51
|
Derudas D, Capraro F, Martinelli G, Cerchione C. How I manage frontline transplant-ineligible multiple myeloma. Hematol Rep 2020; 12:8956. [PMID: 33042505 PMCID: PMC7520858 DOI: 10.4081/hr.2020.8956] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Indexed: 12/24/2022] Open
Abstract
The Multiple Myeloma (MM) is a plasma cells hematological malignancy with a median age of 69 years at diagnosis. The autologous stem cell transplantation is the standard of care for this disease but less than half of newly diagnosed patients are assessed for this treatment due to comorbidities or complications of disease. The management of transplant ineligible MM patients is based on the balance safety and efficacy of the new available regimen and a careful assessment of the frailty status is mandatory to define the goals. In this review we discuss of the clinical dilemmas in the management and define how to manage them based on the evidence from clinical trials and "real life" experience.
Collapse
Affiliation(s)
- Daniele Derudas
- S.C. di Ematologia e C.T.M.O., Ospedale Oncologico di Riferimento Regionale “A. Businco”, Cagliari
| | - Francesca Capraro
- S.C. di Ematologia e C.T.M.O., Ospedale Oncologico di Riferimento Regionale “A. Businco”, Cagliari
| | - Giovanni Martinelli
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola (FC), Italy
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola (FC), Italy
| |
Collapse
|
52
|
Manda S, Yimer HA, Noga SJ, Girnius S, Yasenchak CA, Charu V, Lyons R, Aiello J, Bogard K, Ferrari RH, Cherepanov D, Demers B, Lu V, Whidden P, Kambhampati S, Birhiray RE, Jhangiani HS, Boccia R, Rifkin RM. Feasibility of Long-term Proteasome Inhibition in Multiple Myeloma by in-class Transition From Bortezomib to Ixazomib. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e910-e925. [PMID: 32912820 PMCID: PMC7336931 DOI: 10.1016/j.clml.2020.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/17/2020] [Accepted: 06/30/2020] [Indexed: 10/26/2022]
Abstract
BACKGROUND The ongoing US MM-6 study is investigating in-class transition (iCT) from parenteral bortezomib-based induction to all-oral IRd (ixazomib-lenalidomide-dexamethasone) with the aim of increasing proteasome inhibitor (PI)-based treatment adherence and duration while maintaining patients' health-related quality of life (HRQoL) and improving outcomes. PATIENTS AND METHODS US community sites are enrolling non-transplant-eligible patients with newly diagnosed multiple myeloma (MM) with no evidence of progressive disease after 3 cycles of bortezomib-based therapy to receive IRd (up to 39 cycles or until progression or toxicity). The patients use mobile or wearable digital devices to collect actigraphy (activity and sleep) data and electronically complete HRQoL, treatment satisfaction and medication adherence questionnaires. The primary endpoint is progression-free survival. The key secondary endpoints include response rates and therapy duration. RESULTS At the data cutoff, 84 patients had been treated (median age 73 years; 44% aged ≥ 75 years; 49% men; 15% Black or African American; and 10% Hispanic or Latino). Of the 84 patients, 62% were continuing therapy. The mean duration of total PI therapy was 10.1 months and for the IRd regimen was 7.3 months. With an 8-month median follow-up, the 12-month progression-free survival rate was 86% (95% confidence interval, 73%-93%) from both the start of bortezomib-based treatment and the start of IRd. The overall response rate was 62% (complete response, 4%; very good partial response, 25%; partial response, 33%) after bortezomib-based induction and 70% (complete response, 26%; very good partial response, 29%; partial response, 15%) after iCT. The IRd safety profile was consistent with previous clinical trial data, and HRQoL and treatment satisfaction were maintained. CONCLUSION The patients included in the US MM-6 study are representative of the real-world US MM population. The use of iCT might permit prolonged PI-based therapy with promising efficacy, without impacting patients' HRQoL or treatment satisfaction.
Collapse
Affiliation(s)
- Sudhir Manda
- Arizona Oncology/US Oncology Research, Tucson, AZ
| | | | - Stephen J Noga
- Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | | | | | | | - Roger Lyons
- Texas Oncology/US Oncology Research, San Antonio, TX
| | | | - Kimberly Bogard
- Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Renda H Ferrari
- Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Dasha Cherepanov
- Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Brittany Demers
- Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Vickie Lu
- Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Presley Whidden
- Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | | | - Ruemu E Birhiray
- Hematology Oncology of Indiana/American Oncology Network, Indianapolis, IN
| | | | - Ralph Boccia
- Center for Cancer and Blood Disorders, Bethesda, MD
| | - Robert M Rifkin
- Rocky Mountain Cancer Centers/US Oncology Research, Denver, CO.
| |
Collapse
|
53
|
Ixazomib-based frontline therapy in patients with newly diagnosed multiple myeloma in real-life practice showed comparable efficacy and safety profile with those reported in clinical trial: a multi-center study. Ann Hematol 2020; 99:2589-2598. [PMID: 32892275 PMCID: PMC7474576 DOI: 10.1007/s00277-020-04234-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023]
Abstract
The induction therapy containing ixazomib, an oral proteasome inhibitor, has shown favorable efficacy and safety in clinical trials, but its experience in real-life remains limited. In routine practice, few patients received ixazomib-based induction therapy due to reasons including (1) patients’ preference on oral regimens, (2) concerns on adverse events (AEs) of other intravenous/subcutaneous regimens, (3) requirements for less center visits, and (4) fears of COVID-19 and other infectious disease exposures. With the aim of assessing the real-life effectiveness and safety of ixazomib-based induction therapy, we performed this multi-center, observational study on 85 newly diagnosed multiple myeloma (NDMM) patients from 14 medical centers. Ixazomib-based regimens included ixazomib-lenalidomide-dexamethasone (IRd) in 44.7% of patients, ixazomib-dexamethasone (Id) in 29.4%, and Id plus another agent (doxorubicin, cyclophosphamide, thalidomide, or daratumumab) in 25.9%. Different ixazomib-based therapies were applied due to (1) financial burdens or limitations on local health insurance coverage, (2) concerns on treatment tolerance, and (3) drug accessibility issue. Ten patients received ixazomib maintenance. The median age was 67 years; 43.5% had ISS stage III disease; 48.2% had an Eastern Cooperative Oncology Group performance score ≥ 2; and 17.6% with high-risk cytogenetic abnormalities. Overall response rate for all 85 patients was 95.3%, including 65.9% very good partial response or better and 29.5% complete responses. The median time to response was 30 days. The response rate was similar across different ixazomib-based regimens. Median progression-free survival was not reached. Severe AEs (≥ grade 3) were reported in 29.4% of patients. No grade 3/4 peripheral neuropathy (PN) occurred. Patients received a median of 6 (range 1–20) cycles of ixazomib treatment; 56.6% remained on treatment at data cutoff; 15.3% discontinued treatment due to intolerable AEs. These results support that the ixazomib-based frontline therapy was highly effective with acceptable toxicity in routine practice and the ixazomib oral regimens could be good alternative options for NDMM patients.
Collapse
|
54
|
Quillin J, Patel R, Herzberg E, Alton D, Bikzhanova G, Geisler L, Olson J. A phase 0 analysis of ixazomib in patients with glioblastoma. Mol Clin Oncol 2020; 13:43. [PMID: 32874573 DOI: 10.3892/mco.2020.2114] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/08/2020] [Indexed: 11/06/2022] Open
Abstract
Improving overall survival in recurrent glioblastoma remains a challenge, and drugs acting by unique mechanisms are urgently required. Ixazomib is an orally-administered proteasome inhibitor used in combination with lenalidomide and dexamethasone to treat patients with multiple myeloma who have received at least one prior therapy. However, ixazomib's ability to reach brain tumors has not been studied during its development. The aim of the present study (ClinicalTrials.gov, NCT02630030) was to establish and quantify ixazomib's presence in glioblastoma. The present study investigated 3 patients with recurrent glioblastoma after administration of oral ixazomib citrate (MLN 9708) at a fixed 4.0 mg dose within a 3-hpreoperative window. A total of 2 blood samples were taken from each patient at the time of incision, tumor sampling and closure. Brain tumor samples were collected during tumor resection. These samples were then used to measure the plasma and brain tumor tissue concentration of the biologically-active form of ixazomib (MLN 2238). Patient 1 had plasma concentrations of ixazomib averaging 26.2, 21.8 and 15.3 ng/ml at incision, tumor sampling and closure, respectively. The brain tumor tissue concentration was 7.88 ng/g. Patient 2 had the same interval and brain tumor tissue measurements of 19.0, 18.0 and 8.93 ng/ml, and 2.03 ng/g. Patient 3 had plasma concentration interval measurements of 25.6, 36.2 and 28.7 ng/ml. Multiple brain tumor tissue samples were taken in patient 3, with an average tissue ixazomib concentration of 3.37 ng/g. Ixazomib was found at plasma concentrations commensurate with its previously established pharmacokinetic profile without clinically relevant drug-related adverse events. Ixazomib reaches glioblastoma tissues at measurable concentrations at the time of tumor resection, confirming target tissue delivery. This justifies the phase I study of ixazomib in recurrent glioblastoma currently in development.
Collapse
Affiliation(s)
- Joseph Quillin
- Department of Neurosurgery, Emory University, Atlanta, GA 30322, USA
| | - Rikesh Patel
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | - Denny Alton
- Covance Laboratories, Inc., Madison, WI 53704, USA
| | | | - Lisa Geisler
- Covance Laboratories, Inc., Madison, WI 53704, USA
| | - Jeffrey Olson
- Department of Neurosurgery, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
55
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
56
|
Eluard B, Thieblemont C, Baud V. NF-κB in the New Era of Cancer Therapy. Trends Cancer 2020; 6:677-687. [DOI: 10.1016/j.trecan.2020.04.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 01/06/2023]
|
57
|
Zanwar S, Abeykoon JP, Kapoor P. Challenges and Strategies in the Management of Multiple Myeloma in the Elderly Population. Curr Hematol Malig Rep 2020; 14:70-82. [PMID: 30820879 DOI: 10.1007/s11899-019-00500-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Approximately one half of the patient-population in multiple myeloma (MM) is > 70 years at diagnosis. Despite notable strides in the management and improved survival, MM remains incurable, with an increasing proportion of elderly patients comprising the relapsed-refractory cohort. RECENT FINDINGS The arbitrary age cutoff at 65 years to define the elderly patient-population has evolved to a more nuanced categorization, incorporating a comprehensive assessment for determining frailty prior to commencing treatment. This step is critical in determining the therapy-intensity, including transplant-eligibility, to minimize toxicity. Dose-modifications are crucial, as the merits of continuous therapy are becoming evident in this patient-population. Bortezomib, lenalidomide, and dexamethasone (VRd) combination has emerged as standard of care for newly diagnosed MM. Fixed-duration Rd followed by reduced-dosed continuous R may be considered in select frail patients with standard-risk MM. Herein, we review the unique challenges encountered in elderly MM and discuss strategies for optimal management.
