You are on page 1of 12

Critical Reviews in Oncology/Hematology 99 (2016) 249–260

Contents lists available at ScienceDirect

Critical Reviews in Oncology/Hematology


journal homepage: www.elsevier.com/locate/critrevonc

Immunomodulatory drugs in AL amyloidosis


T. Jelinek a,b , Z. Kufova b , R. Hajek a,b,∗
a
Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
b
Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
2. Mechanism of action of immunomodulatory drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
2.1. Immunomodulatory effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
2.2. Direct antitumor effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
2.3. Anti-angiogenic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
2.4. Anti-inflammatory effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
2.5. Effects on the bone marrow microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
3. Thalidomide in the treatment of AL amyloidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
4. Lenalidomide in the treatment of AL amyloidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
5. Pomalidomide in the treatment of AL amyloidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
6. Problematics of response assessment with IMiDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
7. Treatment strategies in AL amyloidosis and role of IMiDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 258
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 258
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 258
Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260

a r t i c l e i n f o a b s t r a c t

Article history: Immunoglobulin light chain amyloidosis (AL amyloidosis) is indeed a rare plasma cell disorder, yet the
Received 25 October 2014 most common of the systemic amyloidoses. The choice of adequate treatment modality is complicated
Received in revised form and depends dominantly on the risk stratification of these fragile patients. Immunomodulatory drugs
20 November 2015
(IMiDs) are currently used in newly diagnosed patients as well as in salvage therapy in relapsed/refractory
Accepted 12 January 2016
patients. IMiDs have a pleiotropic effect on malignant cells and the exact mechanism of their action has
been described recently. Thalidomide is the most ancient representative, effective but toxic. Lenalidomide
Keywords:
seems to be more effective, nevertheless the toxicity remains high, especially in patients with renal
AL amyloidosis
Immunomodulatory drugs
insufficiency. Pomalidomide is the newest IMiD used in this indication with a good balance between
Thalidomide efficacy and tolerable toxicity and represents the most promising compound. This review is focused on
Lenalidomide the evaluation of all three representatives of IMiDs and their roles in the treatment of this malignant
Pomalidomide disorder.
© 2016 Elsevier Ireland Ltd. All rights reserved.

1. Introduction

Immunoglobulin light chain amyloidosis (AL amyloidosis, AL) is


a low tumor burden plasma cell disorder characterized by the pres-
ence of a small indolent clone of plasmocytes in bone marrow (BM)
synthesizing misfolded light chains. Instead of conforming to the ␣-
∗ Corresponding author at: Department of Haematooncology University Hospital helical configuration of most proteins, these amyloidogenous light
Ostrava 17, Listopadu 1790, 708 52 Ostrava,Czech Republic. Fax: +420 597 37 2092. chains form a ␤-pleated sheet responsible for formatting insoluble
E-mail addresses: tomas.jelinek.md@gmail.com (T. Jelinek), roman.hajek@fno.cz
fibrils causing damage to vital organs (Bhat et al., 2010). Although
(R. Hajek).

http://dx.doi.org/10.1016/j.critrevonc.2016.01.004
1040-8428/© 2016 Elsevier Ireland Ltd. All rights reserved.
250 T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260

AL is a very rare disease, it is the most common systemic amyloi- (Landau et al., 2014). Complete remissions can be expected in
dosis with an incidence of 10 patients per million cases per year, 30–40% of cases using this therapeutical approach (Dispenzieri
resulting in approximately 5000 new patients/year in the European et al., 2013; Gertz et al., 2004; Sanchorawala et al., 2007a; Skinner
Union (Merlini and Palladini, 2013). Due to the nature of the disease, et al., 2004). Only one prospective multicentric randomized clin-
patients usually present with multi-organ dysfunction (involving ical trial comparing ASCT with oral melphalan + dexamethasone
the cardiac, renal, gastrointestinal, peripheral and autonomic ner- was conducted, but the results did not support the superiority of
vous system), which is essential to their prognosis. The presence ASCT. Conversely, median OS was only 22.2 months in the ASCT
and extent of heart involvement is the most important prognostic group vs. 56.9 months in the melphalan + dexamethasone group
determinant, and cardiac dysfunction may be best assessed by the (p = 0.04) (Jaccard et al., 2007). However, these results must be
cardiac biomarkers N-terminal pro-natriuretic peptide type-B (NT- interpreted very carefully because of high peritransplant mortal-
proBNP), brain natriuretic peptide (BNP) and troponins (Gatt and ity (24%) related to the inappropriate choice of transplant eligible
Palladini, 2013). A staging system based on these biomarkers was candidates and some protocol difficulties in the ASCT group.
revised by Kumar et al., (2012a) and allows to discriminate between In 2013 two large retrospective studies supporting the use of
low-risk, intermediate-risk and high-risk patients, who have signif- ASCT in suitable patients with AL amyloidosis were published. An
icantly different prognoses. According to this Mayo Clinic staging European Group for Blood and Marrow Transplantation (EBMT)
system, the physicians should choose the appropriate treatment analysis of 1315 patients with AL showed time progress in treat-
approach based on the fragility of patients. ment results and 1 year OS reached 91% in the period between
The goal of the treatment in AL amyloidosis is the rapid elimi- 2009–2010 in comparison to 65% in the period between 1997–1999
nation of toxic free light chains and the achievement of complete (Hegenbart et al., 2013). Another analysis from The Amyloidosis
hematological remission (Palladini et al., 2012a), which is necessary Center at Boston University with 593 patients with AL treated with
for amelioration of organ function (Merlini and Palladini, 2008). ASCT during last 19 years presented remarkable long-term sur-
Achieving a maximal hematological response is a crucial factor and vival results with a CR rate of 40%, median OS 6.7 years and 25% of
only under these conditions the organ treatment response can be patients living longer than 19 years after ASCT (Sanchorawala et al.,
expected. Reaching organ treatment response may last a long time 2014a). In conclusion, it is possible to state that with appropriate
(1–2 years) and is especially important in patients with advanced patient selection and a risk-adapted treatment approach, ASCT is a
cardiac involvement (Wechalekar et al., 2013). Disease response safe and effective modality for approximately 30% of patients with
and progression criteria are summarized in Table 1 (Comenzo et al., AL. The reported 10 year survival after ASCT is about 43% (Cordes
2012; Gertz et al., 2005; Palladini et al., 2012a). et al., 2012). The already mentioned adequate selection of patients
The conventional treatment of AL amyloidosis is based on a with limited cardiac involvement is indeed the most important fac-
combination of corticosteroids (prednisone, dexamethasone) and tor for the safety of ASCT. Patients who have troponin T > 0.06 ng/ml,
alkylators (melphalan, cyclophosphamide). This combination used NT-proBNP > 5000 pg/ml and systolic blood pressure < 100 mm Hg
to be a standard treatment option for a long time, first used in 1972 at diagnosis should not be considered for high-dose chemotherapy
(Jones et al., 1972). The effects of these two groups of compounds (Gatt and Palladini, 2013; Girnius et al., 2014).
are mutually potentiated. The oral combination of melphalan and Proteasome inhibitors (PIs) represent a very effective group of
prednisone is well tolerated in patients with AL, but for a bet- drugs used in the treatment of AL amyloidosis. The majority of clini-
ter treatment response, dexamethasone is usually used instead of cal studies were conducted with bortezomib. Although there are no
prednisone (Dhodapkar et al., 1997; Kyle et al., 1997; Skinner et al., available results of any prospective randomized clinical trials, data
1996). In the clinical trial combining oral melphalan and high- coming from ongoing clinical trials phase II are very encouraging. In
dose dexamethasone in patients ineligible for autologous stem patients with monotherapy of bortezomib, 67–69% achieved hema-
cell transplantation (ASCT), median progression-free and overall tological response and 24–38% achieved CR (Reece et al., 2011).
survival were 3.8 and 5.1 years, respectively, with an estimated The combination of bortezomib and dexamethasone achieved a
overall response rate (ORR) of 67% including complete remission hematological response in 80–94% of cases (Kastritis et al., 2007).
(CR) in 1/3 of patients. Oral combination of melphalan and high Incorporating cyclophosphamide with bortezomib and dexam-
dose dexamethasone has become widely accepted as a first line ethasone (CVD) proved clinical efficacy with an ORRORR in 81% (CR
treatment option for patients with AL amyloidosis ineligible for 42%) of patients, 2 year progression-free survival (PFS) and OS were
ASCT (Palladini et al., 2007, 2004). Interestingly, the same Italian 67% and 98%, respectively (Venner et al., 2012). These results were
group recently reported the results of a large retrospective study confirmed in many other clinical trials. Rapid treatment response
showing that the treatment outcome of this combination is depen- was also observed (Gatt and Palladini, 2013). Recently, a large ret-
dent on the fraction of patients with advanced cardiac amyloidosis, rospective matched control case study of 184 newly diagnosed
respectively on the doses of dexamethasone they are able to tol- patients with AL was published. The authors compared the out-
erate. Hematological response was achieved in 76% (CR in 31%) of come of 87 patients treated with bortezomib plus melphalan and
patients who were fit enough to receive melphalan with full-dose dexamethasone (BMDex) with that of 87 controls treated with
dexamethasone, while only 51% (CR 12%) of patients responded MDex. A higher proportion of complete responses were observed
to melphalan with attenuated dexamethasone (Palladini et al., in the BMDex cohort (42% vs. 19%), and this resulted in a higher
2014a,b). overall hematological response rate (69% vs. 51%), but it did not
Autologous stem cell transplantation (ASCT) was described for result in the survival improvement in the overall population. How-
the first time in 1998 as an effective treatment modality in AL ever, a significant survival advantage for BMDex was observed in
amyloidosis (Comenzo et al., 1998). The most commonly used patients without severe (New York Heart Association class III or IV)
myeloablative regimen is melphalan 200 mg/m2 , the same as in heart failure and with NT-proBNP < 8500 ng/l. Patients treated with
multiple myeloma patients, but with frequent dose reductions with full-dose dexamethasone had similar response rates and survival
consideration to renal or cardiac insufficiency. Recently, in the whether they received bortezomib or not. So, intermediate-risk
XIVth International Symposium on Amyloidosis, a cohort of 148 patients who are not fit enough to receive high-dose dexametha-
patients who had undergone risk adapted ASCT and had received sone are likely to have the greatest advantage from the addition
melphalan 100, 140 and 200 mg/m2 in 14%, 52% and 34% respec- of bortezomib to MDex (Palladini et al., 2014a,b). In conclusion, no
tively was presented. Interestingly, by multivariate analysis, the phase III or retrospective matched analysis indicates a clear signifi-
melphalan dose had no impact on overall survival () (p = 0.25) cant benefit in the terms of OS/PFS compared to standard treatment
T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260 251

Table 1
Hematological and organ response and progression criteria.

