J Hematol
Journal of Hematology, ISSN 1927-1212 print, 1927-1220 online, Open Access
Article copyright, the authors; Journal compilation copyright, J Hematol and Elmer Press Inc
Journal website https://www.thejh.org

Review

Volume 10, Number 4, August 2021, pages 147-161


Current Updates on the Management of AL Amyloidosis

Marwa Elsayeda, Sara Usherb, Muhammad Hamza Habibc, Nausheen Ahmedd, Jawad Alie, Madeline Begemannb, Syed Ahmed Shabbirf, Leila Shuned, Jaffar Al-Hillig, Furha Cossorb, Brett W. Sperryh, Shahzad Razab, i

aSt Luke’s Hospital of Kansas City, University of Missouri Kansas City, Wornall Rd, Kansas City, MO 64111, USA
bSt Luke’s Cancer Institute, University of Missouri Kansas City, 4321 Washington St, Ste 4000, Kansas City, MO 64111, USA
cRutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA
dUniversity of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
eWest Midland Deanery, 213 Hagley Road, Birmingham, B16 9RG, UK
fUniversity of Kansas, 1450 Jayhawk Boulevard, Lawrence, KS 66045, USA
gUniversity of Missouri Columbia, 1 Hospital Dr, Columbia, MO 65212, USA
hMid America Heart Institute, St Luke’s Hospital of Kansas City, Wornall Rd, Kansas City, MO 64111, USA
iCorresponding Author: Shahzad Raza, St Luke’s Cancer Institute/University of Missouri Kansas City, 4321 Washington St, STE 4000, Kansas City, MO 64111, USA

Manuscript submitted May 24, 2021, accepted June 14, 2021, published online August 4, 2021
Short title: Management of AL Amyloidosis
doi: https://doi.org/10.14740/jh866

Abstract▴Top 

Systemic immunoglobulin light chain (AL) amyloidosis is a rare but fatal disease. It results from clonal proliferation of plasma cells with excessive production of insoluble misfolded proteins that aggregate in the extracellular matrix, causing damage to the normal architecture and function of various organs. For decades, treatment for AL amyloidosis was based mainly on therapeutic agents previously studied for its more common counterpart, multiple myeloma. As the prevalence and incidence of AL amyloidosis have increased, ongoing research has been conducted with treatments typically used in myeloma with varying success. In this review, we focus on current treatment strategies and updates to clinical guidelines and therapeutics for AL amyloidosis.

Keywords: AL amyloidosis; Light chain amyloidosis; AL amyloidosis therapy; Light chain amyloidosis treatment

Introduction▴Top 

Immunoglobulin light chain (AL) amyloidosis is a rare disease characterized by clonal proliferation of plasma cells with over-production of monoclonal light chains that transform into misfolded protein fibrils, with a special configuration making them insoluble and causing large-scale depositions in the extracellular matrix resulting in organ dysfunction. AL deposits most commonly accumulate in the heart and kidneys but can affect other organs, including the peripheral nervous system. This process results in cytotoxicity and ultimately organ dysfunction [1]. Though AL amyloidosis prevalence in the USA is low (40.5 cases per million in 2015) [2], the mortality rate is high, with an average survival ranging 6 - 36 months [3, 4]. AL amyloidosis can affect multiple organs either simultaneously or separately and can manifest with a wide variety of non-specific presenting symptoms, e.g., unexplained heart failure, heavy proteinuria, hepatomegaly, or neuropathy. This heterogeneity of presentations makes it difficult to diagnose AL amyloidosis at earlier stages [5]. The ultimate diagnosis for AL amyloidosis requires tissue and/or bone marrow biopsy confirmation in addition to an extensive laboratory and imaging workup (Table 1) [6], which might cause further delay for the diagnosis [7]. Based on observations from the clinical trials, prognostic factors for risk stratification formulate an individualized treatment plan for each patient and predict morbidity and mortality [6, 8, 9]. It is important to mention that the burden of cardiac involvement is the most important prognostic factor for the treatment outcome and overall survival (OS).

Table 1.
Click to view
Table 1. Diagnostic Workup for AL Amyloidosis
 

Currently, there are no clear guidelines on systemic AL amyloidosis treatment [10]; however, since both AL amyloidosis and multiple myeloma (MM) are monoclonal plasma cell dyscrasias, AL amyloidosis treatment strategies and medications are derived from the anti-plasma cell therapy that is used for MM [11]. Interestingly, lower tumor burdens and cytogenetic abnormalities are associated with better treatment outcomes in AL amyloidosis than in MM [12]. Therapy is aimed at achieving deep and rapid hematological response, which reverses amyloid-mediated organ dysfunction, and improves OS [13-15]. Table 2 [9, 16-44] summarizes therapeutic combinations in the management of AL amyloidosis.

Table 2.
Click to view
Table 2. Therapeutic Combinations in the Management of AL Amyloidosis
 
Autologous Stem Cell Transplantation (ASCT)▴Top 

ASCT with high-dose melphalan conditioning was first described in the literature as a treatment for AL amyloidosis in 1998 [45]. The only randomized controlled trial evaluating the efficacy of ASCT in AL amyloidosis was published in 2007 and compared patients treated with high-dose melphalan followed by ASCT to those who received melphalan and dexamethasone (M-D) without ASCT. Median OS in the patients medically managed was significantly improved compared with the group undergoing ASCT (OS 56.9 months versus 22.2 months, P = 0.04) [46]. This result has withstood several criticisms. First, there was a high treatment-related mortality (TRM) in the ASCT arm, but landmark analysis in patients surviving 6 months showed no difference in outcomes among groups. Second, some higher-risk patients were included in the study. However, an analysis of good-risk patients also showed no difference in outcomes at 3 years (58% OS in ASCT versus 80% OS in M-D, P = 0.13). Third, the duration of response of M-D was questioned versus ASCT, which is thought to offer a more durable response. In a long-term follow-up of the data, the results of ASCT versus M-D were again similar [47]. Both medical therapy for AL amyloidosis and ASCT management have improved since this trial. The reduction in TRM of ASCT in the modern era is primarily due to improved peri-transplant care and better patient selection, including the incorporation of cardiac biomarkers [48]. Due to the commonly late diagnosis of AL amyloidosis, only 20% of patients are found to be eligible for ASCT therapy at the initial diagnosis [9].

High-dose melphalan followed by ASCT is considered the most commonly used first-line therapy for patients who fit the eligibility criteria for ASCT. Oral chemotherapy induction was not found to be helpful and led to disease progression and disqualification for ASCT in some patients [49]. However, bortezomib-based induction with two cycles prior to ASCT was found to improve hematologic remission and OS in a randomized controlled trial and several retrospective studies [50-52]. Landau et al published a pilot study that involved 19 patients newly diagnosed with AL amyloidosis who underwent three phases of therapy, including an induction phase with bortezomib and dexamethasone (BD) for 1 - 3 cycles, followed by risk-adapted ASCT (the induction dose of melphalan was adapted based on the risk stratifications and organ involvement), and ultimately six cycles of consolidation therapy with BD [53]. OS rate and 2-year progression-free survival (PFS) rate were 84% and 64% respectively, with an overall response rate (ORR) of 91% (37% achieved complete response (CR), 37% achieved very good partial response (VGPR), and 21% achieved partial response (PR)). This regimen furthers the hypothesis that proteasome inhibitors (PIs) like bortezomib have a synergistic effect when used in conjunction with alkylator therapy as apoptotic agents that prevent further clonal plasma cell proliferation and augment the effect of ASCT [54, 55]. Cornell et al reported a better outcome of induction therapy with bortezomib prior to ASCT in patients with low burden plasma cells (PCs) (PCs ≤ 10%) when compared to no induction therapy. Bortezomib was able to lower the relapse/progression events (hazard ratio (HR), 0.43; 95% confidence interval (CI), 0.24 to 0.78; P < 0.01) and prolong the PFS rate at 2 years (HR, 0.43; 95% CI, 0.26 to 0.72; P < 0.01) [56]. According to the latest recommendations from the Mayo Clinic, induction therapy with bortezomib is highly recommended, particularly when bone marrow PCs are ≥ 10%, in high-risk cytogenetics (e.g., 17p deletion, t(4;14), t(14;20)), or concurrent myeloma) [16]. In addition, Center for International Blood and Marrow Transplant Research (CIBMTR) registry data showed that patients who received bortezomib-based induction had improved progression ratio, which suggests that pre-transplant bortezomib-based induction is better than proceeding with upfront ASCT. The relevance of minimal residual disease, cytogenetics, and maintenance therapy is being explored.

