1,769
Views
12
CrossRef citations to date
0
Altmetric
Editorial

Novel insights into the mechanism of action of lenalidomide

&
Article: e28386 | Received 01 Feb 2014, Accepted 01 Feb 2014, Published online: 01 Feb 2014

Lenalidomide (Revlimid®) is a synthetic derivative of thalidomide (Thalomid®) currently licensed by the US Food and Drug Administration (FDA) and other international regulatory agencies for the treatment of multiple myeloma (MM) (in combination with dexamethasone)Citation1-Citation3 and low or intermediate-1 risk myelodysplastic syndromes bearing 5q cytogenetic abnormalities (as a standalone agent).Citation4-Citation6 Lenalidomide was originally developed to improve the safety profile of thalidomide, which eventually turned out to be responsible for an estimated number of 10 000–20 000 phocomelic babies worldwide in the 1950–60s.Citation7 As compared with thalidomide, lenalidomide is associated with limited neurotoxic activity, but is not completely devoid of teratogenic effects.Citation8-Citation10 For this reason, both thalidomide (which is also approved for use in MM patients, together with dexamethasone)Citation11 and lenalidomide are commercialized under tightly controlled distribution programs (as per explicit request of the US FDA).Citation8 Recently, a novel thalidomide derivative, pomalidomide (Pomalyst®), has been approved for the treatment of specific subsets of MM patients, including individuals who progressed on or shortly after thalidomide therapy.Citation12,Citation13 At odds with thalidomide and lenalidomide, pomalidomide exerts very limited (if any) teratogenic activity.Citation14

Throughout the 2000s, thalidomide and lenalidomide have been the subjects of an intense wave of investigation, revealing multiple biological effects that could account for their antineoplastic activity.Citation15-Citation17 In particular, lenalidomide has been shown to (1) limit the proliferation of cancer cells and promote their death; (2) interrupt the trophic support provided to malignant cells by the tumor stroma; and (3) operate as a pleiotropic immunomodulator.Citation18,Citation19 Owing to their ability to boost both the innate and adaptive arm of the immune response, thalidomide, lenalidomide, and pomalidomide are collectively referred to as immunomodulatory drugs (IMiDs).Citation20,Citation21

In spite of the acute interest generated by IMiDs throughout the past decade, the actual molecular target of these drugs has been discovered only in 2010, when thalidomide was shown to physically interact with (and hence inhibit) the E3 ubiquitin ligase cereblon (CRBN).Citation22 Zebrafish (Danio rerio) embryos exposed to thalidomide developed limb abnormalities that mimicked human phocomelia, unless they expressed a variant of CRBN that does not bind IMiDs (CRBNYWAA).Citation22 Importantly, developmental limb defects also manifested in CRBN-deficient zebrafish, indicating that the teratogenic effects of thalidomide stem from CRBN inhibition.Citation22,Citation23 Apparently in contrast with this notion, lenalidomide and pomalidomide were subsequently suggested to stabilize CRBN,Citation24 thus decreasing the half-life of several proteins that are normally degraded by the ubiquitin-proteasome system, including interferon-regulatory factor 4 (IRF4).Citation24,Citation25 Of note, the stable depletion of CRBN endowed cultured cancer cells with a pronounced resistance to IMiDs. Moreover, the neoplastic compartment of MM patients who failed to respond to lenalidomide-based therapy was found to express low CRBN levels after treatment.Citation25 These findings indicated that the antineoplastic activity of IMiDs actually requires CRBN, a notion that has recently been confirmed and expanded by 2 independent studies.Citation26,Citation27

By means of 2 distinct experimental approaches, i.e., stable isotope labeling of amino acids in cell culture (SILAC)-based quantitative proteomics and a firefly luciferase-based approach to monitor protein stability, the groups headed by Benjamin L Ebert and William G Kaelin Jr demonstrated that the CRBN-dependent antineoplastic activity of lenalidomide originates from the degradation of 2 lymphoid transcription factors, namely, IKAROS family zinc finger 1 (IKZF1, also known as Ikaros) and IKZF3 (also known as Aiolos).Citation26,Citation27 In particular, lenalidomide (as well as thalidomide and pomalidomide) was found to increase the binding of CRBN to IKZF1 and IKZF3 (but not other Ikaros family members), thus promoting their proteasomal degradation.Citation26,Citation27 In line with previous findings,Citation24,Citation25 such an effect could be abrogated by the depletion of CRBN with short-hairpin RNAs (shRNAs), rendering MM cells resistant to the cytotoxic activity of lenalidomide. Similar results were obtained with CRBN−/− cells as well as with CRBN−/− cells expressing CRBNYWAA.Citation26,Citation27 Conversely, the complementation of CRBN−/− cells with wild-type CRBN restored the sensitivity of IKZF1 and IKZF3 to lenalidomide-triggered proteasomal degradation.Citation27

