+ All Categories
Home > Documents > Lenalidomide: a novel anticancer drug with multiple modalities

Lenalidomide: a novel anticancer drug with multiple modalities

Date post: 11-Dec-2016
Category:
Upload: angus
View: 212 times
Download: 0 times
Share this document with a friend
9
Drug Evaluation 10.1517/14656560802627903 © 2009 Informa UK Ltd ISSN 1465-6566 125 All rights reserved: reproduction in whole or in part not permitted Lenalidomide: a novel anticancer drug with multiple modalities Christine Galustian & Angus Dalgleish St Georges University of London, Division of Cellular and Molecular Medicine, Department of Oncology, Cranmer Terrace, Tooting, London Over the past 5 years, lenalidomide (Revlimid ® , Celgene Co., Summit, NJ, USA), a member of a class of drugs termed immunomodulatory drugs, has emerged as a significant weapon in the arsenal of cancer-therapeutics. It is a lead therapeutic in multiple myeloma and del-5q myelodysplastic syndromes and has also been trialed for acute leukaemia and chronic lymphocytic leukaemia, relapsed or refractory Hodgkin’s lymphoma, T-cell non-Hodgkin’s lymphoma, prostate cancer, non-small cell lung cancer, malignant melanoma, renal cancer, advanced ovarian and peritoneal carcinoma. The most significant development for lenalidomide has been its FDA approval (US and Europe) for previously treated multiple myeloma in combination with dexamethasone. The following review describes key clinical and mechanistic breakthroughs that have made lenalidomide a leading cancer therapeutic. Keywords: anti-angiogenic, anti-metastatic, IMiDs , immunomodulatory drugs, lenalidomide, multiple myeloma, myelodysplastic syndromes Expert Opin. Pharmacother. (2009) 10(1):125-133 1. Overview of the market Current therapies for treatment of multiple myeloma, myelodysplatic syndromes (MDS) and chronic lymphocytic leukaemia (CLL) are based on cytotoxic or antiproliferative agents such as bortezomib, doxorubicin and mephelan for multiple myeloma; chlorambucil, fludarabine, cyclophosphamide and CHOP (cyclophosphamide, doxorubicin (adriamycin) and vincristine combinations) for CLL; and methyltranferase inhibitors (azacitidine) and haematopoiesis stimulators (recombinant erythropoietins) and immunosuppressants (cyclosporine) for MDS. More specifically targeted therapies such as the small-molecule inhibitors, imatinib (ABL) and monoclonal antibodies, rituximab (CD20), and alemtuzumab (CD52) are also available for CLL and multiple myeloma. However, all of the agents have single modes of action, which although effective, target few of the pathways that are involved in these diseases. The requirement for multi-modal therapy in these diseases has resulted in the trial of agents such as thalidomide and its analogues such as lenalidomide, which are anti-metastatic, anti-angiogenic and also augment antitumour immunity. 2. Chemistry and pharmacokinetics of lenalidomide Lenalidomide and the related compound pomalidomide are 4-amino-gultaramide derivatives of thalidomide in which an amino group was added to the fourth carbon of the phthaloyl ring of the parent compound (Figure 1). Both agents also exist as a racemic mixture of the active S(-) and R(+) forms. Like thalidomide, they are both administered in FDA approved daily oral dosing every 21 – 28 days of monthly cycles. Elimination of the drugs is through the renal route; therefore, caution is recommended in dosing patients with impaired creatinine clearance. After oral administration, the half-lifes of lenalidomide and pomalidomide are 1. Overview of the market 2. Chemistry and pharmacokinetics of lenalidomide 3. Clinical efficacy of lenalidomide 4. Regulatory affairs 5. Pharmacodynamics of lenalidomide 6. Expert opinion Expert Opin. Pharmacother. Downloaded from informahealthcare.com by University of Aberdeen on 09/25/13 For personal use only.
Transcript
Page 1: Lenalidomide: a novel anticancer drug with multiple modalities

Drug Evaluation

10.1517/14656560802627903 © 2009 Informa UK Ltd ISSN 1465-6566 125All rights reserved: reproduction in whole or in part not permitted

Lenalidomide:anovelanticancerdrugwithmultiplemodalitiesChristine Galustian† & Angus Dalgleish†St Georges University of London, Division of Cellular and Molecular Medicine, Department of Oncology, Cranmer Terrace, Tooting, London

Over the past 5 years, lenalidomide (Revlimid®, Celgene Co., Summit, NJ, USA), a member of a class of drugs termed immunomodulatory drugs, has emerged as a significant weapon in the arsenal of cancer-therapeutics. It is a lead therapeutic in multiple myeloma and del-5q myelodysplastic syndromes and has also been trialed for acute leukaemia and chronic lymphocytic leukaemia, relapsed or refractory Hodgkin’s lymphoma, T-cell non-Hodgkin’s lymphoma, prostate cancer, non-small cell lung cancer, malignant melanoma, renal cancer, advanced ovarian and peritoneal carcinoma. The most significant development for lenalidomide has been its FDA approval (US and Europe) for previously treated multiple myeloma in combination with dexamethasone. The following review describes key clinical and mechanistic breakthroughs that have made lenalidomide a leading cancer therapeutic.

Keywords: anti-angiogenic, anti-metastatic, IMiDs™, immunomodulatory drugs, lenalidomide, multiple myeloma, myelodysplastic syndromes

Expert Opin. Pharmacother. (2009) 10(1):125-133

1. Overviewofthemarket

Current therapies for treatment of multiple myeloma, myelodysplatic syndromes (MDS) and chronic lymphocytic leukaemia (CLL) are based on cytotoxic or antiproliferative agents such as bortezomib, doxorubicin and mephelan for multiple myeloma; chlorambucil, fludarabine, cyclophosphamide and CHOP (cyclophosphamide, doxorubicin (adriamycin) and vincristine combinations) for CLL; and methyltranferase inhibitors (azacitidine) and haematopoiesis stimulators (recombinant erythropoietins) and immunosuppressants (cyclosporine) for MDS.