Collapse
Affiliation(s)
- Saurabh Zanwar
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Prashant Kapoor
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
58
|
Shi CX, Zhu YX, Bruins LA, Bonolo de Campos C, Stewart W, Braggio E, Stewart AK. Proteasome Subunits Differentially Control Myeloma Cell Viability and Proteasome Inhibitor Sensitivity. Mol Cancer Res 2020; 18:1453-1464. [PMID: 32561655 DOI: 10.1158/1541-7786.mcr-19-1026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/02/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022]
Abstract
We generated eight multiple myeloma cell lines resistant to bortezomib; five acquired PSMB5 mutations. In 1,500 patients such mutations were rare clinically. To better understand disruption of proteasomes on multiple myeloma viability and drug sensitivity, we systematically deleted the major proteasome catalytic subunits. Multiple myeloma cells without PSMB5 were viable. Drug-resistant, PSMB5-mutated cell lines were resensitized to bortezomib by PSMB5 deletion, implying PSMB5 mutation is activating in its drug resistance function. In contrast, PSMB6 knockout was lethal to multiple myeloma cell lines. Depleting PSMB6 prevented splicing of the major catalytic subunits PSMB5, PSMB7, PSMB8, and PSMB10; however, PSMB6 engineered without splicing function or catalytic activity, also restored viability, inferring the contribution of PSMB6 to proteasome structure to be more important than functional activity. Supporting this, bortezomib sensitivity was restored in drug-resistant multiple myeloma cell lines by low level expression of mutated PSMB6 lacking splicing function. Loss of PSMB8 and PSMB9 was neither lethal nor restored bortezomib sensitivity. Significant codependency of PSMB5, PSMB6, and PSMB7 expression was observed. We demonstrated elevated levels of PSMB6 and 7, but not 8 and 9, in some, but not all, serial patient samples exposed to proteasome inhibitors. In summary, we show PSMB6 and PSMB7, but not PSMB5, to be essential for multiple myeloma cell survival, this dependency is structural and that upregulation or activating mutation of PSMB5, 6, and 7 confers proteasome inhibitor resistance, while depletion confers sensitivity. IMPLICATIONS: These findings support modulation of PSMB5, PSMB6, or PSMB7 expression as a new therapeutic strategy.
Collapse
Affiliation(s)
- Chang-Xin Shi
- Department of Hematology, Mayo Clinic in Arizona, Scottsdale, Arizona
| | - Yuan Xiao Zhu
- Department of Hematology, Mayo Clinic in Arizona, Scottsdale, Arizona
| | - Laura A Bruins
- Department of Hematology, Mayo Clinic in Arizona, Scottsdale, Arizona
| | | | - William Stewart
- Department of Hematology, Mayo Clinic in Arizona, Scottsdale, Arizona
| | - Esteban Braggio
- Department of Hematology, Mayo Clinic in Arizona, Scottsdale, Arizona
| | - A Keith Stewart
- Department of Hematology, Mayo Clinic in Arizona, Scottsdale, Arizona. .,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
59
|
Ria R, Melaccio A, Racanelli V, Vacca A. Anti-VEGF Drugs in the Treatment of Multiple Myeloma Patients. J Clin Med 2020; 9:E1765. [PMID: 32517267 PMCID: PMC7355441 DOI: 10.3390/jcm9061765] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
The interaction between the bone marrow microenvironment and plasma cells plays an essential role in multiple myeloma progression and drug resistance. The vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathway in vascular endothelial cells activates and promotes angiogenesis. Moreover, VEGF activates and promotes vasculogenesis and vasculogenic mimicry when it interacts with VEGF receptors expressed in precursor cells and inflammatory cells, respectively. In myeloma bone marrow, VEGF and VEGF receptor expression are upregulated and hyperactive in the stromal and tumor cells. It has been demonstrated that several antiangiogenic agents can effectively target VEGF-related pathways in the preclinical phase. However, they are not successful in treating multiple myeloma, probably due to the vicarious action of other cytokines and signaling pathways. Thus, the simultaneous blocking of multiple cytokine pathways, including the VEGF/VEGFR pathway, may represent a valid strategy to treat multiple myeloma. This review aims to summarize recent advances in understanding the role of the VEGF/VEGFR pathway in multiple myeloma, and mainly focuses on the transcription pathway and on strategies that target this pathway.
Collapse
Affiliation(s)
- Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (A.M.); (V.R.); (A.V.)
| | | | | | | |
Collapse
|
60
|
Rajkumar SV. Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. Am J Hematol 2020; 95:548-567. [PMID: 32212178 DOI: 10.1002/ajh.25791] [Citation(s) in RCA: 505] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022]
Abstract
DISEASE OVERVIEW Multiple myeloma accounts for approximately 10% of hematologic malignancies. DIAGNOSIS The diagnosis requires ≥10% clonal bone marrow plasma cells or a biopsy proven plasmacytoma plus evidence of one or more multiple myeloma defining events (MDE) namely CRAB (hypercalcemia, renal failure, anemia, or lytic bone lesions) features felt related to the plasma cell disorder, bone marrow clonal plasmacytosis ≥60%, serum involved/uninvolved free light chain (FLC) ratio ≥100 (provided involved FLC is ≥100 mg/L), or >1 focal lesion on magnetic resonance imaging (MRI). RISK STRATIFICATION The presence of del(17p), t(4;14), t(14;16), t(14;20), gain 1q, or p53 mutation is considered high-risk multiple myeloma. Presence of any two high risk factors is considered double-hit myeloma; three or more high risk factors is triple-hit myeloma. RISK-ADAPTED INITIAL THERAPY In transplant eligible patients, induction therapy consists of bortezomib, lenalidomide, dexamethasone (VRd) given for approximately 3-4 cycles followed by autologous stem cell transplantation (ASCT). In high-risk patients, daratumumab, bortezomib, lenalidomide, dexamethasone (Dara-VRd) is an alternative to VRd. Selected standard risk patients can get additional cycles of induction, and delay transplant until first relapse. Patients not candidates for transplant are typically treated with VRd for approximately 8-12 cycles followed by lenalidomide; alternatively these patients can be treated with daratumumab, lenalidomide, dexamethasone (DRd). MAINTENANCE THERAPY After ASCT, standard risk patients need lenalidomide maintenance, while bortezomib-based maintenance is needed for patients with high-risk myeloma. MANAGEMENT OF REFRACTORY DISEASE Most patients require a triplet regimen at relapse, with the choice of regimen varying with each successive relapse.
Collapse
|
61
|
Hong L, Zhang C, Jiang Y, Liu H, Huang H, Guo D. Therapeutic status and the prospect of CRISPR/Cas9 gene editing in multiple myeloma. Future Oncol 2020; 16:1125-1136. [PMID: 32338048 DOI: 10.2217/fon-2019-0822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In recent years, CRISPR/Cas9, a novel gene-editing technology, has shown considerable potential in the design of novel research methods and future options for treating multiple myeloma (MM). The use of CRISPR/Cas9 promises faster and more accurate identification and validation of target genes. In this review, we summarize the current research status of the application of CRISPR technology in MM, especially in detecting the expression of MM gene, exploring the mechanism of drug action, screening for drug-resistant genes, developing immunotherapy and screening for new drug targets. Given the tremendous progress that has been made, we believe that CRISPR/Cas9 possesses great potential in MM-related clinical practice.
Collapse
Affiliation(s)
- Lemin Hong
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Chenlu Zhang
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Yijing Jiang
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Haiyan Liu
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Hongming Huang
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Dan Guo
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| |
Collapse
|
62
|
Iron Causes Lipid Oxidation and Inhibits Proteasome Function in Multiple Myeloma Cells: A Proof of Concept for Novel Combination Therapies. Cancers (Basel) 2020; 12:cancers12040970. [PMID: 32295216 PMCID: PMC7226326 DOI: 10.3390/cancers12040970] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/06/2020] [Accepted: 04/10/2020] [Indexed: 12/24/2022] Open
Abstract
Adaptation to import iron for proliferation makes cancer cells potentially sensitive to iron toxicity. Iron loading impairs multiple myeloma (MM) cell proliferation and increases the efficacy of the proteasome inhibitor bortezomib. Here, we defined the mechanisms of iron toxicity in MM.1S, U266, H929, and OPM-2 MM cell lines, and validated this strategy in preclinical studies using Vk*MYC mice as MM model. High-dose ferric ammonium citrate triggered cell death in all cell lines tested, increasing malondialdehyde levels, the by-product of lipid peroxidation and index of ferroptosis. In addition, iron exposure caused dose-dependent accumulation of polyubiquitinated proteins in highly iron-sensitive MM.1S and H929 cells, suggesting that proteasome workload contributes to iron sensitivity. Accordingly, high iron concentrations inhibited the proteasomal chymotrypsin-like activity of 26S particles and of MM cellular extracts in vitro. In all MM cells, bortezomib-iron combination induced persistent lipid damage, exacerbated bortezomib-induced polyubiquitinated proteins accumulation, and triggered cell death more efficiently than individual treatments. In Vk*MYC mice, addition of iron dextran or ferric carboxymaltose to the bortezomib-melphalan-prednisone (VMP) regimen increased the therapeutic response and prolonged remission without causing evident toxicity. We conclude that iron loading interferes both with redox and protein homeostasis, a property that can be exploited to design novel combination strategies including iron supplementation, to increase the efficacy of current MM therapies.
Collapse
|
63
|
Zhang X, Linder S, Bazzaro M. Drug Development Targeting the Ubiquitin-Proteasome System (UPS) for the Treatment of Human Cancers. Cancers (Basel) 2020; 12:cancers12040902. [PMID: 32272746 PMCID: PMC7226376 DOI: 10.3390/cancers12040902] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cells are characterized by a higher rate of protein turnover and greater demand for protein homeostasis compared to normal cells. In this scenario, the ubiquitin-proteasome system (UPS), which is responsible for the degradation of over 80% of cellular proteins within mammalian cells, becomes vital to cancer cells, making the UPS a critical target for the discovery of novel cancer therapeutics. This review systematically categorizes all current reported small molecule inhibitors of the various essential components of the UPS, including ubiquitin-activating enzymes (E1s), ubiquitin-conjugating enzymes (E2s), ubiquitin ligases (E3s), the 20S proteasome catalytic core particle (20S CP) and the 19S proteasome regulatory particles (19S RP), as well as their mechanism/s of action and limitations. We also discuss the immunoproteasome which is considered as a prospective therapeutic target of the next generation of proteasome inhibitors in cancer therapies.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 05 Uppsala, Sweden
| | - Stig Linder
- Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;
- Department of Medical and Health Sciences, Linköping University, SE-58183 Linköping, Sweden
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA;
- Correspondence:
| |
Collapse
|
64
|
Optimizing Immunomodulatory Drug With Proteasome Inhibitor Combinations in Newly Diagnosed Multiple Myeloma. ACTA ACUST UNITED AC 2020; 25:2-10. [PMID: 30694854 DOI: 10.1097/ppo.0000000000000348] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the modern era of multiple myeloma therapeutics, proteasome inhibitor (PI) and immunomodulatory drugs (IMiDs) have replaced chemotherapy regimens for newly diagnosed multiple myeloma patients. Treatment combinations that comprise both first- and next-generation PIs, including bortezomib, carfilzomib, and ixazomib and IMiDs, including thalidomide and lenalidomide, have been evaluated in phases II and III clinical trials and have shown significant efficacy with manageable toxicity profiles. Bortezomib or carfilzomib with lenalidomide and dexamethasone (VRD and KRD) are the most promising regimens resulting in significant survival improvement. Disease and patient characteristics should lead the individualization of treatment, with the eligibility for autologous transplant being of principal importance. The addition of a monoclonal antibody to PI with IMiD combinations is currently under clinical investigation and may lead to further treatment optimization.
