Next Article in Journal
Choice of High-Throughput Proteomics Method Affects Data Integration with Transcriptomics and the Potential Use in Biomarker Discovery
Next Article in Special Issue
Lenalidomide in Multiple Myeloma: Review of Resistance Mechanisms, Current Treatment Strategies and Future Perspectives
Previous Article in Journal
The Dual Blockade of the TIGIT and PD-1/PD-L1 Pathway as a New Hope for Ovarian Cancer Patients
Previous Article in Special Issue
Ixazomib, Lenalidomide and Dexamethasone in Relapsed and Refractory Multiple Myeloma in Routine Clinical Practice: Extended Follow-Up Analysis and the Results of Subsequent Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Immunomodulatory Agents for Multiple Myeloma

by
Jiří Minařík
1,* and
Sabina Ševčíková
2
1
Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, 779 00 Olomouc, Czech Republic
2
Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(23), 5759; https://doi.org/10.3390/cancers14235759
Submission received: 17 November 2022 / Accepted: 18 November 2022 / Published: 23 November 2022
(This article belongs to the Special Issue Immunomodulatory Agents for Multiple Myeloma)

Introduction

The treatment of multiple myeloma (MM) has undergone a significant paradigm shift in the last 20 years, from conventional chemotherapy to more tumor-specific treatments, based on the interference with pathogenesis of the malignant clone as well as the bone microenvironment. Indeed, the introduction of novel drugs with “biological mechanisms of action” into MM treatment has established a worldwide trend towards chemotherapy-free regimens with lower toxicity (or significantly different toxic profiles) and targeted mechanisms of action. Immunomodulatory drugs (IMiDs) represent the cornerstone of the evolution of novel therapy in MM.

1. Thalidomide

The first immunomodulatory drug used for MM was thalidomide. Despite its controversial history, thalidomide showed promising outcomes in relapsed and refractory MM in the late 1990s. Additionally, very soon, it became a part of standard regimens in all phases of MM, including the first-line setting for both transplant-eligible and -ineligible patients, in maintenance after stem cell transplant and of course in relapsed settings of MM [1,2,3,4,5,6,7,8,9]. The MPT regimen (melphalan, prednisone, and thalidomide) became the new golden standard after more than 30 years of the MP (melphalan and prednisone) doublet in transplant-ineligible patients [6]. Novel regimens with thalidomide substituted the dominant role of VAD induction (vincristine, doxorubicin, and dexamethasone) in the majority of transplant-eligible patients [10,11,12,13,14].
The major issue of thalidomide-based therapy was toxicity, including teratogenicity, thromboembolic events, rash, constipation and especially neurotoxicity [15]. Whereas the former toxicities were preventable and manageable, neurotoxicity remained an issue as it caused both sensitivity and motor impairment and was not completely reversible, even after stopping the drug. On the other hand, unlike the majority of other drugs used in MM therapy, hematological toxicity was very rare, making it a useful drug in cytopenic patients [16].

2. IMiDs—Mechanism of Action

Together with attempts to optimize thalidomide-based therapy and reduce long-lasting neuropathy (and possibly teratogenicity), research was carried out in order to reveal the mechanism of action of thalidomide and to synthesize derivates with higher efficacy and lower toxicity. The mechanism of action was initially not fully understood due to its limited in vitro activity. The first reports described thalidomide as having a dual mechanism of action—direct tumor cytotoxicity and indirect interference in the bone marrow microenvironment [2,17]. The known mechanisms included the downregulation of NF-κB signaling, thus inhibiting proliferation and promoting apoptosis, or the induction of c-jun terminal kinase (JNK)-dependent caspase-8-mediated apoptosis [18]. The indirect mechanisms include the TNF-α-induced density of the adhesion molecules ICAM-1 and VCAM-1, as well as the inhibition of IL-6 and vascular endothelial growth factor (VEGF), thus disabling tumor growth and proliferation. Interference with IGF-1 and FGF genes, which are responsible for the activation of integrins and the impairment of AKT signaling, is responsible for the anti-angiogenic effects of thalidomide [19,20]. A very complex mechanism covers the immunomodulatory role of thalidomide via the regulation of the TNF-α-, IL-6-, IL-1β-, IL-12- and CD28-mediated stimulation of CD4+ and CD8+ T-cells.
Soon after the introduction of thalidomide into routine practice, novel IMiDs were developed with similar structures. These were lenalidomide and later pomalidomide. Lenalidomide was first introduced in 2004 by the Celgene company as CC-5013. Unlike thalidomide, lenalidomide does not need metabolic activation, which enabled deeper insights into the IMiDs’ mechanism of action [16]. Most of the effects (both positive and negative), including teratogenic toxicities, were found to be caused by the IMiD-mediated inactivation of cereblon and the degradation of its transcription factors, Ikaros and Aiolos [21,22]. Thalidomide and its derivatives bind to cereblon (lenalidomide and pomalidomide bind more strongly than thalidomide), which leads to the downregulation of IRF4. IRF is a survival factor in MM and its inhibition leads to anti-proliferative effects caused by the induction of the degradation of two essential transcription factors—Ikaros (Ikaros family zinc finger protein 1) and Aiolos (Ikaros family zinc finger protein 3) [23]. The most important pathogenetic findings suggest that the substrate recognition by cereblon depends on the structure of the ligands that bind to cereblon [23]. The modification of the cereblon substrate’s specificity is, however, also the reason behind the teratogenicity of IMiDs [23].
Teratogenicity was the reason for a limited approval of thalidomide in only a few defined conditions, and it is the reason for strict pregnancy-prevention programs in IMiD-based therapies. Recent research identified possible neosubstrates responsible for IMiDs’ teratogenicity, such as promyelocytic leukaemia zinc finger (PLZF)/ZBTB16 protein; still, the mechanism seems to be more complex and probably inseparable from IMiD anti-tumor activity [24].