Regarding melphalan conditioning dose, a higher dose (70 - 200 mg/m2) preceding ASCT was associated with a better hematological response and increased median OS than a lower dose [17, 18]. Moreover, a high melphalan induction dose of 200 mg/m2 was tolerable in patients above the age of 65 with good performance status (Karnofsky Performance status scale of ≥ 90) with no increase in TRM except for a noticeable increase in post-transplant febrile neutrophilia and infections [57]. Low-dose melphalan is used in MM for patients with dialysis-dependent end-stage renal disease (ESRD) with similar efficacy and TRM as melphalan 200 mg/m2 [58]. However, no studies have yet investigated similar approaches in patients with AL amyloidosis.

Consolidation therapy consists of treatment with any therapeutic agent against PCs initiated post-ASCT to maintain or deepen the response of the ASCT; it is usually reserved for patients with more advanced disease stages who failed to achieve a favorable outcome with ASCT therapy. Interestingly, consolidation therapy was found to achieve a better PFS rate and median OS in patients with less than VGPR (22.4 and 125.8 months, respectively) compared to patients with VGPR or better (8.8 and 74.4 months, respectively) [59]. Routine maintenance therapy may not be of benefit, as lenalidomide failed to demonstrate any difference in the mean OS or PFS rate after ASCT in this setting [60].

Though acquired factor X deficiency is not as common in AL amyloidosis as it is in MM, it is associated with a worse outcome with a lower median OS (associated with a median OS of 9.3 months vs. 118.4 months in patients without factor X deficiency) [61]. It is important to note that severe factor X deficiency (< 25% deficiency) was associated with a higher incidence of serious bleeding complications in the peri-transplant period, particularly when combined with other factor deficiencies, and it is important to screen patients with AL amyloidosis for factor X deficiency before offering them ASCT [62].

Alkylating Agents and Steroids▴Top 

Since the first randomized controlled trial in AL amyloidosis in 1978 demonstrated that melphalan and prednisone improve outcomes when compared with placebo, alkylating agents have been used as the mainstay of therapy in this condition [63]. Subsequent studies proved that melphalan and prednisone were superior to colchicine, an anti-inflammatory medication [64]. In a sicker population of patients not eligible for ASCT, Palladini and colleagues in 2007 demonstrated encouraging outcomes with the M-D regimen, of whom 67% were able to achieve a hematological response (CR in 33%) [19]. Even in the prolonged follow-up of the survivors (median follow-up 5 years), the median PFS rate and median OS were 3.8 and 5.1 years, respectively. As noted above, Jaccard et al found that M-D was able to achieve a comparable result and a better TRM profile compared to ASCT [46, 47].

Over time, dexamethasone has replaced prednisone due to stronger glucocorticoid effects and improved survival data in patients with MM [65]. The use of multiple alkylating agents + prednisone was not found to be beneficial versus melphalan + prednisone, suggesting that medications with different and potentially synergistic mechanisms were needed to improve outcomes [66].

Proteasome Inhibitors▴Top 

The proteasome is a cellular complex that participates in protein degradation, which is considered a vital step in cell growth, maturation, and proliferation [67]. Since the FDA approved the first PI, bortezomib, in 2003, the treatment of PC clonal dyscrasias has greatly advanced [68].

Bortezomib

Bortezomib has been used in AL amyloidosis as a single agent or in combination with other chemotherapeutic agents. A retrospective study of 43 patients received bortezomib in combination with cyclophosphamide and dexamethasone (CyBorD) either as upfront or second-line therapy for relapsed disease and found an overall hematological response of 81.4% with a CR of 39.5%; CR was significantly higher in the upfront therapy group vs. the relapsed group (65.0% vs. 21.7%, respectively; P = 0.003) [20]. Moreover, the 1-year and 2-year PFS rates were better in the upfront group compared to the relapsed group (74.5% vs. 70.9% for 1 year, and 66.5% vs. 41.4% for 2 years, respectively). Similarly, Mikhael et al found a rapid and solid hematological response with CyBorD in both newly diagnosed and relapsed/refractory AL amyloidosis patients (time to response was 2 months, and the hematological response was achieved in 94% of patients), and three patients were later able to have ASCT despite initially being classified as ASCT-ineligible [21]. Subsequently, in 2015, Palladini et al investigated the role of CyBorD based on disease severity in a large retrospective study [69]. A total of 230 patients were classified into stage I, II, or III based on the Mayo Cardiac Staging System (Table 3 [6, 70, 71]), with stage III being further divided into stage IIIa or stage IIIb based on the level of amino-terminal pro-natriuretic peptide type-B (NT-proBNP); stage IIIa was below the value of 8,500 ng/L, and stage IIIb was above this value, which indicates a very poor prognosis. The hematological response varied significantly based on the degree of cardiac involvement with overall response rates of 77%, 64%, 69%, and 42% for stage I, II, IIIa, and IIIb, respectively; CR was 33%, 18%, 23%, and 14%, respectively. The 5-year survival rate for the entire population was 55%. Palladini et al published a case-control trial of M-D versus BMDex and found improved rates of complete hematologic response with the bortezomib-based regimen but no change in OS. This was driven by no survival advantage in patients with severe cardiac disease [72]. Similarly, BMDex was found superior to M-D in improving hematological response and OS rates in a recent phase III multicenter, randomized, open-label clinical trial [22]. Sperry et al studied patients with AL amyloidosis presenting with symptomatic heart failure. They found that the regimen of bortezomib, dexamethasone and an alkylating agent (either cyclophosphamide or melphalan) was associated with improved outcomes when compared with other initial regimens [73].

Table 3.
Click to view
Table 3. Prognostic Mayo Staging System for AL Amyloidosis 2012
 

Bortezomib may also be combined with immunomodulatory drugs (IMiDs), particularly the VRD regimen (bortezomib + lenalidomide + dexamethasone). One study showed a favorable hematological response (75.5% after the first cycle with 88% based on intention to treat analysis) and a trend towards a deeper response when compared with CyBorD [23]. Though, this was at the expense of increased toxicity, particularly skin rashes and other non-hematological adverse events (AEs).

Though it has become a backbone treatment for AL amyloidosis, it is important to note that bortezomib therapy is associated with a resistant and less-favorable outcome in AL amyloidosis patients harboring t(11;14) [74]. The most detrimental adverse effect is neuropathy, which has a greater incidence at higher and more prolonged dosing and when the intravenous form is given instead of the subcutaneous form [75, 76].

Carfilzomib

Carfilzomib is a second-generation PI that selectively inhibits chymotrypsin-like (CT-L) activity, a major driver of enhanced cell death in transformed cells with preservation of the proteasome function in non-transformed cells; this is in contrast to bortezomib, which has non-selective PI properties (CT-L, caspase-like (C-L), and trypsin-like (T-L)) causing cytotoxic effects on malignant and non-malignant cells [77]. Carfilzomib showed a comparable result when used in patients with relapsed-refractory AL amyloidosis. A total of 929 patients with refractory AL amyloidosis were recruited between June 20, 2012 and June 30, 2014; 464 were randomly assigned to receive carfilzomib and dexamethasone (with a median follow-up of 11.9 months), while the rest received BD (with a median follow-up of 11.1 months). Carfilzomib therapy was associated with prolonged median PFS compared to bortezomib therapy (18.7 vs. 9.4 months, respectively) [78]. Despite that, carfilzomib is to be used cautiously in cases of refractory AL amyloidosis because the higher doses were associated with more grade 3-4 toxicities, namely cardiac, renal, pulmonary, and hematological toxicities [78, 79]. The only exception is neurotoxicity; carfilzomib was found to be associated with a lower incidence of neurotoxicity, which may make it a better upfront therapy line for patients who have AL amyloidosis associated with neuropathy in contrast to bortezomib; however, the available data are not sufficient to generalize this hypothesis [80]. In fact, in MM, the carfilzomib-lenalidomide-dexamethasone (KRd) regimen was found to have similar PFS compared to bortezomib, lenalidomide, and dexamethasone (VRd) with more toxicity [81]. Further study of carfilzomib in AL amyloidosis is needed.

