A Review of Autologous Stem Cell Transplantation in Amyloidosis

Article

Iuliana Vaxman, MD, and Angela Dispenzieri, MD, review eligibility criteria for ASCT in AL amyloidosis, conditioning dosing, efficacy in terms of hematologic and organ response, and future areas of research.

This content originally appeared on our sister site, Cancer Network.

Abstract

Autologous stem cell transplantation (ASCT) has been an essential part of the treatment armamentarium in light chain (AL) amyloidosis for several decades. Patients who achieve a complete hematologic response following ASCT have a long overall survival. However, only 1 randomized controlled trial compared ASCT with the standard of care used at the time, which was melphalan and dexamethasone, and the results did not support the use of ASCT in AL amyloidosis. These results are of limited significance due to the unexpected high transplant-related mortality (TRM) (24%). TRM is a major concern in AL amyloidosis, but its incidence can be lessened by better patient selection and by patients receiving ASCT in specialized centers. ASCT in AL amyloidosis is performed only in selected patients; approximately 20% of patients with AL amyloidosis are transplant eligible up front or after bortezomib (Velcade) based conditioning. The introduction of newer agents such as bortezomib and daratumumab (Darzalex), which lead to deep responses and have good safety profiles, encourage revisiting the benefit and timing of ASCT in the modern era.

This review provides a comprehensive assessment of eligibility criteria for ASCT in AL amyloidosis, conditioning dosing, efficacy in terms of hematologic and organ response, and future areas of research.


Introduction

In immunoglobulin light chain (AL) amyloidosis, clonal plasma cells in the bone marrow produce light chains that misfold and deposit in various organs.1 These depositions interfere with organs’ function,2 which reduces quality of life (QOL) and shortens survival. The treatment of AL amyloidosis is aimed at quickly clearing the amyloidogenic light chain burden by eliminating the plasma cell clone that produces it.2

The key concepts of effectively treating AL amyloidosis are (1) early diagnosis; (2) accurate typing; (3) a multidisciplinary approach to management; and (4) tailored treatment to tolerance (choosing the safest therapy based on organ involvement and comorbidities). The extent of cardiac involvement drives therapy and outcomes, because patients with advanced cardiac involvement are at increased risk of early death from arrhythmias and progressive heart failure.3 The goal of treatment in AL amyloidosis is for the patient to achieve a response as deep as possible, as quickly as possible.4,54,5

ASCT has been an essential part of the treatment armamentarium in light chain (AL) amyloidosis for several decades because of high response rates and prolonged responses. However, in the past 2 decades, treatment options for AL amyloidosis patients have expanded to include proteosome inhibitor6-10 immunomodulatory drugs,11-15 alkylators,8,10,16 and monoclonal antibodies.17-20 Currently, more than 50% of patients with AL amyloidosis achieve a very good partial response or better (≥VGPR)21-24 to frontline treatments, making the role and the optimal timing of ASCT an area of debate. Moreover, the impact of ASCT on high-risk cytogenetics is unclear.25
6-10 immunomodulatory drugs,11-15 alkylators,8,10,16 and monoclonal antibodies.17-20 Currently, more than 50% of patients with AL amyloidosis achieve a very good partial response or better (≥VGPR)21-24 to frontline treatments, making the role and the optimal timing of ASCT an area of debate. Moreover, the impact of ASCT on high-risk cytogenetics is unclear.25

Induction prior to ASCT

The role of induction therapy prior to ASCT is evolving. Historically, the norm had been no induction prior to ASCT based on 2 justifications. First, a majority of patients with AL amyloidosis had low plasma cell burden, making “debulking” prior to ASCT seemingly unnecessary. Second, induction could render patients who were previously eligible for ASCT ineligible, due to toxicity. Prospective data are quite limited.

In a small trial from China, 56 patients were randomized to receive either 2 cycles of bortezomib and dexamethasone (BD) prior to ASCT, or to move directly to ASCT (Table).26 No attrition occurred in the BD arm prior to ASCT, and 2-year progression-free survival (PFS) was 81% and 50%, respectively, favoring BD induction. Two-year overall survival (OS) was 95% and 69%, respectively. Although the anticipated complete response (CR) rate (defined as normalization of the free light chain level and ratio, negative serum and urine immunofixation) for the noninduced patients was of no surprise, the 2-year OS was much lower than expected, because, based on results of previous larger studies, the anticipated 5-year OS post ASCT is 65% to 75%.26 No attrition occurred in the BD arm prior to ASCT, and 2-year progression-free survival (PFS) was 81% and 50%, respectively, favoring BD induction. Two-year overall survival (OS) was 95% and 69%, respectively. Although the anticipated complete response (CR) rate (defined as normalization of the free light chain level and ratio, negative serum and urine immunofixation) for the noninduced patients was of no surprise, the 2-year OS was much lower than expected, because, based on results of previous larger studies, the anticipated 5-year OS post ASCT is 65% to 75%.

TABLE. Prospective Studies Reporting Induction Regimens Prior to ASCT in AL Amyloidosis

TABLE. Prospective Studies Reporting Induction Regimens Prior to ASCT in AL Amyloidosis

In contrast, the prospective phase 2 study by Minnema et al included 50 patients who were treated with 4 cycles of BD before ASCT.24 The overall hematologic response after induction treatment was 80% (CR, 20%; VGPR, 38%). Fifteen patients (30%) did not proceed to ASCT, mainly due to toxicity induced by the BD treatment; therefore, the primary end point of the study was not met. Treatment-related mortality (TRM) was 0%. In the patients who proceeded to ASCT, overall hematologic responses improved at 6 months after transplant to 86% (CR, 46%; VGPR, 26%).

