Next Article in Journal
The Current Understanding of and Treatment Paradigm for Newly-Diagnosed TP53-Mutated Acute Myeloid Leukemia
Next Article in Special Issue
Learning from Patients: The Interplay between Clinical and Laboratory Research in AL Amyloidosis
Previous Article in Journal
New Targets for PET Imaging of Myeloma
Previous Article in Special Issue
Differences and Similarities in Treatment Paradigms and Goals between AL Amyloidosis and Multiple Myeloma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Treatment in AL Amyloidosis: Moving towards Individualized and Clone-Directed Therapy

by
Ute Hegenbart
1,2,*,
Marc S. Raab
1,3 and
Stefan O. Schönland
1,2
1
Division of Hematology/Oncology, Department of Internal Medicine V, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
2
Amyloidosis Center, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
3
Myeloma Center, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
*
Author to whom correspondence should be addressed.
Submission received: 2 November 2021 / Revised: 27 November 2021 / Accepted: 4 December 2021 / Published: 7 December 2021

Abstract

:
Systemic amyloid light chain (AL) amyloidosis is a rare protein deposition disease caused by a clonal B cell disorder of the bone marrow. The underlying diseases can be plasma cell disorders (monoclonal gammopathy of clinical significance, smoldering or symptomatic myeloma) or B cell non-Hodgkin’s lymphoma (e.g., Waldenstrom’s disease or marginal zone lymphoma) with secretory activity. It is crucial to characterize the underlying disease very precisely as the treatment of AL amyloidosis is directed against the (often small) B cell clone. Finally, the detection of cytogenetic aberrations of the plasma cell clone will likely play an important role for choosing an effective drug in the near future.

1. Introduction

Systemic amyloid light-chain (AL-) amyloidosis is a rare monoclonal B cell disorder with poor prognosis due to the production of free light chains leading to amyloid deposition in nearly all organs [1]. Most patients succumb to advanced heart failure. The only evidence-based and recently approved therapy is cytoreduction of the underlying monoclonal aberrant plasma or B cell population to reduce or even stop the production of amyloid precursors leading to an improvement of organ functions.
In about 90% of AL patients the underlying disease is derived from clonal plasma cells (AL-PC), therefore therapeutic strategies are mostly adapted from established myeloma regimens. In about 10% the underlying clone is a B cell clone [1] and in such cases drugs established in those lymphomas are used.

2. Staging of AL Amyloidosis and Characterization of the Underlying Clone

At diagnosis of AL the first step is to assess organ involvement and to stage and quantify cardiac and renal impairment. These validated staging systems help to choose the appropriate intensity of the chemotherapy [1]. The second step is to characterize the clonal B cell disorder to choose the most efficacious treatment. Table 1 shows the current available (and future) methods to detect, characterize and quantify the clone. In the future, mass spectrometry might become available on a broader scale to quantify the monoclonal light chain in serum more specifically [2].

3. Clone-Directed Treatment in AL Amyloidosis

The goal of the treatment is to avoid further organ damage, to preserve organ function, to maintain quality of life and to prolong survival of these often very sick and fragile patients. This can currently only be achieved by the eradication or reduction of the amyloidogenic clone using several approaches.
Chemotherapy against plasma cells (MGCS, smoldering myeloma, symptomatic myeloma) or against B cells (Waldenstrom’s disease or other secretory active B cell NHLs such as marginal zone lymphoma) is derived from myeloma or lymphoma treatment.
Standard treatment includes alkylating chemotherapy combined with steroids (mostly dexamethasone), proteasome inhibitors and immunomodulating drugs [1]. Monoclonal antibodies are directed against several surface antigens of plasma cells (CD38, SLAM7, BCMA) or B cells (CD20) and are now also widely used in AL patients. There are few data regarding the treatment with Venetoclax targeting the anti-apoptotic bcl2-dependent pathway. Recently bispecific antibodies (BCMA, CD3) and CAR T cell therapies (BCMA, CD19) have been introduced for multiple myeloma and B cell lymphoma and might be applied also for AL amyloidosis in the near future. Targeted treatment is desirable to reduce side effects and to achieve a very rapid reduction of the clonal amyloidogenic light chains in the serum. Figure 1 shows several mechanisms, how malignant plasma or B cells can be attacked.