Ixazomib

Ixazomib is an oral PI that was approved for relapsed/refractory MM treatment in 2015 [82]. The role of ixazomib in AL amyloidosis is not well studied in clinical trials. Initial data from a clinical trial performed on 27 patients who failed the first-line therapy for AL amyloidosis were positive, with an overall hematological response of 52%, including a documented organ response in 56% of patients; the median PFS and 1-year PFS were 14.8 months and 60%, respectively [42]. However, the TOURMALINE-AL1 trial, a multicenter international phase III clinical trial that was designed to evaluate the role of ixazomib in addition to dexamethasone in 248 patients with refractory/relapsed AL amyloidosis, was terminated because ixazomib failed to achieve a desired hematological response compared to other standard regimens [83]. Currently, ixazomib is under investigation in combination with other chemotherapeutic drugs in both upfront and relapsed disease (NCT03236792, NCT01864018, and NCT03283917) [84]. The promise of an oral PI is enticing, given easier administration and less neuropathic side effects. Though, skin rash appears to be more prevalent with ixazomib.

IMiDs▴Top 

In order to maintain their accelerated growth, tumor cells secrete cytokines that suppress natural immune responses and prevent tumor antigens’ recognition by the immune system. One of the well-known mechanisms by which the tumor microenvironment can escape immune system surveillance is by increasing T-regulatory cells, a group of CD4 T cells. T-regulatory cells counteract the normal function of cytotoxic CD8 T cells and natural killer (NK) cells against tumor cells and interfere with their ability to identify tumor cell epitopes. Immunomodulator therapy was found to play a pivotal role in upregulating and modulating this imbalance by co-stimulating CD8 T cells and NK cell production and enhancing their function against tumor cells [85]. Although they achieved a good outcome in MM, IMiDs were associated with higher rates of toxicity in AL amyloidosis patients [86].

Currently, IMiDs are considered second-line therapy in AL amyloidosis [9]. However, they are frequently used as an oral option for maintenance therapy after CR or VGPR has been achieved.

Thalidomide

Thalidomide was initially marketed and developed as a sedative and was used in pregnant women, leading to severe congenital disabilities. It was taken off the market by the FDA in 1961, but subsequent trials in leprosy demonstrated benefit. After its approval in 1998 for this condition, it was tested and showed remarkable benefit on outcomes in MM, leading to its approval for this condition in 2006 [87, 88]. The role of thalidomide role in AL amyloidosis was investigated by Palladini et al [24]. Out of 31 patients who received thalidomide and dexamethasone, 15 patients (48%) were able to achieve a hematological response with CR detected in six patients (19%) with a significant improvement in organ functions. Despite that, only 35% of patients were able to tolerate the full dose of thalidomide because of associated AEs, mainly symptomatic bradycardia (incidence rate of 26%). Adding alkylator-like cyclophosphamide to the previous regimen achieved a hematological response of 74%, including CR in 21% when used in 75 patients with advanced AL amyloidosis [25]. Median OS was 41 months with a 3-year survival rate of 100% in the CR group. TRM was 4%, with 8% of patients experiencing drug toxicity that indicates drug cessation. A slightly better hematological response and CR were observed by Venner et al in 2014 (79.7% and 24.6%, respectively) with substantial concerns related to cardiac toxicity associated with thalidomide therapy [89].

Lenalidomide

Lenalidomide and pomalidomide are associated with a slightly better toxicity profile. Evaluating lenalidomide in combination with dexamethasone (Len-D), particularly in patients with refractory/relapsed disease, did not show consistent results, including one study that showed a total hematological response of 41% only (including 0% CR) [90], and another study that showed a 61% overall hematological response (including 20% CR) [26]. Using lenalidomide as a single agent was associated with a lower efficacy in treating AL amyloidosis [27, 28]. Lenalidomide and dexamethasone have been tested with both alkylators. In a single center prospective clinical trial, using melphalan, lenalidomide and dexamethasone (MLD) in 50 patients newly diagnosed with AL amyloidosis achieved a total hematological response of 68%, including a CR of 18%, with median OS and PFS of 67.5 months and 25.1 months, respectively [91]. Similarly, using a cyclophosphamide, lenalidomide and dexamethasone (CLD) regimen as an upfront therapy in a multicenter prospective clinical trial that included 24 newly diagnosed patients with AL amyloidosis achieved a total hematological response of 46%, including CR in 25% [92]. It is important to note that grade 3-4 AEs may be higher in the MLD regimen.

Pomalidomide

Pomalidomide is another second-generation IMiD approved by the FDA for MM treatment in 2008 [93]. Though the clinical data that support the use of this therapy in AL amyloidosis are based only on phase 1-2 trials, the outcomes are encouraging. A total of 29 patients received pomalidomide between 2009 and 2017 in the UK for AL amyloidosis; only 39% were able to achieve VGPR at 6 months, and no patients achieved CR. However, pomalidomide achieved a rapid response with median OS and PFS of 27 months and 15 months, respectively [94]. On average, the overall hematological response in patients with relapsed/refractory AL amyloidosis who received a combination of pomalidomide and dexamethasone ranged between 48% and 68%, with a very rapid response reported within 1 month [29, 30].

Second-generation IMiDs, particularly pomalidomide, were associated with a favorable outcome in refractory/relapsed AL amyloidosis with a hypothesis that they overcome alkylator/bortezomib resistance in previously treated AL amyloidosis [21, 90]. Pomalidomide and dexamethasone’s role in treating relapsing/refractory AL amyloidosis was recently investigated in a retrospective study for 153 patients, where 93% of them previously received bortezomib, 81% lenalidomide, 75% melphalan, and 24% had ASCT [95]. In combination with dexamethasone, pomalidomide was able to achieve an overall hematological response of 44% after six cycles, with an increase in median OS up to 50 months.

It is important to keep in mind that treatment with IMiDs, particularly when used in AL amyloidosis, can be associated with fluid retention and an increase in NT-proBNP. However, it is not clear if this increase is related to the direct cardiotoxic effect of IMiDs or fluid retention, causing an elevation in NT-proBNP [96]. Additionally, as IMiDs can upregulate parts of the immune system, they should not be used in patients after solid organ transplantation due to the risk of precipitating acute rejection [97].

Monoclonal Antibodies Targeting CD38▴Top 

CD38 is a transmembrane glycoprotein molecule that plays an important role in cell adhesion and is heavily present in clonal PCs [98]. Daratumumab is a humanized monoclonal antibody against CD38 that can initiate antibody-mediated cellular toxicity along with complement-mediated cytotoxicity [99]. After achieving a satisfactory outcome in treating MM, daratumumab has been used in clinical trials of AL amyloidosis treatment recently, mainly for relapsed/refractory disease. Shortly after being approved by the FDA for refractory/relapsed MM in 2015 [100], the efficacy and safety of daratumumab in AL amyloidosis was investigated in a small case series; daratumumab was able to decrease serum-free light chain levels significantly and rapidly [101]. A multicenter phase II clinical trial AMYDARA recruited 40 patients diagnosed with refractory AL amyloidosis who were previously treated (median of three of therapy, range 1 - 5), with more than 50% of patients having equal to or more than two organs involved [102]. A total of 55% of patients achieved a hematological response, including 47.5% with a VGPR or better. As in the previous case series [101], this prospective study demonstrated a very rapid hematological response in patients treated with daratumumab (median hematological response of 1 week). Adding dexamethasone to daratumumab was associated with a better total hematological response of 76%, including CR in 36% when used in 25 patients with previously treated AL amyloidosis, with a median hematological response observed after 1 month [31]. In all cases, daratumumab was associated with a very tolerable AEs profile.

ANDROMEDA was a landmark phase III clinical trial where subcutaneous daratumumab was investigated in combination with CyBorD versus a regimen of CyBorD alone in 388 patients newly diagnosed with AL amyloidosis with equal to or more than one organ involved and with an Eastern Cooperative Oncology Group (ECOG) performance scale of 0 - 2 [32, 33]. The overall hematological response was significantly higher in the daratumumab + CyBorD group (92% including a CR of 53%), compared to the CyBorD group (77% including a CR in 18%). There was also a significant improvement in 6-month organ response, with 42% achieving cardiac response (versus 22%) and 54% achieving renal response (versus 27%).