Two other prospective studies have incorporated BD prior to ASCT; both were single-arm trials27,28 and used bortezomib along with melphalan conditioning, and one of them used bortezomib as consolidation as shown in the Table. Each of these strategies yielded higher-than-expected CR rates, but at the cost of patient attrition prior to ASCT. Moreover, the role of induction cannot be separated from the novel conditioning27,28 and consolidation.27 A similar pattern of patient attrition that occurred when induction was given prior to ASCT was seen in a prospective study published in 2004 that randomized 100 patients newly diagnosed with AL amyloidosis to ASCT up front vs 2 cycles of induction with melphalan and prednisone prior to ASCT. There were no differences in OS, hematologic response, or organ response between the 2 arms, but 16 patients randomized to the induction arm became ineligible for subsequent ASCT.29

A number of retrospective studies suggest that induction, especially bortezomib induction, yields better outcomes. A major problem with these retrospective studies is that they are not designed as intention-to-treat trials, which makes it impossible to know if sicker patients were weeded out and never made it to ASCT; this would leave the noninduction comparator group as a population of higher-risk patients more likely to have higher TRM, which in turn reduces both survival and evaluability for response.

One such retrospective study evaluated 128 patients with AL amyloidosis who were divided into 3 groups: no induction (15.5%), induction with melphalan and prednisone (MP; 19.5%), and induction with novel agents (65%). At day 100 post ASCT, overall hematologic response rates (and CR rates) were 60% (10%), 62% (25%), and 87% (19%) in the no-induction group, MP group, and novel-agents group, respectively.30 Induction with novel agents was predictive of OS in a multivariate analysis. Most of the OS benefit in the novel-agents group occurred in the first 12 months, suggesting a potential weeding-out of sicker patients in the induction group.

Another retrospective study reported outcomes of 63 patients: 34 received bortezomib-based therapy before ASCT, and 29 patients either did not receive induction prior to ASCT or received induction therapy that was not bortezomib based.31 At diagnosis, the bortezomib-treated patients had been more likely to have cardiac involvement and gastrointestinal involvement. Time to transplant was longer in the bortezomib-treated group (median, 226 days vs 150 days). Inexplicably, time from diagnosis to ASCT in the nonbortezomib group was quite long at 5 months. The patients who received bortezomib induction were more likely to receive melphalan 200 mg/m2 (62% vs 45%, respectively), and in these patients, there was a trend toward better CR at 3 months (26% vs 12% respectively). The median OS and the organ responses were also higher in the bortezomib induction group. By 24 months, approximately one-third of the nonbortezomib patients had died in contrast to 6% of the bortezomib-induced group.

The thought process around tumor bulk (≤10% bone marrow plasma cells [BMPC] at presentation) was in part supported by a retrospective publication from the Mayo Clinic in which patients with Mayo 2012 stage I/II disease and BMPC ≤10% did well (5-year OS above 80%), regardless of whether or not they received induction therapy prior to ASCT.32 These findings have been challenged by recent studies. In a retrospective study of 227 patients with <10% BMPC, 124 patients received bortezomib-based induction prior to ASCT, 68 did not receive induction, and 35 patients received other chemotherapeutic induction.33 OS, PFS, overall hematologic response rates, CR rates, and cardiac and renal responses were all higher in patients who received bortezomib-based induction prior to ASCT. Once again, the 2-year mortality was markedly different between the bortezomib induction group vs the patients who received no induction or nonbortezomib induction: 0% vs almost 25%, respectively. On multivariate analysis, only the difference between the involved and uninvolved serum free light chain (dFLC) >50 mg/l and bortezomib induction predicted for OS. Another retrospective study of 45 patients that evaluated induction regimens with novel agents showed a 3-year PFS benefit for patients treated with novel-agent induction vs conventional chemotherapy (79% vs 53%, respectively; P = .05), but no significant OS benefit (95% vs 71%).34

To conclude, most studies support the use of bortezomib-based induction therapy prior to ASCT, even for patients with low disease burden (Figure).

FIGURE. The mSMART Approach to Newly Diagnosed, Transplant-eligible Patients With AL Amyloidosis

FIGURE. The mSMART Approach to Newly Diagnosed, Transplant-eligible Patients With AL Amyloidosis

Which bortezomib-based induction regimen should be used?

Deep responses have been reported with daratumumab therapy in AL amyloidosis; daratumumab is the first FDA approved drug for AL amyloidosis based on the phase 3 ANDROMEDA trial (NCT03201965) that compared daratumumab plus bortezomib, cyclophosphamide, and dexamethasone (VCd) with VCd alone in newly diagnosed patients.17,35 The addition of daratumumab to VCd resulted in a significant improvement in the overall hematologic response (92% vs 77%, respectively), as well as a significant improvement in the rate of complete hematologic response (50% vs 14%). Although ASCT was not a planned part of the trial, 13 patients in the daratumumab + VCd arm and 20 patients in the VCd-alone arm proceeded to ASCT. In the safety run-in report (only of the daratumumab + VCd arm), 5 patients died, including 3 who underwent ASCT.17 The quadruplet resulted in shorter time to response and improved organ response. Because of the deep responses to daratumumab + VCd,35 we recommend using the quadruplet as first-line treatment, if possible (Figure).

If daratumumab/VCd is not available, we recommend using VCd prior to ASCT. A prospective study investigated VCd in 230 newly diagnosed with AL amyloidosis; the overall hematological response rate was 60%, with a VGPR or better in 43%.8 Only 17 patients underwent ASCT, all as part of second-line therapy. The first report to describe the use of VCd in AL amyloidosis was a retrospective study that evaluated VCd as used in 17 patients with AL amyloidosis (bortezomib was given at 1.5 mg/m2 once weekly).36 The overall hematological response rate was 94%, with a CR rate of 71%. The median time to response was 2 months. Five of those patients proceeded to ASCT; 3 of them had initially been considered transplant ineligible and became eligible only after therapy.8 Only 17 patients underwent ASCT, all as part of second-line therapy. The first report to describe the use of VCd in AL amyloidosis was a retrospective study that evaluated VCd as used in 17 patients with AL amyloidosis (bortezomib was given at 1.5 mg/m2 once weekly).36 The overall hematological response rate was 94%, with a CR rate of 71%. The median time to response was 2 months. Five of those patients proceeded to ASCT; 3 of them had initially been considered transplant ineligible and became eligible only after therapy.