Criteria Response Progression

Hematological assessment
CR Normal serum free light chain ratio with negative serum Any detectable monoclonal protein or abnormal FLC ratio
and urinary immunofixation (FLC must double)
VGPR The difference in the free light chains (dFLC) less than
40 mg/l
PR A reduction in the dFLC greater than 50% 50% increase in serum M-protein to >0.5 g/dl or 50%
increase of 24 h urine protein to >200 mg/day
NR A less than 50% response in dFLC FLC increase of 50% to >10 mg/dl

Heart
NT-proBNP >30% and >300 ng/l decrease if baseline >30% and >300 ng/l increase
NT-proBNP ≥650 ng/l
cTn ≥33% increase
NYHA class response ≥two-class decrease if baseline NYHA class 3 or 4 EF (≥10% decrease)

Other organs
Kidney 50% decrease (at least 0.5 g/day) of 24 h urine protein 50% increase (at least 1 g/day) of 24 h urine protein to
(pre-treatment urine protein must be >0.5 g/day) >1 g/day
Creatinine and creatinine clearance must not worsen by OR
25% over baseline 25% worsening of serum creatinine or creatinine clearance
Liver 50% decrease in abnormal ALP value Decrease in liver size 50% increase of ALP above the lowest value
radiographically at least 2 cm
Peripheral nervous system Improvement in electromyogram nerve conduction Progressive neuropathy by electromyography or nerve
velocity (rare) conduction velocity

ALP—alkaline phosphatase; cTn—cardiac troponin; dFLC—difference between involved and uninvolved free light chain; EF—ejection fraction; NR—no response; VGPR—very
good partial response.

regimen and there is still no certainty about the role of upfront although the intensity of each effect may differ in various plasma
bortezomib in the overall patient population with AL amyloidosis cell disorders (Bartlett et al., 2004).
(Dispenzieri, 2014). This question should be answered in the near
future when the results of phase III randomized multicenter clinical
trial EMN03 comparing MDex vs. BMDex (bortezomib, melphalan, 2.1. Immunomodulatory effects
dexamethasone) will be published.
Immunomodulatory drugs are highly effective agents used in Survival of malignant plasma cells, as other tumor cells, is par-
the treatment of multiple myeloma and they have been tested in tially facilitated by the impaired endogenous immune surveillance
other monoclonal gammopathies including AL amyloidosis. This against tumor antigens (Zou, 2005). IMiDs are able to affect the
review is focused on current experiences with three representa- immune system in several ways that lead to the eradication of
tives of immunomodulatory agents: thalidomide, lenalidomide and malignant plasma cells.
the most current, pomalidomide (Table 2). The first effect is co-stimulation of T cells. T cell activation is
mediated via antigen specific T cell receptor (TCR), but also requires
a co-stimulation signal mostly provided by professional antigen-
presenting cells (APC). IMiDs are able to co-stimulate CD3+ T cells
2. Mechanism of action of immunomodulatory drugs that have been partially activated either by anti CD3+ or dendritic
cells (DC). The presence of IMiDs abolishes the requirement of the
Immunomodulatory drugs (IMiDs) are a group of therapeu- secondary co-stimulation signal from APCs (Davies et al., 2001).
tic compounds that are analogs of thalidomide, a glutamic acid IMiDs improve proliferation of both CD4+ and CD8+ T cells and
derivate. Thalidomide was first synthesized by Chemie Grünenthal augment the production of Th2 cytokines–interleukin 2 (IL-2) and
in 1954 and was broadly used as an antiemetic agent for morning interferon ␥ (IFN-␥). The higher production of IL-2 in T cells is due to
sickness during pregnancy. In early 1960s its teratogenic effect was the depletion of IKZF1 and IKZF3, but this exact mechanism will be
recognized and the agent was immediately withdrawn from most discussed later (Krönke et al., 2014). Higher levels of IL-2 and IFN-
markets (Rajkumar, 2004). Nevertheless, around the same time, ␥ increase the number of natural killer cells (NK cells), improve
it was serendipitously discovered that thalidomide is effective in their function and mediate the death of malignant plasma cells
the treatment of erythema nodosum leprosum, a disease charac- (Sedlarikova et al., 2012).
terized by high levels of serum TNF␣ (Iyer et al., 1971). This ability Natural killer T cells (NKT cells) are T lymphocytes which bear
of thalidomide to inhibit production of TNF␣ by activated human natural killer cell markers on their surfaces and they possess a
monocytes was presented in 1991 (Sampaio et al., 1991). Besides direct tumor cytotoxic effect. NK cells have an irreplaceable role in
this anti-inflammatory effect, other mechanisms of action were innate immunity, in killing both tumor and virus infected cells. The
gradually discovered such as its immunomodulatory effect, direct presence of thalidomide, lenalidomide and pomalidomide enhance
antitumor activity, anti-angiogenic activity and its effect on the both NKT and NK cells that lead to the death of clonal plasmocytes
bone marrow microenvironment. Its exact mechanism of action, (Davies et al., 2001; Quach et al., 2010).
however, has remained unclear for a long time. Recent discoveries Another possible effect of IMiDs is the inhibition of regulatory
have brought to light how these drugs exactly work (Krönke et al., T cells (Tregs) that probably play an important role in establishing
2014; Lu et al., 2014). All these properties of IMiDs were dominantly and maintaining immunological unresponsiveness to self-antigens.
investigated in multiple myeloma, the most common malignant Several studies have shown that Tregs are expanded in hema-
monoclonal gammopathy (Quach et al., 2010; Sedlarikova et al., tological malignancies and also in solid tumors and that IMiDs
2012). As AL amyloidosis is also a clonal plasma cell dyscrasia, it is inhibit proliferation of Tregs via decreased FoxP3 mRNA expres-
possible to presume that majority of IMiD’s effects will be identical, sion (Muthu Raja et al., 2011; Sedlarikova et al., 2012). However, as
252 T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260

there are many contradictory results, a conclusion cannot be drawn


at this moment.

2.2. Direct antitumor effects


approvala
EMA/FDA

NO/NO

NO/NO

NO/NO
Immunomodulatory drugs also possess a direct anti-
proliferative effect via the inhibition of the cyclin-dependent

Pomalyst, Imnovid
kinase (CDK) pathway, downregulation of anti-apoptotic proteins
Thalomid, Myrin
Talidex, Talizer,

and activation of FAS-mediated cell death. The IMiD-induced cell


Inmunoprin,
Trade name

cycle arrest of malignant plasmocytes in G1 phase is mediated


Revlimid via increased levels of the p21WAF-1 protein through epigenetic
modifications (Hideshima et al., 2000; Sedlarikova et al., 2012).
Clonal plasma cells are protected against apoptosis by anti-
Prophylaxis of VTE

apoptotic proteins regulated via NF-␬B transcription factor (Wang


et al., 1998). It was demonstrated that treatment with IMiDs
downregulated the activity of this transcription factor in clonal
plasmocytes (Chu et al., 1997).
The exact mechanism of the antitumor activity of IMiDs has been
YES

YES

YES

recently described by several groups of investigators (Krönke et al.,


thrombocytopenia,

thrombocytopenia,

2014; Lu et al., 2014). It seems that IKZF1 (Ikaros) and IKZF3 (Aio-
VTE, fatigue, skin
VTE, obstipation,
polyneuropathy,

los), two lymphoid transcription factors playing central roles in the


Frequent AEs

Neutropenia,

Neutropenia,

biology of B and T cells, are responsible for the antitumor effect


drowsiness
Peripheral

of lenalidomide and all immunomodulatory agents. It was shown


anemia,

that lenalidomide causes selective ubiquitination and degradation


rash

VTE

of these two transcription factors by the CRBN-CRL4 (cereblon-


cullin4A-RING) E3 ubiquitin ligase. IKZF1 and IKZF3 are essential
transcription factors for terminal differentiation of B and T cell lin-
eages. Lenalidomide causes increased binding of IKZF1 and IKZF3
Neuropathy

to cereblon (CRBN) and promotes their ubiquitination and degra-


dation. It seems that CRBN is essential for lenalidomide activity in
+++

multiple myeloma. So, in aggregate it was shown that lenalidomide


+

acts via a previously unknown mechanism of action-enforced bind-


ing of the substrate receptor CRBN to IKZF1 and IKZF3, which results
Teratogenicity

in the degradation and depletion of IKZF1 and IKZF3 (Krönke et al.,


2014; Licht et al., 2014; Lu et al., 2014).
+++

+++

+++

2.3. Anti-angiogenic activity


Chemical formula

All IMiDs possess an anti-angiogenic effect, although it is gen-


erally considered that this activity prevails in thalidomide whereas
C13 H10 N2 O4

C13 H13 N3 O3

C13 H11 N3 O4

lenalidomide and pomalidomide have far greater immune enhanc-


ing effects (Dredge et al., 2002). This property of thalidomide is
probably dominantly responsible for the development of malfor-
mations in children in the late 1950s and early 1960s. It appears
that IMiDs modulate chemotactic factors affecting endothelial cell
migration such as TNF␣, VEGF and ␤FGF secreted by bone marrow
stromal cells (Dredge et al., 2002). It seems that PI3 K/Akt-signalling
pathway plays a key role in anti-angiogenic activity of IMiDs
because immunomodulatory drugs reduce Akt phosphorylation,
thus they affect the expression of these chemotactical factors and
therefore restrain angiogenesis (Dredge et al., 2005; Sedlarikova
et al., 2012).

2.4. Anti-inflammatory effects

The relative contribution of anti-inflammatory properties of


VTE —venous thromboembolism.

IMiDs toward their anti myeloma activity is uncertain (Quach et al.,


In AL amyloidosis indication.
Basic characteristics of IMiDs.

2010). Cyclooxygenase 2 (COX-2), which catalyzes the conversion


Structure

of arachidonic acid to various prostaglandins (PG), is involved in


the pathogenesis of several malignancies including plasma cell
dyscrasias (Masferrer et al., 2000). For instance, PG-E2 promotes
Pomalidomide

tumor induced angiogenesis, production of IL-6 and, of course,


Lenalidomide
Thalidomide

inflammation (Hinson et al., 1996). Thalidomide, lenalidomide and


pomalidomide are able to inhibit the expression of COX-2 via the
Table 2

Drug

shortening of the half life of COX-2 mRNA in a dose dependent


a

manner (Payvandi et al., 2004).