More specifically targeted therapies such as the small-molecule inhibitors, imatinib (ABL) and monoclonal antibodies, rituximab (CD20), and alemtuzumab (CD52) are also available for CLL and multiple myeloma. However, all of the agents have single modes of action, which although effective, target few of the pathways that are involved in these diseases. The requirement for multi-modal therapy in these diseases has resulted in the trial of agents such as thalidomide and its analogues such as lenalidomide, which are anti-metastatic, anti-angiogenic and also augment antitumour immunity.

2. Chemistryandpharmacokineticsoflenalidomide

Lenalidomide and the related compound pomalidomide are 4-amino-gultaramide derivatives of thalidomide in which an amino group was added to the fourth carbon of the phthaloyl ring of the parent compound (Figure 1). Both agents also exist as a racemic mixture of the active S(-) and R(+) forms. Like thalidomide, they are both administered in FDA approved daily oral dosing every 21 – 28 days of monthly cycles. Elimination of the drugs is through the renal route; therefore, caution is recommended in dosing patients with impaired creatinine clearance. After oral administration, the half-lifes of lenalidomide and pomalidomide are

1. Overview of the market

2. Chemistry and pharmacokinetics

of lenalidomide

3. Clinical efficacy of lenalidomide

4. Regulatory affairs

5. Pharmacodynamics of

lenalidomide

6. Expert opinion

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 2: Lenalidomide: a novel anticancer drug with multiple modalities

Lenalidomide

126 ExpertOpin.Pharmacother.(2009) 10(1)

3 and 7 h, respectively [1,2]. In contrast to thalidomide, which has dose limiting toxicity due to somnolence, constipation and peripheral neuropathy, both lenalidomide and pomali-domide have less commonly observed neurosedative toxicity. Dose-limiting neutropenia and thrombocytopenia are the most common toxicities seen with these drugs. Venous thromboembolism has been reported in various trials, and is increased when haemopoeitic agents such as erythropoeitin are also used, but is rarer when the drug is used singly (for example in a recent MDS study in which incidence was only 3.4% [3]). The incidence is markedly reduced when aspirin is given concomitantly.

3. Clinicalefficacyoflenalidomide

3.1 ClinicalstudiesonmultiplemyelomaResults from Phase I and II studies in the relapsed setting show lenalidomide alone to have significant activity with responses seen in 14 – 29% of patients [4]. Activity has been shown to increase markedly when lenalidomide is combined with dexamethasone: two randomised Phase III trials in North America and Europe [5,6] have demonstrated that the combination regimen provides greater efficacy than with dexamethasone alone, with combined response rates of 59 versus 21 – 24% for dexamethasone, and complete response (CR) rates of 13 – 17 versus 1 – 4%. Inclusion criteria in these trials were that patients had had at least 3 cycles of therapy for myeloma. Patients who were resistant to high doses of dexamethasone (200 mg) were excluded from this trial. Median time to progression was doubled from 4.7 to 11.1 months and overall survival times were also increased from 20.2 to 29.6 months. More recent trials

have shown more promising data, for example, in studies with patients in long term therapy (> 4 years). 11 out of 15 patients had partial or complete responses to lenalidomide therapy either given alone, or with dexamethasone [7]. Mono-therapy with lenalidomide seems to give similar percentages of partial/complete responses in this study. A further investigation in newly diagnosed myeloma suggests that low dose dexamethasone plus lenalidomide gives better survival rates after 1 year than standard dose dexamethasone plus lenalidomide (96 versus 88%) [8]. Combination therapy with lenalidomide, bortezomib and dexamethasone in refractory myeloma has shown very promising initial results in 33 patients with > 55% partial response and 36% with very good partial responses or complete responses [9].

The main adverse effects seen in these two Phase III trials were neutropenia, thrombocytopenia, infection and anaemia, which occurred in 25 – 35, 9.7 – 13, 16 and 8 – 13% of patients, respectively. Deep vein thrombosis occurred in 13.5 and 4% of patients in the MM009 and MM010 trials, respectively. Patients at present on combinations of lenalidomide and dexamethasone are given prophylactic aspirin to reduce this effect.

3.2 MdsMDS are disorders in which bone marrow function is abnormal and there is insufficient production of mature blood cells often leading to severe and refractory anaemia (reviewed in [10]). Bone marrow failure can occur in around 30% of patients. Chrosomal (cytogenetic) abnormalities are detected in > 50% of patients with MDS. Patients with low- and intermediate-1-risk MDS with interstitial deletions in the long arm of chromosome 5 (deletion 5Q) alone, or in combination

Thalidomide

O

N

NH

O

O

NH2

Lenalidomide

O

N

NH

O

O

ONH

2

Pomalidomide

N

NH

O

O O

O4

Figure1.Lenalidomideandthe relatedcompoundpomalidomideare4-amino-gultaramidederivativesof thalidomide inwhichanaminogroupwasaddedtothefourthcarbonofthephthaloylringoftheparentcompound.

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 3: Lenalidomide: a novel anticancer drug with multiple modalities

Galustian&Dalgleish

ExpertOpin.Pharmacother.(2009) 10(1) 127

with other cytogenetic abnormalities, represent 20 – 30% of the MDS patient population.

