1
|
Warren KE, Vezina G, Krailo M, Springer L, Buxton A, Peer CJ, Figg WD, William-Hughes C, Kessel S, Fouladi M, Gajjar A, Bowers D. Phase II Randomized Trial of Lenalidomide in Children With Pilocytic Astrocytomas and Optic Pathway Gliomas: A Report From the Children's Oncology Group. J Clin Oncol 2023; 41:3374-3383. [PMID: 37126770 PMCID: PMC10414716 DOI: 10.1200/jco.22.01777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 03/06/2023] [Accepted: 03/15/2023] [Indexed: 05/03/2023] Open
Abstract
PURPOSE Children with low-grade glioma often require long-term therapy and suffer from treatment morbidity. Although targeted agents are promising, tumor targets often encompass normal developmental pathways and long-term effects of inhibition are unknown. Lenalidomide is an immunomodulatory agent with wide-ranging properties. Phase I studies indicated greater tolerability of lenalidomide in children compared with adults and a potential dose-response effect. PATIENTS AND METHODS We performed a phase II trial of lenalidomide in children with pilocytic astrocytomas and optic pathway gliomas who failed initial therapy. Primary objectives included determination of objective response rate of children randomly assigned to regimen A, low-dose (20 mg/m2/dose), or regimen B, high-dose (115 mg/m2/dose) lenalidomide, and assessment for early progression. Secondary objectives included estimation of event-free survival, overall survival, incidence of toxic events, and assessment of plasma lenalidomide concentrations. Lenalidomide was administered once daily × 21 days of each 28-day cycle for each regimen. RESULTS Seventy-four eligible patients were enrolled (n = 37, each arm). The predefined activity level of interest was achieved for both arms. Four objective responses were observed in each arm, and the number of early progressors was low. Eighteen patients completed 26 cycles of therapy (regimen A, n = 12; regimen B, n = 6). The median number of cycles was 14 (range, 2-26) for regimen A and 11 for regimen B (range, 1-26). Of 74 eligible patients who received study drug, 30 required dose reduction for toxicity (regimen A, n = 6; regimen B, n = 24) and 16 discontinued because of toxicity (regimen A, n = 2; regimen B, n = 14). CONCLUSION Lenalidomide demonstrates a sufficient level of activity in children with low-grade glioma to warrant further exploration. Low-dose (20 mg/m2/dose administered once daily × 21 days of each 28-day cycle) lenalidomide appears to have better tolerability with comparable activity.
Collapse
Affiliation(s)
| | | | - Mark Krailo
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA
| | | | - Allen Buxton
- Statistics and Data Center, Children's Oncology Group, Monrovia, CA
| | - Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD
| | | | - Sandy Kessel
- Imaging and Radiation Oncology Core Rhode Island (IROC RI), Lincoln, RI
| | | | | | | |
Collapse
|
2
|
Mogollón P, Díaz-Tejedor A, Algarín EM, Paíno T, Garayoa M, Ocio EM. Biological Background of Resistance to Current Standards of Care in Multiple Myeloma. Cells 2019; 8:cells8111432. [PMID: 31766279 PMCID: PMC6912619 DOI: 10.3390/cells8111432] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/03/2019] [Accepted: 11/09/2019] [Indexed: 12/16/2022] Open
Abstract
A high priority problem in multiple myeloma (MM) management is the development of resistance to administered therapies, with most myeloma patients facing successively shorter periods of response and relapse. Herewith, we review the current knowledge on the mechanisms of resistance to the standard backbones in MM treatment: proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies (mAbs). In some cases, strategies to overcome resistance have been discerned, and an effort should be made to evaluate whether resensitization to these agents is feasible in the clinical setting. Additionally, at a time in which we are moving towards precision medicine in MM, it is equally important to identify reliable and accurate biomarkers of sensitivity/refractoriness to these main therapeutic agents with the goal of having more efficacious treatments and, if possible, prevent the development of relapse.