4. Alkylators

Melphalan and cyclophosphamide (plus corticosteroids) are used as part of combinations with newer agents. Melphalan plus dexamethasone (MDex) have been a standard for transplant ineligible patients [9] and was later on combined with bortezomib (BMDex) in a Phase III trial with high response rates [10]. Gain of 1q21 was identified as negative prognostic marker for AL patients treated with MDex [11]. In multivariate concordance analyses the adverse prognosis carried by gain of 1q21 was an independent prognostic factor (overall survival; OS: p = 0.003, average hazard ratio (AHR) 3.64, hematologic event-free survival; hemEFS: p = 0.008, AHR 2.35), along with the well-established Mayo cardiac staging. Patients with t(11;14) had a longer median OS with 38.2 months versus 17.5 months, though no statistical significance was reached.
The most intense regimen is high-dose chemotherapy with autologous stem cell transplantation (ASCT); the standard conditioning is still melphalan 200 mg/m2 [12]. The use of induction therapy and maintenance is depending on the burden of the plasma cell clone trying to reduce the risk of early progression [13,14].
The prognostic value of cytogenetic aberrations was investigated in 123 AL patients who received high-dose melphalan with ASCT [15]. In multivariate analysis, t(11;14) was confirmed as a favorable prognostic factor regarding hemEFS along with lower values for the difference between involved and uninvolved free light chains. Conversely, deletion13q14, gain of 1q21 and hyperdiploidy had no significant prognostic impact.
The International Society of Amyloidosis (ISA) together with European Hematologic Association (EHA) has currently published their guidelines for high-dose chemotherapy and ASCT [16]. The transplant-related mortality has significantly decreased in the last 20 years in experienced centers, but the selection of eligible patients remains crucial. The rate of complete remission varies between 30–50% and long-term survival can be achieved. A cure of the underlying disease might be possible in a small number of CR patients [17]. Therefore, HDM with ASCT is a very effective treatment for AL amyloidosis in selected patients. A direct comparison with new and clone-directed treatments within randomized clinical trials is desirable, but extremely difficult as patients have denied this in several studies in the past [18]. The best treatment could therefore be a combination of both strategies as it will be likely the case in multiple myeloma, at least for patients not reaching MRD negativity.
Bendamustine-based regimens are mostly used for AL patients with underlying B cell malignancies [19], but can also be used in PC-AL [20].

5. Proteasome Inhibitors (PIs)

Clonal plasma cells in AL amyloidosis are dependent on proteasome integrity. Targeting the proteasome is highly effective in AL amyloidosis. Bortezomib-containing regimens are considered as the primary therapy for AL amyloidosis in most centers, in Europe and US.
Bortezomib–cyclophosphamide–dexamethasone (CyBorD) or bortezomib–Dexamethasone are commonly used today [18]. However, bortezomib cannot be used in AL patients with peripheral polyneuropathy and may be less effective in patients with a translocation t(11;14) [6,21,22]. In multivariable Cox regression models incorporating established hematologic and clinical risk factors, t(11;14) was an independent adverse prognostic marker for hemEFS (HR, 2.94; 95% CI, 1.37 to 6.25; P 0.006) and OS (HR, 3.13; 95% CI, 1.16 to 8.33; P 0.03).
Ixazomib was given in a randomized Phase III trial in relapsed AL patients but the primary endpoints of the study (hematologic response rate and 2-year vital organ deterioration or mortality rate) have unfortunately not been met. The drug is a therapeutic option for proteasome-naïve patients with symptomatic polyneuropathy [23]. Carfilzomib is effective but not yet widely used due to potential cardiac toxicity [24].

6. Immunomodulatory Agents (IMiDs)

The first IMiD thalidomide is not broadly used in AL amyloidosis in standard dosage because of higher rates of polyneuropathy [25]. Lenalidomide is also less well tolerated in AL compared to myeloma patients, the recommended dosage is 15 mg [26]. The main side effects are fluid retention, increase of NT-ProBNP and marked hypotension [27]. However, it is quite effective if used as upfront or relapse treatment in combination with dexamethasone or with alkylating agents [28] or with the monoclonal antibody daratumumab (DRD) [29]. Gain of 1q21 had an adverse impact on treatment results [27,29]. In DRD treated AL patients calculated median hemEFS and OS were 17.4 and 29.1 months, respectively. On univariable analysis, hemEFS was adversely influenced by dFLC > 180 mg/L (HR 2.71, p = 0.027) and gain 1q21 (HR 9.8, p = 0.003). Translocation t(11;14) did not affect outcome. Exploratory multivariable analysis of the two univariably significant factors in combination with albumin-to-creatinine ratio (ACR) > 220 mg/mmol confirmed gain 1q21 as highly significant (HR 8.9, p = 0.005).
Pomalidomide is increasingly used in AL patients and might be the best tolerated IMID in AL patients with some hematologic toxicities. It is an effective relapse treatment in doublet or triplet combination therapies [30].