The haematological activity of lenalidomide in lower-risk MDS, and its potential toxic reactions were initially demonstrated in an open study with 43 patients experiencing transfusion-dependent or symptomatic anaemia [11]. These patients had either failed previous treatment with erythro-poietin or were unlikely to respond to cytokine therapies owing to high erythropoietin and extensive transfusions. Lenalidomide was used at a dose of 25 or 10 mg per day, or 10 mg daily for 21 days out of every 28-day cycle for 16 weeks. The erythroid response rate was 56%, with 24 out of 43 patients responding: 20 of these 24 patients achieved transfusion independence. There was an 83% response rate in patients with the 5q chromosomal deletion compared to 57% in patients with a normal karyotype, and 12% in patients with other cytogenetic abnormalities. In the 20 patients with karyotypic abnormalities, 10 (50%) had a complete remission (with a restoration of normal karyotype and disappearance of dysplasia), including 9 of 12 patients with deletion 5q. Lenalidomide response times were also quicker for the deletion 5q patients (8 versus 11.2 weeks). The most common side events were neutropenia (55%) and thrombocytopenia (44%).

This trial led to multi-centre studies in transfusion-dependent, low- or intermediate-1 risk MDS patients either with deletion 5q (trial MDS-003) [12] or those without the 5q deletion (trial MDS-002) [13]. In the MDS-003 trial, 148 patients received 10 mg of lenalidomide for 21 days every 4 weeks or daily for 24 weeks. The transfusion response rate was 76%, with 99 (67%) patients achieving transfusion-independence. Responding patients increased their haemoglobin to a median of 13.4 g/dl. Transfusion independence was obtained with a median of 4.6 weeks, and the median duration was greater than 104 weeks after follow up. Among 85 evaluable patients, 62 (73%) had cytogenetic improvement and 38 (45%) had complete cytogenetic remission.

In the second multi-centre Phase II trial (MDS-002) in patients without the 5q deletion, with identical doses of lenalidomide, > 80% of study participants had a heavy transfusion burden at the start of the study. Among 215 patients treated, the transfusion response rate was 43%, with 26% achieving transfusion-independence. Based on these results, lenalidomide was approved for use in the US by the FDA in December 2005 for the treatment of IPSS low- or intermediate-1 risk MDS patients with deletion 5q, at a dose of 10 mg daily.

3.3 EffectsoflenalidomideonCLLB-cell CLL is the most common leukaemia in the Western world and results from accumulation of mature B lymphocytes mainly in the blood and bone marrow because of faulty apoptosis [14]. The activity of lenalidomide was studied in patients with relapsed/refractory CLL [15]. Lenalidomide was given at 10 mg daily with dose escalation up to 25 mg daily.

The study showed 3 (7%) patients with CR, 1 nodular partial remission (PR) and 10 patients with PR giving an overall response rate of 32%. When measuring plasma levels of angiogenic factors, inflammatory cytokines and cytokine receptors at baseline, day 7 and day 28, there was a dramatic increase in median IL-6, IL-10, IL-2 and TNF-R1 levels on day 7, whereas no changes were observed in median (VEGF) levels (20 patients studied). Therefore, lenalidomide as a continuous treatment has antitumour activity in heavily pretreated patients with CLL when given continuously. However, in contrast to MDS or multiple myeloma patients in whom 25 mg daily of lenalidomide can be administered, 10 mg daily of lenalidomide is the maximal recommended dose in CLL, as complete responses seen at this dose and higher doses (25 mg daily) can lead to tumour flare reactions, which are not related to clinical responsiveness [16], but this is thought to be manageable when patients are given starting doses of 10 mg daily [17]. Thus, lenalidomide’s tolerability and optimal dose may be disease specific [18]. In a study of patients with CLL, small lymphocytic leukaemia and mantle cell lymphoma, it was also shown that toxicity was decreased by alternating thalidomide with lenalidomide, which allowed longer immunomodualtory drug (IMiD) treatment and good response rates [19]. Other major adverse effects seen in CLL patients include fatigue, neutropenia in 40 – 70% of patients and anaemia [15,20]. The mechanism of action of lenalidomide in CLL patients is still unclear, but there are new data showing that CLL cells can inhibit immune synapse formation in CD4+ and CD8+ T cells, both in autologous patient cell populations and in healthy donor T cells exposed to the CLLs, leading to an impaired response to APCs (antigen-presenting cells), and lenalidomide treatment of CLLs can reverse the suppression of synapse formation by CLLs [21]. Also, there is evidence that lenalidomide does not directly kill CLL cells but does affect the anti-apoptotic effects of nurse-like cells on CLLs in the CLL microenvironment [22].

3.4 OthercancersLenalidomide has been trialed in a number of other cancers with a good degree of success. For example, in a completed Phase I trial of mantle cell lymphoma at 20 mg after 2 cycles, 5 out of 6 patients achieved responses including one CR, one PR, and three minor responses [23]. In a Phase I trial of aggressive relapsed/refractory non-Hodgkin’s lymphoma, objective responses were seen in 28% of patients [24]. Osteonectin, cyclin D1 and p21kip correlate with non-Hodgkin’s lymphoma cell lines sensitivity to lenalidomide [25]. In a study of 18 patients with small lymphocytic lymphoma, 4 (22%) patients had a complete or partial response [26]. In 12 patients with acute leukaemia, CRs were seen in 2 patients [27].

In solid tumour trials, promising data has also been shown: 55% of patients with prostate cancer had reduction of PSA of > 98% [28] and in 18 patients with rapidly

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 4: Lenalidomide: a novel anticancer drug with multiple modalities

Lenalidomide

128 ExpertOpin.Pharmacother.(2009) 10(1)

progressive thyroid cancer, a 67% response was seen with 22% of patients having PR [29].

4. Regulatoryaffairs

As previously mentioned, lenalidomide has been FDA approved in the US and Europe for previously treated multiple myeloma in combination with dexamethasone. It is also approved for use in patients with transfusion-dependent anaemia owing to low or intermediate-1 risk MDS associated with a deletion 5q cytogenetic abnormality with or without further cytogenetic abnormalities. Lenalidomide is available only under a special restricted distribution program, RevAssistSM, similar to the S.T.E.P.S.® program instituted for Thalomid®, to insure no foetal exposure, owing to the teratogenic properties associated with these drugs.

