101
|
Fouquet G, Bories C, Guidez S, Renaud L, Herbaux C, Javed S, Facon T, Leleu X. Pomalidomide for multiple myeloma. Expert Rev Hematol 2014; 7:719-31. [DOI: 10.1586/17474086.2014.966074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
102
|
Angiogenesis in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol Commun 2014; 2:84. [PMID: 25047180 PMCID: PMC4149233 DOI: 10.1186/s40478-014-0084-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/09/2014] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis, the formation of new vessels, is found in Multiple Sclerosis (MS) demyelinating lesions following Vascular Endothelial Growth Factor (VEGF) release and the production of several other angiogenic molecules. The increased energy demand of inflammatory cuffs and damaged neural cells explains the strong angiogenic response in plaques and surrounding white matter. An angiogenic response has also been documented in an experimental model of MS, experimental allergic encephalomyelitis (EAE), where blood–brain barrier disruption and vascular remodelling appeared in a pre-symptomatic disease phase. In both MS and EAE, VEGF acts as a pro-inflammatory factor in the early phase but its reduced responsivity in the late phase can disrupt neuroregenerative attempts, since VEGF naturally enhances neuron resistance to injury and regulates neural progenitor proliferation, migration, differentiation and oligodendrocyte precursor cell (OPC) survival and migration to demyelinated lesions. Angiogenesis, neurogenesis and oligodendroglia maturation are closely intertwined in the neurovascular niches of the subventricular zone, one of the preferential locations of inflammatory lesions in MS, and in all the other temporary vascular niches where the mutual fostering of angiogenesis and OPC maturation occurs. Angiogenesis, induced either by CNS inflammation or by hypoxic stimuli related to neurovascular uncoupling, appears to be ineffective in chronic MS due to a counterbalancing effect of vasoconstrictive mechanisms determined by the reduced axonal activity, astrocyte dysfunction, microglia secretion of free radical species and mitochondrial abnormalities. Thus, angiogenesis, that supplies several trophic factors, should be promoted in therapeutic neuroregeneration efforts to combat the progressive, degenerative phase of MS.
Collapse
|
103
|
Oriol A. Pomalidomide in the treatment of multiple myeloma and perspectives in other hematological malignancies. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Pomalidomide is an immunomodulatory drug recently approved by the US FDA and EMA for the treatment of refractory and relapsed multiple myeloma. Pomalidomide appears to be more potent and less toxic than thalidomide and lenalidomide and has proven to be effective in a proportion of patients already exposed and refractory to these agents. The approved combination of pomalidomide and weekly dexamethasone offers clinical benefit to at least one-third of patients with multiple myeloma that have failed both bortezomib and lenolidomide treatment. This review will focus on the preclinical data and clinical experience in this setting. Off-label use of pomalidomide, mainly its combination with other active drugs for multiple myeloma and the role of pomalidomide in other hematologic malignancies still remain to be explored in depth and are discussed briefly.
Collapse
|
104
|
Panousis K, Nikolaou VS, Tsaganos T, Lallos S, Giamarellos-Bourboulis EJ, Efstathopoulos N. Thalidomide prolongs survival after experimental musculoskeletal injury, through an effect on mononuclear apoptosis. J Surg Res 2014; 188:198-205. [PMID: 24361040 DOI: 10.1016/j.jss.2013.11.1104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/10/2013] [Accepted: 11/21/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND This study was conducted to investigate the effects of intravenous thalidomide administration in an experimental model of musculoskeletal trauma. We hypothesized that because thalidomide inhibits secretion of tumor necrosis factor alpha (TNF-α), survival of animals that received thalidomide would be significantly prolonged. MATERIAL AND METHODS After an open fracture of the right femur, 24 rabbits were randomly assigned to control and thalidomide groups. Intravenous therapy with thalidomide was started 30 min after fracture. Hemodynamic monitoring of all animals was performed for 4 h. Survival was recorded and bacterial growth in blood and organs was measured after animal death or sacrifice. Blood was sampled for TNF-α measurement and for isolation of peripheral blood mononuclear cells (PBMCs). Apoptosis of PBMCs was measured by flow cytometry. RESULTS Survival was significantly prolonged in the thalidomide group. Apoptosis of PBMCs was increased in the control group compared with the thalidomide group at 24 h. There were no differences in vital signs, blood and tissue cultures, and serum TNF-α concentration between the two groups. CONCLUSIONS Intravenous thalidomide prolonged survival in an experimental model of severe musculoskeletal injury in rabbits. Its mechanism of action did not involve TNF-α suppression but prevention of mononuclear apoptosis. In view of these promising results, further research is needed to clarify the immunomodulatory mechanism of action of thalidomide and its potential use for the management of severe trauma.