It is well recognized that, in general, depth of response is prognostic in AL amyloidosis,1 but a retrospective study has demonstrated that response to induction pre-ASCT is also predictive of OS.36 Bortezomib, lenalidomide, and dexamethasone (VRd) is commonly used in patients with multiple myeloma, given the higher response rate they experience compared with lenalidomide and dexamethasone; however, generally, patients with AL amyloidosis have a lower tolerance of lenalidomide, with lenalidomide not infrequently causing hypotension and cardiac decompensation in more fragile patients with AL amyloidosis. We consider using VRd only in patients who do not have cardiac involvement or autonomic dysfunction. Bortezomib with dexamethasone is also an induction option.25,30

Eligibility criteria for ASCT

Organ compromise can lead to significant complications and early mortality, mandating careful patient selection. Carefully selecting who can or cannot receive ASCT heavily influences patients’ outcomes. Selection criteria are not uniform among the specialized centers, but generally, only 20% to 30% of patients with AL amyloidosis are deemed eligible for ASCT.

The early reports of ASCT use in patients with AL amyloidosis elicited important safety issues, with TRM ranging from 13% to 43%.37,38 To lower TRM, clinicians developed risk-adapted approaches that included patient-related factors, such as performance status, and disease-related factors, most importantly the extent of cardiac involvement. Some centers use a more liberal approach to patient selection and use attenuated doses of melphalan conditioning, while others have more strict eligibility criteria aiming to use full-dose melphalan.

At Mayo Clinic, the current consensus criteria for ASCT eligibility of patients with AL amyloidosis include physiologic; age 70 years or younger (determined clinically); performance score of 2 or less; systolic blood pressure 90 mm Hg or greater; troponin T (TnT) lower than 0.06 ng/ml (or high sensitivity -TnT <75 ng/ml); creatinine clearance (CrCl) 30 ml/min or greater (unless on chronic dialysis); and categorized as New York Heart Association Class I/II. Elevated cardiac biomarkers are associated with high rates of early mortality39 and therefore are included in our eligibility criteria. Some centers use less stringent selection criteria for ASCT, ie, they do not include cardiac biomarkers as a tool to exclude patients from ASCT. At Mayo Clinic, dialysis is not considered to be an exclusion criterion if all other eligibility criteria are met.4039 and therefore are included in our eligibility criteria. Some centers use less stringent selection criteria for ASCT, ie, they do not include cardiac biomarkers as a tool to exclude patients from ASCT. At Mayo Clinic, dialysis is not considered to be an exclusion criterion if all other eligibility criteria are met.40

ASCT is safe for selected patients with AL amyloidosis 70 years or older as long as they meet the other criteria listed above. In a retrospective report of 34 patients, TRM was 3% (1 patient died) and 35% completed the entire transplant course outpatient.4141

Stem cell mobilization and collection

Mobilization and collection may also jeopardize patients with AL amyloidosis. In one report of 629 patients, 11 patients died during mobilization and collection42; a retrospective study that included 101 patients reported 4 deaths during mobilization.43 Other adverse effects of stem cell mobilization and collection can include volume overload, thromboembolic events, and bleeding. The susceptible population is mainly patients with cardiac involvement. Plerixafor used to enhance stem cell yield is safe and well tolerated in AL amyloidosis.42; a retrospective study that included 101 patients reported 4 deaths during mobilization.43 Other adverse effects of stem cell mobilization and collection can include volume overload, thromboembolic events, and bleeding. The susceptible population is mainly patients with cardiac involvement. Plerixafor used to enhance stem cell yield is safe and well tolerated in AL amyloidosis.44,45

Conditioning chemotherapy

Conditioning in amyloidosis can be performed with full-intensity melphalan (200 mg/m2) or with reduced melphalan doses (100-140 mg/m2). Dose-reduced melphalan has made ASCT possible for sicker AL amyloidosis patients without excessive TRM. Lower doses of melphalan, however, result in lower CR rates, which translate into lower PFS and OS rates. Given the option of novel agent based therapies6,17 that give CR rates higher than those generally attained with attenuated dose melphalan conditioning, it is our practice at the Mayo Clinic to carefully select patients to whom we can administer full-dose melphalan. Therefore, for patients that are assessed as unable to receive full-dose melphalan, we choose to offer other novel agents based therapies.

The concept of using attenuated melphalan conditioning to transplant sicker patients was introduced at Boston University.46 Although TRM was shown to be reduced, it has since been repeatedly demonstrated that response rate is sacrificed with this maneuver. In a report of 629 patients transplanted at Boston University, hematologic CR was higher in patients receiving 200 mg/m2 than in those receiving 100 to 140 mg/m2 (45% vs 34%, respectively; P = .0091).42 Fifty-six percent of the cohort received melphalan 200 mg/m2, whereas 44% received melphalan 100 to 140 mg/m2; the latter group included patient 65 years or older and those with poor performance status, left ventricular ejection fraction 40% to 45%, or low yield stem cell collection. The reported overall TRM was 7.5% (3.4% after 2005). Hematologic CR rates were higher in patients that received melphalan 200 mg/m2 (45% versus 34%, respectively; P = .0091). The median OS was higher for patients treated with 200 mg/m2 (10.5 vs 5.2 years, respectively; P = .0001). Similar results have been shown at the large amyloidosis transplant centers,47,48 including the Mayo Clinic and Memorial Sloan Kettering. A Center for International Blood and Marrow Transplant Research report, which included 1536 patients with AL amyloidosis undergoing ASCT, demonstrated similar findings; it also showed lower relapse risk with full-dose melphalan (HR, 0.237; 95% CI, 0.118-0.474; P = .001).49

In an effort to include sicker patients as ASCT candidates, but to compensate for lower CR rates, an approach has been to use attenuated melphalan doses followed by consolidative chemotherapy post ASCT for those not achieving a deep response50 (see section below on consolidation and maintenance). In a report of 334 patients who received modified conditioning, the overall hematologic response was 69%, with TRM of 3% after 2010. The median OS was 6.1 years for the entire study population and 13.4 years for the subgroup who achieved hematologic CR.51

As shown in the Table, bortezomib has also been evaluated as part of the conditioning regimen along with high-dose melphalan. Given the small sample size, there is no way to properly judge whether there is any benefit to this approach.32,40

ASCT complications

TRM has declined over the years, due to the refinement of eligibility criteria and
better supportive care.48,49 Specialized centers have superior survival outcomes,49 with TRM less than 5%.48 However, ASCT is associated with many other important potential complications, mainly renal impairment,52,53 fluid accumulation, cardiac arrhythmia, gastrointestinal toxicity,54 and infections.