5. Pharmacodynamicsoflenalidomide

Lenalidomide affects many cell pathways and processes as discussed below (Figure 2).

5.1 AntitumoureffectsInitial efforts to understand the anticancer properties of lenalidomide and its analogues in myeloma and MDS have focused on direct antitumour activities such as increased apoptosis and inhibition of tumour cell proliferation. Lenali-domide and pomalidomide inhibit proliferation of a number of multiple myeloma cell lines including Sultan and RPMI8266 and, furthermore, render doxorubicin and dexamethasone resistant subtypes of these cell lines sensitive to these agents [30]. Lenalidomide and pomalidomide have been shown to inhibit the proliferation of two B-lymphoma cell lines but have no effect on normal B cells and increase proliferation of CD34 progenitors [31]. Lenalidomide, thalidomide and pomalidomide trigger activation of caspase-8, enhance multiple myeloma cell sensitivity to FAS-induced apoptosis and downregulate NF-κB, cellular inhibitor of apoptosis 2 and FLICE inhibitory protein [32]. In MDS del(5)(q31), lenalidomide inhibits growth of del(5q) erythroid progenitors and upregulates SPARC, a tumour suppressor gene, which may play a role in the pathogenesis of the 5q- syndrome and activin A, with a role in apotosis of erythropoietic cells [33].

5.2 EffectsonimmunefunctionThe effects of lenalidomide and its analogues on immune function have been reported since the early 90s and more recently, long-term responses and responses in patients’ low tumoural mass seen with lenalidomide in different trials point toward its immunological mechanism(s).

5.2.1 T-cell costimulationThe IMiDs® immunomodulatory drugs thalidomide, lenali-domide and pomalidomide have been shown to act as T-cell costimulatory molecules. In contrast to their well documented

anti-inflammatory effects on LPS stimulated monocytes [34-36], in which there is a decrease in the secretion of TNF-α, T cells activated through CD3 or IL-2 have enhanced proliferation when co-treated with the IMiDs thalidomide, lenalidomide or pomalidomide [37]. This proliferation is concomitant with increased secretion of proinflammatory cytokines such as IL-2, IFN-γ, and IL-12 from CD4 and CD8 T cells that have been stimulated to become TH1 type T cells (using IL-12).

Lenalidomide alone does not induce cell cycle changes in T cells; however, following CD3 ligation on T cells, lenalidomide enhances G1 to S transition compared with CD3 ligation alone [37]. Lenalidomide also induces significant CD3 T-cell proliferation in the presence of immature DCs, or mature DCs. The mechanisms by which the costimulatory effects are thought to occur are through CD28: CTLA-4–Ig, which blocks costimulation through the B7-CD28 pathway, inhibits T-cell proliferation induced by immature and mature DCs and lenalidomide partially overcomes this inhibitory effect of CTLA-4–Ig. Lenalidomide can induce CD28 phosphorylation and also augments NF-κB activation on anti-CD28 activated T cells. The IMiDs immunomodulatory compounds enhance AP-1-driven transcriptional activity two- to fourfold after 6 h of T-cell stimulation, and their relative potencies for AP-1 activation correlate with potencies for increased IL-2 production in Jurkat T cells and in CD4+ or CD8+ human peripheral blood T cells [38]. IL-2 production from stimulated T cells is increased through rises in PKC-θ activation and enhanced DNA-binding activity of AP-1 but not NF-κB [39].

5.2.2 Effects on T-regulatory cellsThe IMiDs immunomodulatory compounds have been assessed for their activity against naturally occurring regulatory T cells. These cells are of the CD4 lineage and express very high levels of CD25 (Interleukin 2 receptor) [40]. They also are characterised by expression of FOXP3, a protein of the FORKHEAD box family, which leads to inhibition of IL-2 mediated proliferation and renders T cells suppressive to other activatory cell types such as activatory CD4 T-cells, CD8 cytotoxic T-cells and natural killer (NK) cells [41]. Both lenalidomide and pomalidomide, but not thalidomide, inhibit expression and function of these cells [42,43]. This finding is critical as T-regulatory cells are in tumour infiltrating populations in patients with ovarian, breast, pancreatic and colorectal cancers [44-46], and indicate poor prognosis in patients with gastric and oesophageal cancers [47]. Furthermore, in ovarian cancer, a high CD8:T regulatory cell ratio is a good prognostic indicator [48,49]. T regulatory cells are also raised in patients with multiple myeloma [50] non-Hodgkin’s lymphoma [51] and CLL [52], which can respond to lenalidomide treatment.

5.2.3 Effects on NK cells and NKT cellsNK cells comprise around 10% of the lymphocyte population in normal individuals and are important for recognition and removal of cells virally infected or cancer cells. They recognise cells with missing major histocompatibility complex (MHC)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 5: Lenalidomide: a novel anticancer drug with multiple modalities

Galustian&Dalgleish

ExpertOpin.Pharmacother.(2009) 10(1) 129

class I. NK cells are critical in antitumour immunity (reviewed in [53]). Davies et al. have shown that IL-2-primed peripheral blood mononuclear cells (PBMCs) treated with thalidomide/IMiDs immunomodulatory compounds demon-strated significantly increased lysis of multiple myeloma cell lines [78]. More recently, Zhu et al. showed pro-apoptotic effects of lenalidomide and pomalidomide on tumour cells in a co-culture model of PBMC and tumour cells using K562 and RAJI tumour targets, in which increased apoptosis was induced by preincubating the PBMC population with the drugs before addition to the target cells [54]. The NK population within the PBMCs was shown to be responsible for mediating the apoptosis of K562 cells. Lenalidomide can also enhance NK and monocyte cytotoxicity toward target cells through an increase in antibody-dependant cell cytotoxicity (ADCC) when the target cells are treated with rituximab (anti-CD20 monoclonal antibody) [55]. This effect is dependant on the presence of antibody and on IL-2 or IL-12. Lenalidomide and pomalidomide combined with IgG1-isotype antibodies enhance ADCC through enhanced IL-12 cytokine signalling and increased effector cell granzyme B and FasL expression [56]. The effect was seen with non-Hodgkin’s lymphoma target lines in vitro and with B-cell CLLs derived from patients previously treated with fludarabine and cyclophosphamide. Enhanced NK cell Fc-γ receptor signalling is associated with enhanced phosphorylated

extracellular signal-related kinase levels leading to enhanced effector function.