Collapse
Affiliation(s)
- Pedro Mogollón
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Andrea Díaz-Tejedor
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Esperanza M. Algarín
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Teresa Paíno
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Mercedes Garayoa
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Enrique M. Ocio
- Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, 39008 Santander, Spain
- Correspondence: ; Tel.: +34-942202520
| |
Collapse
|
3
|
Uematsu A, Kido K, Manabe E, Takeda H, Takahashi H, Hayashi M, Imai Y, Sawasaki T. DANFIN functions as an inhibitor of transcription factor NF-κB and potentiates the antitumor effect of bortezomib in multiple myeloma. Biochem Biophys Res Commun 2017; 495:2289-2295. [PMID: 29284118 DOI: 10.1016/j.bbrc.2017.12.142] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 12/22/2017] [Indexed: 01/06/2023]
Abstract
Nuclear factor-κB (NF-κB) proteins are transcription factors that play key roles in regulating most immune responses and cell death. Constitutively active NF-κB has been shown to exhibit chemoresistance by inducing anti-apoptosis in tumor cells. Multiple myeloma is known as a constitutive NF-κB activating disease, and the proteasome inhibitor bortezomib is used to treat multiple myeloma and mantle cell lymphoma. We demonstrate here that DANFIN (N,N'-bis-(2,4-dimethyl-phenyl)-ethane-1,2-diamine) functions as an inhibitor of the p65 family proteins and induces chemosensitization to bortezomib in multiple myeloma. DANFIN was found to be an inhibitor of interactions between p65 and IκBα without the inhibition of the DNA binding activity of the p65 protein. In addition, DANFIN affected the IκBα binding region in Rel Homology Domain (RHD) and suppressed the nuclear translocalization of the p65 protein in cells. Furthermore, in multiple myeloma cells, DANFIN suppressed the expression level of NF-κB target genes and induced apoptosis. The combination therapy of DANFIN with bortezomib dramatically enhanced the apoptosis of multiple myeloma cells and indicated a remarkable anti-tumor effect in a multiple-myeloma xenograft mouse model.
Collapse
Affiliation(s)
- Atsushi Uematsu
- Division of Cell-Free Sciences, Proteo-Science Center (PROS), Ehime University, Japan
| | - Kohki Kido
- Division of Cell-Free Sciences, Proteo-Science Center (PROS), Ehime University, Japan
| | - Erika Manabe
- Division of Cell-Free Sciences, Proteo-Science Center (PROS), Ehime University, Japan
| | - Hiroyuki Takeda
- Division of Proteo-Drug-Discovery Sciences, PROS, Ehime University, Japan
| | - Hirotaka Takahashi
- Division of Cell-Free Sciences, Proteo-Science Center (PROS), Ehime University, Japan
| | - Minoru Hayashi
- Department of Materials Science and Biotechnology, Graduate School of Science and Engineering, Ehime University, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, PROS, Ehime University, Japan
| | - Tatsuya Sawasaki
- Division of Cell-Free Sciences, Proteo-Science Center (PROS), Ehime University, Japan.
| |
Collapse
|
4
|
Iwasa M, Miura Y, Fujishiro A, Fujii S, Sugino N, Yoshioka S, Yokota A, Hishita T, Hirai H, Andoh A, Ichinohe T, Maekawa T. Bortezomib interferes with adhesion of B cell precursor acute lymphoblastic leukemia cells through SPARC up-regulation in human bone marrow mesenchymal stromal/stem cells. Int J Hematol 2017; 105:587-597. [DOI: 10.1007/s12185-016-2169-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022]
|
5
|
Kaygusuz I, Toptas T, Aydin F, Uzay A, Firatli-Tuglular T, Bayik M. Bortezomib in patients with renal impairment. ACTA ACUST UNITED AC 2014; 16:200-8. [PMID: 21756535 DOI: 10.1179/102453311x13025568941880] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Renal failure is a common manifestation of multiple myeloma (MM). Bortezomib is primarily metabolized by cytochrome p450 isoforms. It also has a cytochrome-independent metabolism by excretion through the bile and kidney. Based on our observations, we aimed to explore the efficacy and toxicity profiles of bortezomib in 56 patients with MM, 24 of which had moderate to severe renal failure. Overall response and complete response, as well as very good partial response rates, were comparable between patients with normal renal functions and renal impairment. The median overall survivals for patients with estimated glomerular filtration rates of <60 and ≥60 ml/minute were similar. Although there was a tendency for shorter overall survival along lower estimated glomerular filtration rates, this difference did not reach a statistical significance. Overall and severe adverse events, and dose modification and treatment discontinuation rates were higher in patients with renal impairment. Patients with renal failure had more thrombocytopenia and diarrhea. While thrombocytopenia was mild to moderate and manageable, diarrhea, which led to serious adverse events, was more severe in patients with renal failure who received bortezomib as monotherapy. Bortezomib appears to be active; however, when used alone, it may cause more frequent and severe adverse events in patients with MM and renal failure.