Along with the deeper and more rapid hematological response in AL amyloidosis, daratumumab is also attractive given its potential for subcutaneous dosing, saving patients with cardiac involvement from receiving a higher amount of fluid. There was a small risk increase in grade 3-4 toxicity in the daratumumab combination group, particularly lymphopenia (13% with the daratumumab combination vs. 10% with CyBorD alone) and pneumonia (8% vs. 4%, respectively). However, the reported discontinuation rate because of adverse treatment events was only 4% in the daratumumab cohort.

Targeting Amyloid Component Therapy▴Top 

In AL amyloidosis, the burden of symptoms often depends on the burden of amyloid deposition into organs; hence, researchers must not only develop drugs that slow or kill the malignant PCs but also target the products of the misfolded light chain. NEOD001, a monoclonal antibody that targets a specific epitope that is present only on abnormally folded light chains, can bind and neutralize them by facilitating absorption and clearance [103]. NEOD001 was initially evaluated in a phase I/II clinical trial with encouraging results [105]; however, subsequent studies failed to prove a significant improvement in outcomes [34, 104]. Subgroup analysis of VITAL suggested benefit in Mayo Stage IV patients with severe cardiac involvement and a confirmatory trial with this medication (now called birtamimab) is planned in this population [34].

CAEL-101 is another monoclonal antibody that has a strong affinity to kappa and lambda light chains and is a byproduct of monoclonal PC malignant expansion. When it binds to kappa and lambda light chains, it initiates neutrophil-mediated phagocytosis, enhancing the clearance of these light chains. This may decrease the light chain burden on organs and improve overall organ response to AL amyloidosis treatment [35, 36]. An initial analysis of a phase I a/b clinical trial showed an objective cardiac and renal response in 67% of the patients with no reported grade 4/5 AEs [105].

Bendamustine Therapy▴Top 

Bendamustine is a chemotherapeutic agent that has properties of both alkylators and antimetabolite and was approved by the FDA in 2008 for chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL) [106, 107]. A retrospective study of 122 patients with refractory/relapsed AL amyloidosis treated with bendamustine and prednisolone showed a better hematological response in patients with IgM-AL amyloidosis compared to patients with non-IgM-AL amyloidosis (58% vs. 28%, respectively); however, the overall hematological response was 35% only with a median PFS of 9 months [37]. Another multicenter phase II trial investigated the role of bendamustine and dexamethasone (Ben-D) in a total of 31 patients with progressive or persistent AL amyloidosis after receiving at least one therapy or more [108]. The Ben-D combination achieved a PR of 57% or better, and median PFS and median OS of 11.3 and 18.2 months, respectively. Two-thirds of patients developed grade 3-4 toxicities, with myelosuppression and fatigue being the most common. As noted above, the effect of bendamustine in refractory/relapsed AL amyloidosis is unclear and more research is needed in order to investigate its role.

Small Molecules▴Top 

Doxycycline

Doxycycline is an antimicrobial drug that interferes with protein synthesis by binding to certain sites on the ribosomes; perhaps doxycycline decreases the burden of misfolded protein aggregation in amyloidosis through this mechanism [109, 110]. An observational study showed that doxycycline improves the median OS when used as a prophylactic antimicrobial agent for patients who underwent ASCT compared to other patients who received penicillin, and particularly in patients who achieved hematological response [111]. A similar outcome was reported by Wechalekar et al, who compared doxycycline administration along with standard chemotherapeutics for cardiac AL amyloidosis in 30 patients to 73 matched historical controls; the therapy was associated with a better overall hematological response compared to a control group (93% vs. 59%, respectively), as well as better 12- and 24-month survival rates (82% vs. 53% and 82% vs. 40%, respectively) [38]. A recently published prospective phase II trial was able to prove that long-term treatment with doxycycline in patients with newly diagnosed AL amyloidosis was not only associated with a decrease in mortality and a safe profile but also with increased transplant utilization after receiving triple induction chemotherapy with a post-ASCT 100-day mortality rate of 0 [112].

Venteoclax

Venteoclax is a small molecule that has a biological inhibitory effect on the mutated gene t(11;14), which is found in B-cell lymphoma, MM, and in up to 50% of AL amyloidosis patients [74]. Venteoclax was able to achieve a CR in a 67-year-old man with refractory systemic AL amyloidosis harboring t(11;14) after failing a CyBorD regimen [113]. Though a phase III study that was designed to investigate the role of venteoclax in combination with BD in relapsed/refractory MM was suspended due to an increased risk of death in the venteoclax cohort (NCT02755597), the data from a case series using venteoclax in refractory/relapsed AL amyloidosis showed significant results in term of achieving CR without experiencing serious AEs [114, 115]. This is especially important, as bortezomib is often less responsive in patients with this t(11;14) mutation. Venteoclax use remains a notion that could bring hope for refractory/relapsed AL amyloidosis patients, which indeed necessitates more studies to be conducted on safety and efficacy.

Epigallocatechin-3-gallate (ECGC)

Cardiac involvement in AL amyloidosis is considered to be one of the major factors that determine disease severity, choice of initial therapy, and the expected outcome of the treatment. ECGC is a major antioxidant that is found naturally in green tea and has been proven to exert some effects on misfolded amyloid fibrils, rendering them into a more benign form that does not polymerize into insoluble fibrils and does not exert a cytotoxic effect when aggregated in the extracellular matrix [39]. ECGC was introduced as adjuvant therapy in the management of AL amyloidosis, particularly in cardiac AL amyloidosis, based on a favorable result reported in a case reports after a hematologist, Werner Hunstein, treated himself with high doses of this agent [39, 40]. In TAME-AL, a completed phase II randomized clinical trial that was designed to investigate the effect of a 12-month course of ECGC on the left ventricular mass in cardiac AL amyloidosis, the final results have not yet been published (NCT02015312).

Conclusions▴Top 

AL amyloidosis is a systemic disease with a high mortality rate. ASCT has been the preferred treatment in eligible patients; however, randomized clinical trials showing benefit over conventional treatment are lacking. Steroids, alkylating agents, and PIs have emerged as the first-line combination drug therapy in patients with AL amyloidosis, with the most common regimen being CyBorD. Recently, daratumumab has demonstrated improved outcomes when combined with CyBorD with a low side effect profile and should be considered as first-line therapy. IMiDs are less favorable as first-line therapy for AL amyloidosis but should be considered strongly in cases of refractory/relapsed disease or for maintenance therapy. Since treatment of AL amyloidosis is based mainly on experiences in MM, researches focused on a better understanding of AL amyloidosis pathology as well as targeted clinical trials are essential.

Acknowledgments

None to declare.

Financial Disclosure

ME, SU, HH, NA, JA, MB, AS, and LS have no competing financial interests. BS is a consultant for Pfizer and Alnylam. SR participated in advisory board and speaker bureau from Takeda, Incyte, Janssen, Celgene, and Kite.

Conflict of Interest

The authors declare they do not have a conflict of interest.

Author Contributions

ME, NA, BS, and SR involved in the design of the study, literature review, data interpretation, and manuscript writing. SU, HH, JI, AS, MB, and LS involved in critical review of literature, independent data, and manuscript writing.

Data Availability

Any inquiries regarding supporting data availability of this study should be directed to the corresponding author.