3. Lenalidomide

Lenalidomide, the first thalidomide derivative, soon found its largest utility in MM. The initial clinical trials revealed feasibility in the relapsed setting. Still, there were limitations due to its myelosuppressive toxicity, which was not observed in thalidomide trials. In combination with other myelosuppressive drugs (melphalan, cyclophosphamide, and anthracyclines), hematological toxicity seemed to be an issue, though it was manageable [25]. Nevertheless, the therapeutic outcomes were impressive, especially in patients who were able to sustain treatment for a long time, especially as the clinical trials started to be designed “until progression”. The overall response rates (ORR) of RD combination (lenalidomide and dexamethasone) in relapsed multiple myeloma exceeded 60%, with a median progression-free survival (mPFS) of over 11 months [26,27]. The combination enabled lower doses of both drugs with limited toxicity and set up a new gold standard for both relapsed settings and newly diagnosed MM [26,27,28,29,30]. At the moment, RD-based regimens are some of the most effective regimens in relapsed and/or refractory MM when combined with proteasome inhibitors—bortezomib (VRD), carfilzomib (KRD), ixazomib (IRD)—or with monoclonal antibodies—elotuzumab (EloRD) and daratumumab (DRD)—with a consistent ORR over 70% and mPFS over 20 months, which was previously only seen in newly diagnosed patients with standard-risk MM [31,32,33,34,35].
Some of these combinations have been tested in frontline settings and are very likely to become the new preferred option for transplant-ineligible patients (especially DRD regimen) [36]. In transplant-eligible patients, lenalidomide is becoming the cornerstone of induction therapy and maintenance treatment. Lenalidomide maintenance after autologous stem cell transplant added 2 years to the progression-free survival and 2.5 years to overall survival of MM patients, thus shifting the prognosis of MM even further [37].

4. Pomalidomide

Pomalidomide is the “youngest” IMiD routinely used in clinical practice. It is considered the most efficient drug among the IMiDs; however, it is generally used in patients with advanced MM after pretreatment with several drugs, including lenalidomide [38,39]. This might affect the final outcomes despite the evidence of only the minimal cross-resistance of these drugs. Pomalidomide is metabolized predominantly by the liver (unlike lenalidomide) and does not need dose reductions in patients with renal failure [38]. Similarly to lenalidomide, the major toxicities of pomalidomide include neutropenia, anemia and thrombocytopenia, with the most common non-hematologic adverse events being infection (pneumonia, bronchitis). In the registration phase III trial (MM-003), the doublet pomalidomide and dexamethasone versus high-dose dexamethasone in highly pretreated and refractory MM patients lead to significantly better PFS (median 4.0 months vs. 1 month) and OS (12.7 months vs. 8.1 months) [40]. Similarly to lenalidomide, better outcomes were seen in three drug combinations with pomalidomide, such as with bortezomib (PVD), elotuzumab (EloPD), isatuximab (IsaPD) and daratumumab (DaraPD), exceeding an mPFS of 11 months even in a highly pretreated and lenalidomide refractory population [41,42,43,44].