Collapse
Affiliation(s)
- Isik Kaygusuz
- Division of Hematology, Marmara University Hospital, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
6
|
Garcia-Gomez A, Quwaider D, Canavese M, Ocio EM, Tian Z, Blanco JF, Berger AJ, Ortiz-de-Solorzano C, Hernández-Iglesias T, Martens ACM, Groen RWJ, Mateo-Urdiales J, Fraile S, Galarraga M, Chauhan D, San Miguel JF, Raje N, Garayoa M. Preclinical activity of the oral proteasome inhibitor MLN9708 in Myeloma bone disease. Clin Cancer Res 2014; 20:1542-54. [PMID: 24486586 DOI: 10.1158/1078-0432.ccr-13-1657] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE MLN9708 (ixazomib citrate), which hydrolyzes to pharmacologically active MLN2238 (ixazomib), is a next-generation proteasome inhibitor with demonstrated preclinical and clinical antimyeloma activity, but yet with an unknown effect on myeloma bone disease. Here, we investigated its bone anabolic and antiresorptive effects in the myeloma setting and in comparison with bortezomib in preclinical models. EXPERIMENTAL DESIGN The in vitro effect of MLN2238 was tested on osteoclasts and osteoclast precursors from healthy donors and patients with myeloma, and on osteoprogenitors derived from bone marrow mesenchymal stem cells also from both origins. We used an in vivo model of bone marrow-disseminated human myeloma to evaluate MLN2238 antimyeloma and bone activities. RESULTS Clinically achievable concentrations of MLN2238 markedly inhibited in vitro osteoclastogenesis and osteoclast resorption; these effects involved blockade of RANKL (receptor activator of NF-κB ligand)-induced NF-κB activation, F-actin ring disruption, and diminished expression of αVβ3 integrin. A similar range of MLN2238 concentrations promoted in vitro osteoblastogenesis and osteoblast activity (even in osteoprogenitors from patients with myeloma), partly mediated by activation of TCF/β-catenin signaling and upregulation of the IRE1 component of the unfolded protein response. In a mouse model of bone marrow-disseminated human multiple myeloma, orally administered MLN2238 was equally effective as bortezomib to control tumor burden and also provided a marked benefit in associated bone disease (sustained by both bone anabolic and anticatabolic activities). CONCLUSION Given favorable data on pharmacologic properties and emerging clinical safety profile of MLN9708, it is conceivable that this proteasome inhibitor may achieve bone beneficial effects in addition to its antimyeloma activity in patients with myeloma.