T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260 253

Another mechanism of the anti-inflammatory action of IMiDs loidosis. The data of 500 enrolled patients showed that 58% of
is their inhibitory effect on TNF␣ (an inflammatory cytokine pro- patients were treated with a thalidomide-based regimen (mainly
duced by monocytes and macrophages), which is mediated via the CTD), 51% of all patients reached a hematological response with
degradation of TNF␣ mRNA (Sampaio et al., 1991). In compari- a complete response in 14%. The estimated survival at 12, 36 and
son with thalidomide, inhibition of TNF␣ was 2000× more potent 48 months was 67%, 56% and 51%, respectively. Toxicity grade 3
with lenalidomide and 20,000× more potent with pomalidomide or higher was seen in 59% of all patients (Gillmore et al., 2012;
(Muller et al., 1999). Therefore, immunomodulatory agents may Wechalekar et al., 2014). These results show that the toxicity of this
have a therapeutical potential not only in multiple myeloma or regimen remains high and in the UK bortezomib-based regimens
other malignancies, but also in inflammatory conditions such as are being used increasingly. A retrospective matched comparison of
Crohn’s disease or rheumatoid arthritis (Akobeng and Stokkers, cyclophosphamide, bortezomib and dexamethasone (CVD) vs. risk-
2009; Quach et al., 2010). adapted cyclophosphamide, thalidomide and dexamethasone was
published recently by the same London group. The overall response
rates were 71.0% vs. 79.7% in the CVD and CTD arms, respectively,
2.5. Effects on the bone marrow microenvironment
(p = 0.32). A higher CR rate was observed in the CVD arm (40.5%) vs.
CTD (24.6%), (p = 0.046). 1 year OS was 65.2% and 66.7% for CVD and
Osteolytic lesions and back pain are usually the first symptoms
CTD, (p = 0.87) and the median PFS was 28.0 and 14.0 months for
that force patients with multiple myeloma to visit their physicians.
CVD and CTD, respectively (p = 0.039). In summary, CVD correlated
Immunomodulatory drugs possess many properties influencing the
with improved depth of response and superior PFS thus supporting
bone marrow microenvironment and interactions between malig-
its use in the frontline setting, but there was no difference in the
nant plasma cells, including the inhibition of osteoclastogenesis
terms of OS (Venner et al., 2014).
(Breitkreutz et al., 2008) or changes in the expression of adhesive
The toxicity of thalidomide containing regimens is not negli-
molecules important for the proliferation and migration of plas-
gible. Toxicities of grade 3 and higher were present in more than
mocytes (Geitz et al., 1996). In AL amyloidosis, however, osteolytic
50% of treated patients practically in all published studies. Fluid
lesions are extremely rare, so the the detailed description of these
retention, symptomatic bradycardia, peripheral neuropathy and
mechanisms will not be mentioned in this part.
progression of renal failure are the most common (Gillmore et al.,
2012; Palladini et al., 2005; Seldin et al., 2003). Thalidomide in com-
3. Thalidomide in the treatment of AL amyloidosis bination with other drugs shows clinical effectivity but its toxicity
is limiting. These combinations represent the treatment modality
Thalidomide is the first and principal drug of the immunomod- for patients refractory to bortezomib or for patients who tolerate
ulatory agents group. In biochemical nomenclature it is called thalidomide’s toxic effects.
2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (C13 H10 N2 O4 ), a
synthetic derivate of glutamic acid. Thalidomide exists as a racemic
mixture of two optically active enantiomers, S and R, wherein the S 4. Lenalidomide in the treatment of AL amyloidosis
enantiomer is responsible for the teratogenic and anti-tumor effect,
while the R enantiomer possesses a sedative effect (Rajkumar, Lenalidomide,3-(4-amino-l-oxo-l,3-dihydro-2H-isoindol-2-
2004). yl)piperidine-2,6-dione (C13 H13 N3 O3 ), has a preserved chiral
Low dose thalidomide used as monotherapy in the treatment center and occurs as a racemate, in a mixture of two optically
of AL amyloidosis is not very efficient (Dispenzieri et al., 2003; active enantiomers, S and R, similar to thalidomide (Kotla et al.,
Seldin et al., 2003). On the contrary, high doses of thalidomide 2009). Lenalidomide (Revlimid, formerly CC-5013) is ranked
are not well tolerated in this fragile group of patients (Dispenzieri among thalidomide’s analogs (Celgene’s class of immunomodula-
et al., 2003) either. The combination of thalidomide and dexam- tory compounds) with a more favorable toxic profile and increased
ethasone achieved 48% of hematological responses and 19% of efficacy. This second generation IMiD showed approximately
hematological complete remissions in the group of patients with 50–2000× higher effectivity depending on the type of “in vitro”
relapsed/refractory AL (Palladini et al., 2005). The use of thalido- test compared to thalidomide. Unfortunately, its teratogenic
mide and dexamethasone as adjuvant chemotherapy after ASCT in potential is preserved (Quach et al., 2010; Sedlarikova et al., 2012).
patients with newly diagnosed AL, who had not obtained CR after Lenalidomide in monotherapy is not an appropriate treatment
ASCT, was also investigated. The benefits of adjuvant therapy were modality for patients with AL amyloidosis because of low efficacy
seen in 42% (13/31) of patients who had an improvement in hema- and inadequate toxicity, especially in patients with renal insuffi-
tological response at 12 months post-SCT, including seven who ciency. Initial doses of lenalidomide should not exceed 15 mg/day
achieved a CR after having stable disease (SD) or partial remission (Adam et al., 2013; Dispenzieri et al., 2007). In 2007, two phase-
(PR) initially (Cohen et al., 2007). Patients with advanced cardiac AL II studies with a combination of lenalidomide and dexamethasone
amyloidosis (NYHA IV), who received a combination of oral mel- (Len/Dex) were conducted mostly in relapsed/refractory patients
phalan, thalidomide and dexamethasone, reached hematological with AL. The hematological overall response rates ranged from 41%
response in 36%, but 27% of them died during treatment (Palladini to 67% (Dispenzieri et al., 2007; Sanchorawala et al., 2007b). The
et al., 2009). The combination of another alkylator, cyclophos- most common treatment-related adverse event (AE) was myelo-
phamide, with thalidomide and dexamethasone (regimen CTD) suppression in 45% and 35% of patients, respectively. Fatigue, skin
was used in 75 transplant ineligible patients with advanced AL rash and thromboembolic complications represent other frequent
amyloidosis, including 44 patients (56.6%) with clonal relapse to side effects. Another study with Len/Dex was published in 2012 by
prior therapy. Hematological response occurred in 48 out of 65 Palladini et al., in which this combination was used as salvage ther-
(74%) evaluable patients, including a complete response in 14 (21%) apy for 24 patients refractory to bortezomib, melphalan and in 79%
patients. The median estimated OS from commencement of treat- also to thalidomide. Hematological response was observed in 41%
ment was 41 months (Wechalekar et al., 2007). The ALChemy of patients, but 50% of patients experienced severe adverse events
trial, one of the largest prospective observational studies in AL (Palladini et al., 2012b). The most recent study with Len/Dex was
amyloidosis worldwide, is currently being conducted at the UK conducted in 84 AL amyloidosis patients with relapsed/refractory
Amyloidosis Centre in London. Recently, the update of this study clonal disease following prior treatment with thalidomide (76%)
was presented in the XIVth International Symposium on Amy- and/or bortezomib (68%). The overall hematological response rate
254 T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260

was 61%, including 20% of complete responses. While the median symptoms of autonomic neuropathy (14 patients) and infection (8
OS has not been reached, the 2 year OS and PFS rates were 84% and patients). Treatment with MLD was effective and feasible in this
73%, respectively. 16% of patients achieved an organ response at 6 cohort of mostly elderly patients including patients with advanced
months, with a marked improvement in organ responses in patients cardiac involvement (Bochtler et al., 2013).
on long term therapy (median duration 11 months) and 55% achiev- The use of lenalidomide in combination with an alkylator in
ing renal responses by 18 months. The rate of renal responses these pre-treated groups of patients with AL amyloidosis was
among patients who received prolonged treatment was unexpect- frequently associated with toxicities grade 3/4, dominantly with
edly high, raising the possibility that immunomodulatory effects hematological toxicity and fatigue (Bochtler et al., 2013; Kastritis
of lenalidomide therapy might enhance the otherwise slow natural et al., 2012; Kumar et al., 2012a; Palladini et al., 2013; Sanchorawala
regression of amyloid deposits (Mahmood et al., 2014b). The combi- et al., 2013). The higher frequency of severe adverse events is
nation of Len/Dex showed clinical activity in the pre-treated group expectable considering the fragility of this population of patients
of patients with AL amyloidosis and represent a valuable option as and the fact that these chemotherapeutical regimens have not been
salvage therapy, but the treatment is accompanied by significant optimized yet. The results of the recent clinical trial conducted by
toxicity (Gatt and Palladini, 2013; Palladini et al., 2012b). Schönland et al. show that lenalidomide possesses a high therapeu-
The complete response rate remained low (0–29%) with the tic potential in patients with AL amyloidosis. Accompanied toxicity
Len/Dex regimen, therefore a number of phase II studies have may be associated with a different metabolic pathway of lenalido-
focused on the addition of an alkylator to this combination. Three mide in comparison with thalidomide and pomalidomide. The
independent clinical trials with incorporated cyclophospamide pharmacokinetics of lenalidomide is connected with renal func-
were conducted (regimen CLD) (Kastritis et al., 2012; Kumar et al., tion, because the excretion of the drug is provided dominantly by
2012a; Palladini et al., 2013). The hematological response rates the kidneys. The regimens containing lenalidomide may be a good
ranged from 55% to 62% and CR rates remained low—approximately treatment alternative for patients refractory to bortezomib or for
10%. The toxicity was prominent in all these studies with more than patients with polyneuropathy.
60% of patients reporting severe adverse events, including mainly The use of lenalidomide in the first line treatment in AL amy-
hematological toxicity in approximately 40% of cases. Fluid reten- loidosis should be investigated and its use should be optimized in
tion, fatigue, skin rash and gastrointestinal problems rank among clinical trials, especially in patients with no renal insufficiency.
other frequent side effects. Results from another clinical trial with
CLD regimen in 28 newly diagnosed AL amyloidosis patients not
eligible for ASCT were recently presented by Cibeira et al. The
treatment schedule consists of 12 cycles of CLD followed by main- 5. Pomalidomide in the treatment of AL amyloidosis
tentance with low-dose lenalidomide (10 mg) and dexamethasone
until intolerance or disease progression. The overall hematological Pomalidomide (CC-4047, Actimid, Imnovid, Pomalyst) is the
response rate was 46%, including complete response in 14%. The third immunomodulatory agent used in clinical practice. The
organ response rate was 32%. Serious adverse events were reported chemical formula of pomalidomide is 4-amino-2-(2,6-dioxo-3-
in 25% of cases (Cibeira et al., 2014). piperidyl)isoindoline-1,3-dione (C13 H11 N3 O4 ). This compound is
The addition of melphalan to lenalidomide and dexamethasone derived from thalidomide by adding an amino group to position
(regimen MLD) is other logical combination considering that the 4 of the phthaloyl ring and exists in two forms, enantiomers S and
regimen Mel/Dex has been the golden standard of the treatment R (Sedlarikova et al., 2012). This thalidomide analogue possesses
of AL amyloidosis to date. In the study published by Moreau et al., pleiotropic effects on myeloma cells. Most of these mechanisms
26 patients with newly diagnosed AL were treated with the MLD are probably effective also on clonal plasmocytes in AL amyloidosis
regimen, hematological response was observed in 58% of patients (Quach et al., 2010). Pomalidomide inhibits angiogenesis, induces
with CR in 23%. The maximum tolerated dose of lenalidomide was apoptosis of multiple myeloma cells, has strong immunomodula-
15 mg/day and the dose of melphalan was 0.18 mg/kg/day (Moreau tory abilities and is the most effective in TNF-mRNA degradation in
et al., 2010). In another study with MLD in a more pre-treated group comparison to thalidomide and lenalidomide (Dredge et al., 2002;
of patients, hematological partial and complete responses were Muller et al., 1999).
achieved by 43% and 7% of patients, respectively. Grade 3/4 tox- Pomalidomide is the newest immunomodulatory agent used in
icities were experienced by 88% patients, with myelosuppression the treatment of AL amyloidosis. Pomalidomide and dexametha-
being the most common (Sanchorawala et al., 2013). sone (regimen Pom/Dex) were investigated in the treatment of
The largest phase II clinical study using lenalidomide in AL amy- this disease, but only in a limited number of patients. A phase II
loidosis was presented at ASH 2013 by Schönland and his colleagues clinical trial with 33 patients with refractory/relapsed AL amyloi-
from Heidelberg (Bochtler et al., 2013). 50 patients with newly diag- dosis was conducted by investigators from Mayo Clinic Rochester
nosed AL amyloidosis ineligible for HDM-ASCT were enrolled in this (Dispenzieri et al., 2012). Patients were treated with pomalidomide
prospective single center trial. The treatment schedule was 6 cycles 2 mg/day for 28 days (1 cycle) along with orally administered dex-
of oral treatment with lenalidomide 10 mg/day, days 1–21, melpha- amethasone 40 mg/week. A majority (82%) of patients had cardiac
lan 0.15 mg/kg/day, days 1–4 and dexamethasone 20 mg/day, days involvement, 25% had Mayo cardiac stage III disease. The hema-
1–4, every four weeks. Hematological response was achieved in 78% tological response was rapid (median 1.9 months) and was seen
of patients: CR in 9 (20%) and PR in 26 (58%) of 45 evaluable patients, in 48% of patients with 3% of complete response, organ treatment
respectively. Organ response was observed in 5 (10%) patients at response was achieved in 18% of patients. Remarkably, hematolog-
the end of the study (6 months after the end of treatment). In ical responses to pomalidomide were seen in 40–50% of patients
comparison to the historical Mel/Dex group from the same cen- pre-treated with lenalidomide or bortezomib. The median overall
ter (patients with the same inclusion and exclusion criteria), there survival rate was 27.9 months with a 1year OS rate of 76%. Median
was a significantly higher hematological response rate in the MLD and 3 year PFS were 14.1 months and 17%, respectively. The toxicity
group (78% vs. 58%, p = 0.06) and the improvement was also seen in profile of the regimen was manageable, although grade 3 or higher
median OS (not reached vs. 26 months, p = 0.03). Overall, toxicity AEs at least possibly related to treatment were reported in 52%
was manageable in most patients, the most common being hema- (non-hematological) and 91% (hematological) (Dispenzieri et al.,
tological toxicity (neutropenia in 76% patients). The most common 2014, 2012). The NT-proBNP reduction did not follow the hema-
non-hematological AE was the worsening of cardiac function or tological response similar to what has been seen in other studies
T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260 255

Table 3
Results of clinical trials including IMiDs in AL amyloidosi.