In addition, the groups lead by Benjamin L Ebert and William G Kaelin Jr succeeded in mapping the residue of Ikaros family members that is responsible for their differential sensitivity to lenalidomide-triggered degradation. Thus, at odds with their wild-type counterparts, IKZF3Q147H and IKZF4H188Q were shown to be insensitive and sensitive, respectively, to lenalidomide.Citation26,Citation27 Importantly, the shRNA-mediated depletion of IKZF1 or IKZF3 was sufficient to stimulate the demise of lenalidomide-sensitive cells, an effect that was mimicked by a dominant negative variant of IKZF3 lacking the DNA-binding domain. Conversely, the overexpression of wild-type IKZF3 (and more so that of degradation-resistant IKZF mutants, i.e., IKZF1Q146H and IKZF3Q147H) protected MM cells against lenalidomide cytotoxicity.Citation26,Citation27

The degradation of IKZF1 and IKZF3 in IMiD-sensitive cells exposed to lenalidomide was often followed by a decline in the levels of IRF4,Citation26,Citation27 a transcription factor that had previously been involved in the mechanism of action of this drug.Citation25,Citation28,Citation29 However, some cell lines succumbed to lenalidomide in the absence of appreciable IRF4 downregulation, suggesting that the direct antineoplastic effects of IMiDs involve at least another molecular target of Ikaros family members. Finally, T cells exposed to lenalidomide secreted high amounts of interleukin-2, a potent immunostimulatory cytokine,Citation30,Citation31 as a correlate of IKZF1/IKZF3 degradation,Citation26 reinstating the notion that the biological activity of IMiDs is broad and involves a non-negligible immunological component.Citation32,Citation33

In summary, the research units headed by Benjamin L Ebert and William G Kaelin Jr have provided novel insights into the mechanism of action of lenalidomide and other IMiDs, possibly identifying a therapeutic window to definitively discriminate between their teratogenic and therapeutic effects. What remains to be precisely elucidated is to which extent the degradation of IKZF1 and IKZF3 as triggered by lenalidomide mediates antineoplastic effects via cancer cell-intrinsic vs. immunological mechanisms. Moreover, it will be interesting to understand whether polymorphisms in IKZF1, IKZF3, and/or genes encoding their transcriptional targets influence the propensity of individual MM patients to respond to lenalidomide. Well-designed preclinical and clinical studies are required for addressing these hitherto unresolved issues.

Abbreviations:
CRBN=

cereblon

FDA=

Food and Drug Administration

IFR4=

interferon-regulatory factor 4

IKZF=

IKAROS family zinc finger

IMiD=

immunomodulatory drug

MM=

multiple myeloma

shRNA=

short-hairpin RNA

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

Authors are supported by the Ligue contre le Cancer (équipe labelisée); Agence National de la Recherche (ANR); Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; AXA Chair for Longevity Research; Institut National du Cancer (INCa); Fondation Bettencourt-Schueller; Fondation de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).

Citation: Semeraro M, Galluzzi L. Novel insights into the mechanism of action of lenalidomide. OncoImmunology 2014; 3:e28386; 10.4161/onci.28386