Collapse
Affiliation(s)
- Antonio Garcia-Gomez
- Authors' Affiliations: Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC); Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León; Hospital Universitario de Salamanca-IBSAL, Salamanca; Laboratorio de Imagen del Cáncer, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona, Spain; MGH Cancer Center, Massachusetts General Hospital; Dana-Farber Cancer Institute, Harvard Medical School, Boston; Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts, USA; and Departments of Cell Biology and Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Hurchla MA, Garcia-Gomez A, Hornick MC, Ocio EM, Li A, Blanco JF, Collins L, Kirk CJ, Piwnica-Worms D, Vij R, Tomasson MH, Pandiella A, San Miguel JF, Garayoa M, Weilbaecher KN. The epoxyketone-based proteasome inhibitors carfilzomib and orally bioavailable oprozomib have anti-resorptive and bone-anabolic activity in addition to anti-myeloma effects. Leukemia 2012; 27:430-40. [PMID: 22763387 DOI: 10.1038/leu.2012.183] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Proteasome inhibitors (PIs), namely bortezomib, have become a cornerstone therapy for multiple myeloma (MM), potently reducing tumor burden and inhibiting pathologic bone destruction. In clinical trials, carfilzomib, a next generation epoxyketone-based irreversible PI, has exhibited potent anti-myeloma efficacy and decreased side effects compared with bortezomib. Carfilzomib and its orally bioavailable analog oprozomib, effectively decreased MM cell viability following continual or transient treatment mimicking in vivo pharmacokinetics. Interactions between myeloma cells and the bone marrow (BM) microenvironment augment the number and activity of bone-resorbing osteoclasts (OCs) while inhibiting bone-forming osteoblasts (OBs), resulting in increased tumor growth and osteolytic lesions. At clinically relevant concentrations, carfilzomib and oprozomib directly inhibited OC formation and bone resorption in vitro, while enhancing osteogenic differentiation and matrix mineralization. Accordingly, carfilzomib and oprozomib increased trabecular bone volume, decreased bone resorption and enhanced bone formation in non-tumor bearing mice. Finally, in mouse models of disseminated MM, the epoxyketone-based PIs decreased murine 5TGM1 and human RPMI-8226 tumor burden and prevented bone loss. These data demonstrate that, in addition to anti-myeloma properties, carfilzomib and oprozomib effectively shift the bone microenvironment from a catabolic to an anabolic state and, similar to bortezomib, may decrease skeletal complications of MM.
Collapse
Affiliation(s)
- M A Hurchla
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Campbell RA, Berenson JR. Animal models of multiple myeloma and their utility in drug discovery. ACTA ACUST UNITED AC 2012; Chapter 14:Unit 14.9. [PMID: 22294221 DOI: 10.1002/0471141755.ph1409s40] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To evaluate potential new therapies and targets for treating multiple myeloma (MM), reproducible, biologically relevant in vivo models are required. Preclinical in vivo models of human MM allow investigators to evaluate novel therapies alone and in combination and quickly translate these results to the clinic where patients directly benefit, whether in the form of a new clinical trial, new doses and schedules, or new drug combinations. Presented in this unit are protocols for generating and maintaining a human extramedullary MM tumor in mice. Additionally, the extramedullary tumor can be excised and digested into a single-cell suspension and the human MM cells injected into mice subcutaneously, intravenously, or intratibially. Once these tumors are generated, they can be used to evaluate novel anti-MM agents and other therapies.
Collapse
Affiliation(s)
- Richard A Campbell
- Institute for Myeloma & Bone Cancer Research, West Hollywood, California, USA
| | | |
Collapse
|
9
|
Warren KE, Goldman S, Pollack IF, Fangusaro J, Schaiquevich P, Stewart CF, Wallace D, Blaney SM, Packer R, Macdonald T, Jakacki R, Boyett JM, Kun LE. Phase I trial of lenalidomide in pediatric patients with recurrent, refractory, or progressive primary CNS tumors: Pediatric Brain Tumor Consortium study PBTC-018. J Clin Oncol 2010; 29:324-9. [PMID: 21149652 DOI: 10.1200/jco.2010.31.3601] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE A phase I trial of lenalidomide was performed in children with recurrent, refractory, or progressive primary CNS tumors to estimate the maximum-tolerated dose (MTD) and to describe the toxicity profile and pharmacokinetics. PATIENTS AND METHODS Lenalidomide was administered by mouth daily for 21 days, repeated every 28 days. The starting dose was 15 mg/m(2)/d orally, and the dose was escalated according to a modified continuous reassessment method. Correlative studies included pharmacokinetics obtained from consenting patients on course 1, day 1, and at steady-state (between days 7 and 21). RESULTS Fifty-one patients (median age, 10 years; range, 2 to 21 years) were enrolled. Forty-four patients were evaluable for dose finding, and 49 patients were evaluable for toxicity. The primary toxicity was myelosuppression, but the MTD was not defined because doses up to 116 mg/m(2)/d were well-tolerated during the dose-finding period. Two objective responses were observed (one in thalamic juvenile pilocytic astrocytoma and one in optic pathway glioma) at dose levels of 88 and 116 mg/m(2)/d. Twenty-three patients, representing all dose levels, received ≥ six cycles of therapy. Pharmacokinetic analysis demonstrated that the lenalidomide area under the concentration-time curve from 0 to 24 hours and maximum plasma concentration increased with dosage over the range studied. CONCLUSION Lenalidomide was tolerable in children with CNS tumors at doses of 116 mg/m(2)/d during the initial dose-finding period. The primary toxicity is myelosuppression. Antitumor activity, defined by both objective responses and long-term stable disease, was observed, primarily in patients with low-grade gliomas.