However, it is possible that the IMiDs immunomodulatory compounds effects on NK cells is indirect and occurs through increased IL-2 cytokine production in T cells costimulated by the IMiDs immunomodulatory compounds. IMiDs immunomodulatory compounds facilitate nuclear translocation of nuclear factor of activated T cells-2 and activator protein-1 through activation of phosphoinositide-3 kinase signalling, with resulting IL-2 secretion [57]. IMiDs immunomodulatory compounds enhance both NK cell cytotoxicity and ADCC induced by triggering IL-2 production from T cells [57].

Natural killer T (NKT) cells are CD1d-restricted glycolipid reactive innate lymphocytes that provide protection from pathogens and tumours. These cells mediate antitumour effects through direct effects on tumour cells, cytokine production, antiangiogenesis and activation of other cells, notably dendritic cells and NK cells [58]. Lenalidomide enhances antigen-specific expansion of NKT cells in response to the NKT ligand -galactosylceramide α-GalCer) in patients with myeloma and in healthy donors and. NKT cells activated in the presence of lenalidomide have greater ability to secrete interferon-ϒ and patients with multiple myeloma or MDS del5Q treated with lenalidomide have an increase in their NKT cells in vivo [59].

Apoptosis/growth arrestLenalidomide

MonocytesNK cells

T cellsTumour cells e.g. Multiple

myeloma

Apoptosis/necrosis/growth

arrest

Endothelial cellangiogenesis

Inhibition ofangiogenesis

Inhibition ofBFGF/VEGF

Co-stimulation of CD3/increase of IFN-γEnhanced killing of

tumour cells/enhanced host

immunity

Inhibition of TNF-α/PGE2 underinflammatory

settings

Increase ofapoptotic

factors/growtharrest

NKT cells

Inhibition ofinvasion/metastasis

Tumour cells

Figure2.Mechanismsofactionoflenalidomideincell/tissueprocesses.BFGF: Basic fibroblastic-growth factor; NK: Natural killer cell; NKT: Natural killer T cell.

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 6: Lenalidomide: a novel anticancer drug with multiple modalities

Lenalidomide

130 ExpertOpin.Pharmacother.(2009) 10(1)

5.3 EffectsonangiogenesisAngiogenesis, the formation of new blood vessels from existing vasculature, plays a crucial role in tumour progression [60]. New blood vessels are required for tumour growth beyond 1 – 2 mm3; therefore, inhibition of angiogenesis has become an important target for cancer treatment [61]. Increased angiogenesis in the bone marrow of patients with multiple myeloma is a poor prognostic feature [62,63], Neovascularisation in multiple myeloma is mediated by various angiogenic growth factors released by tumour cells and the bone marrow micro-environment such as VEGF, basic fibroblastic-growth factor and hepatocyte-growth factor. IMiDs immunomodulatory compounds can inhibit in vivo angiogenesis in xeno- transplantation models such as Hodgkin’s lymphoma [64], melanoma [65], cervical cancer [66] and colorectal cancer. Lenalidomide has anti-angiogenic activity shown in the rat mesenteric-ring assay [67]. A key inhibitory mechanism in this study has been inhibition of AKT phosphorylation. Other inhibitory mechanisms put forward by the IMiDs immuno-modulatory compounds include inhibition of endothelial cell migration, adhesion and capillary-tube formation, endothelial cell apoptosis in 3D collagen cultures [68], and inhibition of key pro-angiogenic growth factors such as VEGF, basic fibroblastic-growth factor and HIF [69,70]. It is not yet been determined whether inhibition of angiogenesis by lenalidomide and the IMiDs immunomodulatory compounds are crucial to responses in multiple myeloma and other diseases as some studies showed a lack of decreases in proangiogeneic cytokines in thalidomide treated multiple myeloma patients [71,72] and the drugs display anti-angiogenic activity in vitro independently of immunomodulatory effects [73].

5.4 EffectsonmetastasisandcellmigrationMetastasis is critical for cancer progression being present in > 30% of patients at initial diagnosis. The process is complex, involving cell migration from the primary site, cell adhesion and growth in new organs. Thalidomide and

lenalidomide inhibit metastasis in mouse xenograft models of cancers such as Hodgkin’s lymphoma [64] and ocular melanoma [74], The mechanisms for metastasis inhibition are unclear, but so far, with thalidomide, it has been shown that migration [75], inhibition of cell adhesion molecules such as ICAM-1 and LFA-1 [76] and β2 and β3 integrins [75], and gap-junction function are involved [77]. It is believed that increased antitumour responses mediated by NK cells and dendritic cells caused by lenalidomide may augment metastasis inhibition in vivo [64,78], but because direct anti-invasive and anti-adhesive effects occur in vitro, the drug can inhibit steps involved in metastasis irrespective of immune function.