Progressive renal impairment occurring during ASCT can increase TRM, have a crucial impact on the future choice of therapies, and negatively influence QOL. In a retrospective report of 655 patients,52 a subgroup of 87 patients had an estimated glomerular filtration rate (eGFR) of less than 45, and of this subgroup, 16% required dialysis within 100 days of ASCT. Among patients with eGFR of ≥45, 6% required dialysis within 100 days of ASCT. In another retrospective report of 173 patients with AL
amyloidosis who underwent ASCT, 21% developed acute kidney injury (AKI) during ASCT and 5% required dialysis. The risk factors for AKI were low creatinine clearance, high urinary protein, cardiac involvement, and bacteremia during the ASCT course.53 Therefore, at Mayo, we generally refrain from transplanting patients with CrCl ≤30 ml/min (unless they are on chronic dialysis).

Gastrointestinal toxicity may include diarrhea that can increase the length of hospitalization, worsen preexisting hypoalbuminemia, and result in higher use of antibiotics. Age and impaired kidney function are risk factors for severe
diarrhea.54 Cardiac decompensation may result from excessive fluid administration.

Timing of ASCT

The timing of transplant, upfront or at relapse, remains a debatable issue, with limited published data and no prospective trials. Some centers use ASCT only in patients who fail to achieve deep responses to induction,55 while other centers transplant all eligible patients up front. Patients who are not transplant eligible at diagnosis may become eligible if they attain organ response after up-front therapy.

In an attempt to address “intention to treat” in the context of “early and delayed transplant”, all 651 patients with AL amyloidosis whose stem cells were collected at the Mayo Clinic between 2004 and 2018 were included in a retrospective analysis. Patients were divided into 2 groups: those transplanted early (ASCT within 90 days of stem cell collection) and those who had ASCT later. There was no OS difference between early and delayed ASCT.56 In the subgroup that achieved VGPR or better prior to stem cell collection, there was a trend towards superior OS in the early transplant cohort, but the difference was minor (median OS, 14 vs 13 years, respectively). Similar results were reported in a retrospective United Kingdom (UK) study.57

The Amyloidosis Group from Pavia, Italy, has proposed a sequential response-driven approach. The authors reported the outcomes of 139 newly diagnosed patients who were treated with VCd induction. Only those patients who achieved less than a VGPR were given an ASCT.55 The hematologic response to VCd was 68%, with 51% achieving ≥VGPR. Forty percent of the cohort underwent ASCT, and their overall hematologic response rate was 80%, with 65% achieving VGPR or better. There was no OS difference (5-year OS, 86% vs 84%, respectively) nor a significant difference in the duration of response between the 2 groups (60 vs 49 months). Of note, 21 patients (15%) did not achieve VGPR or better but did not undergo ASCT because of refusal or ineligibility. With rescue chemotherapy, they had a 5-year OS of 51%. An interesting UK study reported the outcomes of 22 AL amyloidosis patients who were initially transplant ineligible but became transplant eligible after bortezomib-based therapy. There was no TRM, and 77% achieved VGPR or better and 86% achieved cardiac response.58

What is the role of ASCT if patient achieves a CR with induction?

As induction is used more and more, some patients come to stem cell collection in complete response—fewer, but some, are even minimal residual disease
(MRD) negative.59-61 With VCd, nearly 20% of patients may achieve CR; in
contrast, more than 50% of patients receiving daratumumab + VCd achieve CR. No good guidance exists regarding what to do in these instances. Does one collect and store stem cells? Does one consolidate with transplant? It is known that post ASCT, 34% to 40% of patients achieve CR, and patients achieving CR have a 10-year OS of 75%.62,63 In a retrospective study that explored the characteristics of 186 patients with AL amyloidosis who survived more than 10 years, achieving complete hematologic response was predictive of treatment-free survival, whereas ASCT was not predictive in the multivariate analysis.64

Consolidation and maintenance post ASCT

Consolidation post ASCT is an appealing intervention for those patients who do not achieve deep hematological responses, but prospective data on this topic are in short supply. Using a risk-adapted conditioning approach in 83 patients—post-ASCT thalidomide or bortezomib consolidation—CR rates increased from 24% at day 100 to 48% at 12 months.50

Patients consolidated with bortezomib achieved outstanding CR rates (62%). Patients who achieved CR via ASCT had comparable event-free survival and OS with patients who achieved CR via ASCT and consolidation. In a retrospective series of 471 patients who underwent ASCT at our institution, 15% received consolidation.65 Comparing the patients who received consolidation with those who did not, the no-consolidation patients had inferior PFS and OS; however, focusing on the subset of 91 patients who did not achieve VGPR or better post ASCT, those receiving consolidation had a superior PFS compared with those who did not receive consolidation (22 months vs 9 months, respectively; P <.001). OS was not different between these 2 groups. When the OS analysis was limited to those patients who received consolidation and upgraded their response, this subgroup had better survival compared with those with inadequate response who did not receive consolidation (5-year OS, 79% vs 52%; P = .07). At the Mayo Clinic, we consider patients who have not achieved at least a VGPR as refractory, and we treat them with second-line therapy.

