1
|
Oguegbulu JC, Khalaf AI, Suckling CJ, Harnett MM, Harnett W. Lead optimisation efforts on a molecular prototype of the immunomodulatory parasitic protein ES-62. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2021-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The immunomodulatory property of some parasitic helminths is well documented. The glycoprotein ES-62 from the nematode, acanthocheilonema viteae has been found to possess immunomodulatory properties. Two small molecule analogues (SMA’s) of ES-62 (S3 and S5) were found to mimic its immunomodulatory properties in vivo and were active in animal models of allergic, inflammatory and autoimmune diseases. In this work, new efforts were made to further optimise the activities of compound S3 by making small but directed structural changes. A variety of analogues based on the S3 prototype were simulated by making variations at one position and then screened in silico. The best compounds were selected based on predicted physiochemical properties and medicinal chemistry indices and synthesised. Structural elucidation was done via HNMR, LCMS, FTIR and HRESIMS. The predicted properties were evaluated by HPLC method. A total of 11 novel molecules were synthesised and characterised. Significant correlation was obtained between the predicted physicochemical properties and their HPLC retention times (RT) for eight of our novel compounds. This suggests that these compounds may behave in a physiological environment as closely as computationally predicted. This entails, lesser host toxicity while maintaining good or better activities compared to the earlier prototype. They hence provide a good opportunity for development of drugs for immune conditions such as asthma, inflammation and autoimmune diseases.
Collapse
Affiliation(s)
- Joseph C. Oguegbulu
- Department of Chemical Sciences , Bingham University , PMB 005 , Karu , Nasarawa State , Nigeria
| | - Abedawn I. Khalaf
- Department of Pure and Applied Chemistry , University of Strathclyde , Glasgow G1 1XL , UK
| | - Colin J. Suckling
- Department of Pure and Applied Chemistry , University of Strathclyde , Glasgow G1 1XL , UK
| | - Margaret M. Harnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow , Glasgow G12 8TA , UK
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow G4 0RE , UK
| |
Collapse
|
2
|
Abbiati RA, Pourdehnad M, Carrancio S, Pierce DW, Kasibhatla S, McConnell M, Trotter MWB, Loos R, Santini CC, Ratushny AV. Quantitative Systems Pharmacology Modeling of Avadomide-Induced Neutropenia Enables Virtual Clinical Dose and Schedule Finding Studies. AAPS J 2021; 23:103. [PMID: 34453265 PMCID: PMC8397660 DOI: 10.1208/s12248-021-00623-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023] Open
Abstract
Avadomide is a cereblon E3 ligase modulator and a potent antitumor and immunomodulatory agent. Avadomide trials are challenged by neutropenia as a major adverse event and a dose-limiting toxicity. Intermittent dosing schedules supported by preclinical data provide a strategy to reduce frequency and severity of neutropenia; however, the identification of optimal dosing schedules remains a clinical challenge. Quantitative systems pharmacology (QSP) modeling offers opportunities for virtual screening of efficacy and toxicity levels produced by alternative dose and schedule regimens, thereby supporting decision-making in translational drug development. We formulated a QSP model to capture the mechanism of avadomide-induced neutropenia, which involves cereblon-mediated degradation of transcription factor Ikaros, resulting in a maturation block of the neutrophil lineage. The neutropenia model was integrated with avadomide-specific pharmacokinetic and pharmacodynamic models to capture dose-dependent effects. Additionally, we generated a disease-specific virtual patient population to represent the variability in patient characteristics and response to treatment observed for a diffuse large B-cell lymphoma trial cohort. Model utility was demonstrated by simulating the avadomide effect in the virtual population for various dosing schedules and determining the incidence of high-grade neutropenia, its duration, and the probability of recovery to low-grade neutropenia.
Collapse
Affiliation(s)
- Roberto A Abbiati
- Bristol Myers Squibb, Center for Innovation and Translational Research Europe (CITRE), Seville, Spain.
| | | | | | | | | | | | - Matthew W B Trotter
- Bristol Myers Squibb, Center for Innovation and Translational Research Europe (CITRE), Seville, Spain
| | - Remco Loos
- Bristol Myers Squibb, Center for Innovation and Translational Research Europe (CITRE), Seville, Spain
| | - Cristina C Santini
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | |
Collapse
|
3
|
Chernobrovkin AL, Cázares-Körner C, Friman T, Caballero IM, Amadio D, Martinez Molina D. A Tale of Two Tails: Efficient Profiling of Protein Degraders by Specific Functional and Target Engagement Readouts. SLAS DISCOVERY 2021; 26:534-546. [PMID: 33445986 DOI: 10.1177/2472555220984372] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Targeted protein degradation represents an area of great interest, potentially offering improvements with respect to dosing, side effects, drug resistance, and reaching "undruggable" proteins compared with traditional small-molecule therapeutics. A major challenge in the design and characterization of degraders acting as molecular glues is that binding of the molecule to the protein of interest (PoI) is not needed for efficient and selective protein degradation; instead, one needs to understand the interaction with the responsible ligase. Similarly, for proteasome targeting chimeras (PROTACs), understanding the binding characteristics of the PoI alone is not sufficient. Therefore, simultaneously assessing the binding to both PoI and the E3 ligase as well as the resulting degradation profile is of great value. The cellular thermal shift assay (CETSA) is an unbiased cell-based method, designed to investigate the interaction of compounds with their cellular protein targets by measuring compound-induced changes in protein thermal stability. In combination with mass spectrometry (MS), CETSA can simultaneously evaluate compound-induced changes in the stability of thousands of proteins. We have used CETSA MS to profile a number of protein degraders, including molecular glues (e.g., immunomodulatory drugs) and PROTACs, to understand mode of action and to deconvolute off-target effects in intact cells. Within the same experiment, we were able to monitor both target engagement by observing changes in protein thermal stability as well as efficacy by simultaneous assessment of protein abundances. This allowed us to correlate target engagement (i.e., binding to the PoI and ligases) and functional readout (i.e., degrader induced protein degradation).
Collapse
|
4
|
Piccolomo A, Schifone CP, Strafella V, Specchia G, Musto P, Albano F. Immunomodulatory Drugs in Acute Myeloid Leukemia Treatment. Cancers (Basel) 2020; 12:cancers12092528. [PMID: 32899586 PMCID: PMC7573974 DOI: 10.3390/cancers12092528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/31/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) are analogs of thalidomide. They have immunomodulatory, antiangiogenic and proapoptotic properties and exert a role in regulating the tumor microenvironment. Recently IMiDs have been investigated for their pleiotropic properties and their therapeutic applications in both solid tumors (melanoma, prostate carcinoma and differentiated thyroid cancer) and hematological malignancies. Nowadays, they are applied in de novo and relapsed/refractory multiple myeloma, in myelodysplastic syndrome, in del5q syndrome with specific use of lenalidomide and B-cell lymphoma. Several studies have been conducted in the last few years to explore IMiDs possible use in acute myeloid leukemia treatment. Here we report the mechanisms of action of IMiDs in acute myeloid leukemia and their potential future therapeutic application in this disease.
Collapse
Affiliation(s)
- Antonio Piccolomo
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology and Stem cell Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.P.); (C.P.S.); (V.S.); (P.M.)
| | - Claudia Pia Schifone
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology and Stem cell Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.P.); (C.P.S.); (V.S.); (P.M.)
| | - Vanda Strafella
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology and Stem cell Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.P.); (C.P.S.); (V.S.); (P.M.)
| | - Giorgina Specchia
- Former Full Professor of Hematology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Pellegrino Musto
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology and Stem cell Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.P.); (C.P.S.); (V.S.); (P.M.)
| | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology and Stem cell Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.P.); (C.P.S.); (V.S.); (P.M.)
- Correspondence: ; Tel.: +39-080-5478031
| |
Collapse
|
5
|
Differentiation of antiinflammatory and antitumorigenic properties of stabilized enantiomers of thalidomide analogs. Proc Natl Acad Sci U S A 2015; 112:E1471-9. [PMID: 25775521 DOI: 10.1073/pnas.1417832112] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Therapeutics developed and sold as racemates can exhibit a limited therapeutic index because of side effects resulting from the undesired enantiomer (distomer) and/or its metabolites, which at times, forces researchers to abandon valuable scaffolds. Therefore, most chiral drugs are developed as single enantiomers. Unfortunately, the development of some chirally pure drug molecules is hampered by rapid in vivo racemization. The class of compounds known as immunomodulatory drugs derived from thalidomide is developed and sold as racemates because of racemization at the chiral center of the 3-aminoglutarimide moiety. Herein, we show that replacement of the exchangeable hydrogen at the chiral center with deuterium allows the stabilization and testing of individual enantiomers for two thalidomide analogs, including CC-122, a compound currently in human clinical trials for hematological cancers and solid tumors. Using "deuterium-enabled chiral switching" (DECS), in vitro antiinflammatory differences of up to 20-fold are observed between the deuterium-stabilized enantiomers. In vivo, the exposure is dramatically increased for each enantiomer while they retain similar pharmacokinetics. Furthermore, the single deuterated enantiomers related to CC-122 exhibit profoundly different in vivo responses in an NCI-H929 myeloma xenograft model. The (-)-deuterated enantiomer is antitumorigenic, whereas the (+)-deuterated enantiomer has little to no effect on tumor growth. The ability to stabilize and differentiate enantiomers by DECS opens up a vast window of opportunity to characterize the class effects of thalidomide analogs and improve on the therapeutic promise of other racemic compounds, including the development of safer therapeutics and the discovery of new mechanisms and clinical applications for existing therapeutics.
Collapse
|
6
|
Moros A, Bustany S, Cahu J, Saborit-Villarroya I, Martínez A, Colomer D, Sola B, Roué G. Antitumoral activity of lenalidomide in in vitro and in vivo models of mantle cell lymphoma involves the destabilization of cyclin D1/p27KIP1 complexes. Clin Cancer Res 2013; 20:393-403. [PMID: 24178620 DOI: 10.1158/1078-0432.ccr-13-1569] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Clinical responses to the immmunomodulatory drug lenalidomide have been observed in patients with relapsed/refractory mantle cell lymphoma (MCL), although its mechanism of action remains partially unknown. We investigated whether the expression and subcellular localization of cyclin D1, a major cell-cycle regulator overexpressed in MCL, and the cyclin-dependent kinase inhibitor p27(KIP1), could identify MCL cases sensitive to lenalidomide, and whether the compound could modulate cyclin D1/p27(KIP1) complexes in MCL cells. EXPERIMENTAL DESIGN MCL primary samples and cell lines were analyzed for subcellular levels of cyclin D1/p27(KIP1) complexes by Western blot, immunohistochemistry, immunoprecipitation, and flow cytometry. Activity of lenalidomide in vitro and its effect on cyclin D1/p27(KIP1) complexes were evaluated by real-time PCR, immunoprecipitation, immunofluorescence, and Western blot. In vivo validation was carried out in a mouse xenograft model of human MCL. RESULTS We found cyclin D1 and p27(KIP1) to be coordinately expressed in all the MCL samples tested. Immunoprecipitation analyses and siRNA assays suggested a direct role of cyclin D1 in the regulation of p27(KIP1) levels. The nuclear accumulation of both proteins correlated with MCL cell tumorigenicity in vivo, and sensitivity to lenalidomide activity in vitro and in vivo. Lenalidomide mechanism of action relied on cyclin D1 downregulation and disruption of cyclin D1/p27(KIP1) complexes, followed by cytosolic accumulation of p27(KIP1), cell proliferation arrest, apoptosis, and angiogenesis inhibition. CONCLUSIONS These results highlight a mechanism of action of lenalidomide in MCL cases with increased tumorigenicity in vivo, which is mediated by the dissociation of cyclin D1/p27(KIP1) complexes, and subsequent proliferation blockade and apoptosis induction.
Collapse
Affiliation(s)
- Alexandra Moros
- Authors' Affiliations: Hemato-oncology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Hematopathology Unit, Hospital Clínic, Barcelona, Spain; and Normandie Univ, UNICAEN, MILPAT, Caen, France
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Bagratuni T, Kastritis E, Politou M, Roussou M, Kostouros E, Gavriatopoulou M, Eleutherakis-Papaiakovou E, Kanelias N, Terpos E, Dimopoulos MA. Clinical and genetic factors associated with venous thromboembolism in myeloma patients treated with lenalidomide-based regimens. Am J Hematol 2013; 88:765-70. [PMID: 23757261 DOI: 10.1002/ajh.23504] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 04/29/2013] [Accepted: 05/28/2013] [Indexed: 12/21/2022]
Abstract
Lenalidomide has significant antimyeloma activity but it is associated with a significant risk of venous thromboembolism (VTE). In this study, we assessed clinical and genetic risk factors that may predispose for VTE in myeloma patients who were treated with lenalidomide-based regimens. We analyzed common clinical and selected genetic factors in 200 consecutive, unselected myeloma patients who were treated with lenalidomide-based regimens in a single institution. Twelve patients (6%) developed a VTE (nine deep venous thrombosis and three pulmonary embolism). All VTEs occurred in patients who were receiving aspirin prophylaxis; no patient who received LMWH or acenocoumarol had a VTE. The frequency of VTEs was 9.4% in previously untreated and 4.5% in previously treated patients. VTEs were more frequent in patients >65 years (8.1% vs. 1.6%) especially among patients receiving aspirin as prophylaxis (10.4% vs. 1.8% for patients ≤65 years). In patients who received prophylaxis with low dose aspirin a single-nucleotide polymorphism in NFκB1 (rs3774968) gene was associated with increased risk of VTE (OR 3.76, 95%CI 1-16, P = 0.051). None of the patients who developed VTEs had common genetic variations that are associated with increased risk of VTEs in the general population, such as FVLeiden and FIIG20210A. Our data indicated that LMWH or vitamin K antagonists (with a target INR 2-3) effectively reduce the risk of VTEs. In patients who received prophylaxis with aspirin genetic variants of genes that are involved directly or indirectly in inflammatory response may be associated with increased risk of VTE.
Collapse
Affiliation(s)
- Tina Bagratuni
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| | - Marianna Politou
- Aretaieion Hospital; University of Athens, School of Medicine; Athens; Greece
| | - Maria Roussou
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| | - Efthimios Kostouros
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| | | | - Nikolaos Kanelias
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics; University of Athens; School of Medicine; Athens; Greece
| |
Collapse
|
8
|
Merli M, Ferrario A, Basilico C, Maffioli M, Caramazza D, Appio L, Arcaini L, Passamonti F. Novel agents in indolent lymphomas. Ther Adv Hematol 2013; 4:133-48. [PMID: 23610620 PMCID: PMC3629754 DOI: 10.1177/2040620712466865] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Indolent non-Hodgkin's lymphomas (iNHLs) include follicular lymphomas (FL), marginal-zone lymphoma, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia and small lymphocytic lymphoma. First-line standard therapy in advanced, symptomatic iNHL consists of rituximab-based immunochemotherapy. The recent rediscovery of the 'old' chemotherapeutic agent bendamustine, an alkylating agent with a peculiar mechanism of action, has added a new effective and well-tolerated option to the therapeutic armamentarium in iNHL, increasing response rates and duration. However, patients invariably relapse and subsequent active and well-tolerated agents are needed. In recent years a large number of new targeted agents have been tested in preclinical and clinical experimentation in FL and indolent nonfollicular lymphoma (iNFL), including the new monoclonal antibodies binding CD20 or other surface antigens, immunoconjugates and bispecific antibodies. Moreover novel agents directed against intracellular processes such as proteasome inhibitors, mTOR inhibitors and agents that target the tumour microenvironment, notably the immunomodulatory agent lenalidomide, are under active clinical investigation. The development of these new drugs may change in the near future the approach to iNHL patients, leading to better tolerated and effective therapy regimens.
Collapse
Affiliation(s)
- Michele Merli
- Division of Hematology, Department of Internal Medicine, Ospedale di Circolo and Fondazione Macchi, University of Insubria, Varese, Italy
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Voloshin T, Voest EE, Shaked Y. The host immunological response to cancer therapy: An emerging concept in tumor biology. Exp Cell Res 2013; 319:1687-95. [PMID: 23518388 DOI: 10.1016/j.yexcr.2013.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 02/23/2013] [Accepted: 03/02/2013] [Indexed: 02/04/2023]
Abstract
Almost any type of anti-cancer treatment including chemotherapy, radiation, surgery and targeted drugs can induce host molecular and cellular immunological effects which, in turn, can lead to tumor outgrowth and relapse despite an initial successful therapy outcome. Tumor relapse due to host immunological effects is attributed to angiogenesis, tumor cell dissemination from the primary tumors and seeding at metastatic sites. This short review will describe the types of host cells that participate in this process, the types of factors secreted from the host following therapy that can promote tumor re-growth, and the possible implications of this unique and yet only partially-known process. It is postulated that blocking these specific immunological effects in the reactive host in response to cancer therapy may aid in identifying new host-dependent targets for cancer, which in combination with conventional treatments can prolong therapy efficacy and extend survival. Additional studies investigating this specific research direction-both in preclinical models and in the clinical setting are essential in order to advance our understanding of how tumors relapse and evade therapy.
Collapse
Affiliation(s)
- Tali Voloshin
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine and the Rappaport Institute, Technion-Israel Institute of Technology, 1 Efron Street, Bat Galim, Haifa 31096, Israel
| | - Emile E Voest
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yuval Shaked
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine and the Rappaport Institute, Technion-Israel Institute of Technology, 1 Efron Street, Bat Galim, Haifa 31096, Israel.
| |
Collapse
|
10
|
Fabbri A, Cencini E, Pietrini A, Gozzetti A, Defina M, Fontanelli G, Mazzei MA, Volterrani L, Bocchia M. Impressive activity of lenalidomide monotherapy in refractory angioimmunoblastic T-cell lymphoma: report of a case with long-term follow-up. Hematol Oncol 2012; 31:213-7. [DOI: 10.1002/hon.2038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 10/08/2012] [Accepted: 10/11/2012] [Indexed: 12/23/2022]
Affiliation(s)
- Alberto Fabbri
- Unit of Hematology; “S. Maria alle Scotte” University Hospital; Siena Italy
| | - Emanuele Cencini
- Unit of Hematology; “S. Maria alle Scotte” University Hospital; Siena Italy
| | - Alice Pietrini
- Unit of Hematology; “S. Maria alle Scotte” University Hospital; Siena Italy
| | | | - Marzia Defina
- Unit of Hematology; “S. Maria alle Scotte” University Hospital; Siena Italy
| | | | | | - Luca Volterrani
- Unit of Radiology; “S. Maria alle Scotte” University Hospital; Siena Italy
| | - Monica Bocchia
- Unit of Hematology; “S. Maria alle Scotte” University Hospital; Siena Italy
| |
Collapse
|
11
|
Podar K. Ask the Experts: Deriving new treatment strategies in multiple myeloma. Int J Hematol Oncol 2012. [DOI: 10.2217/ijh.12.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Klaus Podar, MD PhD, is an Associate Professor of Experimental Oncology and Hematology and a hematologist/oncologist at the National Center for Tumor Diseases, University of Heidelberg, and the German Cancer Research Center (DKFZ) in Heidelberg, Germany. He has published more than 100 peer-reviewed manuscripts and keynote reviews, is a member of several scientific societies including the American Association of Cancer Research (AACR), American Society of Hematology (ASH) and European Hematology Association (EHA), and is the recipient of several prestigious awards.
Collapse
Affiliation(s)
- Klaus Podar
- National Center of Tumor Diseases (NCT), University of Heidelberg & German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| |
Collapse
|
12
|
van de Donk NW, Görgün G, Groen RW, Jakubikova J, Mitsiades CS, Hideshima T, Laubach J, Nijhof IS, Raymakers RA, Lokhorst HM, Richardson PG, Anderson KC. Lenalidomide for the treatment of relapsed and refractory multiple myeloma. Cancer Manag Res 2012; 4:253-68. [PMID: 22956884 PMCID: PMC3430086 DOI: 10.2147/cmar.s27087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Lenalidomide is an amino-substituted derivative of thalidomide with direct antiproliferative and cytotoxic effects on the myeloma tumor cell, as well as antiangiogenic activity and immunomodulatory effects. Together with the introduction of bortezomib and thalidomide, lenalidomide has significantly improved the survival of patients with relapsed and refractory myeloma. The most common adverse events associated with lenalidomide include fatigue, skin rash, thrombocytopenia, and neutropenia. In addition, when lenalidomide is combined with dexamethasone or other conventional cytotoxic agents, there is an increase in the incidence of venous thromboembolic events. There is now evidence that continued treatment with lenalidomide has a significant impact on survival by improving the depth and duration of response. This highlights the value of adverse event management and appropriate dose adjustments to prevent toxicity, and of allowing continued treatment until disease progression. In this review, we will discuss the different lenalidomide-based treatment regimens for patients with relapsed/refractory myeloma. This is accompanied by recommendations of how to manage and prevent adverse events associated with lenalidomide-based therapy.
Collapse
Affiliation(s)
- Niels Wcj van de Donk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Schafer PH. Update on immunomodulatory drugs (IMiDs) in hematologic and solid malignancies. Expert Opin Pharmacother 2012; 13:1541-2. [DOI: 10.1517/14656566.2012.692520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|