Abbreviations

MM: multiple myeloma; OS: overall survival; CR: complete response; VGPR: very good partial response; PR: partial response


References▴Top 
  1. Zhang C, Huang X, Li J. Light chain amyloidosis: Where are the light chains from and how they play their pathogenic role? Blood Rev. 2017;31(4):261-270.
    doi pubmed
  2. Quock TP, Yan T, Chang E, Guthrie S, Broder MS. Epidemiology of AL amyloidosis: a real-world study using US claims data. Blood Adv. 2018;2(10):1046-1053.
    doi pubmed
  3. Kyle RA, Gertz MA. Primary systemic amyloidosis: clinical and laboratory features in 474 cases. Semin Hematol. 1995;32(1):45-59.
  4. Weiss BM, Lund SH, Bjorkholm M, Cohen AD, Dember L, Landgren O, et al. Improved survival in AL amyloidosis: a population-based study on 1,430 patients diagnosed in Sweden 1995-2013. Blood. 2016;128(22):4448.
    doi
  5. Desport E, Bridoux F, Sirac C, Delbes S, Bender S, Fernandez B, et al. Al amyloidosis. Orphanet Journal of Rare Diseases. 2012;7(1):1-13.
    doi pubmed
  6. UpToDate. Treatment and prognosis of immunoglobulin light chain (al) amyloidosis and light and heavy chain deposition diseases. 2021. Available from: https://www.uptodate.com/contents/treatment-and-prognosis-of-immunoglobulin-light-chain-al-amyloidosis-and-light-and-heavy-chain-deposition-diseases?search=al%20%20amyloidosis%20source%3Dsearch_result%20selected%20Title%3D2~86%20usage_type%3Ddefault%20display%20rank%3D2.
  7. Rysava R. AL amyloidosis: advances in diagnostics and treatment. Nephrol Dial Transplant. 2019;34(9):1460-1466.
    doi pubmed
  8. Dispenzieri A, Buadi F, Kumar SK, Reeder CB, Sher T, Lacy MQ, Kyle RA, et al. Treatment of immunoglobulin light chain amyloidosis: mayo stratification of myeloma and risk-adapted therapy (mSMART) consensus statement. Mayo Clin Proc. 2015;90(8):1054-1081.
    doi pubmed
  9. Palladini G, Milani P, Merlini G. Management of AL amyloidosis in 2020. Hematology Am Soc Hematol Educ Program. 2020;2020(1):363-371.
    doi pubmed
  10. Network NCC. Systemic light chain amyloidosis NCCN evidence blocks. 2021. Available from: https://www.nccn.org/professionals/physician_gls/pdf/amyloidosis_blocks.pdf.
  11. Palladini G, Merlini G. What is new in diagnosis and management of light chain amyloidosis? Blood. 2016;128(2):159-168.
    doi pubmed
  12. Bochtler T, Hegenbart U, Cremer FW, Heiss C, Benner A, Hose D, Moos M, et al. Evaluation of the cytogenetic aberration pattern in amyloid light chain amyloidosis as compared with monoclonal gammopathy of undetermined significance reveals common pathways of karyotypic instability. Blood. 2008;111(9):4700-4705.
    doi pubmed
  13. Gavriatopoulou M, Musto P, Caers J, Merlini G, Kastritis E, van de Donk N, Gay F, et al. European myeloma network recommendations on diagnosis and management of patients with rare plasma cell dyscrasias. Leukemia. 2018;32(9):1883-1898.
    doi pubmed
  14. Gertz M. Definition of organ involvement and response to treatment in AL amyloidosis: an updated consensus opinion. Amyloid. 2010;17:48-49.
  15. Landau H, Smith M, Landry C, Chou JF, Devlin SM, Hassoun H, Bello C, et al. Long-term event-free and overall survival after risk-adapted melphalan and SCT for systemic light chain amyloidosis. Leukemia. 2017;31(1):136-142.
    doi pubmed
  16. mSMART MC. Mayo consensus on al amyloidosis: Diagnosis, treatment and prognosis 2020. Available from: https://static1.squarespace.com/static/5b44f08ac258b493a25098a3/t/5fad7f650e8bbf646ff5c9cf/1605205862155/Amyloid+Treatment+mSMART+2020+revision+October+2020.pdf.
  17. Casserly LF, Fadia A, Sanchorawala V, Seldin DC, Wright DG, Skinner M, Dember LM. High-dose intravenous melphalan with autologous stem cell transplantation in AL amyloidosis-associated end-stage renal disease. Kidney Int. 2003;63(3):1051-1057.
    doi pubmed
  18. Szalat R, Sarosiek S, Havasi A, Brauneis D, Sloan JM, Sanchorawala V. Organ responses after highdose melphalan and stemcell transplantation in AL amyloidosis. Leukemia. 2021;35(3):916-919.
    doi pubmed
  19. Palladini G, Russo P, Nuvolone M, Lavatelli F, Perfetti V, Obici L, Merlini G. Treatment with oral melphalan plus dexamethasone produces long-term remissions in AL amyloidosis. Blood. 2007;110(2):787-788.
    doi pubmed
  20. Venner CP, Lane T, Foard D, Rannigan L, Gibbs SD, Pinney JH, Whelan CJ, et al. Cyclophosphamide, bortezomib, and dexamethasone therapy in AL amyloidosis is associated with high clonal response rates and prolonged progression-free survival. Blood. 2012;119(19):4387-4390.
    doi pubmed
  21. Mikhael JR, Schuster SR, Jimenez-Zepeda VH, Bello N, Spong J, Reeder CB, Stewart AK, et al. Cyclophosphamide-bortezomib-dexamethasone (CyBorD) produces rapid and complete hematologic response in patients with AL amyloidosis. Blood. 2012;119(19):4391-4394.
    doi pubmed
  22. Kastritis E, Leleu X, Arnulf B, Zamagni E, Cibeira MT, Kwok F, et al. A randomized phase III trial of melphalan and dexamethasone (MDex) versus bortezomib, melphalan and dexamethasone (BMDex) for untreated patients with AL amyloidosis. Blood. 2016;128(22):646.
    doi
  23. Kastritis E, Dialoupi I, Gavriatopoulou M, Roussou M, Kanellias N, Fotiou D, Ntanasis-Stathopoulos I, et al. Primary treatment of light-chain amyloidosis with bortezomib, lenalidomide, and dexamethasone. Blood Adv. 2019;3(20):3002-3009.
    doi pubmed
  24. Palladini G, Perfetti V, Perlini S, Obici L, Lavatelli F, Caccialanza R, Invernizzi R, et al. The combination of thalidomide and intermediate-dose dexamethasone is an effective but toxic treatment for patients with primary amyloidosis (AL). Blood. 2005;105(7):2949-2951.
    doi pubmed
  25. 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;109(2):457-464.
    doi pubmed
  26. Mahmood S, Venner CP, Sachchithanantham S, Lane T, Rannigan L, Foard D, Pinney JH, et al. Lenalidomide and dexamethasone for systemic AL amyloidosis following prior treatment with thalidomide or bortezomib regimens. Br J Haematol. 2014;166(6):842-848.
    doi pubmed
  27. Dispenzieri A, Lacy MQ, Zeldenrust SR, Hayman SR, Kumar SK, Geyer SM, Lust JA, et al. The activity of lenalidomide with or without dexamethasone in patients with primary systemic amyloidosis. Blood. 2007;109(2):465-470.
    doi pubmed
  28. Sanchorawala V, Wright DG, Rosenzweig M, Finn KT, Fennessey S, Zeldis JB, Skinner M, et al. Lenalidomide and dexamethasone in the treatment of AL amyloidosis: results of a phase 2 trial. Blood. 2007;109(2):492-496.
    doi pubmed
  29. Dispenzieri A, Buadi F, Laumann K, LaPlant B, Hayman SR, Kumar SK, Dingli D, et al. Activity of pomalidomide in patients with immunoglobulin light-chain amyloidosis. Blood. 2012;119(23):5397-5404.
    doi pubmed
  30. Palladini G, Milani P, Foli A, Basset M, Russo F, Perlini S, Merlini G. A phase 2 trial of pomalidomide and dexamethasone rescue treatment in patients with AL amyloidosis. Blood. 2017;129(15):2120-2123.
    doi pubmed
  31. Kaufman GP, Schrier SL, Lafayette RA, Arai S, Witteles RM, Liedtke M. Daratumumab yields rapid and deep hematologic responses in patients with heavily pretreated AL amyloidosis. Blood. 2017;130(7):900-902.
    doi pubmed
  32. clinicaltrials.gov. A study to evaluate the efficacy and safety of daratumumab in combination with cyclophosphamide, bortezomib, and dexamethasone (CyBorD) compared to CyBorD alone in newly diagnosed systemic amyloid light-chain (AL) amyloidosis. 2021. Available from: https://clinicaltrials.gov/ct2/show/NCT03201965.
  33. Palladini G, Kastritis E, Maurer MS, Zonder J, Minnema MC, Wechalekar AD, Jaccard A, et al. Daratumumab plus CyBorD for patients with newly diagnosed AL amyloidosis: safety run-in results of ANDROMEDA. Blood. 2020;136(1):71-80.
    doi pubmed
  34. Gertz MA, Cohen AD, Comenzo RL, Du Mond C, Kastritis E, Landau HJ, et al. Results of the phase 3 VITAL study of NEOD001 (Birtamimab) plus standard of care in patients with light chain (AL) amyloidosis suggest survival benefit for mayo stage IV patients. Blood. 2019;134(Supplement_1):3166.
    doi
  35. Hrncic R, Wall J, Wolfenbarger DA, Murphy CL, Schell M, Weiss DT, Solomon A. Antibody-mediated resolution of light chain-associated amyloid deposits. Am J Pathol. 2000;157(4):1239-1246.
    doi
  36. Solomon A, Weiss DT, Wall JS. Therapeutic potential of chimeric amyloid-reactive monoclonal antibody 11-1F4. Clin Cancer Res. 2003;9(10 Pt 2):3831S-3838S.
  37. Milani P, Schonland S, Merlini G, Kimmich C, Foli A, Dittrich T, Basset M, et al. Treatment of AL amyloidosis with bendamustine: a study of 122 patients. Blood. 2018;132(18):1988-1991.
    doi pubmed
  38. Wechalekar AD, Whelan C. Encouraging impact of doxycycline on early mortality in cardiac light chain (AL) amyloidosis. Blood Cancer J. 2017;7(3):e546.
    doi pubmed
  39. Mereles D, Wanker EE, Katus HA. Therapy effects of green tea in a patient with systemic light-chain amyloidosis. Clin Res Cardiol. 2008;97(5):341-344.