Maintenance after ASCT has not been prospectively evaluated, and a trial evaluating ixazomib (Ninlaro) as maintenance therapy post ASCT is ongoing. In a retrospective study, 25 patients received maintenance and 25 didn’t, and maintenance did not impact OS or PFS.66 At the Mayo Clinic, we use maintenance for patients with overt multiple myeloma (patients who fulfilled CRAB criteria at diagnosis) and consider it for patients with >20% BMPC and high-risk genetics.

To do ASCT or not to do ASCT?

ASCT leads to deep and durable responses57 in a disease where the depth of clonal control directly influences long-term organ function and survival. The first report of outcomes of ASCT in 5 AL amyloidosis was published in 1996,67 and ever since several centers gained long-term experience with ASCT in AL patients.42,47 Over the years, results of transplanted patients have become more and more favorable, with complete response (CR) rates as high as 40% and low relapse rates,42 with a good prognosis for long-term long survival among patients who achieve deep response.48,56

A matched case-control study that compared outcomes of 63 patients with AL amyloidosis who underwent ASCT with those of 63 similar patients who were not transplanted showed better OS in the ASCT cohort (71% vs 41%, respectively).68 However, these results were contradicted by those of the only prospective randomized controlled trial that compared ASCT with no ASCT (oral melphalan and dexamethasone) in AL amyloidosis. This study reported inferior survival in the ASCT arm (median OS, 22.2 vs 56.9 months).69 The inferior outcomes were considered by many to be related to the exceptionally high TRM (24%) in the ASCT group. The highest mortality rates occurred at transplant sites that had less experience transplanting patients with AL amyloidosis.70 A prospective study published by our group evaluated ASCT vs chemotherapy based on patient selection. The 3-year PFS (29.1% vs 51.7%) and OS (58.8% vs 83.6%) rates were higher in the ASCT arm, while the hematologic response rates were similar. However, this study did not meet its accrual goal and therefore was underpowered to determine differences in outcomes between the ASCT arm and the non-ASCT arm.71

In the era of novel agents, a retrospective study of 72 patients showed superiority of ASCT. Patients receiving ASCT were more likely to achieve deep responses (≥VGPR) and had higher PFS (not reached vs 9 months), OS (74 vs 9 months), and duration of response (DOR; 31 months vs 7 months).72 A systematic review and meta-analysis published in 2009 failed to show that ASCT improves outcomes vs conventional chemotherapy,73 but the quality of the evidence was low. A recently published retrospective case-matched study compared ASCT with bortezomib and showed no significant difference in any of the following: OS at 2 years, median PFS, time to next treatment, overall hematological responses at 6 months, or organ responses at 12 months.23

Long-term survival data coming from several centers are encouraging. In a long-term report of 159 patients with AL amyloidosis who were transplanted at Mayo Clinic, 47 patients (30%) survived more than 15 years.47 Survival was longer in patients without cardiac involvement, in those who achieved hematological CR, and in patients who received full-dose melphalan. In a report from the UK group, 33% of 264 patients survived at 20 years, and the median OS was 7.2 years.74

In a report from the Boston University group, of 692 patients with AL amyloidosis who underwent ASCT between 1994 and 2014, the median OS was 7.6 years and hematologic CR was achieved in 40% of evaluable patients.42 Thirty percent of the cohort survived more than 20 years. Hematologic relapse occurred in 32% of patients who had achieved CR at a median of 4.3 years, with a median OS in those patients of 8.5 years.75

Future directions

Several open questions remain, and they can be answered only by further clinical trials. Daratumumab therapy has changed the landscape of treatment of AL amyloidosis, generating deep and durable responses and having a very good safety profile. Venetoclax (Venclexta), a well-established therapy for multiple myeloma, is also an appealing treatment option for AL amyloidosis since 50% of this population harbors translocation t(11;14). The question of whether ASCT is superior to the current standard of care remains open, and a prospective randomized controlled trial clarifying this issue in AL amyloidosis is critically needed. Should ASCT be done in patients who achieve MRD negativity after induction, or even CR? Prospective clinical trials that incorporate updated measures of outcome are necessary to evaluate the long-term outcomes of different groups—patients who achieve deep responses prior to transplant vs patients who achieve deep responses after transplant vs patients who do not undergo ASCT—are warranted. Despite these unprecedented advances in treating AL amyloidosis, we still believe ASCT is important in its treatment paradigm.

Financial Disclosure: AD serves on the advisory board and independent review committee for Janssen and data monitoring safety committees for Oncopeptides and Sorrento and receives research funding from Alynlam, Pfizer, Takeda, and Bristol Myers Squibb.