Collapse
Affiliation(s)
- Katherine E Warren
- National Cancer Institute, Pediatric Oncology Branch, Bldg 10 CRC, Rm 1-5750, Bethesda, MD 20892-1104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Ladetto M, Pagliano G, Ferrero S, Cavallo F, Drandi D, Santo L, Crippa C, De Rosa L, Pregno P, Grasso M, Liberati AM, Caravita T, Pisani F, Guglielmelli T, Callea V, Musto P, Cangialosi C, Passera R, Boccadoro M, Palumbo A. Major Tumor Shrinking and Persistent Molecular Remissions After Consolidation With Bortezomib, Thalidomide, and Dexamethasone in Patients With Autografted Myeloma. J Clin Oncol 2010; 28:2077-84. [PMID: 20308672 DOI: 10.1200/jco.2009.23.7172] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose We investigated the effect on minimal residual disease, by qualitative and real-time quantitative polymerase chain reaction (RQ-PCR), of a consolidation regimen that included bortezomib, thalidomide, and dexamethasone (VTD) in patients with multiple myeloma (MM) responding to autologous stem-cell transplantation (auto-SCT). Patients and Methods Patients achieving at least very good partial response who had an available molecular marker based on the immunoglobulin heavy-chain rearrangement received four courses of treatment every month: four infusions per month of bortezomib at 1.6 mg/m2, thalidomide at 200 mg/d, and dexamethasone at 20 mg/d on days 1 to 4, 8 to 11, and 15 to 18. Patients were studied with tumor-clone–specific primers by qualitative nested PCR and RQ-PCR. Results Of 39 patients enrolled, 31 received the four VTD courses. Immunofixation complete responses increased from 15% after auto-SCT to 49% after VTD. Molecular remissions (MRs) were 3% after auto-SCT and 18% after VTD. Median time to maximum response was 3.5 months. So far, no patient in MR has relapsed (median follow-up, 42 months). VTD consolidation induced an additional depletion of 4.14 natural logarithms of tumor burden by RQ-PCR. Patients with a tumor load less than the median value after VTD had outcomes better than those who had tumor loads above the median value after VTD (at median follow-up: progression-free survival, 100% v 57%; P < .001). Conclusion To the best of our knowledge, this study is the first to document the occurrence of persistent MRs in a proportion of MM patients treated without allogeneic transplantation. Moreover, the major reduction in tumor load recorded by RQ-PCR after VTD suggests that unprecedented levels of tumor cell reduction can be achieved in MM thanks to the new nonchemotherapeutic drugs.