    doi pubmed
  40. Hunstein W. Epigallocathechin-3-gallate in AL amyloidosis: a new therapeutic option? Blood. 2007;110(6):2216.
    doi pubmed
  41. Wechalekar AD, Gillmore JD, Bird J, Cavenagh J, Hawkins S, Kazmi M, Lachmann HJ, et al. Guidelines on the management of AL amyloidosis. Br J Haematol. 2015;168(2):186-206.
    doi pubmed
  42. Sanchorawala V, Palladini G, Kukreti V, Zonder JA, Cohen AD, Seldin DC, Dispenzieri A, et al. A phase 1/2 study of the oral proteasome inhibitor ixazomib in relapsed or refractory AL amyloidosis. Blood. 2017;130(5):597-605.
    doi pubmed
  43. Kumar SK, Hayman SR, Buadi FK, Roy V, Lacy MQ, Gertz MA, Allred J, et al. Lenalidomide, cyclophosphamide, and dexamethasone (CRd) for light-chain amyloidosis: long-term results from a phase 2 trial. Blood. 2012;119(21):4860-4867.
    doi pubmed
  44. Parker LT, Rosenthal A, Sanchorawala V, Landau H, Compagnaro E, Kapoor P, et al. A phase II study of Isatuximab (SAR650984) (NSC-795145) for patients with previously treated AL amyloidosis (SWOG S1702; NCT#03499808). Blood. 2020;136(Supplement 1):20-21.
    doi
  45. Comenzo RL, Vosburgh E, Falk RH, Sanchorawala V, Reisinger J, Dubrey S, Dember LM, et al. Dose-intensive melphalan with blood stem-cell support for the treatment of AL (amyloid light-chain) amyloidosis: survival and responses in 25 patients. Blood. 1998;91(10):3662-3670.
    doi pubmed
  46. Jaccard A, Moreau P, Leblond V, Leleu X, Benboubker L, Hermine O, Recher C, et al. High-dose melphalan versus melphalan plus dexamethasone for AL amyloidosis. N Engl J Med. 2007;357(11):1083-1093.
    doi pubmed
  47. Jaccard A, Leblond V, Royer B, Leleu X, Delarue R, Kolb B, et al. Autologous stem cell transplantation (asct) versus oral melphalan and high-dose dexamethasone in patients with al (primary) amyloidosis: long term follow-up of the French Multicentric Randomized Trial. Blood. 2010;116(21):1344.
    doi
  48. Sidiqi MH, Aljama MA, Buadi FK, Warsame RM, Lacy MQ, Dispenzieri A, Dingli D, et al. Stem cell transplantation for light chain amyloidosis: decreased early mortality over time. J Clin Oncol. 2018;36(13):1323-1329.
    doi pubmed
  49. Sanchorawala V, Wright DG, Seldin DC, Falk RH, Finn KT, Dember LM, Berk JL, et al. High-dose intravenous melphalan and autologous stem cell transplantation as initial therapy or following two cycles of oral chemotherapy for the treatment of AL amyloidosis: results of a prospective randomized trial. Bone Marrow Transplant. 2004;33(4):381-388.
    doi pubmed
  50. Gupta VK, Brauneis D, Shelton AC, Quillen K, Sarosiek S, Sloan JM, Sanchorawala V. Induction therapy with bortezomib and dexamethasone and conditioning with high-dose melphalan and bortezomib followed by autologous stem cell transplantation for immunoglobulin light chain amyloidosis: long-term follow-up analysis. Biol Blood Marrow Transplant. 2019;25(5):e169-e173.
    doi pubmed
  51. Huang X, Wang Q, Chen W, Zeng C, Chen Z, Gong D, Zhang H, et al. Induction therapy with bortezomib and dexamethasone followed by autologous stem cell transplantation versus autologous stem cell transplantation alone in the treatment of renal AL amyloidosis: a randomized controlled trial. BMC Med. 2014;12:2.
    doi pubmed
  52. Landau H, Hassoun H, Rosenzweig MA, Maurer M, Liu J, Flombaum C, Bello C, et al. Bortezomib and dexamethasone consolidation following risk-adapted melphalan and stem cell transplantation for patients with newly diagnosed light-chain amyloidosis. Leukemia. 2013;27(4):823-828.
    doi pubmed
  53. Landau H, Lahoud O, Devlin S, Lendvai N, Chung DJ, Dogan A, Landgren CO, et al. Pilot study of bortezomib and dexamethasone pre- and post-risk-adapted autologous stem cell transplantation in AL amyloidosis. Biol Blood Marrow Transplant. 2020;26(1):204-208.
    doi pubmed
  54. Lonial S, Kaufman J, Tighiouart M, Nooka A, Langston AA, Heffner LT, Torre C, et al. A phase I/II trial combining high-dose melphalan and autologous transplant with bortezomib for multiple myeloma: a dose- and schedule-finding study. Clin Cancer Res. 2010;16(20):5079-5086.
    doi pubmed
  55. Mitsiades N, Mitsiades CS, Richardson PG, Poulaki V, Tai YT, Chauhan D, Fanourakis G, et al. The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood. 2003;101(6):2377-2380.
    doi pubmed
  56. Cornell RF, Fraser R, Costa L, Goodman S, Estrada-Merly N, Lee C, Hildebrandt G, et al. Bortezomib-based induction is associated with superior outcomes in light chain amyloidosis patients treated with autologous hematopoietic cell transplantation regardless of plasma cell burden. Transplant Cell Ther. 2021;27(3):264 e261-264 e267.
    doi pubmed
  57. Edwards CV, Sloan JM, Sanchorawala V. Modified high dose versus high dose melphalan conditioning in older patients undergoing autologous stem cell transplantation for immunoglobulin light chain amyloidosis. Transplant Cell Ther. 2021;27(3):S434-S435.
    doi
  58. Badros A, Barlogie B, Siegel E, Roberts J, Langmaid C, Zangari M, Desikan R, et al. Results of autologous stem cell transplant in multiple myeloma patients with renal failure. Br J Haematol. 2001;114(4):822-829.
    doi pubmed
  59. Al Saleh AS, Sidiqi MH, Sidana S, Muchtar E, Dispenzieri A, Dingli D, Lacy MQ, et al. Impact of consolidation therapy post autologous stem cell transplant in patients with light chain amyloidosis. Am J Hematol. 2019;94(10):1066-1071.
    doi pubmed
  60. Ozga M, Zhao Q, Benson D, Elder P, Williams N, Bumma N, Rosko A, et al. AL amyloidosis: the effect of maintenance therapy on autologous stem cell transplantation outcomes. J Clin Med. 2020;9(11):3778.
    doi pubmed
  61. Patel G, Hari P, Szabo A, Rein L, Kreuziger LB, Chhabra S, Dhakal B, et al. Acquired factor X deficiency in light-chain (AL) amyloidosis is rare and associated with advanced disease. Hematol Oncol Stem Cell Ther. 2019;12(1):10-14.
    doi pubmed
  62. Choufani EB, Sanchorawala V, Ernst T, Quillen K, Skinner M, Wright DG, Seldin DC. Acquired factor X deficiency in patients with amyloid light-chain amyloidosis: incidence, bleeding manifestations, and response to high-dose chemotherapy. Blood. 2001;97(6):1885-1887.
    doi pubmed
  63. Kyle RA, Greipp PR. Primary systemic amyloidosis: comparison of melphalan and prednisone versus placebo. Blood. 1978;52(4):818-827.
    doi
  64. Kyle RA, Gertz MA, Greipp PR, Witzig TE, Lust JA, Lacy MQ, Therneau TM. A trial of three regimens for primary amyloidosis: colchicine alone, melphalan and prednisone, and melphalan, prednisone, and colchicine. N Engl J Med. 1997;336(17):1202-1207.
    doi pubmed
  65. Facon T, Mary JY, Pegourie B, Attal M, Renaud M, Sadoun A, Voillat L, et al. Dexamethasone-based regimens versus melphalan-prednisone for elderly multiple myeloma patients ineligible for high-dose therapy. Blood. 2006;107(4):1292-1298.
    doi pubmed
  66. Gertz MA, Lacy MQ, Lust JA, Greipp PR, Witzig TE, Kyle RA. Prospective randomized trial of melphalan and prednisone versus vincristine, carmustine, melphalan, cyclophosphamide, and prednisone in the treatment of primary systemic amyloidosis. J Clin Oncol. 1999;17(1):262-267.
    doi pubmed
  67. Tanaka K. The proteasome: overview of structure and functions. Proc Jpn Acad Ser B Phys Biol Sci. 2009;85(1):12-36.
    doi pubmed
  68. Kane RC, Bross PF, Farrell AT, Pazdur R. Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist. 2003;8(6):508-513.
    doi pubmed
  69. Palladini G, Sachchithanantham S, Milani P, Gillmore J, Foli A, Lachmann H, Basset M, et al. A European collaborative study of cyclophosphamide, bortezomib, and dexamethasone in upfront treatment of systemic AL amyloidosis. Blood. 2015;126(5):612-615.
    doi pubmed
  70. Kumar S, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK, Colby C, Laumann K, 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.
    doi pubmed
  71. Palladini G, Milani P, Merlini G. Predicting survival in light chain amyloidosis. Haematologica. 2019;104(7):1294-1296.
    doi pubmed
  72. Palladini G, Milani P, Foli A, Vidus Rosin M, Basset M, Lavatelli F, Nuvolone M, et al. Melphalan and dexamethasone with or without bortezomib in newly diagnosed AL amyloidosis: a matched case-control study on 174 patients. Leukemia. 2014;28(12):2311-2316.
    doi pubmed
  73. Sperry BW, Ikram A, Hachamovitch R, Valent J, Vranian MN, Phelan D, Hanna M. Efficacy of chemotherapy for light-chain amyloidosis in patients presenting with symptomatic heart failure. J Am Coll Cardiol. 2016;67(25):2941-2948.
    doi pubmed
  74. Bochtler T, Hegenbart U, Kunz C, Granzow M, Benner A, Seckinger A, Kimmich C, et al. Translocation t(11;14) is associated with adverse outcome in patients with newly diagnosed AL amyloidosis when treated with bortezomib-based regimens. J Clin Oncol. 2015;33(12):1371-1378.
    doi pubmed
  75. Meregalli C. An Overview of Bortezomib-Induced Neurotoxicity. Toxics. 2015;3(3):294-303.
    doi pubmed