5. Summary

IMiDs have become the essential part of MM therapy. Their ability to pair with other drugs in a synergistic rather than only additive way makes them ideal partners for combined regimens with manageable toxicities [45]. The introduction of IMiDs has allowed us to gain insights into MM biology and enabled chemotherapy-free regimens with lower toxicities and better therapeutic outcomes in the majority of MM patients. With further research, we are eagerly awaiting new findings with respect to IMiD mechanisms of action as well as mechanisms of resistance in MM cells and their interaction with the tumor microenvironment.
This Special Issue is intended to focus on some aspects of new discoveries and applications of immunomodulatory drugs in basal, preclinical and clinical research.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Singhal, S.; Mehta, J. Thalidomide for relapsed or refractory myeloma. In Immunomodulating Drugs for the Treatment of Cancer; Chanan-Khan, A.A., Ed.; Lippincott Williams and Wilkins: Baltimore, MD, USA, 2011; pp. 51–60. [Google Scholar]
  2. 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, 1565–1571. [Google Scholar] [CrossRef] [Green Version]
  3. Barlogie, B.; Desikan, R.; Eddlemon, P.; Spencer, T.; Zeldis, J.; Munshi, N.; Badros, A.; Zangari, M.; Anaissie, E.; Epstein, J.; et al. Extended survival in advanced and refractory multiple myeloma after single-agent thalidomide: Identification of prognostic factors in a phase 2 study of 169 patients. Blood 2001, 98, 492–494. [Google Scholar] [CrossRef] [Green Version]
  4. Glasmacher, A.; Hahn, C.; Hoffmann, F.; Naumann, R.; Goldschmidt, H.; Lilienfeld-Toal, M.; Orlopp, K.; Schmidt-Wolf, I.; Gorschlüter, M. A systematic review of phase-II trials of thalidomide monotherapy in patients with relapsed or refractory multiple myeloma. Br. J. Haematol. 2006, 132, 584–593. [Google Scholar] [CrossRef]
  5. Fermand, J.-P.; Jaccard, A.; Macro, M.; Uzunhan, Y.; Allard, C.; Castaigne, S.; Divine, M.; Mariette, X.; Leblond, V.; Chevret, S. A Randomized Comparison of Dexamethasone + Thalidomide (Dex/Thal) vs Dex + Placebo (Dex/P) in Patients (pts) with Relapsing Multiple Myeloma (MM). Blood 2006, 108, 3563. [Google Scholar] [CrossRef]
  6. Palumbo, A.; Bringhen, S.; Caravita, T.; Merla, E.; Capparella, V.; Callea, V.; Cangialosi, C.; Grasso, M.; Rossini, F.; Galli, M.; et al. Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: Randomised controlled trial. Lancet 2006, 367, 825–831. [Google Scholar] [CrossRef]
  7. Attal, M.; Harousseau, J.-L.; Leyvraz, S.; Doyen, C.; Hulin, C.; Benboubker, L.; Yakoub-Agha, I.; Bourhis, J.-H.; Garderet, L.; Pegourie, B.; et al. Maintenance therapy with thalidomide improves survival in patients with multiple myeloma. Blood 2006, 108, 3289–3294. [Google Scholar] [CrossRef]
  8. Abdelkefi, A.; Ladeb, S.; Torjman, L.; Ben Othman, T.; Lakhal, A.; Ben Romdhane, N.; El Omri, H.; Elloumi, M.; Belaaj, H.; Jeddi, R.; et al. Single autologous stem-cell transplantation followed by maintenance therapy with thalidomide is superior to double autologous transplantation in multiple myeloma: Results of a multicenter randomized clinical trial. Blood 2008, 111, 1805–1810. [Google Scholar] [CrossRef]
  9. Zemanova, M.; Scudla, V.; Adam, Z.; Gregora, E.; Pour, L.; Minarik, J.; Pavlicek, P.; Pika, T.; Bacovsky, J. Low-dose thalidomide regimens in therapy of relapsed or refractory multiple myeloma. Neoplasma 2008, 55, 345–349. [Google Scholar]