Collapse
|
65
|
Dimopoulos MA, Jakubowiak AJ, McCarthy PL, Orlowski RZ, Attal M, Bladé J, Goldschmidt H, Weisel KC, Ramasamy K, Zweegman S, Spencer A, Huang JSY, Lu J, Sunami K, Iida S, Chng WJ, Holstein SA, Rocci A, Skacel T, Labotka R, Palumbo A, Anderson KC. Developments in continuous therapy and maintenance treatment approaches for patients with newly diagnosed multiple myeloma. Blood Cancer J 2020; 10:17. [PMID: 32054831 PMCID: PMC7018731 DOI: 10.1038/s41408-020-0273-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 12/30/2022] Open
Abstract
The evolving paradigm of continuous therapy and maintenance treatment approaches in multiple myeloma (MM) offers prolonged disease control and improved outcomes compared to traditional fixed-duration approaches. Potential benefits of long-term strategies include sustained control of disease symptoms, as well as continued cytoreduction and clonal control, leading to unmeasurable residual disease and the possibility of transforming MM into a chronic or functionally curable condition. "Continuous therapy" commonly refers to administering a doublet or triplet regimen until disease progression, whereas maintenance approaches typically involve single-agent or doublet treatment following more intensive prior therapy with autologous stem cell transplant (ASCT) or doublet, triplet, or even quadruplet induction therapy. However, the requirements for agents and regimens within these contexts are similar: treatments must be tolerable for a prolonged period of time, should not be associated with cumulative or chronic toxicity, should not adversely affect patients' quality of life, should ideally be convenient with a minimal treatment burden for patients, and should not impact the feasibility or efficacy of subsequent treatment at relapse. Multiple agents have been and are being investigated as long-term options in the treatment of newly diagnosed MM (NDMM), including the immunomodulatory drugs lenalidomide and thalidomide, the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, and the monoclonal antibodies daratumumab, elotuzumab, and isatuximab. Here we review the latest results with long-term therapy approaches in three different settings in NDMM: (1) maintenance treatment post ASCT; (2) continuous frontline therapy in nontransplant patients; (3) maintenance treatment post-frontline therapy in the nontransplant setting. We also discuss evidence from key phase 3 trials. Our review demonstrates how the paradigm of long-term treatment is increasingly well-established across NDMM treatment settings, potentially resulting in further improvements in patient outcomes, and highlights key clinical issues that will need to be addressed in order to provide optimal benefit.
Collapse
Affiliation(s)
- Meletios A Dimopoulos
- Hematology & Medical Oncology, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | | | | | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michel Attal
- Hematology Department, University Hospital Purpan, Toulouse, France
| | - Joan Bladé
- Hematology Department, Hospital Clinic, Institut de Investigacions Biomediques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University Medical Hospital and National Center of Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Katja C Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, VU University Amsterdam, Amsterdam, The Netherlands
| | - Andrew Spencer
- Malignant Haematology and Stem Cell Transplantation Service, Alfred Health-Monash University, Melbourne, Australia
| | | | - Jin Lu
- Department of Hematology, Peking University People's Hospital and Peking University Institute of Hematology, Beijing, China
| | - Kazutaka Sunami
- Department of Hematology, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Wee-Joo Chng
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, and Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alberto Rocci
- Department of Haematology, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
- Faculty of Biology, Medicine and Health, School of Medical Science, Division of Cancer Science, University of Manchester, Manchester, UK
| | - Tomas Skacel
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Richard Labotka
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Antonio Palumbo
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | | |
Collapse
|
66
|
Pinto V, Bergantim R, Caires HR, Seca H, Guimarães JE, Vasconcelos MH. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers (Basel) 2020; 12:E407. [PMID: 32050631 PMCID: PMC7072128 DOI: 10.3390/cancers12020407] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is the second most common blood cancer. Treatments for MM include corticosteroids, alkylating agents, anthracyclines, proteasome inhibitors, immunomodulatory drugs, histone deacetylase inhibitors and monoclonal antibodies. Survival outcomes have improved substantially due to the introduction of many of these drugs allied with their rational use. Nonetheless, MM patients successively relapse after one or more treatment regimens or become refractory, mostly due to drug resistance. This review focuses on the main drugs used in MM treatment and on causes of drug resistance, including cytogenetic, genetic and epigenetic alterations, abnormal drug transport and metabolism, dysregulation of apoptosis, autophagy activation and other intracellular signaling pathways, the presence of cancer stem cells, and the tumor microenvironment. Furthermore, we highlight the areas that need to be further clarified in an attempt to identify novel therapeutic targets to counteract drug resistance in MM patients.
Collapse
Affiliation(s)
- Vanessa Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- FCTUC–Faculty of Science and Technology of the University of Coimbra, 3030-790 Coimbra, Portugal
| | - Rui Bergantim
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hugo R. Caires
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Hugo Seca
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - M. Helena Vasconcelos
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP-Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
67
|
Multiple Myeloma and Thrombosis: Prophylaxis and Risk Prediction Tools. Cancers (Basel) 2020; 12:cancers12010191. [PMID: 31940972 PMCID: PMC7016665 DOI: 10.3390/cancers12010191] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/19/2022] Open
Abstract
Thromboembolism in multiple myeloma (MM) patients remains a common complication that renders the optimization of our thromboprophylaxis practice necessary. This review aims to make clear the need for the development of more accurate risk assessment tools and means of thrombosis prevention. Current clinical practice is guided by available guidelines published by the IMWG in 2014, but the extent to which these are implemented is unclear. Recently, several groups developed clinical scores for thrombosis risk in MM in an attempt to improve risk stratification, but these have not been validated or used in clinical practice so far. Research in this field is increasingly focusing on understanding the unique coagulation profile of the MM patient, and data on potential biomarkers that accurately reflect hypercoagulability is emerging. Finally, promising evidence on the effectiveness of direct oral anticoagulants (DOACs) in the context of thrombosis prevention in MM patients is increasingly becoming available. The critical appraisal of the above research areas will establish the necessity of combining disease-specific clinical risk factors with coagulation biomarkers to allow more effective risk stratification that will eventually lead to the reduction of this significant complication. Results from ongoing clinical trials on the role of DOACs are much anticipated.
Collapse
|
68
|
Abe Y, Ishida T. Immunomodulatory drugs in the treatment of multiple myeloma. Jpn J Clin Oncol 2020; 49:695-702. [PMID: 31187860 DOI: 10.1093/jjco/hyz083] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/26/2019] [Indexed: 11/15/2022] Open
Abstract
The prognosis of multiple myeloma was quite poor in the last century, but it has significantly improved with the incorporation of novel agents, immunomodulatory drugs (IMiDs) and proteasome inhibitors. Thalidomide was first developed as a sedative in 1950s, but it was withdrawn from the market because of teratogenicity. In 1990s, however, thalidomide received attention due to the discovery of its anticancer potential derived from antiangiogenic and immunomodulatory activities, and its therapeutic effect on myeloma. In 2006, the U.S. Food and Drug Administration approved the use of thalidomide under strict control for the treatment of multiple myeloma. After that, two new IMiDs, lenalidomide and pomalidomide, were developed for the sake of more antitumor activity and less adverse events than thalidomide. The molecular mechanism of action of IMiDs remained unclear for a long time until 2010 when the protein cereblon (CRBN) was identified as a primary direct target. IMiDs binds to CRBN and alters the substrate specificity of the CRBN E3 ubiquitin ligase complex, resulting in breakdown of intrinsic downstream proteins such as IKZF1 (Ikaros) and IKZF3 (Aiolos). There are many clinical trials of multiple myeloma using IMiDs under various conditions, and most of them show the efficacy of IMiDs. Nowadays lenalidomide plays a central role in both newly diagnosed and relapsed/refractory settings, mainly in combination with other novel agents such as proteasome inhibitors and monoclonal antibodies. This review presents an overview of recent advances in immunomodulatory drugs in the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Yu Abe
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Tadao Ishida
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| |
Collapse
|
69
|
Chakraborty R, Bin Riaz I, Malik SU, Marneni N, Mejia Garcia A, Anwer F, Khorana AA, Rajkumar SV, Kumar S, Murad MH, Wang Z, Khan SU, Majhail NS. Venous thromboembolism risk with contemporary lenalidomide-based regimens despite thromboprophylaxis in multiple myeloma: A systematic review and meta-analysis. Cancer 2020; 126:1640-1650. [PMID: 31913498 DOI: 10.1002/cncr.32682] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/23/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Thromboprophylaxis is routinely used with lenalidomide-based regimens in multiple myeloma because of a substantial risk of venous thromboembolism (VTE). However, little is known about the incidence of VTE with contemporary lenalidomide-based regimens. The objective of the current study was to estimate the incidence of VTE despite thromboprophylaxis with currently used lenalidomide-based regimens in patients with myeloma. METHODS The Ovid MEDLINE, Embase, and Cochrane databases were queried from study inception to January 2019 for keywords to cover the following concepts: "lenalidomide," "venous thromboembolism," and "multiple myeloma." Phase 1, 2, and 3 clinical trials evaluating lenalidomide-based regimens with thromboprophylaxis were included. The pooled incidence rate of VTE was estimated using a random-effects model. RESULTS The search generated 1372 citations, with 51 clinical trials and 9069 patients included for analysis. The most common thromboprophylaxis agents were aspirin, low-molecular-weight heparin or warfarin, administered either per risk-stratification or at investigators' discretion. The pooled incidence of VTE in trials of patients who had newly diagnosed and relapsed/refractory myeloma was 6.2% (95% CI, 5.4%-7.1%) over median treatment durations ranging from 2 to 34 cycles, which translated into 1.2 VTE events per 100 patient-cycles (95% CI, 0.9-1.7 VTE events per 100 patient-cycles). Among contemporary regimens, the risk of VTE was low with combined lenalidomide and low-dose dexamethasone (0.2 [95% CI, 0.1-0.6] events/100 patient-cycles) and lenalidomide maintenance (0.0 [95% CI, 0.0-0.7] events per 100 patient-cycles). VTE risk was higher with combined lenalidomide and low-dose dexamethasone plus proteasome inhibitors (1.3 [95% CI, 0.7-2.3] events per 100 patient-cycles). CONCLUSIONS Despite adequate thromboprophylaxis, lenalidomide-based regimens have a substantial risk of VTE in controlled clinical trial settings. Further studies are needed on new thromboprophylaxis strategies with regimens that have a high VTE risk.