Regimen Number of patients Hematologic Organ response (%) The most common Median OS (months) or
(%—newly response (%) (CR adverse events grade 1–3 year survival (%)
diagnosed) [%]) 3/4 (%)

Thalidomide containing regimens


Thalidomide Seldin et al., 2003 16 (0%) 25% (0%) 0% Overall 50% Not specified
Fatigue/sedation 38%
Thal/Dex Palladini et al., 2005 31 (0%) 48% (19%) 26% Overall 65% Not specified
Symptomatic bradycardia (26%)
CTD Wechalekar et al., 2007 75 (41%) 74% (21%) 33% Overall 32% 41
MTD Palladini et al., 2009 22 (73%) 36% (5%) 18% Overall 27% 5.3
CTD Venner et al., 2014 69 (100%) 80 (25%) 27% Not specified 67% (1 year OS)

Lenalidomide containing regimens


Len/Dex Dispenzieri et al., 2007 22 (41%) 41% (not specified) 23% Overall 86% Not specified
Neutropenia 45%
Len/Dex Sanchorawala et al., 2007b 34 (9%) 67% (29%) 41% Myelosuppression 35% Not specified
Fatigue 35%
Len/Dex Palladini et al., 2012b 24 (0%) 41% (0%) 4% Overall 50% 14
Thrombocytopenia 17%
Len/Dex Mahmood et al., 2014 84 (0%) 61% (20%) Not specified Not specified 84% (2 year OS)
CLD Kumar et al., 2012b 35 (69%) 60% (11%) 31% Overall 74% 37.8
Neutropenia 40%
CLD Palladini et al., 2013 21 (0%) 62% (5%) 19% Overall 57% 36
CLD Kastritis et al., 2012 37 (65%) 55% (8%) 22% Neutropenia 34% 41% (2 year OS)
CLD Cibeira et al., 2015 28 (100%) 46% (14%) 32% Overall 25% Not specified (study ongoing)
MLD Moreau et al., 2010 26 (100%) 58% (23%) 50% Overall 81% 81% (2 year OS)
Neutropenia 11%
MLD Bochtler et al., 2013 50 (100%) 78% (20%) 10% (at six months) Neutropenia 76% Not achieved
MLD Sanchorawala et al., 2013 16 (69%) 50% (7%) 11% Overall 88% Not achieved
Myelosuppression 57%

Pomalidomide containing regimens


Pom/Dex Dispenzieri et al., 2012 33 (0%) 48% (3%) 15% Hematologic 91% 27.9
Non-hematologic 52%
Pom/Dex Milani et al., 2013 27 (0%) 67% (18%—VGPR) (short-term follow-up) Overall 67% (short-term follow-up)
Neutropenia 30%
Pom/Dex Sanchorawala et al., 2014b 12 (0%) Not specified (22%) Not specified Overall 50% Not specified (study ongoing)

of patients with AL amyloidosis taking immunomodulatory drugs 6. Problematics of response assessment with IMiDs
(Dispenzieri et al., 2010; Gibbs et al., 2009; Tapan et al., 2010).
A second clinical trial with high dose (4 mg/day) pomalidomide NT-proBNP and BNP are important and useful biomarkers of car-
and oral dexamethasone (40 mg/week) was conducted by Milani diac involvement, especially of heart failure. The prognostic value
et al. and its preliminary results were presented at ASH 2013. 27 of these biomarkers in AL amyloidosis is indisputable, but they also
patients with relapsed/refractory AL previously exposed to an alky- serve to assess the organ (cardiac) treatment response. Neverthe-
lator or bortezomib were enrolled in the trial and 41% had also less, the use of immunomodulatory agents may cause an elevation
received an immunomodulatory agent. Overall, 18 patients (67%) of cardiac biomarkers in a significant proportion of patients usu-
achieved a hematological response, with 5 (18%) very good par- ally during the first cycle of chemotherapy. This phenomenon may
tial responses. Moreover, of the 7 patients previously exposed to complicate the assessment of organ treatment response in patients
lenalidomide, 5 responded to pomalidomide and the 3 patients treated with IMiDs.
who were refractory to ixazomib responded as well, indicating that Gibbs et al. described this phenomenon in 51 patients who
pomalidomide can overcome resistance to other immunomodula- achieved complete hematological response measured by the
tory drugs and a second-generation proteasome inhibitor. Overall, decrease of serum free light chains (FLC). Overall, 42 patients were
18 patients (67%) experienced severe (grade 3–4) adverse events: treated with a regimen based on thalidomide (CTD) and 9 patients
neutropenia (n = 8, 30%), fluid retention (n = 7, 26%, all with heart were treated with melphalan and dexamethasone (Mel/Dex). An
involvement), infection (n = 3, 11%), less common were neuropathy increase in NT-proBNP from baseline was seen in 36 (71%) patients
and rash. As previously reported with other immunomodulatory at 6 months. A subsequent fall in their NT-proBNP levels was seen
drugs, pomalidomide was associated with an increase in NT- at 12 months in 33 (92%) of these patients. There was no signifi-
proBNP during cycle 1 (Milani et al., 2013). cant rise in creatinine serum level at 6 months, suggesting that the
The preliminary data of the most recent clinical trial with rise in NT-proBNP was not due to a change in the renal function.
Pom/Dex in 12 patients with relapsed AL were presented at the Importantly, there was no difference in the rate of NT-proBNP rise
XIVth International Symposium on Amyloidosis by Sanchorawala when comparing the CTD regimen to Mel/Dex. It is clearly shown
et al. Hematological complete response was observed in 2 of 9 that NT-proBNP values can rise transiently in the immediate post
evaluable patients (22%), toxicity grade 3 or higher was reported chemotherapy period in nearly three quarters of patients with a
in 6 cases (50%). Other data were not specified but the trial is still complete FLC response in this analysis. The exact mechanism of
ongoing (Sanchorawala et al., 2014b). this phenomenon has remained unclear and it has not impacted
These results show good efficacy of pomalidomide and dex- the rate of clonal relapse or overall survival (Gibbs et al., 2009).
amethasone in patients with refractory AL amyloidosis previously Tapan et al. described the rise of BNP level (> 30% increase from
exposed also to other IMiDs. Further studies to optimize this potent baseline) in 86% of patients treated with lenalidomide and dexam-
rescue regimen are warranted. ethasone (Len/Dex). In 68% of patients the increase occured during
the first cycle of chemotherapy. All the patients with an increase
256
Table 4
Ongoing clinical trials with IMiDs in AL amyloidosis.

Title Regimens Condition Estimated enrollment Identifier

Experimental arm Active comparator

A phase I/II clinical trial of pomalidomide Pomalidomide orally 1 mg/day, days X Untreated systemic AL 54 patients NCT01807286
with melphalan and dexamethasone in 1–21; amyloidosis
patients with newly diagnosed untreated Melphalan orally 9 mg/m2 /day days
systemic AL amyloidosis 1–4;
Dexamethasone orally 40 mg/day days
1–4;
28 days long cycle
An open-label, phase II study of Pomalidomide orally 2–4 mg/day days X Primary amyloidosis of 28 patients NCT01510613
pomalidomide and dexamethasone (PDex) 1–28; light chain type

T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260


for previously treated patients with AL Dexamethasone orally 20–40 mg, days
amyloidosis 1,8,15,22;
28 days long cycle, until progression or
unacceptable toxicity
Phase I study of pomalidomide, Pomalidomide orally days 1–21; X Primary systemic 36 patients NCT01728259
bortezomib, and dexamethasone (PVD) as Bortezomib iv. days 1,8,15; amyloidosis
first-line treatment of AL amyloidosis or Dexamethasone orally 1,8,15,22;
light chain deposition disease 28 days long cycle, until progression or
unacceptable toxicity
A phase 3, randomized, controlled, MLN9708 4 mg orally days 1,8,15; Physician’s choice Relapsed or refractory 248 patients NCT01659658
open-label, multicenter, safety and systemic light chain
efficacy study of dexamethasone plus amyloidosis
MLN9708 or physician’s choice of
treatment administered to patients with
relapsed or refractory systemic light chain Dexamethasone 20 mg/day orally days
(AL) amyloidosis 1,8,15,22;
28 days long cycle
A phase I/II trial of pomalidomide and Pomalidomide orally 2–4 mg/day, days X Previously-treated AL amyloidosis 35 patients NCT01570387
dexamethasone in subjects with 1–28;
previously-treated AL amyloidosis Dexamethasone orally 10–20 mg, days
1,8,15,22;
28 days long cycle, until progression or
unacceptable toxicity
A phase II trial of mrd (melphalan, Dexamethasone 20 mg/day orally days X Primary systemic 35 patients NCT00679367
lenalidomide and dexamethasone) for 1,8,15,22; amyloidosis
patients with AL amyloidosis Lenalidomide 10 mg/day, days 1–21;
Melphalan orally 5 mg/m2 /day days
1–4;
A phase I/II trial of lenalidomide combined Lenalidomide 5–25 mg/day orally, days X Primary (AL) systemic 37 patients NCT00981708
with cyclophosphamide and intermediate 1–21; amyloidosis
dose dexamethasone in patients with Dexamethasone 20 mg/day days 1–4;
primary (AL) systemic amyloidosis Cyclophosphamide 50–100 mg/day
orally days 1–10;
28 days long cycle
A multicentric, phase II trial of Lenalidomide 15 mg/day, day 1–21; X Primary systemic 30 patients NCT01194791
lenalidomide, cyclophosphamide and Cyclophosphamide 300 mg/m2 days amyloidosis (AL) newly
dexamethasone in patients with primary 1,8; diagnosed
systemic amyloidosis (AL) newly Dexamethasone 20 mg/day orally days
diagnosed, not candidates for 1–4 and 9–12;
hematopoietic stem cell transplantation 28 days long cycle, 6 cycles
T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260 257

