − PR + (H,K) − − 

Len indicates lenalidomide.

*

Type of organ response: PR, partial response; H, heart; L, liver; and K, kidney.

Hematologic response (Table 2)

Overall, disregarding the dose of lenalidomide given, on an intent-to-treat basis, a hematologic response was achieved in 15 of 26 cases (58%), complete in 6 patients and partial in 9 patients. Responses were observed at each dose level, but the higher complete response (CR) rate, 42% (5 of 12 cases), was achieved at the dose of 15 mg of lenalidomide per day, defined as MTD. Five patients had stable disease, and 6 did not respond to therapy.

Organ response (Table 2)

Organ response was achieved in 13 of 26 cases (50%), and responses were observed at each dose level. Organ response and hematologic response were strongly correlated: hematologic reponse was observed in all 13 patients experiencing organ response, as compared with 2 cases of hematologic response among the remaining 13 patients (P < .001, Fisher exact test).

Survival and progression

At the cutoff date of March 1, 2010, median follow-up time from diagnosis for surviving patients was 19 months. Twelve patients had progressed or died (Table 2). Five deaths were observed (median time from diagnosis 2 months, 1-7), 4 due to progressive disease (cardiac, liver and gastrointestinal involvement in 1 case, cardiac in 2 cases, and renal involvement in 1 case), and 1 due to cholangiocarcinoma occurring 7 months after starting treatment, with AL amyloidosis responding to therapy. For the 7 patients alive with disease progression: 2 never responded to M-dex-lenalidomide (1 was rescued with bortezomib + dexamethasone), 2 progressed after an initial response (1 was rescued with bortezomib-dexamethasone), and 3 progressed after an initial phase of stable hematologic disease. Estimated 2-year OS and EFS were 80.8% and 53.8%, respectively (Figure 1). Hematologic and organ responses were both associated with superior EFS rates (estimated 2-year EFS of 81.8% and 84.6% in patients with hematologic (CR plus partial response) and organ responses, versus 20 [P = .0001] and 7.7% [P < .0001], respectively (Figure 2). Similarly, a higher EFS was observed in patients whose FLCs decreased by more than 50% during therapy (70% versus 28.6% at 2 years, P = .019, Figure 2).12 

Figure 1

Overall and event-free survival.

Figure 1

Overall and event-free survival.

Close modal
Figure 2

Event-free survival according to response. Event-free survival according to: (A) Hematologic response (complete response plus partial response vs stable disease or no response, P = .0001); (B) Organ response (response vs no response, P < .0001); (C) FLCs decreased by more than 50% during therapy or not, P = .019.

Figure 2

Event-free survival according to response. Event-free survival according to: (A) Hematologic response (complete response plus partial response vs stable disease or no response, P = .0001); (B) Organ response (response vs no response, P < .0001); (C) FLCs decreased by more than 50% during therapy or not, P = .019.

Close modal

Toxicity, treatment exposure

A median of 7 cycles were given (1 to 9) and 11 patients received the planned 9 cycles of combination therapy (Table 2). All 26 patients are assessable for safety. The most commonly reported adverse events (AEs) of any grade by organ class were hematologic events (31% of 302 AEs overall), gastrointestinal events (16% of all AEs consisting mainly of nausea), general disorders such as fatigue and asthenia (12% of all AEs) and infections (19 episodes, 6% of all AEs; Table 3). No case of neuropathy related to therapy was reported. Twenty-one grade 3 or 4 AEs were observed, leading to discontinuations or dose-reductions of therapy in 10 cases (Table 4).

Table 3

Treatment-emergent AEs of any grade (all cycles)

AEs, total N = 302, 26 patientsNumberPercentage
Any  100 
Hematologic 95 31 
    Neutropenia 29  
    Anemia 30  
    Thrombocytopenia 21  
    Lymphopenia 14  
    Eosinophilia  
Gastrointestinal 49 16 
    Nausea 39  
    Constipation  
    Diarrhea  
    Dry mouth  
    Abdominal distension  
General disorders 37 12 
    Fatigue 15  
    Anorexia  
    Pyrexia  
    Asthenia 14  
Dermatologic 29 10 
    Skin rash 11  
    Pruritis  
    Skin atrophy  
    Dry skin  
    Hyperpigmentation  
Cardiovascular 22 
    Left ventricular systolic dysfunction  
    Peripheral edema  
    Hypotension  
    Atrial fibrillation  
    Thrombosis  
    Cardiac ischemia  
Infectious 19 
Others 51 17 
AEs, total N = 302, 26 patientsNumberPercentage
Any  100 
Hematologic 95 31 
    Neutropenia 29  
    Anemia 30  
    Thrombocytopenia 21  
    Lymphopenia 14  
    Eosinophilia  
Gastrointestinal 49 16 
    Nausea 39  
    Constipation  
    Diarrhea  
    Dry mouth  
    Abdominal distension  
General disorders 37 12 
    Fatigue 15  
    Anorexia  
    Pyrexia  
    Asthenia 14  
Dermatologic 29 10 
    Skin rash 11  
    Pruritis  
    Skin atrophy  
    Dry skin  
    Hyperpigmentation  
Cardiovascular 22 
    Left ventricular systolic dysfunction  
    Peripheral edema  
    Hypotension  
    Atrial fibrillation  
    Thrombosis  
    Cardiac ischemia  
Infectious 19 
Others 51 17 
Table 4