6. Expertopinion

Based on the current wealth of data on lenalidomide, its has great potential as a leading cancer drug. Unlike most existing anticancer therapeutics, lenalidomide and its IMiD™ immuno-modulatory compounds analogues have several mechanisms of action encompassing many signalling pathways, which has the advantage of potentially decreasing drug resistance. A range of immunostimulatory effects have been reported in a number of effector-cell types suggesting a powerful mechanism for the drugs antitumour activities. Studies are now focusing on the molecular mechanisms of action of the compound, which is being facilitated by state of the art genomic and proteomic array technologies. These future studies will define the gene and protein targets of the lenalidomide and the IMiD immunomodulatory compounds analogues and allow more focused therapies in combination with other known and novel therapeutics.

Declarationofinterest

A research grant has been awarded to AG Dalgleish from the Celgene corporation.

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 7: Lenalidomide: a novel anticancer drug with multiple modalities

Galustian&Dalgleish

ExpertOpin.Pharmacother.(2009) 10(1) 131

Bibliography1. Bartlett JB, Michael A, Clarke IA, et al.

Phase I study to determine the safety, tolerability and immunostimulatory activity of thalidomide analogue CC-5013 in patients with metastatic malignant melanoma and other advanced cancers. Br J Cancer 2004;90:955-61

2. Schey SA, Fields P, Bartlett JB, et al. Phase I study of an immunomodulatory thalidomide analog, CC-4047, in relapsed or refractory multiple myeloma. J Clin Oncol 2004;22:3269-76

3. Brandenburg NA, Weiss L, Bwire R, et al. Venous thromboembolism in patients with myelodysplastic syndrome treated with lenalidomide: Incidence and risk factors. J Clin Oncol 2008;26:7084 [Meeting Abstracts]

4. Richardson PG, Schlossman RL, Weller E, et al. Immunomodulatory drug CC-5013 overcomes drug resistance and is well tolerated in patients with relapsed multiple myeloma. Blood 2002;100:3063-7

5. Weber DM, Chen C, Niesvizky R, et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med 2007;357:2133-42

6. Dimopoulos M, Spencer A, Attal M, et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 2007;357:2123-32

7. Jagannath S, Richardson PG, Zeldenrust S, et al. Long-term responses observed with lenalidomide therapy for patients with relapsed or refractory multiple myeloma. J Clin Oncol 2008;26:8525 [Meeting Abstracts]

8. Rajkumar SV, Jacobus S, Callander N, et al. Randomized trial of lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone in newly diagnosed myeloma (E4A03), a trial coordinated by the Eastern Cooperative Oncology Group: Analysis of response, survival, and outcome wi. J Clin Oncol 2008;26:8504 [Meeting Abstracts]

9. Anderson KC, Jagannath S, Jakubowiak A, et al. Phase II study of lenalidomide (Len), bortezomib (Bz), and dexamethasone (Dex) in patients (pts) with relapsed or relapsed and refractory multiple myeloma (MM). J Clin Oncol 2008;26:8545 [Meeting Abstracts]

10. Van Etten RA, Shannon KM. Focus on myeloproliferative diseases and

myelodysplastic syndromes. Cancer Cell 2004;6:547-52

11. List A, Kurtin S, Roe DJ, et al. Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med 2005;352:549-57

12. Ortega J, List A. Immunomodulatory drugs in the treatment of myelodysplastic syndromes. Curr Opin Oncol 2007;19:656-9

13. Sokol L, List AF. Immunomodulatory therapy for myelodysplastic syndromes. Int J Hematol 2007;86:301-5

14. Chen J, McMillan NA. Molecular basis of pathogenesis, prognosis and therapy in chronic lymphocytic leukaemia. Cancer Biol Ther 2008;174-9

15. Ferrajoli A, Lee BN, Schlette EJ, et al. Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia. Blood 2008;11:5291-7

16. Andritsos LA, Johnson AJ, Lozanski G, et al. Higher doses of lenalidomide are associated with unacceptable toxicity including life-threatening tumor flare in patients with chronic lymphocytic leukemia. J Clin Oncol 2008;26:2519-25

17. Chanan-Khan AA, Whitworth A, Bangia N, et al. Lenalidomide-Associated Tumor Flare Reaction Is Manageable in Patients With Chronic Lymphocytic Leukemia. J Clin Oncol 2008;26:4851-2

18. Lin TS. Lenalidomide: what is the right dose in CLL? Blood 2008;111:5268

19. Coleman M, Martin P, Ruan J, et al. The THRIL (thalidomide [T], rituximab [R], and lenalidomide [L]) regimen for chronic lymphocytic leukemia, small lymphocytic lymphoma, and mantle cell lymphoma: daily alternating IMiDs and rituximab maintenance. J Clin Oncol 2008;26:7079 [Meeting Abstracts]

20. Chanan-Khan A, Miller KC, Musial L, et al. Clinical efficacy of lenalidomide in patients with relapsed or refractory chronic lymphocytic leukemia: results of a Phase II study. J Clin Oncol 2006;24:5343-9

21. Ramsay AG, Johnson AJ, Lee AM, et al. Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. J Clin Invest 2008;118:2427-37

22. James DF, Betty MR, Mosadeghi R, Kipps TJ. Lenalidomide abrogates the protective influence of nurse-like cells on primary

chronic lymphocytic leukemia cells in vitro. Blood 2007;110:A916-7

23. Wang M, Fayad L, Hagemeister F, et al. A Phase I/II study of lenalidomide (Len) in combination with rituximab (R) in relapsed/refractory mantle cell lymphoma (MCL) with early evidence of efficacy. J Clin Oncol 2007;25:8030 [Meeting Abstracts]

24. Czuczman MS, Reeder CB, Polikoff J, et al. International study of lenalidomide in relapsed/refractory aggressive non-Hodgkin’s lymphoma. J Clin Oncol 2008;26:8509 [Meeting Abstracts]