in BNP were asymptomatic without association of modification in of patients has a poor prognosis dominantly because of exten-
NYHA class congestive heart failure (Tapan et al., 2010). Similarly, sive organ involvement and their treatment remains problematic
the results of clinical trials with pomalidomide showed the discrep- with the use of low-dose corticosteroids in combination with novel
ancy between an increase of cardiac biomarkers and hematological agents (Mahmood et al., 2014a). According to recent data pub-
response (Dispenzieri et al., 2012; Palladini et al., 2013). lished by Wechalekar et al. at EHA 2014, it seems that thalidomide
Even though this phenomenon is transient, it complicates the based regimens (TMD) are slightly beneficial compared to borte-
assessment and interpretation of treatment organ response in AL. zomib based regimens with an estimated survival rate of 75% vs.
It is useful to evaluate the dynamics of FLC levels and levels of 61% at four months for patients with a high-risk Mayo Clinic stage
cardiac biomarkers. Patients with AL amyloidosis receiving IMiDs (Wechalekar et al., 2014).
whose cardiac biomarkers rise should not be assumed to be fail- Immunomodulatory agents showed good efficacy in the treat-
ing therapy without other signs of disease progression, but should ment of AL and they may be used mainly in patients with
be monitored closely. Clinical hematologist-oncologists must care- relapsed/refractory disease but also as a part of first-line ther-
fully consider how to evaluate the organ treatment response, apy. The optimization of the regimen reflecting its toxicity plays
dominantly when the decision of changing therapy is about to be an important role. Due to the lack of randomized clinical trials,
made. Further data is needed for non-thalidomide based regimens it is difficult to decide which immunomodulatory drug is better.
to assess if this has an effect to the same extent. Pomalidomide may be a representative agent of this group with
a good balance between high effectivity and tolerable toxicity.
The summary of practically all published clinical trials containing
7. Treatment strategies in AL amyloidosis and role of IMiDs immunomodulatory agents is in Table 3.
Thalidomide in combination with other drugs showed good clin-
There is no consensus of optimal therapy for patients with ical activity in the treatment of AL, but its use is accompanied by
newly diagnosed AL amyloidosis, because there is minimum data significant toxicity. This IMiD may be a treatment alternative for
available from randomized clinical trials, because of the rarity of patients refractory to bortezomib or may be used also as a first
this disease and the small number of patients possibly enrolled. line treatment if well tolerated. Lenalidomide may be used also in
Often, the treatment strategy is based on expert opinions at a patients refractory to bortezomib or in patients with neuropathy.
national, European or global level. None of the 3 published ran- The toxicity of lenalidomide-containing regimens may be a prob-
domized clinical trials include novel agents such as proteasome lem, especially for patients with renal insufficiency. Pomalidomide
inhibitors or immunomodulatory agents. Thus, the most evident proved to have high efficacy in pilot studies and the lowest toxicity
benefit of novel agents is increased flexibility of our treatment strat- of all immonomodulatory agents and seems to be a very promis-
egy in newly diagnosed patients, because we can switch from a ing drug in the treatment of AL. The appropriate dosage of IMiDs
thalidomide-based to bortezomib-based regimen if not tolerated is important and it is possible to recommend 50–100 mg/day of
and/or effective. Moreover, we have now got several effective treat- thalidomide, 10–15 mg/day of lenalidomide, 2 mg/day or 4 mg/day
ment options in a relapse setting. of pomalidomide (based on available results of clinical studies).
The treatment strategy in patients with AL amyloidosis is shaped Concerning pomalidomide, it is not possible to decide which dose
by background factors including performance status, age, comor- level is better, because only two clinical trials have been conducted
bidities, contraindications to drugs and also patient preference. to date. Combinations of immunomodulatory drugs and a corti-
Risk stratification adopted from the Mayo Clinic staging system is costeroid well tolerated in multiple myeloma may represent an
crucial in the choice of treatment strategy and may be a helpful appropriate treatment option also in AL amyloidosis.
instrument for physicians as many of the patients are very fragile.
According to this risk adapted staging system based on the level of
cardiac biomarkers (NT-proBNP and Troponin T), patients with AL 8. Conclusion
are divided into three groups (Dispenzieri et al., 2004; Kumar et al.,
2012a). ASCT should always be considered as a part of front-line Immunoglobulin light chain amyloidosis is a rare disease, but
therapy in low-risk patients with AL amyloidosis. These patients the most common of the systemic amyloidoses. Patients are often
can be also treated with a combined bortezomib based regimen diagnosed in an advanced stage with multiorgan involvement and
which allows the safe harvest of peripheral stem cells and leaves their prognosis remains poor. Nevertheless, recently published
the performance of ASCT only in the case of not achieving complete data suggests that overall survival of patients eligible for ASCT
remission (Mikhael et al., 2012; Venner et al., 2012). Approximately with preserved cardiac function is surprisingly long and with a
50% of patients with newly diagnosed AL are in the intermediate life expectancy much longer than in multiple myeloma patients.
risk group, these patients are not suitable for undertaking ASCT The combination of melphalan and dexamethasone is still a very
as front line treatment. According to two recently published ret- good treatment option for transplant ineligible patients, but novel
rospective case control studies, there is still no certainty about agents may offer some benefits. Proteasome inhibitors and mainly
the best treatment option for this group of patients. Bortezomib- combined bortezomib based regimens seem to be a good choice in
based regimens yielded higher complete response rates, and these front-line therapy. However, thalidomide based regimens may be
responses were rapidly achieved, but there was no clear improve- used in this setting as well, if well tolerated. Any data of randomized
ment in overall survival. It was only in an Italian study where one clinical trials, which would support the use of one or another group
subgroup of patients with New York Heart Association class of <3 of agents are still not available. Recent trials with immunomodula-
and NT-proBNP of ≤8500 ng/l, who were not fit enough to recieve tory agents, on which this review is focused, show a good clinical
high dose dexamethasone, had a superior OS when bortezomib was efficacy, but the toxicity remains significant in this fragile group
added to Mel/Dex (Palladini et al., 2014a,b; Venner et al., 2014). The of patients. Pomalidomide seems to be a very promising drug with
results of the randomized phase III trial are still awaited. The last preserved high effectivity and favorable toxic profile in patients
interim analysis from ASH 2014 indicates that the addition of borte- with renal insufficiency as well. The development of a new gen-
zomib to Mel/Dex grants more profound hematologic responses eration of proteasome inhibitors, e.g. oral ixazomib (MLN9708),
that should be balanced with a relative increase in toxicity (Merlini gives physicians more options in the decision making process of
et al., 2014). Nevertheless, also thalidomide based regimens may be adequate treatment approach. As AL amyloidosis is a very uncom-
successfuly used in a selected cohort of patients. A high-risk group mon disorder, most patients should be treated within the clinical
258 T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260