References
1. Merlini G, Dispenzieri A, Sanchorawala V, et al. Systemic immunoglobulin light chain amyloidosis. Nat Rev Dis Primers. 2018;4(1):38. doi:10.1038/s41572-018-0034-3
2. Merlini G, Bellotti V. Molecular mechanisms of amyloidosis. N Engl J Med. 2003;349(6):583-596. doi:10.1056/NEJMra023144
3. Wechalekar AD, Schonland SO, Kastritis E, et al. A European collaborative study of treatment outcomes in 346 patients with cardiac stage III AL amyloidosis. Blood. 2013;121(17):3420-3427. doi:10.1182/blood-2012-12-473066
4. Muchtar E, Dispenzieri A, Leung N, et al. Depth of organ response in AL amyloidosis is associated with improved survival: new proposed organ response criteria. Amyloid. 2019;26(suppl 1):101-102. doi:10.1080/13506129.2019.1582486
5. Kastritis E, Fotiou D, Theodorakakou F, et al. Timing and impact of a deep response in the outcome of patients with systemic light chain (AL) amyloidosis. Amyloid. 2021;28(1):3-11. doi:10.1080/13506129.2020.1798224
6. Kastritis E, Leleu X, Arnulf B, et al. Bortezomib, melphalan, and dexamethasone for light-chain amyloidosis. J Clin Oncol. 2020;38(28):3252-3560. doi:10.1200/JCO.20.01285
7. Sanchorawala V, Palladini G, Kukreti V, 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:10.1182/blood.2020005086
8. Palladini G, Sachchithanantham S, Milani P, et al. A European collaborative study of cyclophosphamide, bortezomib, and dexamethasone in upfront treatment of systemic AL amyloidosis. Blood. 2015;126(5):612-615. doi:10.1182/blood-2015-01-620302
9. Reece DE, Hegenbart U, Sanchorawala V, et al. Long-term follow-up from a phase 1/2 study of single-agent bortezomib in relapsed systemic AL amyloidosis. Blood. 2014;124(16):2498-2506. doi:10.1182/blood-2014-04-568329
10. Palladini G, Milani P, Foli A, 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:10.1038/leu.2014.227
11. Palladini G, Milani P, Foli A, et al. A phase 2 trial of pomalidomide and dexamethasone rescue treatment in patients with AL amyloidosis. Blood. 2017;129(15):2120-2123. doi:10.1182/blood-2016-12-756528
12. Cohen OC, Sharpley F, Gillmore JD, et al. Use of ixazomib, lenalidomide and dexamethasone in patients with relapsed amyloid light-chain amyloidosis. Br J Hematol. 2020;189(4):643-649. doi:10.1111/bjh.16401
13. Warsame R, LaPlant B, Kumar SK, et al. Long-term outcomes of IMiD-based trials in patients with immunoglobulin light-chain amyloidosis: a pooled analysis. Blood Cancer J. 2020;10(1):4. doi:10.1038/s41408-019-0266-9
14. Kastritis E, Dialoupi I, Gavriatopoulou M, et al. Primary treatment of light-chain amyloidosis with bortezomib, lenalidomide, and dexamethasone. Blood Adv. 2019;3(20):3002-3009. doi:10.1182/bloodadvances.2019000147
15. Hegenbart U, Bochtler T, Benner A, 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:10.3324/haematol.2016.163246
16. 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;103(8):2936-2938. doi:10.1182/blood-2003-08-2788
17. Palladini G, Kastritis E, Maurer MS, 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:10.1182/blood.2019004460
18. Lecumberri R, Krsnik I, Askari E, et al. Treatment with daratumumab in patients with relapsed/refractory AL amyloidosis: a multicentric retrospective study and review of the literature. Amyloid. 2020;27(3):163-167. doi:10.1080/13506129.2020.1730791
19. Milani P, Fazio F, Basset M, et al. High rate of profound clonal and renal responses with daratumumab treatment in heavily pre-treated patients with light chain (AL) amyloidosis and high bone marrow plasma cell infiltrate. Am J Hematol. 2020;95(8):900-905. doi:10.1002/ajh.25828
20. Roussel M, Merlini G, Chevret S, 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:10.1182/blood.2019004369
21. Vaxman I, Sidiqi MH, Al Saleh AS, et al. Depth of response prior to autologous stem cell transplantation predicts survival in light chain amyloidosis. Bone Marrow Transplant. 2021;56(4):928-935. doi:10.1038/s41409-020-01136-2
22. Sanchorawala V, Brauneis D, Shelton AC, et al. Induction therapy with bortezomib followed by bortezomib-high dose melphalan and stem cell transplantation for light chain amyloidosis: results of a prospective clinical trial. Biol Bone Marrow Transplant. 2015;21(8):1445-1451. doi:10.1016/j.bbmt.2015.04.001
23. Sharpley FA, Manwani R, Petrie A, et al. Autologous stem cell transplantation vs bortezomib based chemotheraphy for the first-line treatment of systemic light chain amyloidosis in the UK. Eur J Haematol. 2021;106(4):537-545. doi:10.1111/ejh.13582
24. Minnema MC, Nasserinejad K, Hazenberg B, et al. Bortezomib-based induction followed by stem cell transplantation in light chain amyloidosis: results of the multicenter HOVON 104 trial. Haematologica. 2019;104(11):2274-2282. doi:10.3324/haematol.2018.213900
25. Bochtler T, Hegenbart U, Kunz C, et al. Prognostic impact of cytogenetic aberrations in AL amyloidosis patients after high-dose melphalan: a long-term follow-up study. Blood. 2016;128(4):594-602. doi:10.1182/blood-2015-10-676361
26. Huang X, Wang Q, Chen W, 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:10.1186/1741-7015-12-2
27. Huang X, Fu C, Chen L, et al. Combination of bortezomib in the induction, conditioning and consolidation with autologous hematopoietic stem cell transplantation in patients with immunoglobulin light chain amyloidosis. Am J Hematol. 2019;94(4):E101-E104. doi:10.1002/ajh.25404
28. Gupta VK, Brauneis D, Shelton AC, et al. 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:10.1016/j.bbmt.2019.01.007
29. Sanchorawala V, Wright DG, Seldin DC, 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:10.1038/sj.bmt.1704346