Collapse
Affiliation(s)
| | - Irbaz Bin Riaz
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Saad Ullah Malik
- Department of Hematology and Oncology, University of Arizona, Tucson, Arizona
| | | | | | - Faiz Anwer
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Alok A Khorana
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | | | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | - M Hassan Murad
- Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota
| | - Zhen Wang
- Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota
| | - Safi U Khan
- Department of Internal Medicine, West Virginia University, Morgantown, West Virginia
| | - Navneet S Majhail
- Blood and Marrow Transplant Program, Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
70
|
Xie SC, Dick LR, Gould A, Brand S, Tilley L. The proteasome as a target for protozoan parasites. Expert Opin Ther Targets 2019; 23:903-914. [PMID: 31679410 DOI: 10.1080/14728222.2019.1685981] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: The proteasome is a multi-subunit enzyme complex responsible for the turnover of short-lived, abnormal or damaged proteins in eukaryotic cells. As organisms that undergo rapid growth and cell division, protozoan parasites exist on the knife-edge of proteotoxic catastrophe and thus rely heavily on their protein quality control machinery for survival. Because of this, the proteasome has recently emerged as a desirable drug target.Area covered: This review focuses on efforts to identify protozoan parasite-specific proteasome inhibitors using substrate profiling, library screening, and in vitro evolution of resistance approaches to inform medicinal chemistry. Targeting the parasite's 20S proteasome chymotrypsin-like (β5) activity and selectively inhibiting protein turnover in parasites compared to human cells are critical properties of potent, selective inhibitors.Expert opinion: Proteasome inhibitors have the potential for rapid action against all stages, all species and all strains of plasmodium and kinetoplastid parasites. Given the high level of conservation of proteasome active sites in eukaryotes, an important challenge is achieving inhibitors that show sufficient selectivity while maintaining properties consistent with drug development.
Collapse
Affiliation(s)
- Stanley C Xie
- Department of Biochemistry and Molecular Biology Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | | | - Alexandra Gould
- Oncology Chemistry, Takeda Pharmaceuticals International Co., Cambridge, MA, USA
| | - Stephen Brand
- Medicines for Malaria Venture, CH-1215 Geneva 15, Switzerland
| | - Leann Tilley
- Department of Biochemistry and Molecular Biology Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
71
|
Skorda A, Sklirou AD, Sakellaropoulos T, Gianniou DD, Kastritis E, Terpos E, Tsitsilonis OE, Florea BI, Overkleeft HS, Dimopoulos MA, Alexopoulos LG, Trougakos IP. Non-lethal proteasome inhibition activates pro-tumorigenic pathways in multiple myeloma cells. J Cell Mol Med 2019; 23:8010-8018. [PMID: 31568628 PMCID: PMC6850931 DOI: 10.1111/jcmm.14653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/17/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a haematological malignancy being characterized by clonal plasma cell proliferation in the bone marrow. Targeting the proteasome with specific inhibitors (PIs) has been proven a promising therapeutic strategy and PIs have been approved for the treatment of MM and mantle‐cell lymphoma; yet, while outcome has improved, most patients inevitably relapse. As relapse refers to MM cells that survive therapy, we sought to identify the molecular responses induced in MM cells after non‐lethal proteasome inhibition. By using bortezomib (BTZ), epoxomicin (EPOX; a carfilzomib‐like PI) and three PIs, namely Rub999, PR671A and Rub1024 that target each of the three proteasome peptidases, we found that only BTZ and EPOX are toxic in MM cells at low concentrations. Phosphoproteomic profiling after treatment of MM cells with non‐lethal (IC10) doses of the PIs revealed inhibitor‐ and cell type‐specific readouts, being marked by the activation of tumorigenic STAT3 and STAT6. Consistently, cytokine/chemokine profiling revealed the increased secretion of immunosuppressive pro‐tumorigenic cytokines (IL6 and IL8), along with the inhibition of potent T cell chemoattractant chemokines (CXCL10). These findings indicate that MM cells that survive treatment with therapeutic PIs shape a pro‐tumorigenic immunosuppressive cellular and secretory bone marrow microenvironment that enables malignancy to relapse.
Collapse
Affiliation(s)
- Aikaterini Skorda
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Aimilia D Sklirou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Despoina D Gianniou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania E Tsitsilonis
- Department of Animal and Human Physiology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Bogdan I Florea
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, Leiden, The Netherlands
| | - Herman S Overkleeft
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, Leiden, The Netherlands
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Leonidas G Alexopoulos
- School of Mechanical Engineering, National Technical University of Athens, Athens, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
72
|
Kurzrock R, Gurski LA, Carlson RW, Ettinger DS, Horwitz SM, Kumar SK, Million L, von Mehren M, Benson AB. Level of evidence used in recommendations by the National Comprehensive Cancer Network (NCCN) guidelines beyond Food and Drug Administration approvals. Ann Oncol 2019; 30:1647-1652. [PMID: 31373348 PMCID: PMC6857604 DOI: 10.1093/annonc/mdz232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND A previous analysis of 113 National Comprehensive Cancer Network® (NCCN®) recommendations reported that NCCN frequently recommends beyond Food and Drug Administration (FDA)-approved indications (44 off-label recommendations) and claimed that the evidence for these recommendations was weak. METHODS In order to determine the strength of the evidence, we carried out an in-depth re-analysis of the 44 off-label recommendations listed in the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). RESULTS Of the 44 off-label recommendations, 14 were later approved by the FDA and/or are supported by randomized controlled trial (RCT) data. In addition, 13 recommendations were either very minor extrapolations from the FDA label (n = 8) or were actually on-label (n = 5). Of the 17 remaining extrapolations, 8 were for mechanism-based agents applied in rare cancers or subsets with few available treatment options (median response rate = 43%), 7 were based on non-RCT data showing significant efficacy (>50% response rates), and 2 were later removed from the NCCN Guidelines because newer therapies with better activity and/or safety became available. CONCLUSION Off-label drug use is a frequent component of care for patients with cancer in the United States. Our findings indicate that when the NCCN recommends beyond the FDA-approved indications, the strength of the evidence supporting such recommendations is robust, with a significant subset of these drugs later becoming FDA approved or supported by RCT. Recommendations without RCT data are often for mechanism-based drugs with high response rates in rare cancers or subsets without effective therapies.
Collapse
Affiliation(s)
- R Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, San Diego, USA.
| | - L A Gurski
- National Comprehensive Cancer Network, Plymouth Meeting, Pennsylvania, USA
| | - R W Carlson
- National Comprehensive Cancer Network, Plymouth Meeting, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Stanford Cancer Institute, Stanford, California, USA
| | - D S Ettinger
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | - S M Horwitz
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - S K Kumar
- Mayo Clinic Cancer Center, Rochester, Minnesota, USA
| | - L Million
- Stanford Cancer Institute, Stanford, California, USA
| | - M von Mehren
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - A B Benson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
73
|
Ludwig H, Poenisch W, Knop S, Egle A, Schreder M, Lechner D, Hajek R, Gunsilius E, Krenosz KJ, Petzer A, Weisel K, Niederwieser D, Einsele H, Willenbacher W, Melchardt T, Greil R, Zojer N. Ixazomib-Thalidomide-Dexamethasone for induction therapy followed by Ixazomib maintenance treatment in patients with relapsed/refractory multiple myeloma. Br J Cancer 2019; 121:751-757. [PMID: 31558804 PMCID: PMC6889132 DOI: 10.1038/s41416-019-0581-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/11/2019] [Accepted: 08/27/2019] [Indexed: 12/16/2022] Open
Abstract
Background Ixazomib-revlimid-dexamethason showed significant activity in relapsed/refractory multiple myeloma (RRMM). Here, we evaluate ixazomib in combination with thalidomide and dexamethasone for induction treatment followed by ixazomib maintenance therapy in RRMM patients. Methods Ninety patients have been included. Ixazomib–thalidomide–dexamethasone (4 mg, day 1, 8, 15; 100 mg daily; and 40 mg weekly) was scheduled for eight cycles followed by maintenance with ixazomib for one year. Results The overall response rate was 51.1%, 23.3% achieved CR or VGPR and 10% MR resulting in a clinical benefit rate of 61.1%. In patients completing ≥2 cycles, the rates were 60.5%, 27.6% and 68.4%, respectively. Median progression-free survival (PFS) was 8.5 months in all, and 9.4 months in those completing ≥2 cycles. Response rates, PFS and overall survival (OS) were similar in patients with and without t(4;14) and/or del(17p), but PFS and OS was significantly shorter in patients with gain of 1q21. Multivariate regression analysis revealed gain of 1q21 as the most important factor associated with OS. Ixazomib maintenance resulted in an upgrade in the depth of response in 12.4% of patients. Grade 3/4 toxicities were relatively rare. Conclusions Ixazomib–thalidomide–dexamethasone followed by ixazomib maintenance therapy is active and well tolerated in patients with RRMM. Trial registration number NCT02410694
Collapse
Affiliation(s)
- Heinz Ludwig
- Wilhelminen Cancer Research Institute, Department of Medicine I, Center for Oncology and Hematology, Wilhelminenspital, Vienna, Austria.