trials. Active clinical trials with immunomodulatory drugs in AL D.G., Skinner, M., 1998. Dose-intensive melphalan with blood stem-cell
amyloidosis are summarized in Table 4. Also, novel therapies tar- support for the treatment of AL (amyloid light-chain) amyloidosis: survival
and responses in 25 patients. Blood 91, 3662–3670.
geted directly on amyloid and not on pre-existing malignant clone Cordes, S., Dispenzieri, A., Lacy, M.Q., Hayman, S.R., Buadi, F.K., Dingli, D., Kumar,
of plasmocytes may play a role in the future. Recently, the clini- S.K., Hogan, W.J., Gertz, M.A., 2012. Ten-year survival after autologous stem
cal trial phase III with NEOD001 has been initialized. NEOD001 is a cell transplantation for immunoglobulin light chain amyloidosis. Cancer 118,
6105–6109, http://dx.doi.org/10.1002/cncr.27660.
monoclonal antibody that targets the amyloid that affects both AL Davies, F.E., Raje, N., Hideshima, T., Lentzsch, S., Young, G., Tai, Y.T., Lin, B., Podar,
and AA amyloidosis. Finally, what seems to be the most important K., Gupta, D., Chauhan, D., Treon, S.P., Richardson, P.G., Schlossman, R.L.,
issue in clinical practise is an increasing awareness of this rare dis- Morgan, G.J., Muller, G.W., Stirling, D.I., Anderson, K.C., 2001. Thalidomide and
immunomodulatory derivatives augment natural killer cell cytotoxicity in
ease among physicians—dominantly cardiologists, nephrologists
multiple myeloma. Blood 98, 210–216.
and also hematologists. Dhodapkar, M.V., Jagannath, S., Vesole, D., Munshi, N., Naucke, S., Tricot, G.,
Barlogie, B., 1997. Treatment of AL-amyloidosis with dexamethasone plus
alpha interferon. Leuk. Lymphoma 27, 351–356, http://dx.doi.org/10.3109/
Conflict of interest 10428199709059690.
Dispenzieri, A., Lacy, M.Q., Rajkumar, S.V., Geyer, S.M., Witzig, T.E., Fonseca, R., Lust,
The authors have no conflict of interest to be disclosed. J.A., Greipp, P.R., Kyle, R.A., Gertz, M.A., 2003. Poor tolerance to high doses of
thalidomide in patients with primary systemic amyloidosis. Amyloid Int. J. Exp.
Clin. Investig. 10, 257–261, http://dx.doi.org/10.3109/13506120309041743.
Acknowledgements Dispenzieri, A., Gertz, M.A., Kyle, R.A., Lacy, M.Q., Burritt, M.F., Therneau, T.M.,
Greipp, P.R., Witzig, T.E., Lust, J.A., Rajkumar, S.V., Fonseca, R., Zeldenrust, S.R.,
McGregor, C.G.A., Jaffe, A.S., 2004. Serum cardiac troponins and N-terminal
TJ—wrote the paper pro-brain natriuretic peptide: a staging system for primary systemic
ZK—proofread the paper amyloidosis. J. Clin. Oncol. 22, 3751–3757, http://dx.doi.org/10.1200/JCO.2004.
03.029.
RH—concept, overall proofread, work coordination Dispenzieri, A., Lacy, M.Q., Zeldenrust, S.R., Hayman, S.R., Kumar, S.K., Geyer, S.M.,
This work was supported by the Moravian-Silesian Region Lust, J.A., Allred, J.B., Witzig, T.E., Rajkumar, S.V., Greipp, P.R., Russell, S.J., Kabat,
grants MSK 02680/2014/RRC and MSK 02692/2014/RRC; grants B., Gertz, M.A., 2007. The activity of lenalidomide with or without
dexamethasone in patients with primary systemic amyloidosis. Blood 109,
by MH CZ-DRO-FNOs/2014; by The Ministry of Education, Youth 465–470, http://dx.doi.org/10.1182/blood-2006-07-032987.
and Sports (Institutional Development Plan of University of Dispenzieri, A., Dingli, D., Kumar, S.K., Rajkumar, S.V., Lacy, M.Q., Hayman, S., Buadi,
Ostrava in 2015); projects SGS01/LF/2014-2015, SGS02/LF/2014- F., Zeldenrust, S., Leung, N., Detweiler-Short, K., Lust, J.A., Russell, S.J., Kyle, R.A.,
Gertz, M.A., 2010. Discordance between serum cardiac biomarker and
2015, SGS03/LF/2015-2016 and by grant IGA of The Ministry of
immunoglobulin-free light-chain response in patients with immunoglobulin
Health15-29667A. The authors want to give special thanks to Eva light-chain amyloidosis treated with immune modulatory drugs. Am. J.
Jarosova for administrative support. Hematol. 85, 757–759, http://dx.doi.org/10.1002/ajh.21822.
Dispenzieri, A., Buadi, F., Laumann, K., LaPlant, B., Hayman, S.R., Kumar, S.K., Dingli,
D., Zeldenrust, S.R., Mikhael, J.R., Hall, R., Rajkumar, S.V., Reeder, C., Fonseca, R.,
References Bergsagel, P.L., Stewart, A.K., Roy, V., Witzig, T.E., Lust, J.A., Russell, S.J., Gertz,
M.A., Lacy, M.Q., 2012. Activity of pomalidomide in patients with
Adam, Z., Sčudla, V., Krejčí, M., Cermáková, Z., Pour, L., Král, Z., 2013. Treatment of immunoglobulin light-chain amyloidosis. Blood 119, 5397–5404, http://dx.doi.
AL amyloidosis in 2012; the benefit of new drugs (bortezomib, thalidomide, org/10.1182/blood-2012-02-413161.
and lenalidomide). Summary of published clinical trials. Vnitr˘ní Lékar˘ství 59, Dispenzieri, A., Seenithamby, K., Lacy, M.Q., Kumar, S.K., Buadi, F.K., Hayman, S.R.,
37–58. Dingli, D., Litzow, M.R., Gastineau, D.A., Inwards, D.J., Micallef, I.N., Ansell, S.M.,
Akobeng, A.K., Stokkers, P.C., 2009. Thalidomide and thalidomide analogues for Johnston, P.B., Porrata, L.F., Patnaik, M.M., Hogan, W.J., Gertz, M.A.A., 2013.
maintenance of remission in Crohn’s disease. Cochrane Database Syst. Rev., Patients with immunoglobulin light chain amyloidosis undergoing autologous
http://dx.doi.org/10.1002/14651858.CD007351.pub2, CD007351. stem cell transplantation have superior outcomes compared with patients with
Bartlett, J.B., Dredge, K., Dalgleish, A.G., 2004. The evolution of thalidomide and its multiple myeloma: a retrospective review from a tertiary referral center. Bone
IMiD derivatives as anticancer agents. Nat. Rev. Cancer 4, 314–322, http://dx. Marrow Transplant 48, 1302–1307, http://dx.doi.org/10.1038/bmt.2013.53.
doi.org/10.1038/nrc1323. Dispenzieri, A., 2014. Still no certainty about the role of upfront bortezomib among
Bhat, A., Selmi, C., Naguwa, S.M., Cheema, G.S., Gershwin, M.E., 2010. Currents patients with AL amyloidosis. Leukemia 28, 2273–2275, http://dx.doi.org/10.
concepts on the immunopathology of amyloidosis. Clin. Rev. Allergy Immunol. 1038/leu.2014.271.
38, 97–106, http://dx.doi.org/10.1007/s12016-009-8163-9. Dispenzieri, A., Buadi, F., Laumann, K., LaPlant, B., Hayman, S.R., Kumar, S.K., Dingli,
Bochtler, T., Benner, A., Gawlik, M., Kimmich, C., Kristen, A.V., Beimler, J., Ho, A.D., D., Zeldenrust, S.R., Mikhael, J.R., Rajkumar, S.V., Reeder, C., Fonseca, R.,
Hegenbart, U., 2013. Lenalidomide/melphalan/dexamethasone chemotherapy Bergsagel, P.L., Stewart, A.K., Roy, V., Lust, J.A., Russell, S.J., Gertz, M.A., Lacy,
in 50 patients with newly diagnosed amyloid light chain amyloidosis: first M.Q., 2014. Long-term results of pomalidomide and dexamethasone for
results of a prospective single center phase 2 study (Leomex). Blood 122, patients with relapsed or refractory AL amyloidosis. Presented at the XIVth
1993–1993. International Symposium on Amyloidosis, Indianapolis.
Breitkreutz, I., Raab, M.S., Vallet, S., Hideshima, T., Raje, N., Mitsiades, C., Chauhan, Dredge, K., Marriott, J.B., Macdonald, C.D., Man, H.-W., Chen, R., Muller, G.W.,
D., Okawa, Y., Munshi, N.C., Richardson, P.G., Anderson, K.C., 2008. Stirling, D., Dalgleish, A.G., 2002. Novel thalidomide analogues display
Lenalidomide inhibits osteoclastogenesis, survival factors and anti-angiogenic activity independently of immunomodulatory effects. Br. J.
bone-remodeling markers in multiple myeloma. Leukemia 22, 1925–1932, Cancer 87, 1166–1172, http://dx.doi.org/10.1038/sj.bjc.6600607.
http://dx.doi.org/10.1038/leu.2008.174. Dredge, K., Horsfall, R., Robinson, S.P., Zhang, L.-H., Lu, L., Tang, Y., Shirley, M.A.,
Chu, Z.L., McKinsey, T.A., Liu, L., Gentry, J.J., Malim, M.H., Ballard, D.W., 1997. 1997: Muller, G., Schafer, P., Stirling, D., Dalgleish, A.G., Bartlett, J.B., 2005. Orally
Suppression of tumor necrosis factor-induced cell death by inhibitor of administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits
apoptosis c-IAP2 is under NF-kappaB control. Proc. Natl. Acad. Sci. U. S. A. 94, endothelial cell migration and Akt phosphorylation in vitro. Microvasc. Res. 69,
10057–10062. 56–63, http://dx.doi.org/10.1016/j.mvr.2005.01.002.
Cibeira, M.T., Oriol, A., Lahuerta, J.J., Mateos, M.V., de la Rubia, J., Hernández, M.T., Gatt, M.E., Palladini, G., 2013. Light chain amyloidosis 2012: a new era. Br. J.
Granell, M., Fernández de Larrea, C., San Miguel, J.F., Bladé, J., 2014. Phase II Haematol. 160, 582–598, http://dx.doi.org/10.1111/bjh.12191.
trial of lenalidomide, dexamethasone and cyclophosphamide (LENDEXAL) for Geitz, H., Handt, S., Zwingenberger, K., 1996. Thalidomide selectively modulates
previously untreated patients with light-chain amyloidosis. Presented at the the density of cell surface molecules involved in the adhesion cascade.
XIVth International Symposium on Amyloidosis, Indianapolis. Immunopharmacology 31, 213–221.
Cohen, A.D., Zhou, P., Chou, J., Teruya-Feldstein, J., Reich, L., Hassoun, H., Levine, B., Gertz, M.A., Lacy, M.Q., Dispenzieri, A., Ansell, S.M., Elliott, M.A., Gastineau, D.A.,
Filippa, D.A., Riedel, E., Kewalramani, T., Stubblefield, M.D., Fleisher, M., Nimer, Inwards, D.J., Micallef, I.N.M., Porrata, L.F., Tefferi, A., Litzow, M.R., 2004.
S., Comenzo, R.L., 2007. Risk-adapted autologous stem cell transplantation Risk-adjusted manipulation of melphalan dose before stem cell transplantation
with adjuvant dexamethasone + —thalidomide for systemic light-chain in patients with amyloidosis is associated with a lower response rate. Bone
amyloidosis: results of a phase II trial. Br. J. Haematol. 139, 224–233, http://dx. Marrow Transplant 34, 1025–1031, http://dx.doi.org/10.1038/sj.bmt.1704691.
doi.org/10.1111/j.1365-2141.2007.06783.x. Gertz, M.A., Comenzo, R., Falk, R.H., Fermand, J.P., Hazenberg, B.P., Hawkins, P.N.,
Comenzo, R.L., Reece, D., Palladini, G., Seldin, D., Sanchorawala, V., Landau, H., Falk, Merlini, G., Moreau, P., Ronco, P., Sanchorawala, V., Sezer, O., Solomon, A.,
R., Wells, K., Solomon, A., Wechalekar, A., Zonder, J., Dispenzieri, A., Gertz, M., Grateau, G., 2005. Definition of organ involvement and treatment response in
Streicher, H., Skinner, M., Kyle, R.A., Merlini, G., 2012. Consensus guidelines for immunoglobulin light chain amyloidosis (AL): a consensus opinion from the
the conduct and reporting of clinical trials in systemic light-chain amyloidosis. 10th International Symposium on Amyloid and Amyloidosis, Tours, France,
Leukemia 26, 2317–2325, http://dx.doi.org/10.1038/leu.2012.100. 18–22 April 2004. Am. J. Hematol. 79, 319–328, http://dx.doi.org/10.1002/ajh.
Comenzo, R.L., Vosburgh, E., Falk, R.H., Sanchorawala, V., Reisinger, J., Dubrey, S., 20381.
Dember, L.M., Berk, J.L., Akpek, G., LaValley, M., O’hara, C., Arkin, C.F., Wright,
T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260 259