7. Monoclonal Antibodies

Currently combination treatments with daratumumab, isatuximab and elotuzumab or antibody-conjugates such as belantamab-mafodotin are approved to treat multiple myeloma [31] and are also used for treatment in AL patients [29,32].
The ANDROMEDA trial included 388 patients with newly diagnosed AL. Patients have been randomized to receive either CyBorD or CyBorD + daratumumab 1800 mg s.c. [28]. After a median follow-up of 11.4 months the CR rate in the study arm was significantly higher than in the standard arm (53.3% vs. 18.1%). Furthermore, at 6 months cardiac and renal responses occurred more frequently in the daratumumab arm (41.5% vs. 22.2% and 53% vs. 23.9%) [33]. Recently, Daratumumab in combination with CyBorD was approved in the US and Europe for newly diagnosed patients with AL amyloidosis.
Data on daratumumab treatment in Rel/Ref-AL have been recently reviewed [34]. This treatment is very effective in daratumumab-naïve AL patients and generally well tolerated. However, the efficacy is reduced in patients with nephrotic range albuminuria [32] and gain of 1q21 [29].
AL patients with an underlying B cell malignancy can be treated with combinations of anti-CD20 monoclonal antibodies in combination with chemotherapy [35].
Table 2 shows the targets and ongoing clinical trials for AL patients.

8. Bispecific Antibodies and CAR-T-Cells

Bispecific antibodies and CAR-T cells are intensively studied in B cell lymphoma and multiple myeloma [31] and might also be investigated in AL patients in the near future. Given their highly specific mechanism of action against individual cell types with respective marker profiles such as CD19, BCMA or CD38, these novel immune-therapeutic approaches might be ideal to eradicate the underlying clonal cell population. However, side effects such as cytokine release syndrome or neurotoxicities might limit their use in certain subsets of patient with renal, neurologic or cardiac involvement.

9. Pathway-Inhibition

The characteristic cytogenetic aberration of the clonal plasma cells in AL amyloidosis is the translocation t(11;14), which can be detected in about 50% of AL patients [36]. Venetoclax is a drug targeting the anti-apoptotic bcl2-dependent pathway and is very effective in plasma cells harboring t(11;14). Venetoclax is active and approved in several diseases such as CLL [37] and AML [38]. Several studies are ongoing in multiple myeloma where venetoclax is used in combination with other anti-myeloma drugs [39] but currently stopped due to increased mortality in the venetoclax arm.
Venetoclax has recently been evaluated in AL. The first report came from the Mayo clinic [40] and afterwards an international group collected 43 pretreated (median 3 regimen) patients to analyze the efficacy and side effects of venetoclax in AL [41]. A total of 31 out of 42 patients carried the t(11;14) The hematologic response rate (≥VGPR) was higher in patients with the translocation (81% vs. 40%). There are currently no open clinical trials using bcl2 inhibitors in AL.
In opposite to Waldenstrom’s disease it has been shown in a small case series that ibrutinib as monotherapy is not well tolerated in AL patients [42] and is not leading to a deep remission.
Table 3 shows the advantages and disadvantages of the different treatment approaches.

10. Anti-Amyloid Treatment

Many AL patients are diagnosed in an advanced stage of organ damage which leads to rapid organ failure and/or shortened survival. Most of these patients do not benefit from reduction of amyloidogenic light chains as the amyloid organ damage is not reversible anymore. Therefore, it would be desirable to include anti-amyloid drugs which not only inhibit amyloid formation or deposition during or after the clone directed treatment, but actively remove amyloid deposits. There have been a few approaches with several drugs in the past, but these have not been successful yet. However, two anti-amyloid antibodies are currently investigated in prospective clinical trials (Table 4).

11. Summary

Patients with systemic AL amyloidosis represent a very fragile population. Goal of therapy is a rapid and deep reduction of the toxic light chain in the blood using a feasible approach. The intensity of the therapy is relying on highly validated risk adaption using cardiac and renal biomarkers. In a next step the underlying disorder is characterized as plasma or B cell clone and respective effective regimen are chosen. In PC-AL it is highly recommended to use iFISH for the detection of cytogenetic aberrations. Recent publications from different centers suggest that t(11;14) can be used to select the most effective therapies. Translocation t(11;14) favors melphalan treatment (either conventional or as high-dose) and venetoclax in opposite to bortezomib. Gain of 1q21 might be a poor prognostic factor irrespective the given therapy. Finally, new immunotherapies in combination with established drugs will further enhance anti-clonal efficacy leading to an improvement of overall survival in the majority of the patients. For patients in advanced stages of the disease anti-amyloid strategies are investigated in prospective clinical trials.

Author Contributions

U.H., M.S.R. and S.O.S.: Writing—original draft preparation; writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was in part funded by the Deutsche Forschungsgemeinschaft (Research Unit FOR 2969, Projects HE 8472/1-1, SCHO 1364/2-1).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

U.H. has received travel grants from Janssen, Prothena, and Pfizer; served on the advisory boards for Pfizer and Prothena; and has received honoraria from Janssen, Pfizer, Alnylam, and Akcea. M.S.R. served as a consultant (includes expert testimony) for Amgen, BMS and AbbVie, took part in research funding for Sanofi and Novartis, received honoraria from AbbVie and GSK and is/was a member on an entity’s board of directors or advisory committees: Amgen, BMS, Janssen, GSK and Sanofi. S.O.S. has received travel grants from Janssen, MSD, Prothena and Takeda; served on the advisory boards for Janssen and Prothena; has received honoraria from Janssen, Prothena, and Takeda; and received research funding from Sanofi and Janssen.