  10. Lokhorst, H.M.; Schmidt-Wolf, I.; Sonneveld, P.; Van Der Holt, B.; Martin, H.; Barge, R.; Bertsch, U.; Schlenzka, J.; Bos, G.M.; Croockewit, S.; et al. Thalidomide in induction treatment increases the very good partial response rate before and after high-dose therapy in previously untreated multiple myeloma. Haematologica 2008, 93, 124–127. [Google Scholar] [CrossRef]
  11. Morgan, G.J.; Davies, F.E.; Gregory, W.M.; Bell, S.E.; Szubert, A.J.; Coy, N.N.; Cook, G.; Feyler, S.; Johnson, P.R.E.; Rudin, C.; et al. Cyclophosphamide, thalidomide, and dexamethasone as induction therapy for newly diagnosed multiple myeloma patients destined for autologous stem-cell transplantation: MRC Myeloma IX randomized trial results. Haematologica 2012, 97, 442–450. [Google Scholar] [CrossRef] [Green Version]
  12. Zervas, K.; Mihou, D.; Katodritou, E.; Pouli, A.; Mitsouli, C.; Anagnostopoulos, A.; Delibasi, S.; Kyrtsonis, M.; Terpos, E.; Zikos, P.; et al. VAD-doxil versus VAD-doxil plus thalidomide as initial treatment for multiple myeloma: Results of a multicenter randomized trial of the Greek myeloma study group. Ann. Oncol. 2007, 18, 1369–1375. [Google Scholar] [CrossRef]
  13. Minarik, J.; Sandecka, V.; Maisnar, V.; Gregora, E.; Spicka, I.; Starostka, D.; Plonkova, H.; Jarkovsky, J.; Walterová, L.; Wróbel, M.; et al. 10 years of experience with thalidomide in multiple myeloma patients: Report of the Czech Myeloma Group. Leuk. Res. 2013, 37, 1063–1069. [Google Scholar] [CrossRef]
  14. Krejci, M.; Gregora, E.; Straub, J.; Minarik, J.; Scudla, V.; Adam, Z.; Krivanova, A.; Pour, L.; Zahradova, L.; Buchler, T.; et al. Similar efficacy of thalidomide- and bortezomib-based regimens for first relapse of multiple myeloma. Ann. Hematol. 2011, 90, 1441–1447. [Google Scholar] [CrossRef]
  15. Mileshkin, L.; Stark, R.; Day, B.; Seymour, J.F.; Zeldis, J.B.; Prince, H.M. Development of Neuropathy in Patients With Myeloma Treated With Thalidomide: Patterns of Occurrence and the Role of Electrophysiologic Monitoring. J. Clin. Oncol. 2006, 24, 4507–4514. [Google Scholar] [CrossRef]
  16. Kumar, S.; Rajkumar, S.V. Thalidomide and lenalidomide in the treatment of multiple myeloma. Eur. J. Cancer 2006, 42, 1612–1622. [Google Scholar] [CrossRef]
  17. D’Amato, R.J.; Loughnan, M.S.; Flynn, E.; Folkman, J. Thalidomide is an inhibitor of angiogenesis. Proc. Natl. Acad. Sci. USA 1994, 91, 4082–4085. [Google Scholar] [CrossRef] [Green Version]
  18. Anderson, K.C. Lenalidomide and Thalidomide: Mechanisms of Action—Similarities and Differences. Semin. Hematol. 2005, 42, S3–S8. [Google Scholar] [CrossRef]
  19. Stephens, T.D.; Bunde, C.J.; Fillmore, B.J. Mechanism of action in thalidomide teratogenesis. Biochem. Pharmacol. 2000, 59, 1489–1499. [Google Scholar] [CrossRef]
  20. Vallet, S.; Palumbo, A.; Raje, N.; Boccadoro, M.; Anderson, K.C. Thalidomide and lenalidomide: Mechanism-based potential drug combinations. Leuk. Lymphoma 2008, 49, 1238–1245. [Google Scholar] [CrossRef]
  21. 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, 1345–1350. [Google Scholar] [CrossRef] [Green Version]
  22. Vrábel, D.; Vrábel, D.; Pour, L.; Pour, L.; Ševčíková, S.; Ševčíková, S. The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma. Blood Rev. 2019, 34, 56–66. [Google Scholar] [CrossRef]
  23. Ito, T.; Handa, H. Molecular mechanisms of thalidomide and its derivatives. Proc. Jpn. Acad. Ser. B 2020, 96, 189–203. [Google Scholar] [CrossRef]