30. Afrough A, Saliba RM, Hamdi A, et al. Impact of induction therapy on the outcome of immunoglobulin light chain amyloidosis after autologous hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2018;24(11):2197-2203. doi:10.1016/j.bbmt.2018.07.010
31. Jain T, Kosiorek HE, Kung ST, et al. Treatment with bortezomib-based therapy, followed by autologous stem cell transplantation, improves outcomes in light chain amyloidosis: a retrospective study. Clin Lymphoma Myeloma Leuk. 2018;18(7):486-492.e1. doi:10.1016/j.clml.2018.04.006
32. Hwa YL, Kumar SK, Gertz MA, et al. Induction therapy pre-autologous stem cell transplantation in immunoglobulin light chain amyloidosis: a retrospective evaluation. Am J Hematol. 2016;91(10):984-988. doi:10.1002/ajh.24453
33. Huang X, Ren G, Chen W, et al. The role of induction therapy before autologous stem cell transplantation in low disease burden AL amyloidosis patients. Amyloid. 2021;28(2):75-83. doi:10.1080/13506129.2020.1835635
34. Cowan AJ, Klippel ZK, Stevenson PA, et al. Pre-transplantation novel agent induction predicts progression-free survival for patients with immunoglobulin light-chain amyloidosis undergoing high-dose melphalan and autologous stem cell transplantation. Amyloid. 2016;23(4):254-259. doi:10.1080/13506129.2016.1258356
35. Kastritis E, Palladini G, Monique C, et al. Subcutaneous daratumumab + cyclophosphamide, bortezomib, and dexamethasone (CYBORD) in patietns with newly diagnosed light chain (AL) amyloidosis: primary results from phase 3 ANDROMEDA study. Presented at: 2020 European Hematology Association Virtual Congress; June 9-17, 2020. Abstract LB2604. https://bit.ly/3rgOgvv
36. Mikhael JR, Schuster SR, Jimenez-Zepeda VH, et al. Cyclophosphamide-bortezomib-dexamethasone (CyBorD) produces rapid and complete hematologic response in patients with AL amyloidosis. Blood. 2012;119(19):4391-4394. doi:10.1182/blood-2011-11-390930
37. Moreau P, Leblond V, Bourquelot P, et al. Prognostic factors for survival and response after high-dose therapy and autologous stem cell transplantation in systemic AL amyloidosis: a report on 21 patients. Br J Haematol. 1998;101(4):766-769. doi:10.1046/j.1365-2141.1998.00772.x
38. Skinner M, Sanchorawala V, Seldin DC, et al. High-dose melphalan and autologous stem-cell transplantation in patients with AL amyloidosis: an 8-year study. Ann Intern Med. 2004;140(2):85-93. doi:10.7326/0003-4819-140-2-200401200-00008
39. Gertz MA, Lacy MQ, Dispenzieri A, et al. Refinement in patient selection to reduce treatment-related mortality from autologous stem cell transplantation in amyloidosis. Bone Marrow Transplant. 2013;48(4):557-561. doi:10.1038/bmt.2012.170
40. Batalini F, Econimo L, Quillen K, et al. High-dose melphalan and stem cell transplantation in patients on dialysis due to immunoglobulin light-chain amyloidosis and monoclonal immunoglobulin deposition disease. Biol Blood Marrow Transplant. 2018;24(1):127-132. doi:10.1016/j.bbmt.2017.08.031
41. Sidiqi MH, Aljama MA, Muchtar E, et al. Autologous stem cell transplant for immunoglobulin light chain amyloidosis patients aged 70 to 75. Biol Blood Marrow Transplant. 2018;24(10):2157-2159. doi:10.1016/j.bbmt.2018.06.017
42. Sanchorawala V, Sun F, Quillen K, Sloan JM, Berk JL, Seldin DC. Long-term outcome of patients with AL amyloidosis treated with high-dose melphalan and stem cell transplantation: 20-year experience. Blood. 2015;126(20):2345-2347. doi:10.1182/blood-2015-08-662726
43. Yeh JC, Shank BR, Milton DR, Qazilbash MH. Adverse prognostic factors for morbidity and mortality during peripheral blood stem cell mobilization in patients with light chain amyloidosis. Biol Blood Marrow Transplant. 2018;24(4):815-819. doi:10.1016/j.bbmt.2017.11.040
44. Badar T, Dhakal B, Szabo A, et al. An updated single center experience with plerixafor and granulocyte colony-stimulating factor for stem cell mobilization in light chain amyloidosis. J Clin Apher. 2019;34(6):686-691. doi:10.1002/jca.21747
45. Lee SY, Sanchorawala V, Seldin DC, Sloan JM, Andrea N, Quillen K. Plerixafor-augmented peripheral blood stem cell mobilization in AL amyloidosis with cardiac involvement: a case series. Amyloid. 2014;21(3):149-153. doi:10.3109/13506129.2014.900486
46. Comenzo RL, Gertz MA. Autologous stem cell transplantation for primary systemic amyloidosis. Blood. 2002;99(12):4276-4282. doi:10.1182/blood.v99.12.4276
47. Sidana S, Sidiqi MH, Dispenzieri A, et al. Fifteen year overall survival rates after autologous stem cell transplantation for AL amyloidosis. Am J Hematol. 2019;94(9):1020-1026. doi:10.1002/ajh.25566.48
48. Sidiqi MH, Aljama MA, Buadi FK, et al. Stem cell transplantation for light chain amyloidosis: decreased early mortality over time. J Clin Oncol. 2018;36(13):1323-1329. doi:10.1200/JCO.2017.76.9554
49. D’Souza A, Dispenzieri A, Wirk B, et al. Improved outcomes after autologous hematopoietic cell transplantation for light chain amyloidosis: a Center for International Blood and Marrow Transplant Research study. J Clin Oncol. 2015;33(32):3741-3749. doi:10.1200/JCO.2015.62.4015
50. Landau H, Smith M, Landry 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:10.1038/leu.2016.229
51. Nguyen VP, Landau H, Quillen K, et al. Modified high-dose melphalan and autologous stem cell transplantation for immunoglobulin light chain amyloidosis. Biol Blood Marrow Transplant. 2018;24(9):1823-1827. doi:10.1016/j.bbmt.2018.06.018
52. Sidiqi MH, Nadiminti K, Al Saleh AS, et al. Autologous stem cell transplantation in patients with AL amyloidosis with impaired renal function. Bone Marrow Transplant. 2019;54(11):1775-1779. doi:10.1038/s41409-019-0524-2
53. Fadia A, Casserly LF, Sanchorawala V, et al. Incidence and outcome of acute renal failure complicating autologous stem cell transplantation for AL amyloidosis. Kidney Int. 2003;63(5):1868-1873. doi:10.1046/j.1523-1755.2003.00936.x