  76. Velasco R, Alberti P, Bruna J, Psimaras D, Argyriou AA. Bortezomib and other proteosome inhibitors-induced peripheral neurotoxicity: from pathogenesis to treatment. J Peripher Nerv Syst. 2019;24(Suppl 2):S52-S62.
    doi
  77. Parlati F, Lee SJ, Aujay M, Suzuki E, Levitsky K, Lorens JB, Micklem DR, et al. Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome. Blood. 2009;114(16):3439-3447.
    doi pubmed
  78. Dimopoulos MA, Moreau P, Palumbo A, Joshua D, Pour L, Hajek R, Facon T, et al. Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study. Lancet Oncol. 2016;17(1):27-38.
    doi
  79. Cohen AD, Scott EC, Liedtke M, Kaufman JL, Landau H, Vesole DH, et al. A phase I dose-escalation study of carfilzomib in patients with previously-treated systemic light-chain (AL) amyloidosis. Blood. 2014;124(21):4741.
    doi
  80. Manwani R, Mahmood S, Sachchithanantham S, Lachmann HJ, Gillmore JD, Yong K, Rabin N, et al. Carfilzomib is an effective upfront treatment in AL amyloidosis patients with peripheral and autonomic neuropathy. Br J Haematol. 2019;187(5):638-641.
    doi pubmed
  81. Kumar SK, Jacobus SJ, Cohen AD, Weiss M, Callander N, Singh AK, Parker TL, et al. Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2020;21(10):1317-1330.
    doi
  82. Shirley M. Ixazomib: First Global Approval. Drugs. 2016;76(3):405-411.
    doi pubmed
  83. Clinicaltrials.gov. Study of dexamethasone plus ixazomib (mln9708) or physicians choice of treatment in relapsed or refractory systemic light chain (al) amyloidosis. 2020. Available from: https://clinicaltrials.gov/ct2/show/NCT01659658.
  84. Cook J, Muchtar E, Warsame R. Updates in the diagnosis and management of AL amyloidosis. Curr Hematol Malig Rep. 2020;15(3):155-167.
    doi pubmed
  85. Quach H, Ritchie D, Stewart AK, Neeson P, Harrison S, Smyth MJ, Prince HM. Mechanism of action of immunomodulatory drugs (IMiDS) in multiple myeloma. Leukemia. 2010;24(1):22-32.
    doi pubmed
  86. Vaxman I, Dispenzieri A, Muchtar E, Gertz M. New developments in diagnosis, risk assessment and management in systemic amyloidosis. Blood Rev. 2020;40:100636.
    doi pubmed
  87. Vincent Rajkumar S. Thalidomide: tragic past and promising future. Mayo Clin Proc. 2004;79(7):899-903.
  88. Singhal S, Mehta J, Desikan R, Ayers D, Roberson P, Eddlemon P, et al. Erratum: Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med. 2000;342(5):364.
  89. Venner CP, Gillmore JD, Sachchithanantham S, Mahmood S, Lane T, Foard D, Rannigan L, et al. A matched comparison of cyclophosphamide, bortezomib and dexamethasone (CVD) versus risk-adapted cyclophosphamide, thalidomide and dexamethasone (CTD) in AL amyloidosis. Leukemia. 2014;28(12):2304-2310.
    doi pubmed
  90. Palladini G, Russo P, Foli A, Milani P, Lavatelli F, Obici L, Nuvolone M, et al. Salvage therapy with lenalidomide and dexamethasone in patients with advanced AL amyloidosis refractory to melphalan, bortezomib, and thalidomide. Ann Hematol. 2012;91(1):89-92.
    doi pubmed
  91. Hegenbart U, Bochtler T, Benner A, Becker N, Kimmich C, Kristen AV, Beimler J, et al. Lenalidomide/melphalan/dexamethasone in newly diagnosed patients with immunoglobulin light chain amyloidosis: results of a prospective phase 2 study with long-term follow up. Haematologica. 2017;102(8):1424-1431.
    doi pubmed
  92. Cibeira MT, Oriol A, Lahuerta JJ, Mateos MV, de la Rubia J, Hernandez MT, Granell M, et al. A phase II trial of lenalidomide, dexamethasone and cyclophosphamide for newly diagnosed patients with systemic immunoglobulin light chain amyloidosis. Br J Haematol. 2015;170(6):804-813.
    doi pubmed
  93. Raedler LA. Pomalyst (Pomalidomide) received a new indication for patients with relapsed and/or refractory multiple myeloma. Am Health Drug Benefits. 2016;9(Spec Feature):111-114.
  94. Sharpley FA, Manwani R, Mahmood S, Sachchithanantham S, Lachmann H, Gilmore J, Whelan C, et al. Real world outcomes of pomalidomide for treatment of relapsed light chain amyloidosis. Br J Haematol. 2018;183(4):557-563.
    doi pubmed
  95. Milani P, Sharpley F, Schonland SO, Basset M, Mahmood S, Nuvolone M, Kimmich C, et al. Pomalidomide and dexamethasone grant rapid haematologic responses in patients with relapsed and refractory AL amyloidosis: a European retrospective series of 153 patients. Amyloid. 2020;27(4):231-236.
    doi pubmed
  96. Dispenzieri A, Dingli D, Kumar SK, Rajkumar SV, Lacy MQ, Hayman S, Buadi F, 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;85(10):757-759.
    doi pubmed
  97. Qualls DA, Lewis GD, Sanchorawala V, Staron A. Orthotopic heart transplant rejection in association with immunomodulatory therapy for AL amyloidosis: A case series and review of the literature. Am J Transplant. 2019;19(11):3185-3190.
    doi pubmed
  98. Roccatello D, Fenoglio R, Sciascia S, Naretto C, Rossi D, Ferro M, Barreca A, et al. CD38 and anti-CD38 monoclonal antibodies in AL amyloidosis: targeting plasma cells and beyond. Int J Mol Sci. 2020;21(11):4129.
    doi pubmed
  99. Kriegsmann K, Dittrich T, Neuber B, Awwad MHS, Hegenbart U, Goldschmidt H, Hillengass J, et al. Quantification of number of CD38 sites on bone marrow plasma cells in patients with light chain amyloidosis and smoldering multiple myeloma. Cytometry B Clin Cytom. 2018;94(5):611-620.
    doi pubmed
  100. Bhatnagar V, Gormley NJ, Luo L, Shen YL, Sridhara R, Subramaniam S, Shen G, et al. FDA approval summary: daratumumab for treatment of multiple myeloma after one prior therapy. Oncologist. 2017;22(11):1347-1353.
    doi pubmed
  101. Sher T, Fenton B, Akhtar A, Gertz MA. First report of safety and efficacy of daratumumab in 2 cases of advanced immunoglobulin light chain amyloidosis. Blood. 2016;128(15):1987-1989.
    doi pubmed
  102. Roussel M, Merlini G, Chevret S, Arnulf B, Stoppa AM, Perrot A, Palladini G, et al. A prospective phase 2 trial of daratumumab in patients with previously treated systemic light-chain amyloidosis. Blood. 2020;135(18):1531-1540.
    doi pubmed
  103. Wall JS, Kennel SJ, Williams A, Richey T, Stuckey A, Huang Y, Macy S, et al. AL amyloid imaging and therapy with a monoclonal antibody to a cryptic epitope on amyloid fibrils. PLoS One. 2012;7(12):e52686.
    doi pubmed
  104. Gertz MA, Landau H, Comenzo RL, Seldin D, Weiss B, Zonder J, Merlini G, et al. First-in-Human Phase I/II Study of NEOD001 in Patients With Light Chain Amyloidosis and Persistent Organ Dysfunction. J Clin Oncol. 2016;34(10):1097-1103.
    doi pubmed
  105. Edwards CV, Gould J, Langer AL, Mapara M, Radhakrishnan J, Maurer MS, Raza S, et al. Interim analysis of the phase 1a/b study of chimeric fibril-reactive monoclonal antibody 11-1F4 in patients with AL amyloidosis. Amyloid. 2017;24(sup1):58-59.
    doi pubmed
  106. Cheson BD, Leoni L. Bendamustine: mechanism of action and clinical data. Clin Adv Hematol Oncol. 2011;9(8 Suppl 19):1-11.
  107. Dennie TW, Kolesar JM. Bendamustine for the treatment of chronic lymphocytic leukemia and rituximab-refractory, indolent B-cell non-Hodgkin lymphoma. Clin Ther. 2009;31(Pt 2):2290-2311.
    doi pubmed
  108. Lentzsch S, Lagos GG, Comenzo RL, Zonder JA, Osman K, Pan S, Bhutani D, et al. Bendamustine With Dexamethasone in Relapsed/Refractory Systemic Light-Chain Amyloidosis: Results of a Phase II Study. J Clin Oncol. 2020;38(13):1455-1462.
    doi pubmed
  109. Montagna G, Cazzulani B, Obici L, Uggetti C, Giorgetti S, Porcari R, Ruggiero R, et al. Benefit of doxycycline treatment on articular disability caused by dialysis related amyloidosis. Amyloid. 2013;20(3):173-178.
    doi pubmed
  110. Ward JE, Ren R, Toraldo G, Soohoo P, Guan J, O'Hara C, Jasuja R, et al. Doxycycline reduces fibril formation in a transgenic mouse model of AL amyloidosis. Blood. 2011;118(25):6610-6617.
    doi pubmed
  111. Kumar SK, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK, Dingli D, et al. Doxycycline used as post transplant antibacterial prophylaxis improves survival in patients with light chain amyloidosis undergoing autologous stem cell transplantation. Blood. 2012;120(21):3138.
    doi
  112. D'Souza A, Szabo A, Flynn KE, Dhakal B, Chhabra S, Pasquini MC, Weihrauch D, et al. Adjuvant doxycycline to enhance anti-amyloid effects: Results from the dual phase 2 trial. EClinicalMedicine. 2020;23:100361.
    doi pubmed
  113. Leung N, Thome SD, Dispenzieri A. Venetoclax induced a complete response in a patient with immunoglobulin light chain amyloidosis plateaued on cyclophosphamide, bortezomib and dexamethasone. Haematologica. 2018;103(3):e135-e137.
    doi pubmed
  114. Premkumar V, Comenzo R, Lentzsch S. Venetoclax in Immunoglobulin Light Chain Amyloidosis: Is This the Beginning or the End? Clin Lymphoma Myeloma Leuk. 2019;19(10):686-688.
    doi pubmed
  115. Sidiqi MH, Al Saleh AS, Leung N, Jevremovic D, Aljama MA, Gonsalves WI, Buadi FK, et al. Venetoclax for the treatment of translocation (11;14) AL amyloidosis. Blood Cancer J. 2020;10(5):55.
    doi pubmed