  24. Yamanaka, S.; Murai, H.; Saito, D.; Abe, G.; Tokunaga, E.; Iwasaki, T.; Takahashi, H.; Takeda, H.; Suzuki, T.; Shibata, N.; et al. Thalidomide and its metabolite 5-hydroxythalidomide induce teratogenicity via the cereblon neosubstrate PLZF. EMBO J. 2021, 40, e105375. [Google Scholar] [CrossRef]
  25. Palumbo, A.; Falco, P.; Corradini, P.; Falcone, A.; Di Raimondo, F.; Giuliani, N.; Crippa, C.; Ciccone, G.; Omedè, P.; Ambrosini, M.T.; et al. Melphalan, Prednisone, and Lenalidomide Treatment for Newly Diagnosed Myeloma: A Report From the GIMEMA—Italian Multiple Myeloma Network. J. Clin. Oncol. 2007, 25, 4459–4465. [Google Scholar] [CrossRef]
  26. Weber, D.M.; Chen, C.; Niesvizky, R.; Wang, M.; Belch, A.; Stadtmauer, E.A.; Siegel, D.; Borrello, I.; Rajkumar, S.V.; Chanan-Khan, A.A.; et al. Lenalidomide plus Dexamethasone for Relapsed Multiple Myeloma in North America. N. Engl. J. Med. 2007, 357, 2133–2142. [Google Scholar] [CrossRef]
  27. Dimopoulos, M.; Spencer, A.; Attal, M.; Prince, H.M.; Harousseau, J.-L.; Dmoszynska, A.; Miguel, J.S.; Hellmann, A.; Facon, T.; Foà, R.; et al. Lenalidomide plus Dexamethasone for Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2007, 357, 2123–2132. [Google Scholar] [CrossRef] [Green Version]
  28. Gay, F.; Hayman, S.R.; Lacy, M.Q.; Buadi, F.; Gertz, M.A.; Kumar, S.; Dispenzieri, A.; Mikhael, J.R.; Bergsagel, P.L.; Dingli, D.; et al. Lenalidomide plus dexamethasone versus thalidomide plus dexamethasone in newly diagnosed multiple myeloma: A comparative analysis of 411 patients. Blood 2010, 115, 1343–1350. [Google Scholar] [CrossRef] [Green Version]
  29. Rajkumar, S.V.; Hayman, S.R.; Lacy, M.Q.; Dispenzieri, A.; Geyer, S.M.; Kabat, B.; Zeldenrust, S.R.; Kumar, S.; Greipp, P.R.; Fonseca, R.; et al. Combination therapy with lenalidomide plus dexamethasone (Rev/Dex) for newly diagnosed myeloma. Blood 2005, 106, 4050–4053. [Google Scholar] [CrossRef] [Green Version]
  30. Rajkumar, S.V.; Jacobus, S.; Callander, N.S.; Fonseca, R.; Vesole, D.H.; E Williams, M.; Abonour, R.; Siegel, D.S.; Katz, M.; Greipp, P.R. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: An open-label randomised controlled trial. Lancet Oncol. 2010, 11, 29–37. [Google Scholar] [CrossRef] [Green Version]
  31. Van Beurden-Tan, C.H.Y.; Franken, M.G.; Blommestein, H.M.; Groot, C.A.U.-D.; Sonneveld, P. Systematic Literature Review and Network Meta-Analysis of Treatment Outcomes in Relapsed and/or Refractory Multiple Myeloma. J. Clin. Oncol. 2017, 35, 1312–1319. [Google Scholar] [CrossRef]
  32. Richardson, P.G.; Xie, W.; Jagannath, S.; Jakubowiak, A.; Lonial, S.; Raje, N.S.; Alsina, M.; Ghobrial, I.M.; Schlossman, R.L.; Munshi, N.C.; et al. A phase 2 trial of lenalidomide, bortezomib, and dexamethasone in patients with relapsed and relapsed/refractory myeloma. Blood 2014, 123, 1461–1469. [Google Scholar] [CrossRef]
  33. Stewart, A.K.; Rajkumar, S.V.; Dimopoulos, M.A.; Masszi, T.; Špička, I.; Oriol, A.; Hájek, R.; Rosiñol, L.; Siegel, D.S.; Mihaylov, G.G.; et al. Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma. New Engl. J. Med. 2015, 372, 142–152. [Google Scholar] [CrossRef]
  34. Lonial, S.; Dimopoulos, M.; Palumbo, A.; White, D.; Grosicki, S.; Spicka, I.; Walter-Croneck, A.; Moreau, P.; Mateos, M.V.; Magen, H.; et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2015, 373, 621–631. [Google Scholar] [CrossRef] [Green Version]