| | - Wolfram Poenisch
- Department of Hematology, University of Leipzig, Leipzig, Germany
| | - Stefan Knop
- Division of Hematology and Medical Oncology, Department of Internal Medicine II, Wuerzburg University Medical Center, Wuerzburg, Germany
| | - Alexander Egle
- Department of Internal Medicine III with Haematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Salzburg, Cancer Cluster Salzburg, Austria
| | - Martin Schreder
- Department of Medicine I, Center for Oncology and Hematology, Wilhelminenspital, Vienna, Austria
| | - Daniel Lechner
- Department of Medicine I-Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Roman Hajek
- Fakultní Nemocnice Ostrava, Ostrava, Czech Republic
| | - Eberhard Gunsilius
- Department of Internal Medicine V, Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Karl Jochen Krenosz
- Department of Internal Medicine 3, Kepler Universitaetsklinikum GmbH, Med Campus III, Linz, Austria
| | - Andreas Petzer
- Department of Internal Medicine I, Ordensklinikum Linz-Barmherzige Schwestern, Linz, Austria
| | - Katja Weisel
- University Medical Center of Hamburg-Eppendorf, Hamburg, Germany and University of Tuebingen, Tuebingen, Germany
| | | | - Hermann Einsele
- Division of Hematology and Medical Oncology, Department of Internal Medicine II, Wuerzburg University Medical Center, Wuerzburg, Germany
| | - Wolfgang Willenbacher
- Department of Internal Medicine V, Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria.,Oncotyrol, Center for personalized Cancer Medicine, Innsbruck, Austria
| | - Thomas Melchardt
- Department of Internal Medicine III with Haematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Salzburg, Cancer Cluster Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Salzburg, Cancer Cluster Salzburg, Austria
| | - Niklas Zojer
- Department of Medicine I, Center for Oncology and Hematology, Wilhelminenspital, Vienna, Austria
| |
Collapse
|
74
|
Tzogani K, Florez B, Markey G, Caleno M, Olimpieri OM, Melchiorri D, Hovgaard DJ, Sarac SB, Penttilä K, Lapveteläinen T, Salmonson T, Bergh J, Gisselbrecht C, Pignatti F. European Medicines Agency review of ixazomib (Ninlaro) for the treatment of adult patients with multiple myeloma who have received at least one prior therapy. ESMO Open 2019; 4:e000570. [PMID: 31555488 PMCID: PMC6735670 DOI: 10.1136/esmoopen-2019-000570] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/04/2019] [Indexed: 11/24/2022] Open
Abstract
On 21 November 2016, the European Commission issued a marketing authorisation valid throughout the European Union for ixazomib in combination with lenalidomide and dexamethasone for the treatment of adult patients with multiple myeloma who have received at least one prior therapy. Ixazomib was evaluated in one, randomised, double-blind, phase III study comparing ixazomib plus lenalidomide and dexamethasone (n=360; ixazomib arm) versus placebo plus lenalidomide and dexamethasone (n=362; placebo arm) in adult patients with relapsed and/or refractory multiple myeloma who had received at least one prior therapy. The median progression-free survival (PFS) in the intent-to-treat population was 20.6 months in patients treated with ixazomib compared with 14.7 months for patients in the placebo arm (stratified HR=0.742, 95% CI 0.587 to 0.939, stratified p-value=0.012). The most frequently reported adverse reactions (≥20%) within the ixazomib and placebo arms were diarrhoea (42% vs 36%), constipation (34% vs 25%), thrombocytopaenia (28% vs 14%), peripheral neuropathy (28% vs 21%), nausea (26% vs 21%), peripheral oedema (25% vs 18%), vomiting (22% vs 11%) and back pain (21% vs 16%). The scientific review concluded that the gain in PFS of 5.9 months observed with ixazomib was considered clinically meaningful. Concerning the possible uncertainty about the magnitude of the effect, this uncertainty was acceptable given the favourable toxicity profile, and considering that ixazomib is the first agent to allow oral triple combination therapy in this patient population which represents a therapeutic innovation in terms of convenience for patients. Therefore, the benefit-risk for ixazomib in combination with lenalidomide and dexamethasone was considered positive, although the efficacy evidence was not as comprehensive as normally required.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniela Melchiorri
- Dip. Physiology and Pharmacology, V. Erspamer, University of Rome La Sapienza, Roma, Italy
| | | | | | | | | | | | - Jonas Bergh
- Radiumhemmet Microbiology and Tumorbiology Center, Karolinska University Hospital, Stockholm, Sweden
| | - Christian Gisselbrecht
- Institut d'Hématologie, Hôpital Saint Louis Paris Diderot Université, Hospital Saint-Louis, Paris, France
| | | |
Collapse
|
75
|
Dhakal B, D'Souza A, Hamadani M, Arce-Lara C, Schroeder K, Chhabra S, Shah NN, Gauger K, Keaton T, Pasquini M, Hari P. Phase I/II trial of bendamustine, ixazomib, and dexamethasone in relapsed/refractory multiple myeloma. Blood Cancer J 2019; 9:56. [PMID: 31358733 PMCID: PMC6663939 DOI: 10.1038/s41408-019-0219-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/22/2019] [Indexed: 01/08/2023] Open
Abstract
In this phase I/II trial, BID, bendamustine (70, 80, or 90 mg/m2), ixazomib (4 mg), and dexamethasone (40 mg), was administered to 28 patients with relapsed and/or refractory multiple myeloma (RRMM) exposed to bortezomib and lenalidomide and refractory to at least one. A 3 + 3 dose escalation based on dose-limiting toxicities (DLTs) was employed in phase I (total 15); 2/6 patients developed DLTs (neutropenia and thrombocytopenia) at dose level 3 establishing the recommended phase II dose as bendamustine 80 mg/m2, ixazomib 4 mg, and dexamethasone 40 mg. The median age was 67 years (range, 42–72), and 43% were females. Patients received a median of 4 (range, 4–9) prior lines of therapy, of which ~50% were double refractory. In phase II, total 19 patients were treated. With a median follow-up of 17 months, 11% achieved very good partial response, 50% achieved partial response, and 27% achieved stable disease. Median progression free (PFS) and overall (OS) survival were 5.2 months (95% CI, 1.96–8.3) and 23.2 months (95% CI 16.3–30.07). The most frequent adverse events were anemia, thrombocytopenia, leukopenia, nausea, diarrhea, and infections. Peripheral neuropathy was infrequent. BID is a well-tolerated and effective combination therapy for patients with RRMM.
Collapse
Affiliation(s)
- Binod Dhakal
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Anita D'Souza
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mehdi Hamadani
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carlos Arce-Lara
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Katrina Schroeder
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nirav N Shah
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Katelyn Gauger
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Taylor Keaton
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marcelo Pasquini
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Parameswaran Hari
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
76
|
Baljevic M, Orlowski RZ. Pharmacodynamics and pharmacokinetics of proteasome inhibitors for the treatment of multiple myeloma. Expert Opin Drug Metab Toxicol 2019; 15:459-473. [PMID: 31104525 PMCID: PMC10393465 DOI: 10.1080/17425255.2019.1621839] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/17/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Multiple myeloma (MM) is the second most commonly diagnosed hematologic malignancy and has an increasing incidence and prevalence globally, and proteasome inhibitors (PIs) form the backbone of some of our most effective regimens for all phases of this disease in fit and frail patients. Areas covered: Strong understanding of the proteasome complex is increasingly important as the rapid development of new PIs and innovative myeloma therapies complicate the use of old and new combination regimens. We focus herein on the pharmacodynamics and pharmacokinetics of the approved PIs and others in development, including their safety and efficacy in corresponding clinical studies. Expert opinion: Advancements such as the first oral PI, ixazomib, with a more convenient route of administration and improved toxicity profile led to an improved quality of life, patient compliance, and all-oral combination regimens which are efficacious for long-term management of standard and high-risk MM. Novel pan-PIs, such as marizomib, hold the promise of superior clinical activity due to irreversible targeting of all multicatalytic proteinase complex subunits. Development of clinically validated biomarkers of PI sensitivity/resistance is required to inform utilization of the most optimal and effective, rationally targeted PI treatments for all MM patients.
Collapse
Affiliation(s)
- Muhamed Baljevic
- The University of Nebraska Medical Center, Division of Hematology and Oncology, Omaha, NE, USA
| | - Robert Z. Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
77
|
Varga C, Titus SE, Toskic D, Comenzo RL. Use of novel therapies in the treatment of light chain amyloidosis. Blood Rev 2019; 37:100581. [PMID: 31167719 DOI: 10.1016/j.blre.2019.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/26/2019] [Accepted: 05/20/2019] [Indexed: 12/22/2022]
Abstract
Immunoglobulin light-chain (AL) amyloidosis is a rare life-threatening disease caused by light chains that are toxic to vital organs such as the heart, kidneys, liver and peripheral nervous system, and that misfold and assemble as amyloid fibrils and deposit both in affected organs and systemically in the vasculature and other tissues. Patients afflicted by this disease have B-cell disorders, almost always related to clonal plasma cells in the bone marrow, the burden of which can range from small clones involving 5% or less of marrow cells to frank multiple myeloma. The goal of therapy is to eliminate the clonal plasma cells producing these toxic light chains to halt and possibly reverse symptomatic organ damage. While autologous stem cell transplantation can be a very effective treatment modality in AL, it has a limited role due to the frailty of this particular population. Conservative treatment in the form of chemotherapy has become the backbone of therapy. Bortezomib combined with alkylators has proven quite successful in inducing hematologic responses. However, despite these advances, tolerability and resistance continue to be an ongoing issue. Novel anti-plasma cell therapies such as ixazomib, carfilzomib, lenalidomide and pomalidomide are actively being combined and evaluated in clinical trials for efficacy and toxicity in this challenging patient population. Other approaches, such as monoclonal antibodies targeting surface proteins and amyloid deposits, are being tested and combined with novel agents. In this review, we will provide an overview of the clinical trials that have led to current treatment algorithms and will also discuss monoclonal antibodies currently under investigation and in various stages of clinical development.
Collapse
Affiliation(s)
- Cindy Varga
- The John Conant Davis Myeloma and Amyloid Program, Division of Hematology-Oncology, Tufts Medical Center, 800 Washington St., Boston, MA 02111, USA; Department of Medicine, Tufts Medical Center, 800 Washington St, Boston, MA 02111, USA.
| | - Stephanie E Titus
- Department of Medicine, Tufts Medical Center, 800 Washington St, Boston, MA 02111, USA.
| | - Denis Toskic
- The John Conant Davis Myeloma and Amyloid Program, Division of Hematology-Oncology, Tufts Medical Center, 800 Washington St., Boston, MA 02111, USA
| | - Raymond L Comenzo
- The John Conant Davis Myeloma and Amyloid Program, Division of Hematology-Oncology, Tufts Medical Center, 800 Washington St., Boston, MA 02111, USA; Department of Medicine, Tufts Medical Center, 800 Washington St, Boston, MA 02111, USA.
| |
Collapse
|
78
|
Dimopoulos MA, Laubach JP, Echeveste Gutierrez MA, Grzasko N, Hofmeister CC, San-Miguel JF, Kumar S, Labotka R, Lu V, Berg D, Byrne C, Teng Z, Liu G, van de Velde H, Richardson PG. Ixazomib maintenance therapy in newly diagnosed multiple myeloma: An integrated analysis of four phase I/II studies. Eur J Haematol 2019; 102:494-503. [PMID: 30943323 DOI: 10.1111/ejh.13231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/18/2019] [Accepted: 03/20/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To evaluate the safety and efficacy of maintenance therapy with the oral proteasome inhibitor ixazomib in patients with newly diagnosed multiple myeloma (NDMM) not undergoing transplantation. METHODS Data were pooled from four NDMM phase I/II studies; patients received induction therapy with once- or twice-weekly ixazomib plus lenalidomide-dexamethasone (IRd), melphalan-prednisone (IMP), or cyclophosphamide-dexamethasone (ICd), followed by single-agent ixazomib maintenance, given at the last tolerated dose during induction, until disease progression, death, or unacceptable toxicity. RESULTS A total of 121 patients achieved stable disease or better after induction (weekly IRd, n = 25; twice-weekly IRd, n = 18; weekly or twice-weekly IMP, n = 35; weekly ICd, n = 43) and received ≥ 1 dose of ixazomib maintenance. Grade ≥ 3 drug-related adverse events occurred in 24% of patients during maintenance; each event was reported in ≤2% of patients. Rates of complete response were 22% after induction and 35% after maintenance. A total of 28 patients (23%) improved their response during maintenance. CONCLUSIONS Ixazomib maintenance following ixazomib-based induction is associated with deepening of responses and a positive safety profile with no cumulative toxicity in patients with NDMM not undergoing transplantation, suggesting that ixazomib is feasible for long-term administration. Phase III investigation of ixazomib maintenance is ongoing.