Grade 3 or 4 AEs (all cycles)

Any grade 3 or 4 AETotal N = 21, in 26 patients
Cohort 5 mg  
    Thombosis 
    Eosinophilia 
Cohort 10 mg  
    Congestive heart failure 
    Neutropenia 
    Gastrointestinal hemorrhage 
    Atrial fibrillation 
    Cholangiocarcinoma 
    Urinary retention 
Cohort 15 mg  
    Cardiac ischemia 
    Congestive heart failure 
    Delirium 
    Hypoxia 
    Liver dysfunction 
Cohort 20 mg  
    Anemia 
    Neutropenia 
    Cerebrovascular ischemia 
    Muscle pain 
    Congestive heart failure 
    Renal failure 
Any grade 3 or 4 AETotal N = 21, in 26 patients
Cohort 5 mg  
    Thombosis 
    Eosinophilia 
Cohort 10 mg  
    Congestive heart failure 
    Neutropenia 
    Gastrointestinal hemorrhage 
    Atrial fibrillation 
    Cholangiocarcinoma 
    Urinary retention 
Cohort 15 mg  
    Cardiac ischemia 
    Congestive heart failure 
    Delirium 
    Hypoxia 
    Liver dysfunction 
Cohort 20 mg  
    Anemia 
    Neutropenia 
    Cerebrovascular ischemia 
    Muscle pain 
    Congestive heart failure 
    Renal failure 

This study is the first reporting results of the combination of M-dex + lenalidomide in patients with newly diagnosed AL-amyloidosis. We were able to define the MTD of lenalidomide as follows: 15 mg once daily on days 1 to 21 of a 28-day cycle. This dose was recommended in relapse when patients received lenalidomide alone or in combination with dexamethasone, without addition of melphalan.8,9  In the 2 series already reported of lenalidomide in relapsed AL amyloidosis, most patients were previously treated with alkylating agents including high-dose melphalan in some cases, that could preclude the administration of higher doses of lenalidomide. In first-line treatment, 15 mg of lenalidomide + melphalan and dexamethasone could be given without significant toxicity.

The goal of the combination is to increase both the hematologic and organ response rates, which are associated with longer survival. Note that our dose-escalation schedule tends to underestimate response rates. In our series, on an intent-to-treat basis, hematologic response rates was 58% and organ response rates was 50%. It is sometimes difficult to compare the results of clinical studies in AL amyloidosis due to the wide variability of patient cohorts from trial to trial. For example, the median age of patients in our series is 57 years, which is approximately 10 years younger than the age of patients in the community. Nevertheless, although the number of patients was small, the 42% CR rate achieved at MTD compares favorably with the CR rate achieved with melphalan and dexamethasone without lenalidomide. In the first experience of M-dex reported by the Italian group, the CR rate was 33%.13  Recently updated results on a larger number of patients (159 cases) showed a CR rate achieved with M-dex alone of 25%.14  The 2-year OS estimated to 80% also compared favorably with the OS achieved with M-dex alone. In our recent prospective trial comparing ASCT with M-dex, the 2-year survival in the M-dex arm was 65% in 50 patients.4 

Lenalidomide is not the only novel agent effective in AL amyloidosis. Thalidomide was the first novel agent tested in this disease with significant activity, mostly in combination with cyclophosphamide and dexamethasone.15  Bortezomib has been increasingly used with promising results, both in relapse and frontline treatment.16-18  A recent phase 2 trial of M-dex + bortezomib yielded high response rates, including a 45% CR rate.19  It is important to note that M-dex + lenalidomide can be delivered safely in patients with neuropathy, as opposed to combination using bortezomib or thalidomide. Another difference with bortezomib-based therapy is oral availability. Recently pomalidomide has also shown effectiveness in relapse setting but has only been tested in combination with dexamethasone.20 

In fit and good-risk patients with adequate organ function, low NT-proBNP and troponin T, high-dose therapy with ASCT may be considered as front-line treatment.2,11,21-23  Nevertheless, this group of patients only represents 15% to 20% of the total AL-amyloidosis population.3  Thus, for most patients and given the poor prognosis of this disease, determining a new effective oral combination therapy, such as M-dex lenalidomide without neurotoxicity as defined in this trial, is an important step forward.

This work has been presented previously at the 51st American Society of Hematology annual meeting, New Orleans, December 7, 2009, oral presentation, abstract 427; and the XIIth International Symposium on Amyloidosis, Rome, Italy, April 21, 2010, oral presentation, abstract 86.

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.

Contribution: P.M., A.J., and J.P.F. designed the research; P.M., A.J., L.B., B.R., X.L., F.B., G.S., V.L., M.R., O.H., J.L.H., and J.P.F. performed the research; P.M. collected data; P.M., A.J., and J.P.F. analyzed and interpreted data; L.P. performed statistical analysis; P.M., A.J., and J.P.F. wrote the paper; and all authors reviewed and approved the paper.