References

  • Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau JL, Dmoszynska A, San Miguel J, Hellmann A, Facon T, Foà R, et al, Multiple Myeloma (010) Study Investigators. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 2007; 357:2123 - 32; http://dx.doi.org/10.1056/NEJMoa070594; PMID: 18032762
  • Weber DM, Chen C, Niesvizky R, Wang M, Belch A, Stadtmauer EA, Siegel D, Borrello I, Rajkumar SV, Chanan-Khan AA, et al, Multiple Myeloma (009) Study Investigators. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med 2007; 357:2133 - 42; http://dx.doi.org/10.1056/NEJMoa070596; PMID: 18032763
  • Qian J, Yi Q. DKK1 as a novel target for myeloma immunotherapy. Oncoimmunology 2012; 1:756 - 8; http://dx.doi.org/10.4161/onci.19655; PMID: 22934273
  • List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman E, Powell B, Greenberg P, Thomas D, Stone R, et al, Myelodysplastic Syndrome-003 Study Investigators. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med 2006; 355:1456 - 65; http://dx.doi.org/10.1056/NEJMoa061292; PMID: 17021321
  • List A, Kurtin S, Roe DJ, Buresh A, Mahadevan D, Fuchs D, Rimsza L, Heaton R, Knight R, Zeldis JB. Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med 2005; 352:549 - 57; http://dx.doi.org/10.1056/NEJMoa041668; PMID: 15703420
  • Mailloux AW, Epling-Burnette PK. Effector memory regulatory T-cell expansion marks a pivotal point of immune escape in myelodysplastic syndromes. Oncoimmunology 2013; 2:e22654; http://dx.doi.org/10.4161/onci.22654; PMID: 23524348
  • Curran WJ. The thalidomide tragedy in Germany: the end of a historic medicolegal trial. N Engl J Med 1971; 284:481 - 2; http://dx.doi.org/10.1056/NEJM197103042840906; PMID: 5100423
  • Bwire R, Freeman J, Houn F. Managing the teratogenic risk of thalidomide and lenalidomide: an industry perspective. Expert Opin Drug Saf 2011; 10:3 - 8; http://dx.doi.org/10.1517/14740338.2011.527331; PMID: 21121869
  • Christian MS, Laskin OL, Sharper V, Hoberman A, Stirling DI, Latriano L. Evaluation of the developmental toxicity of lenalidomide in rabbits. Birth Defects Res B Dev Reprod Toxicol 2007; 80:188 - 207; http://dx.doi.org/10.1002/bdrb.20115; PMID: 17570132
  • Sharma RA, Steward WP, Daines CA, Knight RD, O’Byrne KJ, Dalgleish AG. Toxicity profile of the immunomodulatory thalidomide analogue, lenalidomide: phase I clinical trial of three dosing schedules in patients with solid malignancies. Eur J Cancer 2006; 42:2318 - 25; http://dx.doi.org/10.1016/j.ejca.2006.05.018; PMID: 16899362
  • Singhal S, Mehta J, Desikan R, Ayers D, Roberson P, Eddlemon P, Munshi N, Anaissie E, Wilson C, Dhodapkar M, et al. Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med 1999; 341:1565 - 71; http://dx.doi.org/10.1056/NEJM199911183412102; PMID: 10564685
  • Leleu X, Attal M, Arnulf B, Moreau P, Traulle C, Marit G, Mathiot C, Petillon MO, Macro M, Roussel M, et al, Intergroupe Francophone du Myélome. Pomalidomide plus low-dose dexamethasone is active and well tolerated in bortezomib and lenalidomide-refractory multiple myeloma: Intergroupe Francophone du Myélome 2009-02. Blood 2013; 121:1968 - 75; http://dx.doi.org/10.1182/blood-2012-09-452375; PMID: 23319574
  • Richardson PG, Siegel D, Baz R, Kelley SL, Munshi NC, Laubach J, Sullivan D, Alsina M, Schlossman R, Ghobrial IM, et al. Phase 1 study of pomalidomide MTD, safety, and efficacy in patients with refractory multiple myeloma who have received lenalidomide and bortezomib. Blood 2013; 121:1961 - 7; http://dx.doi.org/10.1182/blood-2012-08-450742; PMID: 23243282
  • Mahony C, Erskine L, Niven J, Greig NH, Figg WD, Vargesson N. Pomalidomide is nonteratogenic in chicken and zebrafish embryos and nonneurotoxic in vitro. Proc Natl Acad Sci U S A 2013; 110:12703 - 8; http://dx.doi.org/10.1073/pnas.1307684110; PMID: 23858438
  • Davies F, Baz R. Lenalidomide mode of action: linking bench and clinical findings. Blood Rev 2010; 24:Suppl 1 S13 - 9; http://dx.doi.org/10.1016/S0268-960X(10)70004-7; PMID: 21126632