Collapse
Affiliation(s)
- Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Norbert Grzasko
- Department of Hematology, St. John's Cancer Center and Medical University of Lublin, Lublin, Poland
| | - Craig C Hofmeister
- The Ohio State University, Columbus, Ohio.,Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Jesus F San-Miguel
- Clinica Universidad de Navarra, Centro Investigación Medica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | | | - Vickie Lu
- Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Deborah Berg
- Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | - Zhaoyang Teng
- Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Guohui Liu
- Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | | |
Collapse
|
79
|
Hypertension in hematologic malignancies and hematopoietic cell transplantation: An emerging issue with the introduction of novel treatments. Blood Rev 2019; 35:51-58. [DOI: 10.1016/j.blre.2019.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/29/2018] [Accepted: 03/12/2019] [Indexed: 12/12/2022]
|
80
|
Joshua DE, Bryant C, Dix C, Gibson J, Ho J. Biology and therapy of multiple myeloma. Med J Aust 2019; 210:375-380. [DOI: 10.5694/mja2.50129] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | | | - Joy Ho
- Royal Prince Alfred Hospital Sydney NSW
| |
Collapse
|
81
|
Smolewski P, Rydygier D. Ixazomib: an investigational drug for the treatment of lymphoproliferative disorders. Expert Opin Investig Drugs 2019; 28:421-433. [DOI: 10.1080/13543784.2019.1596258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Piotr Smolewski
- Department of Experimental Hematology, Medical University of Lodz, Lodz,
Poland
| | - Dominika Rydygier
- Department of Experimental Hematology, Medical University of Lodz, Lodz,
Poland
| |
Collapse
|
82
|
Gupta N, Hanley MJ, Xia C, Labotka R, Harvey RD, Venkatakrishnan K. Clinical Pharmacology of Ixazomib: The First Oral Proteasome Inhibitor. Clin Pharmacokinet 2019; 58:431-449. [PMID: 30117017 PMCID: PMC6397141 DOI: 10.1007/s40262-018-0702-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ixazomib, the first oral proteasome inhibitor, is approved in combination with lenalidomide and dexamethasone for the treatment of patients with multiple myeloma (MM) who have received at least one prior therapy. Ixazomib is a selective, potent, and reversible inhibitor of the 20S proteasome, and preferentially binds to and inhibits the β5 chymotrypsin-like proteolytic site. Ixazomib absorption is rapid, with a median time to reach maximum plasma concentration of approximately 1 h post-dose. Ixazomib pharmacokinetics (PK) are adequately described by a three-compartment model (terminal half-life of 9.5 days) with first-order linear absorption (oral bioavailability of 58%). Plasma exposures of ixazomib increase in a dose-proportional manner. A high-fat meal decreases both the rate and extent of ixazomib absorption, supporting administration on an empty stomach. Population PK analyses demonstrated that no dose adjustment is required based on age, body size/weight, race, sex, mild-to-moderate renal impairment, or mild hepatic impairment. Results from dedicated studies indicate that a reduced starting dose (from 4 to 3 mg) is appropriate for patients with severe renal impairment, end-stage renal disease requiring dialysis, or moderate-to-severe hepatic impairment. Non-cytochrome P450 (CYP)-mediated metabolism appears to be the major clearance mechanism for ixazomib. Drug-drug interaction studies have shown no meaningful effects of strong inhibitors of CYP3A on ixazomib PK; however, the strong inducer rifampin caused a clinically relevant reduction in ixazomib exposure, supporting the recommendation to avoid concomitant administration of ixazomib with strong CYP3A inducers. Exposure-response analyses of data from the phase III TOURMALINE-MM1 registrational study demonstrate a favorable benefit-risk profile for the approved dose and regimen of weekly ixazomib 4 mg on days 1, 8, and 15 of each 28-day cycle.
Collapse
Affiliation(s)
- Neeraj Gupta
- Millennium Pharmaceuticals, Inc., a Wholly Owned Subsidiary of Takeda Pharmaceutical Company Limited, 40 Landsdowne Street, Cambridge, MA, 02139, USA.
| | - Michael J Hanley
- Millennium Pharmaceuticals, Inc., a Wholly Owned Subsidiary of Takeda Pharmaceutical Company Limited, 40 Landsdowne Street, Cambridge, MA, 02139, USA
| | - Cindy Xia
- Millennium Pharmaceuticals, Inc., a Wholly Owned Subsidiary of Takeda Pharmaceutical Company Limited, 40 Landsdowne Street, Cambridge, MA, 02139, USA
| | - Richard Labotka
- Millennium Pharmaceuticals, Inc., a Wholly Owned Subsidiary of Takeda Pharmaceutical Company Limited, 40 Landsdowne Street, Cambridge, MA, 02139, USA
| | - R Donald Harvey
- Departments of Hematology and Medical Oncology and Pharmacology, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Karthik Venkatakrishnan
- Millennium Pharmaceuticals, Inc., a Wholly Owned Subsidiary of Takeda Pharmaceutical Company Limited, 40 Landsdowne Street, Cambridge, MA, 02139, USA
| |
Collapse
|
83
|
Gupta N, Hanley MJ, Diderichsen PM, Yang H, Ke A, Teng Z, Labotka R, Berg D, Patel C, Liu G, van de Velde H, Venkatakrishnan K. Model-Informed Drug Development for Ixazomib, an Oral Proteasome Inhibitor. Clin Pharmacol Ther 2019; 105:376-387. [PMID: 29446068 PMCID: PMC6585617 DOI: 10.1002/cpt.1047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/26/2018] [Accepted: 02/12/2018] [Indexed: 12/27/2022]
Abstract
Model-informed drug development (MIDD) was central to the development of the oral proteasome inhibitor ixazomib, facilitating internal decisions (switch from body surface area (BSA)-based to fixed dosing, inclusive phase III trials, portfolio prioritization of ixazomib-based combinations, phase III dose for maintenance treatment), regulatory review (model-informed QT analysis, benefit-risk of 4 mg dose), and product labeling (absolute bioavailability and intrinsic/extrinsic factors). This review discusses the impact of MIDD in enabling patient-centric therapeutic optimization during the development of ixazomib.
Collapse
Affiliation(s)
- Neeraj Gupta
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Michael J. Hanley
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | | | - Huyuan Yang
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Alice Ke
- Certara USA, Inc.PrincetonNew JerseyUSA
| | - Zhaoyang Teng
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Richard Labotka
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Deborah Berg
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Chirag Patel
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Guohui Liu
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Helgi van de Velde
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| | - Karthik Venkatakrishnan
- Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited
| |
Collapse
|
84
|
Kumar SK, Berdeja JG, Niesvizky R, Lonial S, Laubach JP, Hamadani M, Stewart AK, Hari P, Roy V, Vescio R, Kaufman JL, Berg D, Liao E, Rajkumar SV, Richardson PG. Ixazomib, lenalidomide, and dexamethasone in patients with newly diagnosed multiple myeloma: long-term follow-up including ixazomib maintenance. Leukemia 2019; 33:1736-1746. [PMID: 30696949 PMCID: PMC6755968 DOI: 10.1038/s41375-019-0384-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/14/2018] [Accepted: 12/24/2018] [Indexed: 11/10/2022]
Abstract
Triplet combinations containing a proteasome inhibitor are a standard of care in newly diagnosed multiple myeloma (NDMM). We examined the long-term efficacy and safety of the all-oral combination of weekly ixazomib plus lenalidomide-dexamethasone (IRd), followed by single-agent ixazomib maintenance in NDMM patients. Of 65 enrolled patients, 53 received ixazomib 4 mg (days 1, 8, and 15) plus lenalidomide 25 mg (days 1–21) and dexamethasone 40 mg (days 1, 8, 15, and 22) for up to twelve 28-day induction cycles. Twenty-three patients discontinued induction for stem cell transplantation (SCT). In the remaining 42 patients, overall response rate was 80%, including 63% ≥very good partial response (VGPR) and 32% complete responses. At a median follow-up of 56 months, median progression-free survival (PFS) was 35.4 months in the total population. Twenty-five patients received ixazomib maintenance; eight deepened their response (76% ≥VGPR), and median PFS was 37.2 months in this subgroup. Nine of 42 patients who did not proceed to SCT (14% of total population) had an adverse event requiring discontinuation. Ixazomib (median ≥ 96%) and lenalidomide (median 88–94%) relative dose intensities were maintained throughout treatment. Weekly IRd, followed by ixazomib maintenance, was highly active with acceptable toxicity, enabling long-term administration with no evidence of cumulative toxicities.
Collapse
Affiliation(s)
| | | | - Ruben Niesvizky
- Myeloma Center, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Mehdi Hamadani
- West Virginia University, Mary Babb Randolph Cancer Center, Morgantown, WV, USA
| | | | - Parameswaran Hari
- Division of Hematology Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Robert Vescio
- Cedars-Sinai Outpatient Cancer Center at the Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Deborah Berg
- Millennium Pharmaceuticals Inc. (a wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, MA, USA
| | - Eileen Liao
- Millennium Pharmaceuticals Inc. (a wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, MA, USA.,Biogen Inc., Cambridge, MA, USA
| | | | | |
Collapse
|
85
|
Abstract
Multiple myeloma is diagnosed in over 100,000 patients each year worldwide, has an increasing incidence and prevalence in many regions, and follows a relapsing course, making it a significant and growing healthcare challenge. Recent basic, translational, and clinical studies have expanded our therapeutic armamentarium, which now consists of alkylating agents, corticosteroids, deacetylase inhibitors, immunomodulatory agents, monoclonal antibodies, and proteasome inhibitors. New drugs in these categories, and additional agents, including both small and large molecules, as well as cellular therapies, are under development that promise to further expand our capabilities and bring us closer to the cure of this plasma cell dyscrasia.
Collapse
Affiliation(s)
- Chutima Kunacheewa
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Robert Z. Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
86
|
Xie J, Wan N, Liang Z, Zhang T, Jiang J. Ixazomib – the first oral proteasome inhibitor. Leuk Lymphoma 2019; 60:610-618. [DOI: 10.1080/10428194.2018.1523398] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jingmei Xie
- College of Pharmacy, Jinan University, Guangzhou, PR China
| | - Ning Wan
- Department of Pharmacy, Guangzhou General Hospital of Guangzhou Military Command, PR China
- Guangzhou Huabo Biopharmaceutical Research Institute, PR China
| | - Zhuoru Liang
- College of Pharmacy, Jinan University, Guangzhou, PR China
| | - Tiantian Zhang
- College of Pharmacy, Jinan University, Guangzhou, PR China
| | - Jie Jiang
- College of Pharmacy, Jinan University, Guangzhou, PR China
- Dongguan Institute of Jinan University, Dongguan, PR China
| |
Collapse
|
87
|
|
88
|
Fuchs O. Treatment of Lymphoid and Myeloid Malignancies by Immunomodulatory Drugs. Cardiovasc Hematol Disord Drug Targets 2019; 19:51-78. [PMID: 29788898 DOI: 10.2174/1871529x18666180522073855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/05/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Thalidomide and its derivatives (lenalidomide, pomalidomide, avadomide, iberdomide hydrochoride, CC-885 and CC-90009) form the family of immunomodulatory drugs (IMiDs). Lenalidomide (CC5013, Revlimid®) was approved by the US FDA and the EMA for the treatment of multiple myeloma (MM) patients, low or intermediate-1 risk transfusion-dependent myelodysplastic syndrome (MDS) with chromosome 5q deletion [del(5q)] and relapsed and/or refractory mantle cell lymphoma following bortezomib. Lenalidomide has also been studied in clinical trials and has shown promising activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). Lenalidomide has anti-inflammatory effects and inhibits angiogenesis. Pomalidomide (CC4047, Imnovid® [EU], Pomalyst® [USA]) was approved for advanced MM insensitive to bortezomib and lenalidomide. Other IMiDs are in phases 1 and 2 of clinical trials. Cereblon (CRBN) seems to have an important role in IMiDs action in both lymphoid and myeloid hematological malignancies. Cereblon acts as the substrate receptor of a cullin-4 really interesting new gene (RING) E3 ubiquitin ligase CRL4CRBN. This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1α (CK1α) and marks them for degradation in proteasomes. Both these transcription factors (IKZF1 and IKZF3) stimulate proliferation of MM cells and inhibit T cells. Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Argonaute-2 seems to be an important drug target against IMiDs resistance in MM cells. Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. The CK1α gene (CSNK1A1) is located on 5q32 in commonly deleted region (CDR) in del(5q) MDS. Inhibition of CK1α sensitizes del(5q) MDS cells to lenalidomide. CK1α mediates also survival of malignant plasma cells in MM. Though, inhibition of CK1α is a potential novel therapy not only in del(5q) MDS but also in MM. High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. While transfusion independence (TI) after lenalidomide treatment is more than 60% in MDS patients with del(5q), only 25% TI and substantially shorter duration of response with occurrence of neutropenia and thrombocytopenia were achieved in lower risk MDS patients with normal karyotype treated with lenalidomide. Shortage of the biomarkers for lenalidomide response in these MDS patients is the main problem up to now.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| |
Collapse
|
89
|
Alahmadi M, Masih-Khan E, Atenafu EG, Chen C, Kukreti V, Tiedemann R, Trudel S, Reece DE. Addition of Cyclophosphamide "On Demand" to Lenalidomide and Corticosteroids in Patients With Relapsed/Refractory Multiple Myeloma-A Retrospective Review of a Single-center Experience. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 19:e195-e203. [PMID: 30723035 DOI: 10.1016/j.clml.2018.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/30/2018] [Accepted: 12/12/2018] [Indexed: 10/27/2022]
Abstract
INTRODUCTION The combination of lenalidomide and dexamethasone (Len-Dex) is an established regimen for patients with relapsed or refractory myeloma. To prolong the benefit of this effective regimen, the Myeloma Program at Princess Margaret Cancer Centre has routinely added a third agent, oral cyclophosphamide (Cy) given weekly, to Len-Dex at disease progression. PATIENTS AND METHODS In the present report, we describe the cases of 53 patients who had received Len-Dex-Cy for a minimum of 4 weeks from January 2007 to December 2014 after progression with Len-Dex alone. The dose of added Cy ranged from 250 to 500 mg weekly. The median number of previous regimens, including Len-Dex, was 2 (range, 2-4); 80% of patients had undergone previous autologous stem cell transplantation. RESULTS The overall rate of response equal to or greater than a partial response was 34%, and clinical benefit (stable disease or better) was observed in 87% of the patients. The median duration of Len-Dex-Cy therapy was 6.9 months (range, 0.9-55.1 months). The median progression-free survival was 6.1 months (range, 4.2-8.1 months) from the addition of Cy and 24.1 months (range, 15.9-32.0 months) from start of Len-Dex. CONCLUSION The addition of Cy for patients with myeloma developing progression with Len-Dex was an inexpensive option with manageable toxicity that resulted in a clinically meaningful extension of disease control.