Gibbs, S.D.J., De Cruz, M., Sattianayagam, P.T., Lachmann, H.J., Gillmore, J.D., Lu, G., Middleton, R.E., Sun, H., Naniong, M., Ott, C.J., Mitsiades, C.S., Wong, K.-K.,
Hawkins, P.N., Wechalekar, A.D., (2009). Transient Post Chemotherapy Rise in Bradner, J.E., Kaelin, W.G., 2014. The myeloma drug lenalidomide promotes the
NT Pro-BNP in AL Amyloidosis: Implications for Organ Response Assessment. cereblon-dependent destruction of Ikaros proteins. Science 343, 305–309,
[WWW Document]. BLOOD. URL http://discovery.ucl.ac.uk/93113/ (accessed http://dx.doi.org/10.1126/science.1244917.
8.20.14). Mahmood, S., Palladini, G., Sanchorawala, V., Wechalekar, A., 2014a. Update on
Gillmore, J.D., Lane, T., Rannigan, L., Foard, D., Gibbs, S.D.J., Pinney, J.H., Venner, C.P., treatment of light chain amyloidosis. Haematologica 99, 209–221, http://dx.
Banypersad, S., Lachmann, H.J., Wechalekar, A.D., Hawkins, P.N., 2012. doi.org/10.3324/haematol.2013.087619.
ALchemy—a large prospective real world study of chemotherapy in AL Mahmood, S., Venner, C.P., Sachchithanantham, S., Lane, T., Rannigan, L., Foard, D.,
amyloidosis Julian D Gillmore, Thirusha Lane, Lisa Rannigan, Darren Foard, Pinney, J.H., Gibbs, S.D.J., Whelan, C.J., Lachmann, H.J., Gillmore, J.D., Hawkins,
Simon DJ Gibbs, Jennifer H Pinney, Christopher P Venner, Sanjay Banypersad, P.N., Wechalekar, A.D., 2014b. Lenalidomide and dexamethasone for systemic
Helen J Lachmann, Ashutosh D Wechalekar Philip N Hawkins. Presented at the AL amyloidosis following prior treatment with thalidomide or bortezomib
XIIIth International Symposium on Amyloidosis. regimens. Br. J. Haematol. 166, 842–848, http://dx.doi.org/10.1111/bjh.12973.
Girnius, S., Seldin, D.C., Meier-Ewert, H.K., Sloan, J.M., Quillen, K., Ruberg, F.L., Berk, Masferrer, J.L., Leahy, K.M., Koki, A.T., Zweifel, B.S., Settle, S.L., Woerner, B.M.,
J.L., Doros, G., Sanchorawala, V., 2014. Safety and efficacy of high-dose Edwards, D.A., Flickinger, A.G., Moore, R.J., Seibert, K., 2000. Antiangiogenic and
melphalan and auto-SCT in patients with AL amyloidosis and cardiac antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res. 60,
involvement. Bone Marrow Transplant 49, 434–439, http://dx.doi.org/10.1038/ 1306–1311.
bmt.2013.192. Merlini, G., Palladini, G., 2008. Amyloidosis: is a cure possible? Ann. Oncol. Off. J.
Hegenbart, U., Iacobelli, S., Hoek, J., Rovira, M., Imhoff, G., van Bandini, G., Eur. Soc. Med. Oncol. ESMO 19 (Suppl. (4)), 63–66, http://dx.doi.org/10.1093/
Mellqvist, U.-H., Leblond, V., Carlson, K., Zver, S., Blaise, D., Alessandrino, E.P., annonc/mdn200.
Garderet, L., Kröger, N., 2013. Center experience and calendar year of Merlini, G., Palladini, G., 2013. Light chain amyloidosis: the heart of the problem.
transplantation strongly influence short term survival after autologous Haematologica 98, 1492–1495, http://dx.doi.org/10.3324/haematol.2013.
peripheral blood transplantation in 1315 patients with light chain 094482.
amyloidosis: an EBMT analysis. Blood 122, 417–417. Merlini, G., Leleu, X., Arnulf, B., Zamagi, E., Mollee, P., Cibeira, T.M., Schönland, S.O.,
Hideshima, T., Chauhan, D., Shima, Y., Raje, N., Davies, F.E., Tai, Y.T., Treon, S.P., Lin, Moreau, P., Hajek, R., Jaccard, A., Nicolas-Virelizier, E., Filshie, R., Augustson, B.,
B., Schlossman, R.L., Richardson, P., Muller, G., Stirling, D.I., Anderson, K.C., Mateos, M.-V., Milani, P., Dimopoulos, M.A., Hachula, E., Fermand, J.-P., Foli, A.,
2000. Thalidomide and its analogs overcome drug resistance of human Palumbo, A., Sonneveld, P., Johnsen, H.E., Kastritis, E., Palladini, G., 2014. A
multiple myeloma cells to conventional therapy. Blood 96, 2943–2950. randomized phase III trial of melphalan and dexamethasone (MDex) versus
Hinson, R.M., Williams, J.A., Shacter, E., 1996. Elevated interleukin 6 is induced by bortezomib, melphalan and dexamethasone (BMDex) for untreated patients
prostaglandin E2 in a murine model of inflammation: possible role of with al amyloidosis, in: 56th ash annual meeting and exposition. Presented at
cyclooxygenase-2. Proc. Natl. Acad. Sci. U. S. A. 93, 4885–4890. the 56th ASH Annual Meeting and Exposition, San Francisco.
Iyer, C.G., Languillon, J., Ramanujam, K., Tarabini-Castellani, G., De las Aguas, J.T., Mikhael, J.R., Schuster, S.R., Jimenez-Zepeda, V.H., Bello, N., Spong, J., Reeder, C.B.,
Bechelli, L.M., Uemura, K., Martinez Dominguez, V., Sundaresan, T., 1971. WHO Stewart, A.K., Bergsagel, P.L., Fonseca, R., 2012.
co-ordinated short-term double-blind trial with thalidomide in the treatment Cyclophosphamide-bortezomib-dexamethasone (CyBorD) produces rapid and
of acute lepra reactions in male lepromatous patients. Bull. World Health complete hematologic response in patients with AL amyloidosis. Blood 119,
Organ 45, 719–732. 4391–4394, http://dx.doi.org/10.1182/blood-2011-11-390930.
Jaccard, A., Moreau, P., Leblond, V., Leleu, X., Benboubker, L., Hermine, O., Recher, Milani, P., Rosin, M.V., Foli, A., Merlini, G., 2013. High-dose pomalidomide and
C., Asli, B., Lioure, B., Royer, B., Jardin, F., Bridoux, F., Grosbois, B., Jaubert, J., dexamethasone induce rapid responses in patients with AL amyloidosis
Piette, J.-C., Ronco, P., Quet, F., Cogne, M., Fermand, J.-P., 2007. Myélome exposed to alkylators, immune modulatory drugs, and proteasome inhibitors.
autogreffe (MAG) and intergroupe francophone du myélome (IFM) intergroup, Blood 122, 288–288.
2007. High-dose melphalan versus melphalan plus dexamethasone for AL Moreau, P., Jaccard, A., Benboubker, L., Royer, B., Leleu, X., Bridoux, F., Salles, G.,
amyloidosis. N. Engl. J. Med., http://dx.doi.org/10.1056/NEJMoa070484. Leblond, V., Roussel, M., Alakl, M., Hermine, O., Planche, L., Harousseau, J.-L.,
Jones, N.F., Hilton, P.J., Tighe, J.R., Hobbs, J.R., 1972. Treatment of primary renal Fermand, J.-P., 2010. Lenalidomide in combination with melphalan and
amyloidosis with melphalan. Lancet 2, 616–619. dexamethasone in patients with newly diagnosed AL amyloidosis: a
Kastritis, E., Anagnostopoulos, A., Roussou, M., Toumanidis, S., Pamboukas, C., multicenter phase 1/2 dose-escalation study. Blood 116, 4777–4782, http://dx.
Migkou, M., Tassidou, A., Xilouri, I., Delibasi, S., Psimenou, E., Mellou, S., Terpos, doi.org/10.1182/blood-2010-07-294405.
E., Nanas, J., Dimopoulos, M.A., 2007. Treatment of light chain (AL) amyloidosis Muller, G.W., Chen, R., Huang, S.Y., Corral, L.G., Wong, L.M., Patterson, R.T., Chen, Y.,
with the combination of bortezomib and dexamethasone. Haematologica 92, Kaplan, G., Stirling, D.I., 1999. Amino-substituted thalidomide analogs: potent
1351–1358, http://dx.doi.org/10.3324/haematol.11325. inhibitors of TNF-alpha production. Bioorg. Med. Chem. Lett. 9, 1625–1630.
Kastritis, E., Terpos, E., Roussou, M., Gavriatopoulou, M., Pamboukas, C., Boletis, I., Muthu Raja, K.R., Kovárová, L., Stossová, J., Hájek, R., 2011. Flow cytometric
Marinaki, S., Apostolou, T., Nikitas, N., Gkortzolidis, G., Michalis, E., Delimpasi, phenotyping and analysis of T regulatory cells in multiple myeloma patients.
S., Dimopoulos, M.A., 2012. A phase 1/2 study of lenalidomide with low-dose Klin. Onkol. Cas. Ceské Slov. Onkol. Spolecnosti 24 (Suppl), S30–33.
oral cyclophosphamide and low-dose dexamethasone (RdC) in AL amyloidosis. Palladini, G., Perfetti, V., Obici, L., Caccialanza, R., Semino, A., Adami, F., Cavallero,
Blood 119, 5384–5390, http://dx.doi.org/10.1182/blood-2011-12-396903. G., Rustichelli, R., Virga, G., Merlini, G., 2004. Association of melphalan and
Kotla, V., Goel, S., Nischal, S., Heuck, C., Vivek, K., Das, B., Verma, A., 2009. high-dose dexamethasone is effective and well tolerated in patients with AL
Mechanism of action of lenalidomide in hematological malignancies. J. (primary) amyloidosis who are ineligible for stem cell transplantation. Blood
Hematol. Oncol. 2, 36, http://dx.doi.org/10.1186/1756-8722-2-36. 103, 2936–2938, http://dx.doi.org/10.1182/blood-2003-08-2788.
Krönke, J., Udeshi, N.D., Narla, A., Grauman, P., Hurst, S.N., McConkey, M., Svinkina, Palladini, G., Perfetti, V., Perlini, S., Obici, L., Lavatelli, F., Caccialanza, R., Invernizzi,
T., Heckl, D., Comer, E., Li, X., Ciarlo, C., Hartman, E., Munshi, N., Schenone, M., R., Comotti, B., Merlini, G., 2005. The combination of thalidomide and
Schreiber, S.L., Carr, S.A., Ebert, B.L., 2014. Lenalidomide causes selective intermediate-dose dexamethasone is an effective but toxic treatment for
degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, patients with primary amyloidosis (AL). Blood 105, 2949–2951, http://dx.doi.
301–305, http://dx.doi.org/10.1126/science.1244851. org/10.1182/blood-2004-08-3231.
Kumar, S., Dispenzieri, A., Lacy, M.Q., Hayman, S.R., Buadi, F.K., Colby, C., Laumann, Palladini, G., Russo, P., Nuvolone, M., Lavatelli, F., Perfetti, V., Obici, L., Merlini, G.,
K., Zeldenrust, S.R., Leung, N., Dingli, D., Greipp, P.R., Lust, J.A., Russell, S.J., Kyle, 2007. Treatment with oral melphalan plus dexamethasone produces
R.A., Rajkumar, S.V., Gertz, M.A., 2012a. Revised prognostic staging system for long-term remissions in AL amyloidosis. Blood 110, 787–788, http://dx.doi.
light chain amyloidosis incorporating cardiac biomarkers and serum free light org/10.1182/blood-2007-02-076034.
chain measurements. J. Clin. Oncol. 30, 989–995, http://dx.doi.org/10.1200/ Palladini, G., Russo, P., Lavatelli, F., Nuvolone, M., Albertini, R., Bosoni, T., Perfetti,
JCO.2011.38.5724. V., Obici, L., Perlini, S., Moratti, R., Merlini, G., 2009. Treatment of patients with
Kumar, S.K., Hayman, S.R., Buadi, F.K., Roy, V., Lacy, M.Q., Gertz, M.A., Allred, J., advanced cardiac AL amyloidosis with oral melphalan, dexamethasone, and
Laumann, K.M., Bergsagel, L.P., Dingli, D., Mikhael, J.R., Reeder, C.B., Stewart, thalidomide. Ann. Hematol. 88, 347–350, http://dx.doi.org/10.1007/s00277-
A.K., Zeldenrust, S.R., Greipp, P.R., Lust, J.A., Fonseca, R., Russell, S.J., Rajkumar, 008-0600-y.
S.V., Dispenzieri, A., 2012b. Lenalidomide, cyclophosphamide, and Palladini, G., Dispenzieri, A., Gertz, M.A., Kumar, S., Wechalekar, A., Hawkins, P.N.,
dexamethasone (CRd) for light-chain amyloidosis: long-term results from a Schönland, S., Hegenbart, U., Comenzo, R., Kastritis, E., Dimopoulos, M.A.,
phase 2 trial. Blood 119, 4860–4867, http://dx.doi.org/10.1182/blood-2012- Jaccard, A., Klersy, C., Merlini, G., 2012a. New criteria for response to treatment
01-407791. in immunoglobulin light chain amyloidosis based on free light chain
Kyle, R.A., Gertz, M.A., Greipp, P.R., Witzig, T.E., Lust, J.A., Lacy, M.Q., Therneau, T.M., measurement and cardiac biomarkers: impact on survival outcomes. J. Clin.
1997. A trial of three regimens for primary amyloidosis: colchicine alone, Oncol. 30, 4541–4549, http://dx.doi.org/10.1200/JCO.2011.37.7614.
melphalan and prednisone, and melphalan, prednisone, and colchicine. N. Engl. Palladini, G., Russo, P., Foli, A., Milani, P., Lavatelli, F., Obici, L., Nuvolone, M.,
J. Med. 336, 1202–1207, http://dx.doi.org/10.1056/NEJM199704243361702. Brugnatelli, S., Invernizzi, R., Merlini, G., 2012b. Salvage therapy with
Landau, H., Smith, M., Chou, J., Comenzo, R.L., Sean, M.D., Hassoun, H., Bello, C., lenalidomide and dexamethasone in patients with advanced AL amyloidosis
Giralt, S., 2014. Event-free and overall survival following risk-adapted refractory to melphalan, bortezomib, and thalidomide. Ann. Hematol. 91,
melphalan stem cell transplant and consolidation for systemic light chain 89–92, http://dx.doi.org/10.1007/s00277-011-1244-x.
amyloidosis. Presented at the XIVth International Symposium on Amyloidosis, Palladini, G., Russo, P., Milani, P., Foli, A., Lavatelli, F., Nuvolone, M., Perlini, S.,
Indianapolis. Merlini, G., 2013. A phase II trial of cyclophosphamide, lenalidomide and
Licht, J.D., Shortt, J., Johnstone, R., 2014. From anecdote to targeted therapy: the dexamethasone in previously treated patients with AL amyloidosis.
curious case of thalidomide in multiple myeloma. Cancer Cell 25, 9–11, http:// Haematologica 98, 433–436, http://dx.doi.org/10.3324/haematol.2012.073593.
dx.doi.org/10.1016/j.ccr.2013.12.019.
260 T. Jelinek et al. / Critical Reviews in Oncology/Hematology 99 (2016) 249–260