54. Gordillo CA, Parmar S, Blanco M, et al. Gastrointestinal toxicity of high-dose melphalan in autologous hematopoietic stem cell transplantation: identification of risk factors and a benchmark for experimental therapies. Ann Hematol. 2021;100(7):1863-1870. doi:10.1007/s00277-020-04378-8
55. Basset M, Milani P, Nuvolone M, et al. Sequential response-driven bortezomib-based therapy followed by autologous stem cell transplant in AL amyloidosis. Blood Adv. 2020;4(17):4175-4179. doi:10.1182/bloodadvances.2020002219
56. Abdallah N, Sidana S, Dispenzieri A, et al. Outcomes with early vs. deferred stem cell transplantation in light chain amyloidosis. Bone Marrow Transplant. 2020;55(7):1297-1304. doi:10.1038/s41409-020-0964-8
57. Venner CP, Gillmore JD, Sachchithanantham S, et al. Stringent patient selection improves outcomes in systemic light-chain amyloidosis after autologous stem cell transplantation in the upfront and relapsed setting. Haematologica. 2014;99(12):e260-e263. doi:10.3324/haematol.2014.108191
58. Manwani R, Hegenbart U, Mahmood S, et al. Deferred autologous stem cell transplantation in systemic AL amyloidosis. Blood Cancer J. 2018;8(11):101. doi:10.1038/s41408-018-0137-9
59. Kastritis E, Kostopoulos IV, Theodorakakou F, et al. Next generation flow cytometry for MRD detection in patients with AL amyloidosis. Amyloid. 2021;28(1):19-23. doi:10.1080/13506129.2020.1802713
60. Palladini G, Paiva B, Wechalekar A, et al. Minimal residual disease negativity by next-generation flow cytometry is associated with improved organ response in AL amyloidosis. Blood Cancer J. 2021;11(2):34. doi:10.1038/s41408-021-00428-0
61. Staron A, Burks EJ, Lee JC, Sarosiek S, Sloan JM, Sanchorawala V. Assessment of minimal residual disease using multiparametric flow cytometry in patients with AL amyloidosis. Blood Adv. 2020;4(5):880-884. doi:10.1182/bloodadvances.2019001331
62. Dispenzieri A, Seenithamby K, Lacy MQ, et al. Patients with immunoglobulin light chain amyloidosis undergoing autologous stem cell transplantation have superior outcomes compared with patients with multiple myeloma: a retrospective review from a tertiary referral center. Bone Marrow Transplant. 2013;48(10):1302-1307. doi:10.1038/bmt.2013.53
63. Cibeira MT, Sanchorawala V, Seldin DC, et al. Outcome of AL amyloidosis after high-dose melphalan and autologous stem cell transplantation: long-term results in a series of 421 patients. Blood. 2011;118(16):4346-4352. doi:10.1182/blood-2011-01-330738
64. Muchtar E, Gertz MA, Lacy MQ, et al. Ten-year survivors in AL amyloidosis: characteristics and treatment pattern. Br J Haematol. 2019;187(5):588-594. doi: 10.1111/bjh.16096
65. Al Saleh AS, Sidiqi MH, Sidana S, 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:10.1002/ajh.25572
66. Ozga M, Zhao Q, Benson D, et al. AL Amyloidosis: the effect of maintenance therapy on autologous stem cell transplantation outcomes. J Clin Med. 2020;9(11):3778. doi:10.3390/jcm9113778
67. Comenzo RL, Vosburgh E, Simms RW, et al. Dose-intensive melphalan with blood stem cell support for the treatment of AL amyloidosis: one-year follow-up in five patients. Blood. 1996;88(7):2801-2806.
68. Dispenzieri A, Kyle RA, Lacy MQ, et al. Superior survival in primary systemic amyloidosis patients undergoing peripheral blood stem cell transplantation: a case-control study. Blood. 2004;103(10):3960-3963. doi:10.1182/blood-2003-12-4192
69. Jaccard A, Moreau P, Leblond V, et al; Myélome Autogreffe (MAG) and Intergroupe Francophone du Myélome (IFM) Intergroup. High-dose melphalan versus melphalan plus dexamethasone for AL amyloidosis. N Engl J Med. 2007;357(11):1083-1093. doi:10.1056/NEJMoa070484
70. Mehta J. High-dose melphalan versus melphalan plus dexamethasone for AL amyloidosis. The N Engl J Med. 2008;358(1):91; author reply 92-93. doi:10.1056/NEJMoa070484
71. Gertz MA, Lacy MQ, Dispenzieri A, et al. Stem cell transplantation compared with melphalan plus dexamethasone in the treatment of immunoglobulin light-chain amyloidosis. Cancer. 2016;122(14):2197-2205. doi:10.1002/cncr.30051
72. Oke O, Sethi T, Goodman S, et al. Outcomes from autologous hematopoietic cell transplantation versus chemotherapy alone for the management of light chain amyloidosis. Biol Blood Marrow Transplant. 2017;23(9):1473-1477. doi:10.1016/j.bbmt.2017.05.020
73. Mhaskar R, Kumar A, Behera M, Kharfan-Dabaja MA, Djulbegovic B. Role of high-dose chemotherapy and autologous hematopoietic cell transplantation in primary systemic amyloidosis: a systematic review. Biol Blood Marrow Transplant. 2009;15(8):893-902. doi:10.1016/j.bbmt.2009.01.022
74. Sharpley FA, Petrie A, Mahmood S, et al. A 24-year experience of autologous stem cell transplantation for light chain amyloidosis patients in the United Kingdom. Br J Haematol. 2019;187(5):642-652. doi:10.1111/bjh.16143
75. Browning S, Quillen K, Sloan JM, Doros G, Sarosiek S, Sanchorawala V. Hematologic relapse in AL amyloidosis after high-dose melphalan and stem cell transplantation. Blood. 2017;130(11):1383-1386. doi:10.1182/blood-2017-06-788729
Related Videos
Genovefa (Zenia) Papanicolaou, MD, an infectious diseases specialist at Memorial Sloan Kettering Cancer Center
Akshay Sharma, MBBS, a bone marrow transplant physician at St. Jude Children’s Research Hospital
John DiPersio, MD, PhD, the director of the Center for Gene and Cellular Immunotherapy at Washington University School of Medicine
Aude Chapuis, MD, an associate professor in the Translational Science and Therapeutics Division at Fred Hutch Cancer Center
Amar Kelkar, MD, a stem cell transplantation physician at the Dana-Farber Cancer Institute
David Porter, MD
© 2024 MJH Life Sciences

All rights reserved.