Collapse
Affiliation(s)
- Majed Alahmadi
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Esther Masih-Khan
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Eshetu G Atenafu
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Christine Chen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Vishal Kukreti
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Rodger Tiedemann
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Suzanne Trudel
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Donna E Reece
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
90
|
Paner A, Okwuosa TM, Richardson KJ, Libby EN. Triplet therapies - the new standard of care for multiple myeloma: how to manage common toxicities. Expert Rev Hematol 2018; 11:957-973. [PMID: 30339769 DOI: 10.1080/17474086.2018.1538777] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Multiple three drug combination regimens have been approved for the treatment of multiple myeloma in the last few years. Triplets have become the new standard of care for transplant eligible and ineligible patients with newly diagnosed as well as relapsed multiple myeloma. Novel agents have a unique profile of side effects. The management of toxicities is important to maintain quality of life and maximize treatment duration and benefit. Areas covered: This article reviews efficacy data, incidence of key adverse events and provide recommendations and expert opinion regarding how to manage common toxicities in triplet therapies. Relevant publications and abstracts were searched in PubMed, ASH, ASCO and EHA meetings. Guidelines from IMWG, NCCN, ESMO and ASCO, published trial protocols and prescribing information were used to formulate recommendations for the management of toxicities. Expert commentary: Side effects are a critical factor guiding the selection of optimal chemotherapy regimens for multiple myeloma. The majority of toxicities encountered with triplet therapies are reversible and can be readily managed with supportive care and dose modifications.
Collapse
Affiliation(s)
- Agne Paner
- a Division of Hematology, Department of Internal Medicine , Rush University Medical Center , Chicago , IL , USA
| | - Tochukwu M Okwuosa
- b Division of Cardiology, Department of Internal Medicine , Rush University Medical Center , Chicago , IL , USA
| | - Kristin J Richardson
- a Division of Hematology, Department of Internal Medicine , Rush University Medical Center , Chicago , IL , USA
| | - Edward N Libby
- c Division of Medical Oncology, Department of Medicine , University of Washington , Seattle , WA , USA
| |
Collapse
|
91
|
Dimopoulos MA, Grosicki S, Jędrzejczak WW, Nahi H, Gruber A, Hansson M, Gupta N, Byrne C, Labotka R, Teng Z, Yang H, Grzasko N, Kumar S. All-oral ixazomib, cyclophosphamide, and dexamethasone for transplant-ineligible patients with newly diagnosed multiple myeloma. Eur J Cancer 2018; 106:89-98. [PMID: 30471652 DOI: 10.1016/j.ejca.2018.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/07/2018] [Accepted: 09/15/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Novel efficacious treatments with long-term tolerability are needed for transplant-ineligible, newly diagnosed multiple myeloma (NDMM) patients. This phase 2 study evaluated the safety and efficacy of all-oral ixazomib-cyclophosphamide-dexamethasone (ICd) followed by single-agent ixazomib maintenance. PATIENTS AND METHODS Patients were randomised (1:1) to receive 4.0 mg of ixazomib, 300 (Arm A) or 400 (Arm B) mg/m2 of cyclophosphamide (days 1, 8, and 15), and 40 mg of dexamethasone (days 1, 8, 15, and 22) as induction (up to 13 × 28-day cycles), followed by single-agent ixazomib maintenance (28-day cycles) until progressive disease, death, or unacceptable toxicity. Primary end-point was complete response (CR) + very good partial response (VGPR) rate for ICd induction. RESULTS Seventy patients were enrolled (n = 36 Arm A; n = 34 Arm B); median age was 73 years (range, 61-87). At data cut-off, 66% of patients had completed 13 induction cycles followed by ixazomib maintenance. Median overall treatment duration was 19 cycles (range, 1-29); 21% of patients discontinued treatment during induction and 3% during maintenance due to adverse events (AEs). During induction, among 67 response-evaluable patients, CR+VGPR rate was 25%, and overall response rate (ORR) was 73%. Including the maintenance phase, CR+VGPR rate was 33%, and ORR was 76%. Median progression-free survival was 23.5 months (median follow-up: 26.1 months). The most common all-grade AE was neutropenia (31%). Grade ≥3 AEs were reported by 73% of patients. Five on-study deaths occurred (not treatment-related). CONCLUSIONS ICd treatment followed by ixazomib maintenance is tolerable and active in elderly, transplant-ineligible NDMM patients. TRIAL REGISTRATION NUMBER NCT02046070.
Collapse
Affiliation(s)
- Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | - Sebastian Grosicki
- Department of Cancer Prevention, Silesian Medical University, Katowice, Poland.
| | - Wiesław W Jędrzejczak
- Department of Haematology and Oncology, Medical University of Warsaw, Warsaw, Poland.
| | - Hareth Nahi
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Astrid Gruber
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Markus Hansson
- Hematology Clinic, Skåne University Hospital, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| | - Neeraj Gupta
- Millennium Pharmaceuticals Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA.
| | - Catriona Byrne
- Millennium Pharmaceuticals Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA.
| | - Richard Labotka
- Millennium Pharmaceuticals Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA.
| | - Zhaoyang Teng
- Millennium Pharmaceuticals Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA.
| | - Huyuan Yang
- Millennium Pharmaceuticals Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA.
| | - Norbert Grzasko
- Department of Haematology, St. John's Cancer Centre, Lublin, Poland; Department of Experimental Haemato-oncology, Medical University of Lublin, Lublin, Poland(1).
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
92
|
Kumar SK, Grzasko N, Delimpasi S, Jedrzejczak WW, Grosicki S, Kyrtsonis MC, Spencer A, Gupta N, Teng Z, Byrne C, Labotka R, Dimopoulos MA. Phase 2 study of all-oral ixazomib, cyclophosphamide and low-dose dexamethasone for relapsed/refractory multiple myeloma. Br J Haematol 2018; 184:536-546. [PMID: 30460684 DOI: 10.1111/bjh.15679] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
Abstract
There is a need for efficacious, convenient treatments with long-term tolerability for patients with relapsed/refractory multiple myeloma (RRMM). This phase 2 study evaluated the all-oral combination of ixazomib, cyclophosphamide and dexamethasone (ICd). Patients with RRMM received ixazomib 4 mg and cyclophosphamide 300 mg/m2 on days 1, 8 and 15, and dexamethasone 40 mg on days 1, 8, 15 and 22 in 28-day cycles. The primary endpoint was overall response rate (ORR). Seventy-eight patients were enrolled (median age 63·5 years). At data cut-off, patients had received a median of 12 treatment cycles; 31% remained on treatment. ORR was 48% [16% very good partial response or better (≥VGPR)]. ORR was 64% and 32% in patients aged ≥65 and <65 years (25% and 16% ≥VGPR), respectively. At a median follow-up of 15·2 months, median progression-free survival (PFS) was 14·2 months, with a trend towards better PFS in patients aged ≥65 years vs. <65 years (median 18·7 months vs. 12·0 months; hazard ratio 0·62, P = 0·14). ICd was well tolerated. The most common treatment-emergent adverse events were diarrhoea (33%), nausea (24%), upper respiratory tract infection (24%), and thrombocytopenia (22%); 10 patients (13%) had peripheral neuropathy (one grade 3). This study is registered at ClinicalTrials.gov (NCT02046070).
Collapse
Affiliation(s)
- Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Norbert Grzasko
- Department of Haematology, St. John's Cancer Centre, Lublin, Poland.,Department of Experimental Haemato-oncology, Medical University of Lublin, Lublin, Poland
| | - Sosana Delimpasi
- Department of Haematology, Evangelismos Hospital, Athens, Greece
| | - Wieslaw W Jedrzejczak
- Department of Haematology and Oncology, Medical University of Warsaw, MTZ Clinical Research, Warsaw, Poland
| | - Sebastian Grosicki
- Department of Cancer Prevention, Silesian Medical University, Katowice, Poland
| | - Marie-Christine Kyrtsonis
- Haematology Section - 1st Department of Propaedeutic Internal Medicine, Laikon University Hospital, Athens, Greece
| | - Andrew Spencer
- Malignant Haematology and Stem Cell Transplantation Service, The Alfred Hospital/MONASH University/Australian Centre for Blood Diseases, Melbourne, Australia
| | - Neeraj Gupta
- Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Zhaoyang Teng
- Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | | | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
93
|
Guerrero-Garcia TA, Gandolfi S, Laubach JP, Hideshima T, Chauhan D, Mitsiades C, Anderson KC, Richardson PG. The power of proteasome inhibition in multiple myeloma. Expert Rev Proteomics 2018; 15:1033-1052. [PMID: 30427223 DOI: 10.1080/14789450.2018.1543595] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Proteasome inhibitors (PIs) are therapeutic backbones of multiple myeloma treatment, with PI-based therapies being standards of care throughout the treatment algorithm. Proteasome inhibition affects multiple critical signaling pathways in myeloma cells and interacts synergistically with mechanisms of action of other conventional and novel agents, resulting in substantial anti-myeloma activity and at least additive effects. Areas covered: This review summarizes the biologic effects of proteasome inhibition in myeloma and provides an overview of the importance of proteasome inhibition to the current treatment algorithm. It reviews key clinical data on three PIs, specifically bortezomib, carfilzomib, and ixazomib; assesses ongoing phase 3 trials with these agents; and looks ahead to the increasingly broad role of both approved PIs and PIs under investigation in the frontline and relapsed settings. Expert commentary: Progress to date with PIs in multiple myeloma has been impressive, but there remain unmet needs and challenges, as well as increasing opportunities to optimize the use of these agents. Understanding discrepancies between PIs in terms of efficacy and safety profile is a key goal of ongoing research, along with proteomics-based efforts to identify potential biomarkers of sensitivity and resistance, thereby enabling increasingly personalized treatment approaches in the future.