25. Zhang L, Schafer P, Muller G, et al. The ratio of cyclin D1/p21kip baseline gene expression and SPARC gene expression can be potential predictors of non-Hodgkin’s lymphoma (NHL) patient response to lenalidomide therapy. J Clin Oncol 2008;26:22150 [Meeting Abstracts]

26. Witzig TE, Vose JM, Justice G, et al. Lenalidomide oral monotherapy in relapsed/refractory small lymphocytic non-Hodgkin’s lymphoma. J Clin Oncol 2008;26:8573 [Meeting Abstracts]

27. Blum WG, Andritsos LA, Johnson AJ, et al. Preliminary results of a Phase I trial with dose escalated lenalidomide (L) in separate cohorts of relapsed acute leukemia (AL) or chronic lymphocytic leukemia (CLL): evidence of activity in acute myeloid leukemia (AML) and toxicity in CLL at starting dose. J Clin Oncol 2008;26:7075 [Meeting Abstracts]

28. Garcia JA, Triozzi P, Elson P, et al. Clinical activity of ketoconazole and lenalidomide in castrate progressive prostate carcinoma (CPPCA): Preliminary results of a Phase II trial. J Clin Oncol 2008;26:5143 [Meeting Abstracts]

29. Ain KB, Lee C, Holbrook KM, et al. Phase II study of lenalidomide in distantly metastatic, rapidly progressive, and radioiodine-unresponsive thyroid carcinomas: preliminary results. J Clin Oncol 2008;26:6027 [Meeting Abstracts]

30. Hideshima T, Chauhan D, Shima Y, et al. Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy. Blood 2000;96:2943-50

31. Verhelle D, Corral LG, Wong K, et al. Lenalidomide and CC-4047 inhibit the proliferation of malignant B cells while

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 8: Lenalidomide: a novel anticancer drug with multiple modalities

Lenalidomide

132 ExpertOpin.Pharmacother.(2009) 10(1)

expanding normal CD34+ progenitor cells. Cancer Res 2007;67:746-55

32. Mitsiades N, Mitsiades CS, Poulaki V, et al. Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications. Blood 2002;99:4525-30

33. Pellagatti A, Jadersten M, Forsblom AM, et al. Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients. Proc Natl Acad Sci USA 2007;104:11406-11

34. Sampaio EP, Sarno EN, Galilly R, et al. Thalidomide selectively inhibits tumor necrosis factor alpha production by stimulated human monocytes. J Exp Med 1991;173:699-703

35. Corral LG, Kaplan G. Immunomodulation by thalidomide and thalidomide analogues. Ann Rheum Dis 1999;58(Suppl 1):I107-13

36. Corral LG, Muller GW, Moreira AL, et al. Selection of novel analogs of thalidomide with enhanced tumor necrosis factor alpha inhibitory activity. Mol Med 1996;2:506-15

37. LeBlanc R, Hideshima T, Catley LP, et al. Immunomodulatory drug costimulates T cells via the B7-CD28 pathway. Blood 2004;103:1787-90

38. Schafer PH, Gandhi AK, Loveland MA, et al. Enhancement of cytokine production and AP-1 transcriptional activity in T cells by thalidomide-related immunomodulatory drugs. J Pharmacol Exp Ther 2003;305:1222-32

39. Payvandi F, Wu L, Naziruddin SD, et al. Immunomodulatory drugs (IMiDs) increase the production of IL-2 from stimulated T cells by increasing PKC-theta activation and enhancing the DNA-binding activity of AP-1 but not NF-kappaB, OCT-1, or NF-AT. J Interferon Cytokine Res 2005;25:604-16

40. Jones E, hm-Vicker M, Golgher D, Gallimore A. CD25+ regulatory T cells and tumor immunity. Immunol Lett 2003;85:141-3

41. Hori S, Sakaguchi S. Foxp3: a critical regulator of the development and function of regulatory T cells. Microbes Infect 2004;6:745-51

42. Galustian C, Klaschka DC, Meyer B, et al. Lenalidomide (Revlimid(R), CC-5013) and ActimidTM (CC-4047) inhibit the function and expansion of T regulatory (Treg) cells in vitro: Implications for

anti-tumor activity in vivo AACR 2006:1147 [Meeting Abstracts]

43. Galustian C, Meyer B, Labarthe MC, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol Immunother 2008 (In Press)

44. Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004;10:942-9

45. Liyanage UK, Moore TT, Joo HG, et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol 2002;169:2756-61

46. Diederichsen ACP, Zeuthen J, Christensen PB, Kristensen T. Characterisation of tumour infiltrating lymphocytes and correlations with immunological surface molecules in colorectal cancer. Eur J Cancer 1999;35:721-6

47. Ichihara F, Kono K, Takahashi A, et al. Increased Populations of Regulatory T Cells in Peripheral Blood and Tumor-Infiltrating Lymphocytes in Patients with Gastric and Esophageal Cancers. Clin Cancer Res 2003;9:4404-8

48. Sato E, Olson SH, Ahn J, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. PNAS 2005;102:18538-43

49. Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol 2006;6:295-307

50. Beyer M, Kochanek M, Giese T, et al. In vivo peripheral expansion of naive CD4+CD25high FOXP3+ regulatory T cells in patients with multiple myeloma. Blood 2006;107:3940-9

51. Yang ZZ, Novak AJ, Stenson MJ, et al. Intratumoral CD4+CD25+ regulatory T-cell-mediated suppression of infiltrating CD4+ T cells in B-cell non-Hodgkin lymphoma. Blood 2006;107:3639-46

52. Motta M, Rassenti L, Shelvin BJ, et al. Increased expression of CD152 (CTLA-4) by normal T lymphocytes in untreated patients with B-cell chronic lymphocytic leukemia. Leukemia 2005;19:1788-93

53. Smyth MJ, Hayakawa Y, Takeda K, Yagita H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat Rev Cancer 2002;2:850-61