Conflict-of-interest disclosure: P.M., A.J., X.L., G.S., M.R., J.L.H., and J.P.F. have received honoraria from Celgene. M.A. is employee of Celgene. The remaining authors declare no competing financial interests.

Correspondence: Philippe Moreau, Hematology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Place A.Ricordeau, 44093, Nantes, France; e-mail: philippe.moreau@chu-nantes.fr.

1
Merlini
 
G
Bellotti
 
V
Molecular mechanisms of amyloidosis.
N Engl J Med
2003
, vol. 
349
 
6
(pg. 
583
-
596
)
2
Comenzo
 
RL
How I treat amyloidosis.
Blood
2009
, vol. 
114
 
15
(pg. 
3147
-
3157
)
3
Palladini
 
G
Merlini
 
G
Current treatment of AL amyloidosis.
Haematologica
2009
, vol. 
94
 
8
(pg. 
1044
-
1048
)
4
Jaccard
 
A
Moreau
 
P
Leblond
 
V
, et al. 
High-dose melphalan versus melphalan plus dexamethasone for AL amyloidosis.
N Engl J Med
2007
, vol. 
357
 
11
(pg. 
9
-
19
)
5
Dimopoulos
 
M
Spencer
 
A
Attal
 
M
, et al. 
Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma.
N Engl J Med
2007
, vol. 
357
 
21
(pg. 
2123
-
2132
)
6
Rajkumar
 
SV
Jacobus
 
S
Callander
 
NS
, et al. 
Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial.
Lancet Oncol
2010
, vol. 
11
 
1
(pg. 
29
-
37
)
7
Palumbo
 
A
Falco
 
P
Corradini
 
P
, et al. 
Melphalan, prednisone, and lenalidomide treatment for newly diagnosed myeloma: a report from the GIMEMA, Italian Multiple Myeloma Network.
J Clin Oncol
2007
, vol. 
25
 
28
(pg. 
4459
-
4465
)
8
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
)
9
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
)
10
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.
Am J Hematol
2005
, vol. 
79
 
4
(pg. 
319
-
328
)
11
Dispenzieri
 
A
Lacy
 
MQ
Kyle
 
RA
, et al. 
Eligibility for hematopoietic stem-cell transplantation for primary systemic amyloidosis is a favourable prognostic factor for survival.
J Clin Oncol
2001
, vol. 
19
 
14
(pg. 
3350
-
3356
)
12
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
)
13
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
, vol. 
107
 
10
(pg. 
3854
-
3858
)
14
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
)
15
Palladini
 
G
Foli
 
A
Milani
 
P
, et al. 
Oral melphalan and dexamethasone for Al amyloidosis: efficacy, prognostic factors and response criteria.
Amyloid
2010
, vol. 
17
 
Suppl 1
(pg. 
81
-
82
)
16
Wechalekar
 
AD
Goodman
 
HJ
Lachmann
 
HJ
, et al. 
Safety and efficacy of risk-adapted cyclophosphamide, thalidomide, and dexamethasone in systemic AL amyloidosis.
Blood
2007
, vol. 
109
 
2
(pg. 
457
-
464
)
17
Kastritis
 
E
Anagnostopoulos
 
A
Roussou
 
M
, et al. 
Treatment of light chain (AL) amyloidosis with the combination of bortezomib and dexamethasone.
Haematologica
2007
, vol. 
92
 
10
(pg. 
1351
-
1358
)
18
Reece
 
DE
Sanchorawala
 
V
Hegenbart
 
U
, et al. 
Weekly and twice-weekly bortezomib in patients with systemicAL amyloidosis: results of a phase 1 dose-escalation study.
Blood
2009
, vol. 
114
 
8
(pg. 
1489
-
1497
)
19
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
)
20
Zonder
 
JA
Sanchorawala
 
V
Snyder
 
RM
, et al. 
Rapid haematologic and organ responses in patients with AL amyloidosis treated with bortezomib plus melphalan and dexamethasone.
Amyloid
2010
, vol. 
17
 
suppl 1
(pg. 
86
-
87
)
21
Dispenzieri
 
A
Gertz
 
MA
Hayman
 
SR
, et al. 
Pomalidomide and dexamethasone for previously treated AL: a phase 2 study.
Amyloid
2010
, vol. 
17
 
suppl 1
pg. 
87
 
22
Dispenzieri
 
A
Gertz
 
MA
Kyle
 
RA
, et al. 
Prognostication of survival using cardiac troponins and N-terminal pro-brain natriuretic peptide in patients with primary systemic amyloidosis undergoing peripheral blood stem cell transplantation.
Blood
2004
, vol. 
104
 
6
(pg. 
1881
-
1887
)
23
Sanchorawala
 
V
Skinner
 
M
Quillen
 
K
, et al. 
Long-term outcome of patients with AL amyloidosis treated with high-dose melphalan and stem-cell transplantation.
Blood
2007
, vol. 
110
 
10
(pg. 
3561
-
3563
)
Sign in via your Institution