Collapse
Affiliation(s)
| | - Sara Gandolfi
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Jacob P Laubach
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Teru Hideshima
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | | | | | | | - Paul G Richardson
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| |
Collapse
|
94
|
Richardson PG, Zweegman S, O’Donnell EK, Laubach JP, Raje N, Voorhees P, Ferrari RH, Skacel T, Kumar SK, Lonial S. Ixazomib for the treatment of multiple myeloma. Expert Opin Pharmacother 2018; 19:1949-1968. [DOI: 10.1080/14656566.2018.1528229] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Paul G. Richardson
- Division of Hematologic Malignancy, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Jacob P. Laubach
- Division of Hematologic Malignancy, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Noopur Raje
- Department of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Peter Voorhees
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Renda H. Ferrari
- Global Medical Affairs, Millennium Pharmaceuticals, Inc, Cambridge, MA, USA
| | - Tomas Skacel
- Global Medical Affairs, Millennium Pharmaceuticals, Inc, Cambridge, MA, USA
| | | | - Sagar Lonial
- Hematology & Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
95
|
Xie SC, Gillett DL, Spillman NJ, Tsu C, Luth MR, Ottilie S, Duffy S, Gould AE, Hales P, Seager BA, Charron CL, Bruzzese F, Yang X, Zhao X, Huang SC, Hutton CA, Burrows JN, Winzeler EA, Avery VM, Dick LR, Tilley L. Target Validation and Identification of Novel Boronate Inhibitors of the Plasmodium falciparum Proteasome. J Med Chem 2018; 61:10053-10066. [PMID: 30373366 PMCID: PMC6257627 DOI: 10.1021/acs.jmedchem.8b01161] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
The Plasmodium proteasome
represents a potential
antimalarial drug target for compounds with activity against multiple
life cycle stages. We screened a library of human proteasome inhibitors
(peptidyl boronic acids) and compared activities against purified P. falciparum and human 20S proteasomes. We chose four hits
that potently inhibit parasite growth and show a range of selectivities
for inhibition of the growth of P. falciparum compared
with human cell lines. P. falciparum was selected
for resistance in vitro to the clinically used
proteasome inhibitor, bortezomib, and whole genome sequencing was
applied to identify mutations in the proteasome β5 subunit.
Active site profiling revealed inhibitor features that enable retention
of potent activity against the bortezomib-resistant line. Substrate
profiling reveals P. falciparum 20S proteasome active
site preferences that will inform attempts to design more selective
inhibitors. This work provides a starting point for the identification
of antimalarial drug leads that selectively target the P.
falciparum proteasome.
Collapse
Affiliation(s)
- Stanley C Xie
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Melbourne , VIC 3010 , Australia
| | - David L Gillett
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Melbourne , VIC 3010 , Australia
| | - Natalie J Spillman
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Melbourne , VIC 3010 , Australia
| | - Christopher Tsu
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Madeline R Luth
- Host-Microbe Systems and Therapeutics Division , UC San Diego School of Medicine , La Jolla , California 92093 , United States
| | - Sabine Ottilie
- Host-Microbe Systems and Therapeutics Division , UC San Diego School of Medicine , La Jolla , California 92093 , United States
| | - Sandra Duffy
- Griffith Institute for Drug Discovery , Griffith University , Brisbane , QLD 4111 , Australia
| | - Alexandra E Gould
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Paul Hales
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Benjamin A Seager
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Melbourne , VIC 3010 , Australia
| | - Carlie L Charron
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Melbourne , VIC 3010 , Australia
| | - Frank Bruzzese
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Xiaofeng Yang
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Xiansi Zhao
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Shih-Chung Huang
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Craig A Hutton
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Melbourne , VIC 3010 , Australia
| | | | - Elizabeth A Winzeler
- Host-Microbe Systems and Therapeutics Division , UC San Diego School of Medicine , La Jolla , California 92093 , United States
| | - Vicky M Avery
- Griffith Institute for Drug Discovery , Griffith University , Brisbane , QLD 4111 , Australia
| | - Lawrence R Dick
- Oncology Clinical R&D , Takeda Pharmaceuticals International Co. , Cambridge , Massachusetts 02139 , United States
| | - Leann Tilley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Melbourne , VIC 3010 , Australia
| |
Collapse
|
96
|
Zheleznyak A, Shokeen M, Achilefu S. Nanotherapeutics for multiple myeloma. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1526. [PMID: 29701006 PMCID: PMC6185771 DOI: 10.1002/wnan.1526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/19/2018] [Accepted: 03/23/2018] [Indexed: 01/11/2023]
Abstract
Multiple myeloma (MM) is an age-related hematological malignancy with an estimated 30,000 new cases and 13,000 deaths per year. A disease of antibody-secreting malignant plasma B-cells that grow primarily in the bone marrow (BM), MM causes debilitating fractures, anemia, renal failure, and hypercalcemia. In addition to the abnormal genetic profile of MM cells, the permissive BM microenvironment (BMM) supports MM pathogenesis. Although advances in treatment options have significantly enhanced survival in MM patients, transient perfusion of small-molecule drugs in the BM does not provide sufficient residence to enhance MM cell-drug interaction, thus allowing some myeloma cells to escape the first line of treatment. As such, there remains a crucial need to develop advanced drug delivery systems that can navigate the complex BMM and effectively reach the myeloma cells. The high vascular density and spongy nature of bone structure suggest that nanoparticles (NPs) can serve as smart drug-delivery systems capable of extravasation and retention in various BM compartments to exert a durable therapeutic effect. In this focus article, we first summarize the pathophysiology of MM, emphasizing how the BM niche presents serious challenges for effective treatment of MM with small-molecule drugs. We then pivot to current efforts to develop NP-based drug carriers and intrinsically therapeutic nanotherapeutics. The article concludes with a brief perspective on the opportunities and challenges in developing and translating nanotherapeutics to improve the treatment outcomes of MM patients. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Monica Shokeen
- Departments of Radiology, and Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Samuel Achilefu
- Departments of Radiology, Biomedical Engineering, and Biochemistry & Molecular Biophysics, Washington University, St. Louis, MO, USA
| |
Collapse
|
97
|
Okayama M, Kitabatake S, Sato M, Fujimori K, Ichikawa D, Matsushita M, Suto Y, Iwasaki G, Yamada T, Kiuchi F, Hirao M, Kunieda H, Osada M, Okamoto S, Hattori Y. A novel derivative (GTN024) from a natural product, komaroviquinone, induced the apoptosis of high-risk myeloma cells via reactive oxygen production and ER stress. Biochem Biophys Res Commun 2018; 505:787-793. [DOI: 10.1016/j.bbrc.2018.09.177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/28/2018] [Indexed: 01/16/2023]
|
98
|
Yue D, Sun X. Ixazomib promotes CHOP-dependent DR5 induction and apoptosis in colorectal cancer cells. Cancer Biol Ther 2018; 20:284-294. [PMID: 30359552 DOI: 10.1080/15384047.2018.1529095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ixazomib (Ninlaro), a novel proteasome inhibitor, has been developed for the treatment of many cancers and has demonstrated anti-tumor efficacy against various malignancies. However, the mechanism of the anti-tumor effect of ixazomib in colorectal cancer (CRC) cells remains unclear. METHODS MTS and flow cytometry were performed to determine the effect of ixazomib on CRC cells. Western blotting and real-time RT-PCR were performed to detect ixazomib-induced DR5 upregulation. ChIP was performed to detect CHOP binding to DR5 promoter. Finally, xenograft experiments were carried out to measure the antitumor effect of ixazomib in vivo. RESULTS In this study, we revealed the mechanism by which ixazomib inhibits the growth of CRC cells. Our findings indicated that ixazomib treatment induces CHOP-dependent DR5 induction, irrespective of p53 status. Furthermore, DR5 is necessary for ixazomib-mediated apoptosis. Ixazomib also synergized with TRAIL to induce marked apoptosis via DR5 in CRC cells. CONCLUSIONS Our findings further suggested that ixazomib sensitizes TRAIL/death receptor signaling pathway-targeted CRC and suggested that DR5 induction could be a valuable indicator of ixazomib sensitivity.
Collapse
Affiliation(s)
- Dan Yue
- a Department of Laboratory Medicine , ShengJing Hospital of China Medical University , Shenyang , China
| | - Xun Sun
- b Department of Immunology , China Medical University , Shenyang , China
| |
Collapse
|
99
|
|
100
|
Multiple myeloma: 2018 update on diagnosis, risk‐stratification, and management. Am J Hematol 2018; 93:981-1114. [PMID: 30400719 PMCID: PMC6223128 DOI: 10.1002/ajh.25117] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/22/2022]
Abstract
Disease overview Multiple myeloma accounts for approximately 10% of hematologic malignancies. Diagnosis The diagnosis requires ≥10% clonal bone marrow plasma cells or a biopsy proven plasmacytoma plus evidence of one or more multiple myeloma defining events (MDE): CRAB (hyperc alcemia, r enal failure, a nemia, or lytic b one lesions) features felt related to the plasma cell disorder, bone marrow clonal plasmacytosis ≥60%, serum involved/uninvolved free light chain (FLC) ratio ≥100 (provided involved FLC is ≥100 mg/L), or >1 focal lesion on magnetic resonance imaging. Risk stratification Patients with del(17p), t(14;16), and t(14;20) have high-risk multiple myeloma. Patients with t(4;14) translocation and gain(1q) have intermediate-risk. All others are considered standard-risk. Risk-adapted initial therapy Initial treatment consists of bortezomib, lenalidomide, dexamethasone (VRd). In high-risk patients, carfilzomib, lenalidomide, dexamethasone (KRd) is an alternative to VRd. In eligible patients, initial therapy is given for approximately 3–4 cycles followed by autologous stem cell transplantation (ASCT). Standard risk patients can opt for delayed ASCT at first relapse. Patients not candidates for transplant are treated with VRd for approximately 8–12 cycles followed by lenalidomide or lenalidomide plus dexamethasone. Maintenance therapy After ASCT, lenalidomide maintenance is recommended for standard risk patients, while maintenance with a bortezomib-based regimen is needed for patients with intermediate or high-risk disease. Management of refractory disease Most patients require a triplet regimen at relapse, with the choice of regimen varying with each successive relapse. Aggressive relapse with extramedullary plasmacytomas or plasma cell leukemia may require anthracycline containing combination chemotherapy regimens.
Collapse
|