References

  1. Palladini, G.; Milani, P.; Merlini, G. Management of AL amyloidosis in 2020. Blood 2020, 136, 2620–2627. [Google Scholar] [CrossRef] [PubMed]
  2. Murray, D.L.; Dasari, S. Clinical mass spectrometry approaches to myeloma and amyloidosis. Clin. Lab. Med. 2021, 41, 203–219. [Google Scholar] [CrossRef] [PubMed]
  3. Puig, N.; Paiva, B.; Lasa, M.; Burgos, L.; Perez, J.J.; Merino, J.; Moreno, C.; Vidriales, M.-B.; Toboso, D.G.; Cedena, M.-T.; et al. Flow cytometry for fast screening and automated risk assessment in systemic light-chain amyloidosis. Leukemia 2019, 33, 1256–1267. [Google Scholar] [CrossRef]
  4. Lisenko, K.; Schönland, S.; Jauch, A.; Andrulis, M.; Röcken, C.; Ho, A.D.; Goldschmidt, H.; Hegenbart, U.; Hundemer, M. Flow cytometry-based characterization of underlying clonal B and plasma cells in patients with light chain amyloidosis. Cancer Med. 2016, 5, 1464–1472. [Google Scholar] [CrossRef]
  5. Bochtler, T.; Merz, M.; Hielscher, T.; Granzow, M.; Hoffmann, K.; Krämer, A.; Raab, M.-S.; Hillengass, J.; Seckinger, A.; Kimmich, C.; et al. Cytogenetic intraclonal heterogeneity of plasma cell dyscrasia in AL amyloidosis as compared with multiple myeloma. Blood Adv. 2018, 2, 2607–2618. [Google Scholar] [CrossRef] [Green Version]
  6. Bochtler, T.; Hegenbart, U.; Kunz, C.; Granzow, M.; Benner, A.; Seckinger, A.; Kimmich, C.; Goldschmidt, H.; Ho, A.D.; Hose, D.; 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, 1371–1378. [Google Scholar] [CrossRef]
  7. Bertamini, L.; D’Agostino, M.; Gay, F. MRD assessment in multiple myeloma: Progress and challenges. Curr. Hematol. Malig. Rep. 2021, 16, 162–171. [Google Scholar] [CrossRef]
  8. Bagratuni, T.; Ntanasis-Stathopoulos, I.; Gavriatopoulou, M.; Mavrianou-Koutsoukou, N.; Liacos, C.; Patseas, D.; Kanellias, N.; Migkou, M.; Ziogas, D.C.; Eleutherakis-Papaiakovou, E.; et al. Detection of MYD88 and CXCR4 mutations in cell-free DNA of patients with IgM monoclonal gammopathies. Leukemia 2018, 32, 2617–2625. [Google Scholar] [CrossRef]
  9. Palladini, G.; Milani, P.; Foli, A.; Obici, L.; Lavatelli, F.; Nuvolone, M.; Caccialanza, R.; Perlini, S.; Merlini, G. Oral melphalan and dexamethasone grants extended survival with minimal toxicity in AL amyloidosis: Long-term results of a risk-adapted approach. Haematologica 2014, 99, 743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kastritis, E.; Leleu, X.; Arnulf, B.; Zamagni, E.; Cibeira, M.T.; Kwok, F.; Mollee, P.; Hájek, R.; Moreau, P.; Jaccard, A.; et al. Bortezomib, melphalan, and dexamethasone for light-chain amyloidosis. J. Clin. Oncol. 2020, 38, 3252–3260. [Google Scholar] [CrossRef]
  11. Bochtler, T.; Hegenbart, U.; Kunz, C.; Benner, A.; Seckinger, A.; Dietrich, S.; Granzow, M.; Neben, K.; Goldschmidt, H.; Ho, A.D.; et al. Gain of chromosome 1q21 is an independent adverse prognostic factor in light chain amyloidosis patients treated with melpha-lan/dexamethasone. Amyloid 2014, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  12. Sanchorawala, V. High-dose melphalan and autologous peripheral blood stem cell transplantation in AL amyloidosis. Acta Haematol. 2020, 143, 381–387. [Google Scholar] [CrossRef] [PubMed]
  13. Hwa, Y.L.; Kumar, S.K.; Gertz, M.A.; Lacy, M.Q.; Buadi, F.K.; Kourelis, T.V.; Gonsalves, W.I.; Rajkumar, S.V.; Go, R.S.; Leung, N.; et al. Induction therapy preautologous stem cell transplantation in immunoglobulin light chain amyloidosis: A retrospective evaluation. Am. J. Hematol. 2016, 91, 984–988. [Google Scholar] [CrossRef] [PubMed]
  14. Afrough, A.; Saliba, R.M.; Hamdi, A.; Honhar, M.; Varma, A.; Cornelison, A.M.; Rondon, G.; Parmar, S.; Shah, N.D.; Bashir, Q.; 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, 2197–2203. [Google Scholar] [CrossRef] [Green Version]