54. Zhu D, Corral LG, Fleming YW, Stein B. Immunomodulatory drugs Revlimid((R)) (lenalidomide) and CC-4047 induce apoptosis of both hematological and solid tumor cells through NK cell activation. Cancer Immunol Immunother 2008;57:1849-59

55. Wu L, Adams M, Carter T, et al. Lenalidomide Enhances Natural Killer Cell and Monocyte-Mediated Antibody-Dependent Cellular Cytotoxicity of Rituximab-Treated CD20+ Tumor Cells. Clin Cancer Res 2008;14:4650-7

56. Wu L, Adams M, Schafer P, et al. Effect of lenalidomide and pomalidomide combined with IgG1-isotype antibodies on antibody-dependent cellular cytotoxicity (ADCC) via cytokine signaling and effector cell granzyme B and FasL expression. J Clin Oncol 2008;26:3058 [Meeting Abstracts]

57. Hayashi T, Hideshima T, Akiyama M, et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. Br J Haematol 2005;128:192-203

58. Kronenberg M. Toward an understanding of NKT cell biology: progress and paradoxes. Annu Rev Immunol 2005;23:877-900

59. Chang DH, Liu N, Klimek V, et al. Enhancement of ligand dependent activation of human Natural Killer T cells by Lenalidomide: Therapeutic Implications. Blood 2006;108:2005-10

60. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971;285:1182-6

61. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature 2000;407:249-57

62. Eleutherakis-Papaiakovou V, Karali M, Kokkonouzis I, et al. Bone marrow angiogenesis and progression in multiple myeloma: clinical significance and therapeutic approach. Leuk Lymphoma 2003;44:937-48

63. Xu JL, Lai R, Kinoshita T, et al. Proliferation, apoptosis, and intratumoral vascularity in multiple myeloma: correlation with the clinical stage and cytological grade. J Clin Pathol 2002;55:530-4

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.

Page 9: Lenalidomide: a novel anticancer drug with multiple modalities

Galustian&Dalgleish

ExpertOpin.Pharmacother.(2009) 10(1) 133

64. Reddy N, Hernandez-Ilizaliturri FJ, Deeb G, et al. Immunomodulatory drugs stimulate natural killer-cell function, alter cytokine production by dendritic cells, and inhibit angiogenesis enhancing the anti-tumour activity of rituximab in vivo Br J Haematol 2008;140:36-45

65. Heere-Ress E, Boehm J, Thallinger C, et al. Thalidomide enhances the anti-tumor activity of standard chemotherapy in a human melanoma xenotransplatation model. J Invest Dermatol 2005;125:201-6

66. Downs LS Jr, Rogers LM, Yokoyama Y, Ramakrishnan S. Thalidomide and angiostatin inhibit tumor growth in a murine xenograft model of human cervical cancer. Gynecol Oncol 2005;98:203-10

67. Dredge K, Horsfall R, Robinson SP, et al. Orally administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell migration and Akt phosphorylation in vitro. Microvasc Res 2005;69:56-63

68. Fujita K, Asami Y, Tanaka K, et al. Anti-angiogenic effects of thalidomide: expression of apoptosis-inducible active-caspase-3 in a three-dimensional collagen gel culture of aorta. Histochem Cell Biol 2004;122:27-33

69. Li X, Liu X, Wang J, et al. Thalidomide down-regulates the expression of VEGF and bFGF in cisplatin-resistant human

lung carcinoma cells. Anticancer Res 2003;23:2481-7

70. Li X, Liu X, Wang J, et al. Effects of thalidomide on the expression of angiogenesis growth factors in human A549 lung adenocarcinoma cells. Int J Mol Med 2003;11:785-90

71. Neben K, Moehler T, Kraemer A, et al. Response to thalidomide in progressive multiple myeloma is not mediated by inhibition of angiogenic cytokine secretion. Br J Haematol 2001;115:605-8

72. Rosinol L, Cibeira MT, Segarra M, et al. Response to thalidomide in multiple myeloma: impact of angiogenic factors. Cytokine 2004;26:145-8

73. Dredge K, Marriott JB, Macdonald CD, et al. Novel thalidomide analogues display anti-angiogenic activity independently of immunomodulatory effects. Br J Cancer 2002;87:1166-72

74. Mangiameli DP, Blansfield JA, Kachala S, et al. Combination therapy targeting the tumor microenvironment is effective in a model of human ocular melanoma. J Trans Med 2007;5:38

75. McCarty MF. Thalidomide may impede cell migration in primates by down-regulating integrin beta-chains: potential therapeutic utility in solid malignancies, proliferative retinopathy, inflammatory disorders, neointimal

hyperplasia, and osteoporosis. Med Hypotheses 1997;49:123-31

76. Settles B, Stevenson A, Wilson K, et al. Down-regulation of cell adhesion molecules LFA-1 and ICAM-1 after in vitro treatment with the anti-TNF-alpha agent thalidomide. Cell Mol Biol (Noisy le grand) 2001;47:1105-14

77. Onat D, Stahl W, Sies H. Stimulation of gap junctional intercellular communication by thalidomide and thalidomide analogs in human fetal skin fibroblasts (HFFF2) and in rat liver epithelial cells (WB-F344). Biochem Pharmacol 2001;62:1081-6

78. Davies FE, Raje N, Hideshima T, et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood 2001;98:210-6

AffiliationChristine Galustian† & Angus Dalgleish†Author for correspondenceSt Georges University of London, Division of Cellular and Molecular Medicine, Department of Oncology, Cranmer Terrace, Tooting, London Tel: +44 208 725 0954; Fax: +44 208 725 0158; E-mail: [email protected]

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Abe

rdee

n on

09/

25/1

3Fo

r pe

rson

al u

se o

nly.


Recommended