This article is distributed under the terms of the Creative Commons Attribution Non-Commercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.


Journal of Hematology is published by Elmer Press Inc.

 

Browse  Journals  

     

Journal of Clinical Medicine Research

Journal of Endocrinology and Metabolism

Journal of Clinical Gynecology and Obstetrics

World Journal of Oncology

Gastroenterology Research

Journal of Hematology

Journal of Medical Cases

Journal of Current Surgery

Clinical Infection and Immunity

Cardiology Research

World Journal of Nephrology and Urology

Cellular and Molecular Medicine Research

Journal of Neurology Research

International Journal of Clinical Pediatrics

 

 

 

 

 

Journal of Hematology, bimonthly, ISSN 1927-1212 (print), 1927-1220 (online), published by Elmer Press Inc.                            
The content of this site is intended for health care professionals.
This is an open-access journal distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License, which permits unrestricted
non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Creative Commons Attribution license (Attribution-NonCommercial 4.0 International CC BY-NC 4.0)



This journal follows the International Committee of Medical Journal Editors (ICMJE) recommendations for manuscripts submitted to biomedical journals,
the Committee on Publication Ethics (COPE) guidelines, and the Principles of Transparency and Best Practice in Scholarly Publishing.

website: www.thejh.org    editorial contact: editor@thejh.org
Address: 9225 Leslie Street, Suite 201, Richmond Hill, Ontario, L4B 3H6, Canada
 

© Elmer Press Inc. All Rights Reserved.


Disclaimer: The views and opinions expressed in the published articles are those of the authors and do not necessarily reflect the views or opinions of the editors and Elmer Press Inc. This website is provided for medical research and informational purposes only and does not constitute any medical advice or professional services. The information provided in this journal should not be used for diagnosis and treatment, those seeking medical advice should always consult with a licensed physician.