Palladini, G., Milani, P., Foli, A., Obici, L., Lavatelli, F., Nuvolone, M., Caccialanza, R., treatment with lenalidomide in AL amyloidosis. Blood 116, 5071–5072, http://
Perlini, S., Merlini, G., 2014a. Oral melphalan and dexamethasone grants dx.doi.org/10.1182/blood-2010-09-305136.
extended survival with minimal toxicity in AL amyloidosis: long-term results Venner, C.P., Lane, T., Foard, D., Rannigan, L., Gibbs, S.D.J., Pinney, J.H., Whelan, C.J.,
of a risk-adapted approach. Haematologica 99, 743–750, http://dx.doi.org/10. Lachmann, H.J., Gillmore, J.D., Hawkins, P.N., Wechalekar, A.D., 2012.
3324/haematol.2013.095463. Cyclophosphamide, bortezomib, and dexamethasone therapy in AL
Palladini, G., Milani, P., Foli, A., Vidus Rosin, M., Basset, M., Lavatelli, F., Nuvolone, amyloidosis is associated with high clonal response rates and prolonged
M., Obici, L., Perlini, S., Merlini, G., 2014b. Melphalan and dexamethasone with progression-free survival. Blood 119, 4387–4390, http://dx.doi.org/10.1182/
or without bortezomib in newly diagnosed AL amyloidosis: a matched blood-2011-10-388462.
case-control study on 174 patients. Leukemia 28, 2311–2316, http://dx.doi. Venner, C.P., Gillmore, J.D., Sachchithanantham, S., Mahmood, S., Lane, T., Foard, D.,
org/10.1038/leu.2014.227. Rannigan, L., Gibbs, S.D.J., Pinney, J.H., Whelan, C.J., Lachmann, H.J., Hawkins,
Payvandi, F., Wu, L., Haley, M., Schafer, P.H., Zhang, L.-H., Chen, R.S., Muller, G.W., P.N., Wechalekar, A.D., 2014. A matched comparison of cyclophosphamide,
Stirling, D.I., 2004. Immunomodulatory drugs inhibit expression of bortezomib and dexamethasone (CVD) versus risk-adapted
cyclooxygenase-2 from TNF-alpha IL-1beta, and LPS-stimulated human PBMC cyclophosphamide, thalidomide and dexamethasone (CTD) in AL amyloidosis.
in a partially IL-10-dependent manner. Cell. Immunol. 230, 81–88, http://dx. Leukemia 28, 2304–2310, http://dx.doi.org/10.1038/leu.2014.218.
doi.org/10.1016/j.cellimm.2004.09.003. Wang, C.Y., Mayo, M.W., Korneluk, R.G., Goeddel, D.V., Baldwin, A.S., 1998.
Quach, H., Ritchie, D., Stewart, A.K., Neeson, P., Harrison, S., Smyth, M.J., Prince, NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and
H.M., 2010. Mechanism of action of immunomodulatory drugs (IMiDS) in c-IAP2 to suppress caspase-8 activation. Science 281, 1680–1683.
multiple myeloma. Leukemia 24, 22–32, http://dx.doi.org/10.1038/leu.2009. Wechalekar, A.D., Goodman, H.J.B., Lachmann, H.J., Offer, M., Hawkins, P.N.,
236. Gillmore, J.D., 2007. Safety and efficacy of risk-adapted cyclophosphamide,
Rajkumar, S.V., 2004. Thalidomide: tragic past and promising future. Mayo Clin. thalidomide, and dexamethasone in systemic AL amyloidosis. Blood 109,
Proc. 79, 899–903, http://dx.doi.org/10.1016/S0025-6196(11) 62157-5. 457–464, http://dx.doi.org/10.1182/blood-2006-07-035352.
Reece, D.E., Hegenbart, U., Sanchorawala, V., Merlini, G., Palladini, G., Bladé, J., Wechalekar, A.D., Schonland, S.O., Kastritis, E., Gillmore, J.D., Dimopoulos, M.A.,
Fermand, J.-P., Hassoun, H., Heffner, L., Vescio, R.A., Liu, K., Enny, C., Esseltine, Lane, T., Foli, A., Foard, D., Milani, P., Rannigan, L., Hegenbart, U., Hawkins, P.N.,
D.-L., Velde van de, H., Cakana, A., Comenzo, R.L., 2011. Efficacy and safety of Merlini, G., Palladini, G., 2013. A European collaborative study of treatment
once-weekly and twice-weekly bortezomib in patients with relapsed systemic outcomes in 346 patients with cardiac stage III AL amyloidosis. Blood 121,
AL amyloidosis: results of a phase 1/2 study. Blood 118, 865–873, http://dx.doi. 3420–3427, http://dx.doi.org/10.1182/blood-2012-12-473066.
org/10.1182/blood-2011-02-334227. Wechalekar, A., Foard, D., Rannigan, L., Lane, T., Sachachtanatham, S., Mahmood, S.,
Sampaio, E.P., Sarno, E.N., Galilly, R., Cohn, Z.A., Kaplan, G., 1991. Thalidomide Sayed, R., Patel, K., Whelan, C., Lachmann, H., Hawkins, P., Gillmore, J., 2014.
selectively inhibits tumor necrosis factor alpha production by stimulated Characteristics and outcomes of 714 patients with systemic AL
human monocytes. J. Exp. Med. 173, 699–703. amyloidosis—analysis of a prospective study (ALChemy study). Presented at
Sanchorawala, V., Doros, G., Quillen, K., Sloan, J.M., Shelton, A.C., Brauneis, D., Finn, the XIVth International Symposium on Amyloidosis, Indianapolis.
K.T., Seldin, D.C., 2014a. Long-term outcome of patients with AL amyloidosis Zou, W., 2005. Immunosuppressive networks in the tumour environment and their
treated with high-dose melphalan and stem cell transplantation: 19 year therapeutic relevance. Nat. Rev. Cancer 5, 263–274, http://dx.doi.org/10.1038/
experience at a single center. Biol. Blood Marrow Transpl. 20, S47–S48, http:// nrc1586.
dx.doi.org/10.1016/j.bbmt.2013.12.045.
Sanchorawala, V., Patel, J.M., Sloan, J.M., Shelton, A.C., Zeldis, J.B., Seldin, D.C., 2013.
Melphalan, lenalidomide and dexamethasone for the treatment of Biographies
immunoglobulin light chain amyloidosis: results of a phase II trial.
Haematologica 98, 789–792, http://dx.doi.org/10.3324/haematol.2012.075192.
Sanchorawala, V., Shelton, A., Stephen, L., Sloan, J.M., Seldin, D.C., 2014b. Tomas Jelinek is a physician at the Department of Haematooncology at the
Pomalidomide and dexamethasone in patients with relapsed light-chain University Hospital Ostrava, Czech Republic. In 2011 he earned his degree at
amyloidosis (AL): results of a phase 1 study. Presented at the XIVth Masaryk University in Brno, Czech Republic. During his studies at university he
International Symposium on Amyloidosis, Indianapolis. conducted internships at Hospital Anna Costa, Santos, Brazil; Universidade de Coim-
Sanchorawala, V., Skinner, M., Quillen, K., Finn, K.T., Doros, G., Seldin, D.C., 2007a. bra, Portugal and Autonomous University of Zacatecas, Mexico. He started his
Long-term outcome of patients with AL amyloidosis treated with high-dose career at the Department of Internal Medicine—Hematology and Oncology, Uni-
melphalan and stem-cell transplantation. Blood 110, 3561–3563, http://dx.doi. versity Hospital Brno and since 2013 he has been working at the Intensive Care and
org/10.1182/blood-2007-07-099481. Transplant Unit at University Hospital Ostrava. Dr. Jelinek is a member of Czech Soci-
Sanchorawala, V., Wright, D.G., Rosenzweig, M., Finn, K.T., Fennessey, S., Zeldis, J.B.,
ety of Hematology and Czech Myeloma Group. He has several publications in the
Skinner, M., Seldin, D.C., 2007b. Lenalidomide and dexamethasone in the
field of hematological malignancies. His current research interest includes multiple
treatment of AL amyloidosis: results of a phase 2 trial. Blood 109, 492–496,
myeloma and other plasma cell disorders. He is also interested in modern diagnostic
http://dx.doi.org/10.1182/blood-2006-07-030544.
Sedlarikova, L., Kubiczkova, L., Sevcikova, S., Hajek, R., 2012. Mechanism of methods, dominantly flow cytometry and its clinical and scientific application.
immunomodulatory drugs in multiple myeloma. Leuk. Res. 36, 1218–1224,
Roman Hajek is Professor of Oncology and Vice-rector at the Ostrava University
http://dx.doi.org/10.1016/j.leukres.2012.05.010.
and Head of the Department of Haematooncology at the University Hospital Ostrava,
Seldin, D.C., Choufani, E.B., Dember, L.M., Wiesman, J.F., Berk, J.L., Falk, R.H., O’Hara,
C., Fennessey, S., Finn, K.T., Wright, D.G., Skinner, M., Sanchorawala, V., 2003. Czech Republic. After qualifying in medicine in 1988 from J.E. Purkyne (Masaryk)
Tolerability and efficacy of thalidomide for the treatment of patients with light University, Faculty of Medicine Brno, Dr. Hajek received a PhD in hematology in
chain-associated (AL) amyloidosis. Clin. Lymphoma 3, 241–246. 1995 and completed degrees in Internal Medicine in 1996 and in Medical Oncology
Skinner, M., Anderson, J., Simms, R., Falk, R., Wang, M., Libbey, C., Jones, L.A., Cohen, in 1998. Dr. Hajek was a founder member of the Czech Myeloma Group. He is an
A.S., 1996. Treatment of 100 patients with primary amyloidosis: a randomized active chairman of the Czech Myeloma Group, main investigator of the randomized
trial of melphalan, prednisone, and colchicine versus colchicine only. Am. J. clinical trials phase III of Czech Myeloma Group and phase I/II/III of other clinical
Med. 100, 290–298. trials in MM including experimental immunotherapy and main investigator of more
Skinner, M., Sanchorawala, V., Seldin, D.C., Dember, L.M., Falk, R.H., Berk, J.L., than 30 national grants. He is a member of several scientific societies, including the
Anderson, J.J., O’Hara, C., Finn, K.T., Libbey, C.A., Wiesman, J., Quillen, K., Swan, European Hematology Association and the Amercian Society of Hematology. He has
N., Wright, D.G., 2004. High-dose melphalan and autologous stem-cell published over 300 scientific articles and book chapters and he is an Editor of the
transplantation in patients with AL amyloidosis: an 8-year study. Ann. Intern. European Journal of Hematology. His research interests include multiple myeloma,
Med. 140, 85–93.
bone marrow transplantation and amyloidosis.
Tapan, U., Seldin, D.C., Finn, K.T., Fennessey, S., Shelton, A., Zeldis, J.B.,
Sanchorawala, V., 2010. Increases in B-type natriuretic peptide (BNP) during

You might also like