  35. Dimopoulos, M.A.; San-Miguel, J.; Belch, A.; White, D.; Benboubker, L.; Cook, G.; Leiba, M.; Morton, J.; Ho, P.J.; Kim, K.; et al. Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone in relapsed or refractory multiple myeloma: Updated analysis of POLLUX. Haematologica 2018, 103, 2088–2096. [Google Scholar] [CrossRef] [Green Version]
  36. Facon, T.; Kumar, S.; Plesner, T.; Orlowski, R.Z.; Moreau, P.; Bahlis, N.; Basu, S.; Nahi, H.; Hulin, C.; Quach, H.; et al. Daratumumab plus Lenalidomide and Dexamethasone for Untreated Myeloma. N. Engl. J. Med. 2019, 380, 2104–2115. [Google Scholar] [CrossRef]
  37. Attal, M.; Lauwers-Cances, V.; Marit, G.; Caillot, D.; Moreau, P.; Facon, T.; Stoppa, A.M.; Hulin, C.; Benboubker, L.; Garderet, L.; et al. Lenalidomide Maintenance after Stem-Cell Transplantation for Multiple Myeloma. N. Engl. J. Med. 2012, 366, 1782–1791. [Google Scholar] [CrossRef] [Green Version]
  38. Zhu, Y.X.; Kortuem, K.M.; Stewart, A.K. Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk. Lymphoma 2012, 54, 683–687. [Google Scholar] [CrossRef]
  39. McCurdy, A.R.; Lacy, M.Q. Pomalidomide and its clinical potential for relapsed or refractory multiple myeloma: An update for the hematologist. Ther. Adv. Hematol. 2013, 4, 211–216. [Google Scholar] [CrossRef] [Green Version]
  40. Miguel, J.S.; Weisel, K.; Moreau, P.; Lacy, M.; Song, K.; Delforge, M.; Karlin, L.; Goldschmidt, H.; Banos, A.; Oriol, A.; et al. Pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): A randomised, open-label, phase 3 trial. Lancet Oncol. 2013, 14, 1055–1066. [Google Scholar] [CrossRef] [Green Version]
  41. Richardson, P.G.; Oriol, A.; Beksac, M.; Liberati, A.M.; Galli, M.; Schjesvold, F.; Lindsay, J.; Weisel, K.; White, D.; Facon, T.; et al. Pomalidomide, bortezomib, and dexamethasone for patients with relapsed or refractory multiple myeloma previously treated with lenalidomide (OPTIMISMM): A randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 781–794. [Google Scholar] [CrossRef]
  42. Dimopoulos, M.A.; Dytfeld, D.; Grosicki, S.; Moreau, P.; Takezako, N.; Hori, M.; Leleu, X.; Leblanc, R.; Suzuki, K.; Raab, M.S.; et al. Elotuzumab plus Pomalidomide and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2018, 379, 1811–1822. [Google Scholar] [CrossRef]
  43. Attal, M.; Richardson, P.G.; Rajkumar, S.V.; San-Miguel, J.; Beksac, M.; Spicka, I.; Leleu, X.; Schjesvold, F.; Moreau, P.; Dimopoulos, M.A.; et al. Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): A randomised, multicentre, open-label, phase 3 study. Lancet 2019, 394, 2096–2107. [Google Scholar] [CrossRef]
  44. Chari, A.; Suvannasankha, A.; Fay, J.W.; Arnulf, B.; Kaufman, J.L.; Ifthikharuddin, J.J.; Weiss, B.M.; Krishnan, A.; Lentzsch, S.; Comenzo, R.; et al. Daratumumab plus pomalidomide and dexamethasone in relapsed and/or refractory multiple myeloma. Blood 2017, 130, 974–981. [Google Scholar] [CrossRef]
  45. Holstein, S.A.; McCarthy, P.L. Immunomodulatory Drugs in Multiple Myeloma: Mechanisms of Action and Clinical Experience. Drugs 2017, 77, 505–520. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Minařík, J.; Ševčíková, S. Immunomodulatory Agents for Multiple Myeloma. Cancers 2022, 14, 5759. https://doi.org/10.3390/cancers14235759

AMA Style

Minařík J, Ševčíková S. Immunomodulatory Agents for Multiple Myeloma. Cancers. 2022; 14(23):5759. https://doi.org/10.3390/cancers14235759

Chicago/Turabian Style

Minařík, Jiří, and Sabina Ševčíková. 2022. "Immunomodulatory Agents for Multiple Myeloma" Cancers 14, no. 23: 5759. https://doi.org/10.3390/cancers14235759

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop