Overcoming the Immunosuppressive Tumor Microenvironment in Multiple Myeloma

Total Page:16

File Type:pdf, Size:1020Kb

Overcoming the Immunosuppressive Tumor Microenvironment in Multiple Myeloma cancers Review Overcoming the Immunosuppressive Tumor Microenvironment in Multiple Myeloma Fatih M. Uckun 1,2,3 1 Norris Comprehensive Cancer Center and Childrens Center for Cancer and Blood Diseases, University of Southern California Keck School of Medicine (USC KSOM), Los Angeles, CA 90027, USA; [email protected] 2 Department of Developmental Therapeutics, Immunology, and Integrative Medicine, Drug Discovery Institute, Ares Pharmaceuticals, St. Paul, MN 55110, USA 3 Reven Pharmaceuticals, Translational Oncology Program, Golden, CO 80401, USA Simple Summary: This article provides a comprehensive review of new and emerging treatment strategies against multiple myeloma that employ precision medicines and/or drugs capable of improving the ability of the immune system to prevent or slow down the progression of multiple myeloma. These rationally designed new treatment methods have the potential to change the therapeutic landscape in multiple myeloma and improve the long-term survival outcome. Abstract: SeverFigurel cellular elements of the bone marrow (BM) microenvironment in multiple myeloma (MM) patients contribute to the immune evasion, proliferation, and drug resistance of MM cells, including myeloid-derived suppressor cells (MDSCs), tumor-associated M2-like, “alter- natively activated” macrophages, CD38+ regulatory B-cells (Bregs), and regulatory T-cells (Tregs). These immunosuppressive elements in bidirectional and multi-directional crosstalk with each other inhibit both memory and cytotoxic effector T-cell populations as well as natural killer (NK) cells. Immunomodulatory imide drugs (IMiDs), protease inhibitors (PI), monoclonal antibodies (MoAb), Citation: Uckun, F.M. Overcoming the Immunosuppressive Tumor adoptive T-cell/NK cell therapy, and inhibitors of anti-apoptotic signaling pathways have emerged as Microenvironment in Multiple promising therapeutic platforms that can be employed in various combinations as part of a rationally Myeloma. Cancers 2021, 13, 2018. designed immunomodulatory strategy against an immunosuppressive tumor microenvironment https://doi.org/10.3390/ (TME) in MM. These platforms provide the foundation for a new therapeutic paradigm for achieving cancers13092018 improved survival of high-risk newly diagnosed as well as relapsed/refractory MM patients. Here we review the scientific rationale and clinical proof of concept for each of these platforms. Academic Editor: David Wong Keywords: tumor microenvironment (TME); multiple myeloma (MM); immunomodulatory imide Received: 27 February 2021 drugs (IMiDs); autologous hematopoietic stem cell transplantation (ASCT); immune-checkpoint Accepted: 20 April 2021 receptors; spleen tyrosine kinase (SYK); transforming growth factor beta (TGF-β); bispecific T-cell Published: 22 April 2021 engagers (BiTEs); chimeric antigen receptor (CAR)-T Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affil- 1. Introduction iations. The tumor microenvironment (TME) is one of the main contributors of a marked im- munobiological and clinical heterogeneity as well as clonal evolution in multiple myeloma (MM) [1–3]. Several cellular elements of the bone marrow (BM) microenvironment in MM patients contribute to the immune evasion, proliferation, and drug resistance of MM Copyright: © 2021 by the author. Licensee MDPI, Basel, Switzerland. cells [4–6], including myeloid-derived suppressor cells (MDSCs), tumor-associated M2-like, + This article is an open access article “alternatively activated”, macrophages, CD38 regulatory B-cells (Bregs), and regulatory distributed under the terms and T-cells (Tregs) [4–6] (Figure1A). These immunosuppressive elements in bidirectional and conditions of the Creative Commons multi-directional crosstalk with each other inhibit both memory and cytotoxic effector Attribution (CC BY) license (https:// T-cell populations as well as natural killer (NK) cells [4–6]. MDSCs along with MM cell- creativecommons.org/licenses/by/ derived interleukin 10 (IL-10) [7–10], TGF-β, and IL-6 are also known to impair dendritic 4.0/). cell (DC) maturation and their antigen-presenting function, which further accentuates Cancers 2021, 13, 2018. https://doi.org/10.3390/cancers13092018 https://www.mdpi.com/journal/cancers Cancers 2021, 13, x 2 of 15 Cancers 2021, 13, 2018 2 of 14 with MM cell‐derived interleukin 10 (IL‐10) [7–10], TGF‐, and IL‐6 are also known to impair dendritic cell (DC) maturation and their antigen‐presenting function, which fur‐ thether immunosuppression accentuates the immunosuppression [6]. In addition to [6]. their In addition immune-suppressive to their immune activity‐suppressive mediated ac‐ bytivity their mediated secretion by of their IL-10 secretion (an activator of IL‐10 of Tregs(an activator and M2 of macrophages), Tregs and M2 and macrophages), TGF-β (an inhibitorand TGF‐ of (an both inhibitor cytotoxic of T-cellsboth cytotoxic and NK T cells),‐cells and M2 macrophagesNK cells), M2 alsomacrophages promote MMalso pro cell‐ proliferation,mote MM cell angiogenesis, proliferation, and angiogenesis, chemotherapy and resistance chemotherapy [11–13 ]resistance (Figure1A). [11–13] Nonclinical (Figure studies1A). Nonclinical in animal studies models in of animal MM have models demonstrated of MM have that demonstrated exosomes serve that asexosomes regulators serve of theas regulators signaling networksof the signaling within networks the BM microenvironment, within the BM microenvironment, activate anti-apoptotic activate mech- anti‐ anismsapoptotic promoted mechanisms by oncogenic promoted proteins by oncogenic such asproteins the signal such transducer as the signal and transducer activator and of transcriptionactivator of transcription 3 (STAT3), and 3 (STAT3), cause immunosuppression and cause immunosuppression by facilitating theby growthfacilitating of MD- the SCsgrowth [14– of19 ].MDSCs Exosome-activated [14–19]. Exosome MDSCs‐activated have beenMDSCs implicated have been in implicated development in develop of other‐ immunosuppressivement of other immunosuppressive cells, such as Tregs, cells, tumor-promoting such as Tregs, tumor angiogenesis,‐promoting proliferation angiogenesis, of MMproliferation cells, and of increased MM cells, osteoclast and increased activity osteoclast contributing activity to thecontributing lytic bone to lesions the lytic [14 –bone19]. Complementinglesions [14–19]. theComplementing immunosuppressive the immunosuppressive TME, T-cell exhaustion TME, combined T‐cell exhaustion with high-level com‐ expressionbined with ofhigh immune-checkpoint‐level expression of ligands immune on‐checkpoint MM cells ligands are the mainon MM contributors cells are the to main the immunecontributors evasion to the of immune MM cells evasion [20–23]. of MM cells [20–23]. Figure 1. ImmunosuppressiveFigure TME 1. inImmunosuppressive MM. (A) MM cells TMEsecrete in several MM. (A cytokines) MM cells including secrete several IL‐6, TGF cytokines‐ (TGF), including and IL‐ IL-6, 10 [7–10] that inhibit DCs, CTLs,TGF-β but(TGF), stimulate and IL-10 Tregs. [7– 10MDSCs] that inhibit are stimulated DCs, CTLs, by butM2 stimulatemacrophages Tregs. via MDSCs IL‐6 and are stimulate stimulated by M2 macrophages as well asM2 Tregs macrophages via IL‐10. MDSC via IL-6 inhibit and stimulate CTLs and M2 NK macrophages cells via IL‐ as10. well See astext Tregs for detailed via IL-10. discussion. MDSC inhibit CTLs and NK cells via IL-10. See text for detailed discussion. (B) Lactate shuttle in TME. Lactate derived from MM cells stimulates Tregs and M2 macrophages, but it inhibits TILs, CTL, NK cells, and γδT cells. See text for detailed discussion. Cancers 2021, 13, 2018 3 of 14 2. Immunomodulatory Imide Drugs (IMiDs) to Overcome the Immunosuppressive TME in MM IMiDs augment the activity of cereblon E3 ubiquitin ligase complex and enhance the effector functions as well as the proliferative expansion of cytotoxic T-cells and NK cells through cereblon-dependent proteolytic degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3), increased production of IL-2 and IFN-γ with concomitant reduction in IL-10 production and suppression of Tregs [24–27]. They also block the pro- inflammatory cytokines TNF-α and IL-6, thereby decreasing the contributions of MDSC and M2 macrophages to the immunosuppressive milieu of the TME. Notably, in a IMiD- conditioned TME, T-cell activation is facilitated, as it no longer requires co-stimulation by antigen-presenting DCs [24–27]. In addition, IMiDs trigger caspase-mediated apoptosis in MM cells and impair their interaction with the protective BM stroma [28]. Efforts are under- way to further enhance the clinical potency of IMiDs by combining them with modulators of cereblon E3-ligase, such as iberdomide, that accelerate IMiD-mediated degradation of IKZF1/IKZF3 [26,27]. Due to the considerable added benefit of the IMiD thalidomide, a combination of bortezomib, thalidomide, and dexamethasone (VTD) has become a stan- dard induction regimen [29]. New generation IMiDs, including lenalidomide (Revlimid; Celgene) and pomalidomide (Pomalyst; Celgene) have potent immune modulatory effects and have significantly improved the survival outcomes of newly diagnosed as well as R/R MM patients [30]. IMiDs are used in combination with Proteasome inhibitors (PIs), such as bortezomib, carfilzomib, or ixazomib, in standard of care for MM to achieve optimal anti-tumor activity as well as inhibit angiogenesis within the TME [30]. Recent studies in- dicated that PIs also
Recommended publications
  • Us 8530498 B1 3
    USOO853 0498B1 (12) UnitedO States Patent (10) Patent No.: US 8,530,498 B1 Zeldis (45) Date of Patent: *Sep. 10, 2013 (54) METHODS FORTREATING MULTIPLE 5,639,476 A 6/1997 OShlack et al. MYELOMAWITH 5,674,533 A 10, 1997 Santus et al. 3-(4-AMINO-1-OXO-1,3-DIHYDROISOINDOL- 395 A 22 N. 2-YL)PIPERIDINE-2,6-DIONE 5,731,325 A 3/1998 Andrulis, Jr. et al. 5,733,566 A 3, 1998 Lewis (71) Applicant: Celgene Corporation, Summit, NJ (US) 5,798.368 A 8, 1998 Muller et al. 5,874.448 A 2f1999 Muller et al. (72) Inventor: Jerome B. Zeldis, Princeton, NJ (US) 5,877,200 A 3, 1999 Muller 5,929,117 A 7/1999 Muller et al. 5,955,476 A 9, 1999 Muller et al. (73) Assignee: Celgene Corporation, Summit, NJ (US) 6,020,358 A 2/2000 Muller et al. - 6,071,948 A 6/2000 D'Amato (*) Notice: Subject to any disclaimer, the term of this 6,114,355 A 9, 2000 D'Amato patent is extended or adjusted under 35 SS f 1939. All et al. U.S.C. 154(b) by 0 days. 6,235,756 B1 5/2001 D'Amatoreen et al. This patent is Subject to a terminal dis- 6,281.230 B1 8/2001 Muller et al. claimer 6,316,471 B1 1 1/2001 Muller et al. 6,326,388 B1 12/2001 Man et al. 6,335,349 B1 1/2002 Muller et al. (21) Appl. No.: 13/858,708 6,380.239 B1 4/2002 Muller et al.
    [Show full text]
  • Revlimid U.S. Full Prescribing Information
    HIGHLIGHTS OF PRESCRIBING INFORMATION • FL or MZL: 20 mg once daily orally on Days 1-21 of repeated 28-day cycles for up to These highlights do not include all the information needed to use REVLIMID® safely 12 cycles (2.4). and effectively. See full prescribing information for REVLIMID. • Renal impairment: Adjust starting dose based on the creatinine clearance value (2.6). • For concomitant therapy doses, see Full Prescribing Information (2.1, 2.4, 14.1, 14.4). REVLIMID (lenalidomide) capsules, for oral use Initial U.S. Approval: 2005 ------------------------- DOSAGE FORMS AND STRENGTHS ------------------------- Capsules: 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg, and 25 mg (3). WARNING: EMBRYO-FETAL TOXICITY, HEMATOLOGIC TOXICITY, -------------------------------- CONTRAINDICATIONS -------------------------------- and VENOUS and ARTERIAL THROMBOEMBOLISM • Pregnancy (Boxed Warning, 4.1, 5.1, 8.1). See full prescribing information for complete boxed warning. • Demonstrated severe hypersensitivity to lenalidomide (4.2, 5.9, 5.15). EMBRYO-FETAL TOXICITY --------------------------- WARNINGS AND PRECAUTIONS --------------------------- • Lenalidomide, a thalidomide analogue, caused limb abnormalities in a developmental monkey study similar to birth defects caused by thalidomide • Increased Mortality: serious and fatal cardiac adverse reactions occurred in patients in humans. If lenalidomide is used during pregnancy, it may cause birth with CLL treated with REVLIMID (lenalidomide) (5.5). defects or embryo-fetal death. • Second Primary Malignancies (SPM): Higher incidences of SPM were observed in • Pregnancy must be excluded before start of treatment. Prevent pregnancy controlled trials of patients with MM receiving REVLIMID (5.6). during treatment by the use of two reliable methods of contraception (5.1). • Increased Mortality: Observed in patients with MM when pembrolizumab was added REVLIMID is available only through a restricted distribution program, called the to dexamethasone and a thalidomide analogue (5.7).
    [Show full text]
  • Cereblon and Its Downstream Substrates As Molecular Targets of Immunomodulatory Drugs
    Int J Hematol (2016) 104:293–299 DOI 10.1007/s12185-016-2073-4 PROGRESS IN HEMATOLOGY Mechanisms of action of novel drugs in multiple myeloma and those responsible for the acquired resistance Cereblon and its downstream substrates as molecular targets of immunomodulatory drugs Takumi Ito1,2 · Hiroshi Handa1 Received: 15 June 2016 / Revised: 19 July 2016 / Accepted: 19 July 2016 / Published online: 26 July 2016 © The Japanese Society of Hematology 2016 Abstract Thalidomide was first developed as a sedative History of immunomodulatory drugs (IMiDs) around 60 years ago, but exhibited teratogenicity, leading to serious defects such as limb deformities. Nevertheless, Immunomodulatory drugs (IMiDs) are a new class of anti- thalidomide is now recognized as a therapeutic drug for the cancer drugs for which the parent molecule is thalidomide. treatment of Hansen’s disease and myeloma. Immunomod- Thalidomide (Fig. 1) was developed as a sedative in 1950s ulatory drugs (IMiDs), a new class of anti-cancer drug by the German pharmaceutical company Grunenthal. derived from thalidomide, have also been developed and Experiments using rodents initially suggested it to be safe exert potent anti-cancer effects. Although the molecular for use in humans, and the drug was sold over 40 countries, mechanism of thalidomide and IMiDs remained unclear for including Japan. However, as is widely known, thalidomide a long time, cereblon, a substrate receptor of the CRL4 E3 was found to have serious teratogenic effects. Use during ubiquitin ligase was identified as a primary direct target by pregnancy is associated with developmental defects of the a new affinity technique. A growing body of evidence sug- limbs and ears.
    [Show full text]
  • Characterization of Ocular Adverse Events in Patients Receiving
    Characterization of Ocular Adverse Events in Patients Receiving Belantamab Mafadotin for ≥12 Months: Post-Hoc Analysis of DREAMM-2 Study in Relapsed/Refractory Multiple Myeloma S. LONIAL1; A.K. NOOKA1; P. THULASI2; A.Z. BADROS3; B.H. JENG3; N.S. CALLANDER4; D. SBOROV5; B.E. ZAUGG6; R. POPAT7; S. DEGLI ESPOSTI8; J. BARON9; A. DOHERTY9; E. LEWIS10; J. OPALINSKA9; P. PAKA9; T. PIONTEK9; I. GUPTA9; A.V. FAROOQ11; A. JAKUBOWIAK11 | 1Emory University, Winship Cancer Institute, Atlanta, GA, USA; 2Emory Eye Center, Emory University, Atlanta, GA, USA; 3University of Maryland School of Medicine, Baltimore, MD, USA; 4University of Wisconsin, Carbone Cancer Center, Madison, WI, USA; 5Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; 6Moran Eye Center, University of Utah, Salt Lake City, UT, USA; 7University College London Hospitals, NHS Foundation Trust, London, UK; 8NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK; 9GlaxoSmithKline, Collegeville, PA, USA; 10GlaxoSmithKline, Research Triangle Park, NC, USA; 11University of Chicago Medical Center, Chicago, IL, USA Dose delays and reductions related to ocular adverse events INTRODUCTION RESULTS All 14 patients required ≥2 dose delays, with dose reduction (to 1.92 mg/kg) in CONCLUSIONS Belantamab mafodotin (belamaf; GSK2857916) is a first-in-class, monomethyl auristatin F (MMAF)-containing 12 patients (86%). Demographics, efficacy, and overall safety information for patients treated with belamaf In this subset of 14 patients from the DREAMM-2 study, the median duration of response was antibody–drug conjugate (ADC) that binds to B-cell maturation antigen (BCMA) and eliminates multiple myeloma cells by for ≥12 months ● Patients experienced a mean of 3.6 dose delays over the ≥12 months of treatment a multimodal mechanism of action.1 (median: 3.5, range: 2–6).
    [Show full text]
  • Computational Approaches for Drug Design and Discovery: an Overview
    Review Article Computational Approaches for Drug Design and Discovery: An Overview Baldi A College of Pharmacy, Dr. Shri R.M.S. Institute of Science & Technology, Bhanpura, Dist. Mandsaur (M.P.), India ARTICLE INFO ABSTRACT Article history: The process of drug discovery is very complex and requires an interdisciplinary effort to design Received 12 July 2009 effective and commercially feasible drugs. The objective of drug design is to find a chemical Accepted 21 July 2009 compound that can fit to a specific cavity on a protein target both geometrically and chemically. Available online 04 February 2010 After passing the animal tests and human clinical trials, this compound becomes a drug available Keywords: to patients. The conventional drug design methods include random screening of chemicals Computer-aided drug design found in nature or synthesized in laboratories. The problems with this method are long design Combinatorial chemistry cycle and high cost. Modern approach including structure-based drug design with the help Drug of informatic technologies and computational methods has speeded up the drug discovery Structure-based drug deign process in an efficient manner. Remarkable progress has been made during the past five years in almost all the areas concerned with drug design and discovery. An improved generation of softwares with easy operation and superior computational tools to generate chemically stable and worthy compounds with refinement capability has been developed. These tools can tap into cheminformation to shorten the cycle of drug discovery, and thus make drug discovery more cost-effective. A complete overview of drug discovery process with comparison of conventional approaches of drug discovery is discussed here.
    [Show full text]
  • Opportunities and Challenges in Phenotypic Drug Discovery: an Industry Perspective
    PERSPECTIVES Nevertheless, there are still challenges in OPINION prospectively understanding the key success factors for modern PDD and how maximal Opportunities and challenges in value can be obtained. Articles published after the analysis by Swinney and Anthony have re-examined the contribution of PDD phenotypic drug discovery: an to new drug discovery6,7 and have refined the conditions for its successful application8. industry perspective Importantly, it is apparent on closer examination that the classification of drugs John G. Moffat, Fabien Vincent, Jonathan A. Lee, Jörg Eder and as ‘phenotypically discovered’ is somewhat Marco Prunotto inconsistent6,7 and that, in fact, the majority of successful drug discovery programmes Abstract | Phenotypic drug discovery (PDD) approaches do not rely on knowledge combine target knowledge and functional of the identity of a specific drug target or a hypothesis about its role in disease, in cellular assays to identify drug candidates contrast to the target-based strategies that have been widely used in the with the most advantageous molecular pharmaceutical industry in the past three decades. However, in recent years, there mechanism of action (MoA). Although there is clear evidence that phenotypic has been a resurgence in interest in PDD approaches based on their potential to screening can be an attractive proposition address the incompletely understood complexity of diseases and their promise for efficiently identifying functionally of delivering first-in-class drugs, as well as major advances in the tools for active hits that lead to first-in-class drugs, cell-based phenotypic screening. Nevertheless, PDD approaches also have the gap between a screening hit and an considerable challenges, such as hit validation and target deconvolution.
    [Show full text]
  • Medicinal Chemistry for Drug Discovery | Charles River
    Summary Medicinal chemistry is an integral part of bringing a drug through development. Our medicinal chemistry approach enables clients to benefit from efficient navigation of the early drug discovery process through to delivery of preclinical candidates. DISCOVERY Click to learn more Medicinal Chemistry for Drug Discovery Medicinal Chemistry A Proven Track Record in Drug Discovery Services: Our medicinal chemistry team has experience in progressing small molecule drug discovery programs across a broad range • Target identification of therapeutic areas and gene families. Our scientists are skilled in the design and synthesis of novel pharmacologically active - Capture Compound® mass compounds and understand the challenges facing modern drug discovery. Together, they are cited as inventors on over spectrometry (CCMS) 350 patents and have identified 80 preclinical candidates for client organizations across a variety of therapeutic areas. As • Hit-finding strategies project leaders, our chemists are fundamental in driving the program strategy and have consistently empowered our clients’ - Optimizing high-throughput success. A high proportion of candidates regularly progress to the clinic, and our first co-invented drug, Belinostat, received screening (HTS) hits marketing approval in 2015. As an organization, Charles River has worked on 85% of the therapies approved in 2018. • Hit-to-lead We have a deep understanding of the factors that drive medicinal chemistry design: structure-activity relationship (SAR), • Lead optimization biology, physical chemistry, drug metabolism and pharmacokinetics (DMPK), pharmacokinetic/pharmacodynamic (PK/PD) • Patent strategy modelling, and in vivo efficacy. Charles River scientists are skilled in structure-based and ligand-based design approaches • Preparation for IND filing utilizing our in-house computer-aided drug design (CADD) expertise.
    [Show full text]
  • Revlimid-INN Lenalidomide
    ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS 1 This medicinal product is subject to additional monitoring. This will allow quick identification of new safety information. Healthcare professionals are asked to report any suspected adverse reactions. See section 4.8 for how to report adverse reactions. 1. NAME OF THE MEDICINAL PRODUCT Revlimid 2.5 mg hard capsules Revlimid 5 mg hard capsules Revlimid 7.5 mg hard capsules Revlimid 10 mg hard capsules Revlimid 15 mg hard capsules Revlimid 20 mg hard capsules Revlimid 25 mg hard capsules 2. QUALITATIVE AND QUANTITATIVE COMPOSITION Revlimid 2.5 mg hard capsules Each capsule contains 2.5 mg of lenalidomide. Excipient(s) with known effect Each capsule contains 73.5 mg of lactose (as anhydrous lactose). Revlimid 5 mg hard capsules Each capsule contains 5 mg of lenalidomide. Excipient(s) with known effect Each capsule contains 147 mg of lactose (as anhydrous lactose). Revlimid 7.5 mg hard capsules Each capsule contains 7.5 mg of lenalidomide. Excipient(s) with known effect Each capsule contains 144.5 mg of lactose (as anhydrous lactose). Revlimid 10 mg hard capsules Each capsule contains 10 mg of lenalidomide. Excipient(s) with known effect Each capsule contains 294 mg of lactose (as anhydrous lactose). Revlimid 15 mg hard capsules Each capsule contains 15 mg of lenalidomide. Excipient(s) with known effect Each capsule contains 289 mg of lactose (as anhydrous lactose). Revlimid 20 mg hard capsules Each capsule contains 20 mg of lenalidomide. Excipient(s) with known effect Each capsule contains 244.5 mg of lactose (as anhydrous lactose).
    [Show full text]
  • In Relapsed Or Refractory Diffuse
    Dührsen et al. Blood Cancer Journal (2021) 11:95 https://doi.org/10.1038/s41408-021-00485-5 Blood Cancer Journal CORRESPONDENCE Open Access Phase I/II trial of lenalidomide, methotrexate, leucovorin, cytarabine, and rituximab (LeMLAR) in relapsed or refractory diffuse large B cell lymphoma Ulrich Dührsen 1, Mareike Tometten2,FrankKroschinsky3, Arnold Ganser4,StefanIbach5, Stefanie Bertram6 and Andreas Hüttmann 1 Dear Editor, ≥2.5 mg/dl, serum aspartate [AST] or alanin amino- Lenalidomide has moderate activity in relapsed or transferase [ALT] >4× upper limit of normal [ULN]), refractory diffuse large B cell lymphoma (r/rDLBCL)1. active hepatitis B or C, human immunodeficiency virus Based on the COMLA regimen used in the 1970s and infection, any other uncontrolled infection, as well as 1980s2, we combined lenalidomide with methotrexate pregnancy and nursing period. All patients gave written (plus leucovorin), cytarabine, and rituximab (LeMLAR) in informed consent. The LeMLAR regimen (Fig. 1) com- a phase I/II trial. Lenalidomide induces apoptosis and cell prised up to six 28-day cycles consisting of lenalidomide cycle arrest in the G0/G1 phase3. After discontinuation, (day 1–21), methotrexate (5–10 min i.v. bolus; day 1, 8, surviving cells start to proliferate and become susceptible 15), leucovorin (four oral 45 mg doses six hours apart, to the S-phase-specific agents methotrexate and cytar- starting 24 h after methotrexate), cytarabine (5-10 min i.v. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; abine administered in the subsequent cycle. Because of bolus; day 1, 8, 15), and rituximab (375 mg/m², day 1). All – low toxicity for immune effector cells4 6, these drugs are patients received prophylactic enoxaparin (40 mg s.c.).
    [Show full text]
  • Drug Discovery - Yesterday and Tomorrow: the Common Approaches in Drug Design and Cancer
    Cell & Cellular Life Sciences Journal Drug Discovery - Yesterday and Tomorrow: The Common Approaches in Drug Design and Cancer 1,2 1 3 Hamad ON *, Amran SIB and Sabbah AM Mini Review 1Faculty of Bioscience & Medical Engineering, Malaysia Volume 3 Issue 1 2University of Wasit, College of Medicine, Iraq Received Date: February 27, 2018 3Forensic DNA for research and training Centre, Al Nahrain University, Iraq Published Date: April 03, 2018 *Corresponding author: Oras Naji Hamad, Faculty of Bioscience & Medical Engineering, University of Technology, Malaysia, Tel: 01121715960; E-mail: [email protected] Abstract The process of drug discovery has undergone radical changes and development over years. Traditionally, the drugs were discovered by employing chemistry and pharmacology-based cautious approach. When natural products were the most important source of drugs or drug precursors, but the conventional randomized drug research phenomenon was no longer effective at that time due to many negatives of these approaches like: high expenses of discovering new drugs, time-consuming and reduced success guarantee. Thus, with the development of the era, the concept of “Rational Drug Design” has enabled drug target identification and validation to be more specific. In addition, several novel technologies and approaches have been introducing economics, proteomics and other omics areas such as 3D QSAR, pharmacophore modeling and other, which playing a promising role in accelerating the pace of drug discovery process. Their view of the current
    [Show full text]
  • Safety and Tolerability of Lenalidomide Maintenance in Post-Transplant
    www.nature.com/bmt ARTICLE OPEN Safety and tolerability of lenalidomide maintenance in post-transplant acute myeloid leukemia and high-risk myelodysplastic syndrome ✉ Brian Pham 1 , Rasmus Hoeg1, Rajeev Krishnan2, Carol Richman1, Joseph Tuscano1 and Mehrdad Abedi 1 © The Author(s) 2021 Relapse after allogeneic stem cell transplant in unfavorable-risk acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS) portends a poor prognosis. We conducted a single-center phase I dose-escalation study with lenalidomide maintenance in high-risk MDS and AML patients after allogeneic transplantation. Sixteen patients enrolled in a “3 + 3” study design starting at lenalidomide 5 mg daily, increasing in increments of 5 mg up to 15 mg. Lenalidomide was given for 21 days of a 28-day cycle for a total of six cycles. Most common dose-limiting toxicities were lymphopenia, diarrhea, nausea, and neutropenia. Two patients had acute graft-versus-host disease (GVHD), and five patients developed chronic GVHD. The maximum tolerated dose was 10 mg, after dose-limiting toxicities were seen in the 15 mg group. Two dose-limiting toxicities were seen from development of acute GVHD and grade III diarrhea. Limitations of the study include time to initiation at 6 months post transplant, as many high-risk patients will have relapsed within this time frame before starting maintenance lenalidomide. Overall, lenalidomide was well tolerated with minimal GVHD and low rates of relapse rates, warranting further study. Bone Marrow Transplantation; https://doi.org/10.1038/s41409-021-01444-1 INTRODUCTION mutations, the role for post-transplant maintenance is less clear. Allogeneic stem cell transplantation (allo-SCT) remains the best Hypomethylating agents have been studied for post allo-SCT option for cure in most patients with unfavorable-risk acute maintenance therapy.
    [Show full text]
  • REVLIMID (Lenalidomide) 5 Mg, 10 Mg, 15 Mg and 25 Mg Capsules
    ® 1 REVLIMID (lenalidomide) 2 5 mg, 10 mg, 15 mg and 25 mg capsules 3 WARNINGS: 4 1. POTENTIAL FOR HUMAN BIRTH DEFECTS 5 2. HEMATOLOGIC TOXICITY (NEUTROPENIA AND 6 THROMBOCYTOPENIA) 7 3. DEEP VENOUS THROMBOSIS AND PULMONARY EMBOLISM 8 9 POTENTIAL FOR HUMAN BIRTH DEFECTS 10 WARNING: POTENTIAL FOR HUMAN BIRTH DEFECTS 11 LENALIDOMIDE IS AN ANALOGUE OF THALIDOMIDE. THALIDOMIDE IS 12 A KNOWN HUMAN TERATOGEN THAT CAUSES SEVERE LIFE­ 13 THREATENING HUMAN BIRTH DEFECTS. IF LENALIDOMIDE IS TAKEN 14 DURING PREGNANCY, IT MAY CAUSE BIRTH DEFECTS OR DEATH TO AN 15 UNBORN BABY. FEMALES SHOULD BE ADVISED TO AVOID PREGNANCY 16 WHILE TAKING REVLIMID® (lenalidomide). 17 Special Prescribing Requirements 18 BECAUSE OF THIS POTENTIAL TOXICITY AND TO AVOID FETAL 19 EXPOSURE TO REVLIMID® (lenalidomide), REVLIMID® (lenalidomide) IS 20 ONLY AVAILABLE UNDER A SPECIAL RESTRICTED DISTRIBUTION 21 PROGRAM. THIS PROGRAM IS CALLED "RevAssist®." UNDER THIS 22 PROGRAM, ONLY PRESCRIBERS AND PHARMACISTS REGISTERED WITH 23 THE PROGRAM CAN PRESCRIBE AND DISPENSE THE PRODUCT. IN 24 ADDITION, REVLIMID® (lenalidomide) MUST ONLY BE DISPENSED TO 25 PATIENTS WHO ARE REGISTERED AND MEET ALL THE CONDITIONS OF 26 THE RevAssist® PROGRAM. 27 PLEASE SEE THE FOLLOWING INFORMATION FOR PRESCRIBERS, 28 FEMALE PATIENTS, AND MALE PATIENTS ABOUT THIS RESTRICTED 29 DISTRIBUTION PROGRAM. 30 RevAssist® PROGRAM DESCRIPTION 31 Prescribers 32 REVLIMID® (lenalidomide) can be prescribed only by licensed prescribers who are 33 registered in the RevAssist® program and understand the potential risk of teratogenicity if 34 lenalidomide is used during pregnancy. 1 35 Effective contraception must be used by female patients of childbearing potential for at 36 least 4 weeks before beginning REVLIMID® (lenalidomide) therapy, during 37 REVLIMID® (lenalidomide) therapy, during dose interruptions and for 4 weeks 38 following discontinuation of REVLIMID® (lenalidomide) therapy.
    [Show full text]