Collapse
Affiliation(s)
- Marco Ladetto
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Gloria Pagliano
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Simone Ferrero
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Federica Cavallo
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Daniela Drandi
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Loredana Santo
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Claudia Crippa
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Luca De Rosa
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Patrizia Pregno
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Mariella Grasso
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Anna Marina Liberati
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Tommaso Caravita
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Francesco Pisani
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Tommasina Guglielmelli
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Vincenzo Callea
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Pellegrino Musto
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Clotilde Cangialosi
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Roberto Passera
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Mario Boccadoro
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| | - Antonio Palumbo
- From the Department of Experimental Medicine and Oncology, Division of Hematology, University of Torino, S. Giovanni Battista Hospital; Division of Nuclear Medicine, University of Torino, S. Giovanni Battista Hospital, Torino; Department of Hematology, Spedali Civili, Brescia; Hematology and Bone Marrow Transplantation Unit, San Camillo-Forlanini Hospital; Department of Hematology, S. Eugenio Hospital, Tor Vergata University; Department of Hematology, Regina Elena National Cancer Institute, Roma
| |
Collapse
|
11
|
Reske T, Fulciniti M, Munshi NC. Mechanism of action of immunomodulatory agents in multiple myeloma. Med Oncol 2010; 27 Suppl 1:S7-13. [PMID: 20405248 DOI: 10.1007/s12032-010-9527-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 03/31/2010] [Indexed: 12/17/2022]
Abstract
Immunomodulatory agents (IMiD's) have become an important drug category in the treatment of multiple myeloma. The agents have a complex mechanism of action that influence the microenvironment in the bone marrow. The microenvironment is an essential promotor of disease progression and therefore important in targeting the disease. The article reviews mechanism of action and essential pathways of IMiD's that are important in disease treatment.
Collapse
|
12
|
Lonial S. Future directions in immunomodulatory therapy. Med Oncol 2009; 27 Suppl 1:S62-6. [PMID: 20012563 DOI: 10.1007/s12032-009-9376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 11/19/2009] [Indexed: 11/30/2022]
Abstract
The role of immunomodulatory-based therapy with thalidomide or lenalidomide is clearly established in the management of patients with myeloma in all phases of their disease. Recent preclinical and clinical works have demonstrated that in addition to combination therapy with dexamethasone, there is significant activity when combined with the proteasome inhibitor bortezomib. More recent clinical studies have also demonstrated significant activity when combined with akt inhibitors, HDAC inhibitors, and even monoclonal antibodies. Further clinical development of immunomodulatory agents should continue to be based on preclinical rationale, which has resulted in a number of promising and clinically active combinations.
Collapse
Affiliation(s)
- Sagar Lonial
- Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
13
|
Costa A, Osório C, Dias S. MicroRNA expression profiling in bone marrow: implications in hematological malignancies. Biotechnol J 2009; 4:88-97. [PMID: 19156746 DOI: 10.1002/biot.200800194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNA (miRNA) have been recently attributed a crucial role in the control of gene expression in numerous physiological and pathological processes including growth, differentiation and even oncogenesis. Besides detailed mechanistic studies on their generation and function, there has been a great deal of interest in the study of miRNA as surrogate markers of disease. Numerous studies have attempted to define miRNA profiles as predictors of disease outcome, or for the classification/diagnosis of different pathologies. In the present review, we summarize the main studies describing the involvement of miRNA in bone marrow (BM) diseases and in normal BM function during hematopoiesis.
Collapse
Affiliation(s)
- Ana Costa
- Angiogenesis Laboratory, CIPM, Portuguese Institute of Oncology, Lisboa, Portugal
| | | | | |
Collapse
|
14
|
Abstract
Multiple myeloma is characterised by clonal proliferation of malignant plasma cells, and mounting evidence indicates that the bone marrow microenvironment of tumour cells has a pivotal role in myeloma pathogenesis. This knowledge has already expanded treatment options for patients with multiple myeloma. Prototypic drugs thalidomide, bortezomib, and lenalidomide have each been approved for the treatment of this disease by targeting both multiple myeloma cells and the bone marrow microenvironment. Although benefit was first shown in relapsed and refractory disease, improved overall response, duration of response, and progression-free and overall survival can be achieved when these drugs are part of first-line regimens. This treatment framework promises to improve outcome not only for patients with multiple myeloma, but also with other haematological malignancies and solid tumours.