  15. Bochtler, T.; Hegenbart, U.; Kunz, C.; Benner, A.; Kimmich, C.; Seckinger, A.; Hose, D.; Goldschmidt, H.; Granzow, M.; Dreger, P.; et al. Prognostic impact of cytogenetic aberrations in AL amyloidosis patients after high-dose melphalan: A long-term follow-up study. Blood 2016, 128, 594–602. [Google Scholar] [CrossRef] [Green Version]
  16. Sanchorawala, V.; Boccadoro, M.; Gertz, M.; Hegenbart, U.; Kastritis, E.; Landau, H.; Mollee, P.; Wechalekar, A.; Palladini, G. Guidelines for high dose chemotherapy and stem cell transplantation for systemic AL amyloidosis: EHA-ISA working group guidelines. Amyloid 2021, 1–7. [Google Scholar] [CrossRef] [PubMed]
  17. Sanchorawala, V.; Sun, F.; Quillen, K.; Sloan, J.M.; Berk, J.L.; Seldin, D.C. Long-term outcome of patients with AL amyloidosis treated with high-dose melphalan and stem cell transplantation: 20-year experience. Blood 2015, 126, 2345–2347. [Google Scholar] [CrossRef] [Green Version]
  18. Minnema, M.C.; Nasserinejad, K.; Hazenberg, B.; Hegenbart, U.; Vlummens, P.; Ypma, P.F.; Kröger, N.; Wu, K.L.; Kersten, M.J.; Schaafsma, M.R.; et al. Bortezomib-based induction followed by stem cell transplantation in light chain amyloidosis: Results of the multicenter HOVON 104 trial. Haematologica 2019, 104, 2274–2282. [Google Scholar] [CrossRef]
  19. Milani, P.; Schönland, S.; Merlini, G.; Kimmich, C.; Foli, A.; Dittrich, T.; Basset, M.; Müller-Tidow, C.; Bochtler, T.; Palladini, G.; et al. Treatment of AL amyloidosis with bendamustine: A study of 122 patients. Blood 2018, 132, 1988–1991. [Google Scholar] [CrossRef] [Green Version]
  20. Lentzsch, S.; Lagos, G.G.; Comenzo, R.L.; Zonder, J.A.; Osman, K.; Pan, S.; Bhutani, D.; Pregja, S.; Sanchorawala, V.; Landau, H. Bendamustine with dexamethasone in relapsed/refractory systemic light-chain amyloidosis: Results of a phase II study. J. Clin. Oncol. 2020, 38, 1455–1462. [Google Scholar] [CrossRef]
  21. Palladini, G.; Sachchithanantham, S.; Milani, P.; Gillmore, J.; Foli, A.; Lachmann, H.; Basset, M.; Hawkins, P.; Merlini, G.; Wechalekar, A.D. A European collaborative study of cyclophosphamide, bortezomib, and dexamethasone in upfront treatment of systemic AL amyloidosis. Blood 2015, 126, 612–615. [Google Scholar] [CrossRef] [Green Version]
  22. Muchtar, E.; Dispenzieri, A.; Kumar, S.K.; Ketterling, R.P.; Dingli, D.; Lacy, M.Q.; Buadi, F.K.; Hayman, S.R.; Kapoor, P.; Leung, N.; et al. Interphase fluorescence in situ hybridization in untreated AL amyloidosis has an independent prognostic impact by abnormality type and treatment category. Leukemia 2017, 31, 1562–1569. [Google Scholar] [CrossRef] [PubMed]
  23. Dispenzieri, A.; Kastritis, E.; Wechalekar, A.D.; Schönland, S.O.; Kim, K.; Sanchorawala, V.; Landau, H.J.; Kwok, F.; Suzuki, K.; Comenzo, R.L.; et al. A randomized phase 3 study of ixazomib-dexamethasone versus physician’s choice in relapsed or refractory AL amyloidosis. Leukemia 2021, 1–11. [Google Scholar] [CrossRef]
  24. Manwani, R.; Mahmood, S.; Sachchithanantham, S.; Lachmann, H.J.; Gillmore, J.D.; Yong, K.; Rabin, N.; Popat, R.; Kyriakou, C.; Worthington, S.; et al. Carfilzomib is an effective upfront treatment in AL amyloidosis patients with peripheral and autonomic neuropathy. Br. J. Haematol. 2019, 187, 638–641. [Google Scholar] [CrossRef]
  25. Wechalekar, A.D.; Goodman, H.J.B.; Lachmann, H.; Offer, M.; Hawkins, P.N.; Gillmore, J.D. Safety and efficacy of risk-adapted cyclophosphamide, thalidomide, and dexamethasone in systemic AL amyloidosis. Blood 2007, 109, 457–464. [Google Scholar] [CrossRef] [Green Version]