  • Hsu A, Ritchie DS, Neeson P. Are the immuno-stimulatory properties of Lenalidomide extinguished by co-administration of Dexamethasone?. Oncoimmunology 2012; 1:372 - 4; http://dx.doi.org/10.4161/onci.18963; PMID: 22737619
  • Martiniani R, Di Loreto V, Di Sano C, Lombardo A, Liberati AM. Biological activity of lenalidomide and its underlying therapeutic effects in multiple myeloma. Adv Hematol 2012; 2012:842945; http://dx.doi.org/10.1155/2012/842945; PMID: 22919394
  • Semeraro M, Vacchelli E, Eggermont A, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Lenalidomide-based immunochemotherapy. Oncoimmunology 2013; 2:e2OK6494; http://dx.doi.org/10.4161/onci.26494; PMID: 24482747
  • Viel S, Charrier E, Marçais A, Rouzaire P, Bienvenu J, Karlin L, Salles G, Walzer T. Monitoring NK cell activity in patients with hematological malignancies. Oncoimmunology 2013; 2:e26011; http://dx.doi.org/10.4161/onci.26011; PMID: 24327939
  • McDaniel JM, Pinilla-Ibarz J, Epling-Burnette PK. Molecular action of lenalidomide in lymphocytes and hematologic malignancies. Adv Hematol 2012; 2012:513702; http://dx.doi.org/10.1155/2012/513702; PMID: 22888354
  • McMillin DW, Mitsiades CS. High-throughput approaches to discover novel immunomodulatory agents for cancer. Oncoimmunology 2012; 1:1406 - 8; http://dx.doi.org/10.4161/onci.21058; PMID: 23243609
  • Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, Yamaguchi Y, Handa H. Identification of a primary target of thalidomide teratogenicity. Science 2010; 327:1345 - 50; http://dx.doi.org/10.1126/science.1177319; PMID: 20223979
  • Ito T, Ando H, Handa H. Teratogenic effects of thalidomide: molecular mechanisms. Cell Mol Life Sci 2011; 68:1569 - 79; http://dx.doi.org/10.1007/s00018-010-0619-9; PMID: 21207098
  • Lopez-Girona A, Mendy D, Ito T, Miller K, Gandhi AK, Kang J, Karasawa S, Carmel G, Jackson P, Abbasian M, et al. Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Leukemia 2012; 26:2326 - 35; http://dx.doi.org/10.1038/leu.2012.119; PMID: 22552008
  • Zhu YX, Braggio E, Shi CX, Bruins LA, Schmidt JE, Van Wier S, Chang XB, Bjorklund CC, Fonseca R, Bergsagel PL, et al. Cereblon expression is required for the antimyeloma activity of lenalidomide and pomalidomide. Blood 2011; 118:4771 - 9; http://dx.doi.org/10.1182/blood-2011-05-356063; PMID: 21860026
  • Krönke J, Udeshi ND, Narla A, Grauman P, Hurst SN, McConkey M, Svinkina T, Heckl D, Comer E, Li X, et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 2014; 343:301 - 5; http://dx.doi.org/10.1126/science.1244851; PMID: 24292625
  • Lu G, Middleton RE, Sun H, Naniong M, Ott CJ, Mitsiades CS, Wong KK, Bradner JE, Kaelin WG Jr.. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 2014; 343:305 - 9; http://dx.doi.org/10.1126/science.1244917; PMID: 24292623
  • Lopez-Girona A, Heintel D, Zhang LH, Mendy D, Gaidarova S, Brady H, Bartlett JB, Schafer PH, Schreder M, Bolomsky A, et al. Lenalidomide downregulates the cell survival factor, interferon regulatory factor-4, providing a potential mechanistic link for predicting response. Br J Haematol 2011; 154:325 - 36; http://dx.doi.org/10.1111/j.1365-2141.2011.08689.x; PMID: 21707574
  • Li S, Pal R, Monaghan SA, Schafer P, Ouyang H, Mapara M, Galson DL, Lentzsch S. IMiD immunomodulatory compounds block C/EBPbeta translation through eIF4E down-regulation resulting in inhibition of MM. Blood 2011; 117:5157 - 65; http://dx.doi.org/10.1182/blood-2010-10-314278; PMID: 21389327
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2013; 2:e24850; http://dx.doi.org/10.4161/onci.24850; PMID: 24073369
  • Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2012; 1:493 - 506; http://dx.doi.org/10.4161/onci.20459; PMID: 22754768
  • Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013; 39:74 - 88; http://dx.doi.org/10.1016/j.immuni.2013.06.014; PMID: 23890065
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215 - 33; http://dx.doi.org/10.1038/nrd3626; PMID: 22301798