Collapse
Affiliation(s)
- Marc S Raab
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
15
|
Sung B, Kunnumakkara AB, Sethi G, Anand P, Guha S, Aggarwal BB. Curcumin circumvents chemoresistance in vitro and potentiates the effect of thalidomide and bortezomib against human multiple myeloma in nude mice model. Mol Cancer Ther 2009; 8:959-70. [PMID: 19372569 PMCID: PMC2694943 DOI: 10.1158/1535-7163.mct-08-0905] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Curcumin (diferuloylmethane), a yellow pigment in turmeric, has been shown to inhibit the activation of nuclear factor-kappaB (NF-kappaB), a transcription factor closely linked to chemoresistance in multiple myeloma cells. Whether curcumin can overcome chemoresistance and enhance the activity of thalidomide and bortezomib, used to treat patients with multiple myeloma, was investigated in vitro and in xenograft model in nude mice. Our results show that curcumin inhibited the proliferation of human multiple myeloma cells regardless of their sensitivity to dexamethasone, doxorubicin, or melphalan. Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Furthermore, in a nude mice model, we found that curcumin potentiated the antitumor effects of bortezomib (P<0.001, vehicle versus bortezomib+curcumin; P<0.001, bortezomib versus bortezomib+curcumin), and this correlated with suppression of Ki-67 (P<0.001 versus control), CD31 (P<0.001 versus vehicle), and vascular endothelial growth factor (P<0.001 versus vehicle) expression. Collectively, our results suggest that curcumin overcomes chemoresistance and sensitizes multiple myeloma cells to thalidomide and bortezomib by down-regulating NF-kappaB and NF-kappaB-regulated gene products.
Collapse
Affiliation(s)
- Bokyung Sung
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Ajaikumar B. Kunnumakkara
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Gautam Sethi
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Preetha Anand
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Sushovan Guha
- Department of Gastrointestinal Medicine and Nutrition, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Bharat B. Aggarwal
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
16
|
Chen Z, Ricker JL, Malhotra PS, Nottingham L, Bagain L, Lee TL, Yeh NT, Van Waes C. Differential bortezomib sensitivity in head and neck cancer lines corresponds to proteasome, nuclear factor-kappaB and activator protein-1 related mechanisms. Mol Cancer Ther 2008; 7:1949-60. [PMID: 18645005 PMCID: PMC2743084 DOI: 10.1158/1535-7163.mct-07-2046] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Head and neck squamous cell carcinomas (HNSCC) exhibit constitutive activation of transcription factors nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1), which are modulated by the proteasome and promote resistance to cell death. HNSCC show variable sensitivity to the proteasome inhibitor bortezomib in vitro as well as in murine xenografts and patient tumors in vivo, and the mechanisms are not well understood. To address this question, the sensitivities of nine HNSCC cell lines to bortezomib were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, and the potential relationship between the sensitivity and bortezomib effects on biological processes was examined in HNSCC lines of differential bortezomib sensitivity. The most sensitive cell line (UM-SCC-11B) underwent cell death at 10(-9) mol/L in vitro and tumor regression at a maximally tolerated dose of bortezomib in a murine xenograft model. The differential sensitivity between UM-SCC-11A and UM-SCC-11B cells corresponded to differences in the extent of suppression of proteasome activity, ubiquitinated protein degradation, and NF-kappaB and AP-1 activation. Lower concentrations of bortezomib transiently increased NF-kappaB and sustained AP-1 activation in UM-SCC-11A cells. AP-1 reporter activity and cell density of UM-SCC-11A were suppressed when bortezomib was combined with c-Jun NH(2)-terminal kinase and p38 kinase pathways inhibitors. Thus, the differential sensitivities to bortezomib corresponded to dissimilar effects on the proteasome, NF-kappaB and AP-1 activities. Inhibition of c-Jun NH(2)-terminal kinase and p38 pathways blocked AP-1 activity and enhanced the antitumor effects. These findings revealed molecular mechanisms of bortezomib sensitivity and resistance, which are under development as biomarkers for clinical trials in patients with HNSCC.
Collapse
Affiliation(s)
- Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, 10/5D55, MSC-1419, Bethesda, MD 20892-1419, USA
| | | | | | | | | | | | | | | |
Collapse
|