  26. Sanchorawala, V.; Wright, D.G.; Rosenzweig, M.; Finn, K.T.; Fennessey, S.; Zeldis, J.B.; Skinner, M.; Seldin, D.C. Lenalidomide and dexamethasone in the treatment of AL amyloidosis: Results of a phase 2 trial. Blood 2007, 109, 492–496. [Google Scholar] [CrossRef]
  27. Basset, M.; Kimmich, C.R.; Schreck, N.; Krzykalla, J.; Dittrich, T.; Veelken, K.; Goldschmidt, H.; Seckinger, A.; Hose, D.; Jauch, A.; et al. Lenalidomide and dexamethasone in relapsed/refractory immunoglobulin light chain (AL) amyloidosis: Results from a large cohort of patients with long follow-up. Br. J. Haematol. 2021, 195, 230–243. [Google Scholar] [CrossRef]
  28. Hegenbart, U.; Bochtler, T.; Benner, A.; Becker, N.; Kimmich, C.; Kristen, A.V.; Beimler, J.; Hund, E.; Zorn, M.; Freiberger, 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, 1424–1431. [Google Scholar] [CrossRef]
  29. Kimmich, C.R.; Terzer, T.; Benner, A.; Hansen, T.; Carpinteiro, A.; Dittrich, T.; Veelken, K.; Jauch, A.; Huhn, S.; Basset, M.; et al. Daratumumab, lenalidomide, and dexamethasone in systemic light-chain amyloidosis: High efficacy, relevant toxicity and main adverse effect of gain 1q21. Am. J. Hematol. 2021, 96, E253–E257. [Google Scholar] [CrossRef] [PubMed]
  30. Milani, P.; Sharpley, F.; Schönland, S.O.; Basset, M.; Mahmood, S.; Nuvolone, M.; Kimmich, C.; Foli, A.; Sachchithanantham, S.; Merlini, G.; 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, 231–236. [Google Scholar] [CrossRef] [PubMed]
  31. Rasche, L.; Wäsch, R.; Munder, M.; Goldschmidt, H.; Raab, M.S. Novel immunotherapies in multiple myeloma—Chances and challenges. Haematologica 2021, 106, 2555–2565. [Google Scholar] [CrossRef] [PubMed]
  32. Kimmich, C.R.; Terzer, T.; Benner, A.; Dittrich, T.; Veelken, K.; Carpinteiro, A.; Hansen, T.; Gold-schmidt, H.; Seckinger, A.; Hose, D.; et al. Daratumumab for systemic AL amyloidosis: Prognostic factors and adverse outcome with nephrotic-range albuminuria. Blood 2020, 135, 1517–1530. [Google Scholar] [CrossRef]
  33. Kastritis, E.; Palladini, G.; Minnema, M.C.; Wechalekar, A.D.; Jaccard, A.; Lee, H.C.; Sanchorawala, V.; Gibbs, S.; Mollee, P.; Venner, C.P.; et al. Daratumumab-based treatment for immunoglobulin light-chain amyloidosis. N. Engl. J. Med. 2021, 385, 46–58. [Google Scholar] [CrossRef] [PubMed]
  34. Palladini, G.; Milani, P.; Malavasi, F.; Merlini, G. Daratumumab in the treatment of light-chain (AL) amyloidosis. Cells 2021, 10, 545. [Google Scholar] [CrossRef]
  35. Godara, A.; Palladini, G. Monoclonal antibody therapies in systemic light-chain amyloidosis. Hematol. Oncol. Clin. N. Am. 2020, 34, 1145–1159. [Google Scholar] [CrossRef]
  36. Bochtler, T.; Hegenbart, U.; Heiss, C.; Benner, A.; Moos, M.; Seckinger, A.; Pschowski-Zuck, S.; Kirn, D.; Neben, K.; Bartram, C.R.; et al. Hyperdiploidy is less frequent in AL amyloidosis compared with monoclonal gammopathy of undetermined significance and inversely associated with translocation t(11;14). Blood 2011, 117, 3809–3815. [Google Scholar] [CrossRef]
  37. Perini, G.F.; Feres, C.C.P.; Teixeira, L.L.C.; Hamerschlak, N. BCL-2 Inhibition as treatment for chronic lymphocytic leukemia. Curr. Treat. Options Oncol. 2021, 22, 1–9. [Google Scholar] [CrossRef]
  38. Othman, T.A.; Azenkot, T.; Moskoff, B.N.; Tenold, M.E.; Jonas, B.A. Venetoclax-based combinations for the treatment of newly diagnosed acute myeloid leukemia. Future Oncol. 2021, 17, 2989–3005. [Google Scholar] [CrossRef] [PubMed]
  39. Sidiqi, M.H.; Al Saleh, A.S.; Kumar, S.K.; Leung, N.; Jevremovic, D.; Muchtar, E.; Gonsalves, W.I.; Kou-relis, T.V.; Warsame, R.; Buadi, F.K.; et al. Venetoclax for the treatment of multiple myeloma: Outcomes outside of clinical trials. Am. J. Hematol. 2021, 96, 1131–1136. [Google Scholar] [CrossRef]
  40. Sidiqi, M.H.; Al Saleh, A.S.; Leung, N.; Jevremovic, D.; Aljama, M.A.; Gonsalves, W.I.; Buadi, F.K.; Kourelis, T.V.; Warsame, R.; Muchtar, E.; et al. Venetoclax for the treatment of translocation (11;14) AL amyloidosis. Blood Cancer J. 2020, 10, 1–4. [Google Scholar] [CrossRef]
  41. Premkumar, V.J.; Lentzsch, S.; Pan, S.; Bhutani, D.; Richter, J.; Jagannath, S.; Liedtke, M.; Jaccard, A.; Wechalekar, A.D.; Comenzo, R.; et al. Venetoclax induces deep hematologic remissions in t(11;14) relapsed/refractory AL amyloidosis. Blood Cancer J. 2021, 11, 10. [Google Scholar] [CrossRef] [PubMed]
  42. Pika, T.; Hegenbart, U.; Flodrova, P.; Maier, B.; Kimmich, C.; Schönland, S.O. First report of ibrutinib in IgM-related amyloidosis: Few responses, poor tolerability, and short survival. Blood 2018, 131, 368–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Hegenbart, U.; aus dem Siepen, F.; Benner, A.; Wink, M.; Buss, S.; Mereles, D.; Aurich, M.; Kimmich, C.; Andre, F.; Ho, A.D.; et al. EGCG treatment in patients with cardiac AL amyloidosis: First results of a randomized and placebo-controlled clinical trial of Germany. The XVI. Int. Symp. Amyloidosis 2018, 2018, e287. [Google Scholar]
  44. D’Souza, A.; Szabo, A.; Flynn, K.E.; Dhakal, B.; Chhabra, S.; Pasquini, M.C.; Weihrauch, D.; Hari, P.N. Adjuvant doxycycline to enhance anti-amyloid effects: Results from the dual phase 2 trial. EClinicalMedicine 2020, 23, 100361. [Google Scholar] [CrossRef] [PubMed]
  45. Shen, K.N.; Fu, W.J.; Wu, Y.; Dong, Y.J.; Huang, Z.X.; Wei, Y.Q.; Li, C.R.; Sun, C.Y.; Chen, Y.; Miao, H.L.; et al. Doxycycline combined with bortezomib-cyclophosphamide-dexamethasone chemotherapy for newly diagnosed cardiac light-chain amyloidosis: A multicenter randomized controlled trial. Circulation 2021. [CrossRef] [PubMed]
  46. Richards, D.B.; Cookson, L.M.; Berges, A.C.; Barton, S.V.; Lane, T.; Ritter, J.M.; Fontana, M.; Moon, J.C.; Pin-zani, M.; Gillmore, J.D.; et al. Therapeutic clearance of amyloid by antibodies to serum amyloid P component. N. Engl. J. Med. 2015, 373, 1106–1114. [Google Scholar] [CrossRef] [Green Version]
  47. Gertz, M.A.; Landau, H.J.; Weiss, B.M. Organ response in patients with AL amyloidosis treated with NEOD001, an amyloid-directed monoclonal antibody. Am. J. Hematol. 2016, 91, E506–E508. [Google Scholar] [CrossRef]
  48. Edwards, C.V.; Rao, N.; Bhutani, D.; Mapara, M.Y.; Radhakrishnan, J.; Shames, S.; Maurer, M.S.; Leng, S.; Solomon, A.; Lentzsch, S.; et al. Phase 1a/b study of monoclonal antibody CAEL-101 (11-1F4) in patients with AL amyloidosis. Blood 2021. [CrossRef]
Figure 1. Mechanisms of drugs to attack the clonal cells in AL amyloidosis.
Figure 1. Mechanisms of drugs to attack the clonal cells in AL amyloidosis.
Hemato 02 00050 g001
Table 1. Methods to detect, characterize and quantify the clone.
Table 1. Methods to detect, characterize and quantify the clone.
MethodSignificance
Measurement of immunofixation in serum and urine as well as free light chains in serumStandard non-invasive method to evaluate monoclonal gammopathy in the daily practice without sampling error
Bone marrow examination
CytologyNumber of cells, phenotype
BiopsyNumber of clonal cells (light chain restriction), phenotype
FACSPhenotype, differentiation between clonal plasma and B cells, measurement of minimal residual disease [3,4]
iFISHDetection of cytogenetic aberrations (gain of 1,5,15,19; deletion 13 and 17, translocations) [5,6]
Serum mass spectrometryQualitative detection of small amounts of amyloidogenic proteins in serum [2]
Measurement of minimal residual disease [7]
MYD88 L265P, CXCR4Clonal markers for IgM gammopathies [8]
Table 2. Lists of recruiting clinical trials using monoclonal antibodies in AL.
Table 2. Lists of recruiting clinical trials using monoclonal antibodies in AL.
IdentifierTargetDrug CombinationsTypeInstitution
NCT04895917CD38Dara + pomalidomideSecond lineAmyloidosis Center Pavia, Italy
NCT04474938CD38Dara-bortezomib–dexamethasonUpfront, Mayo Stage 4University Beijing, China
NCT04131309CD38Dara-bortezomib–dexamethasonUpfront, Mayo Stage IIIbMulticenter Europe
NCT03283917CD38Dara-ixazomib–dexamethasonUpfront and relapsed/refractoryMD Anderson, Houston, USA
NCT04270175CD38Dara-pomalidomide–dexamethasonrelapsed/refractoryMulticenter USA
NCT04754945CD38IsatuximabHigh-riskMulticenter USA
NCT04617925BCMABelantamab–mafodotinPhase II, pre-treatedMulticenter Europe
noneSLAMF7Elotuzumab
noneCD20Rituximab
Table 3. Pro’s and con’s of main treatments.
Table 3. Pro’s and con’s of main treatments.
DrugProCon
High-dose melphalan and ASCTHigh rate of hematologic remission and organ response
Long-term efficacy, probably cure in some patients
Only available for selected patients
Increased treatment-related mortality and morbidity
Risk of secondary malignancies
Proteasome-inhibitorsFast reduction of the tumor burden
Subcutaneous application
Risk of polyneuropathy
Reduced efficacy in patients with t(11;14)
IMID’sEffective in combination therapies
Oral application
Reduced efficacy in patients with gain 1q21
Cardiac and renal side effects
CD38 antibodiesHigh rate of CR/VGPR and organ responses in combination with chemotherapy
Approved therapy in many countries
Few side effects
No long term data available
Bcl-2 inhibitorsHigh efficacy in patients with t(11;14)No long term data available
Table 4. Summary of drugs for anti-amyloid treatment.
Table 4. Summary of drugs for anti-amyloid treatment.
Drug (and Reference)Mechanism of ActionCurrent Status
Epigallocatechinegallat (EGCG), green tea substance [43]Inhibition of amyloid formation and/or degradationClinical trial not successful.
Compound cannot be applied in high dosages due to liver toxicity.
Poor intestinal resorption.
Doxycycline [44]Inhibition of fibril formationOne clinical trial ongoing (NCT03474458), no effect in one randomized clinical trial [45]
Miridesap + dezamizumab [46]SAP depletion in plasma + SAP removal from the tissuePhase III trial was closed due to toxicity issues
Birtamimab [47]Immunoglobulin G1 kappa monoclonal antibody, binds to light chain aggregatesTwo clinical trials closed due to missing efficacy in interim analysis; one ongoing
prospective Phase III trial in patients with cardiac stage Mayo IV
CAEL-101 [48]Chimeric immunoglobulin G1 kappa monoclonal antibody binds to a cryptic epitope at light chain proteins that adopt a non-native structureTwo ongoing prospective Phase III trials in patients with cardiac AL patients, ongoing (NCT02245867)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hegenbart, U.; Raab, M.S.; Schönland, S.O. Treatment in AL Amyloidosis: Moving towards Individualized and Clone-Directed Therapy. Hemato 2021, 2, 739-747. https://0-doi-org.brum.beds.ac.uk/10.3390/hemato2040050

AMA Style

Hegenbart U, Raab MS, Schönland SO. Treatment in AL Amyloidosis: Moving towards Individualized and Clone-Directed Therapy. Hemato. 2021; 2(4):739-747. https://0-doi-org.brum.beds.ac.uk/10.3390/hemato2040050

Chicago/Turabian Style

Hegenbart, Ute, Marc S. Raab, and Stefan O. Schönland. 2021. "Treatment in AL Amyloidosis: Moving towards Individualized and Clone-Directed Therapy" Hemato 2, no. 4: 739-747. https://0-doi-org.brum.beds.ac.uk/10.3390/hemato2040050

Article Metrics

Back to TopTop