Collapse
Affiliation(s)
| | | | - Thomas Tsaganos
- 4th Department of Internal Medicine, University of Athens, Medical School, Greece
| | - Stergios Lallos
- 2nd Department of Orthopaedics, University of Athens, Medical School, Greece
| | | | | |
Collapse
|
105
|
Shortt J, Hsu A, Martin B, Doggett K, Matthews G, Doyle M, Ellul J, Jockel T, Andrews D, Hogg S, Reitsma A, Faulkner D, Bergsagel P, Chesi M, Heath J, Denny W, Thompson P, Neeson P, Ritchie D, McArthur G, Johnstone R. The Drug Vehicle and Solvent N-Methylpyrrolidone Is an Immunomodulator and Antimyeloma Compound. Cell Rep 2014; 7:1009-19. [DOI: 10.1016/j.celrep.2014.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/12/2014] [Accepted: 04/07/2014] [Indexed: 01/03/2023] Open
|
106
|
Godin AM, Araújo DP, Menezes RR, Brito AMS, Melo ISF, Coura GME, Soares DG, Bastos LFS, Amaral FA, Ribeiro LS, Boff D, Santos JRA, Santos DA, Teixeira MM, de Fátima Â, Machado RR, Coelho MM. Activities of 2-phthalimidethanol and 2-phthalimidethyl nitrate, phthalimide analogs devoid of the glutarimide moiety, in experimental models of inflammatory pain and edema. Pharmacol Biochem Behav 2014; 122:291-8. [PMID: 24780502 DOI: 10.1016/j.pbb.2014.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/08/2014] [Accepted: 04/18/2014] [Indexed: 12/17/2022]
Abstract
The reintroduction of thalidomide in the pharmacotherapy greatly stimulated the interest in the synthesis and pharmacological evaluation of phthalimide analogs with new and improved activities and also greater safety. In the present study, we evaluated the activities of two phthalimide analogs devoid of the glutarimide ring, namely 2-phthalimidethanol (PTD-OH) and 2-phthalimidethyl nitrate (PTD-NO), in experimental models of inflammatory pain and edema in male C57BL/6J mice. Intraplantar (i.pl.) injection of carrageenan (300 μg) induced mechanical allodynia and this response was inhibited by previous per os (p.o.) administration of PTD-OH and PTD-NO (750 mg/kg) and also by thalidomide (500 or 750 mg/kg). The edema induced by carrageenan was also inhibited by previous p.o. administration of PTD-OH (500 and 750 mg/kg) and PTD-NO (125, 250, 500 or 750 mg/kg), but not by thalidomide. Carrageenan increased tumor necrosis factor (TNF)-α and CXCL1 concentrations and also the number of neutrophils in the paw tissue. Previous p.o. administration of PTD-NO (500 mg/kg) reduced all the parameters, while PTD-OH (500 mg/kg) reduced only the accumulation of neutrophils. Thalidomide, on the other hand, was devoid of effect on these biochemical parameters. Plasma concentrations of nitrite were increased after p.o. administration of the phthalimide analog coupled to a NO donor, PTD-NO (500 mg/kg), but not after administration of PTD-OH or thalidomide. In conclusion, our results show that small molecules, structurally much simpler than thalidomide or many of its analogs under investigation, exhibit similar activities in experimental models of pain and inflammation. Finally, as there is evidence that the glutarimide moiety contributes to the teratogenic effect of many thalidomide analogs, our results indicate that phthalimide analogs devoid of this functional group could represent a new class of analgesic and anti-inflammatory candidates with potential greater safety.
Collapse
Affiliation(s)
- Adriana M Godin
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Débora P Araújo
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Raquel R Menezes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Ana Mercy S Brito
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Ivo S F Melo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Giovanna M E Coura
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Darly G Soares
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Leandro F S Bastos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Flávio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Lucas S Ribeiro
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Daiane Boff
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Julliana R A Santos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Daniel A Santos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Ângelo de Fátima
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Renes R Machado
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Márcio M Coelho
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Pampulha CEP 31270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
107
|
Goto M, Nogam R, Okano Y, Gidoh M, Yotsu R, Ishida Y, Kitajima S, Kai M, Ishii N, Ozaki M, Hatano K. [Guidelines for the treatment of Hansen's disease in Japan (third edition)]. ACTA ACUST UNITED AC 2014; 82:143-84. [PMID: 24579462 DOI: 10.5025/hansen.82.143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
ad hoc committee of Japanese Leprosy Association recommends revised standard treatment protocol of leprosy in Japan, which is a modification of World Health Organization's multidrug therapy (WHO/MDT, 2010). For paucibacillary (PB) leprosy, 6 months treatment by rifampicin and dapsone (MDT/PB) is enough. However, for high bacterial load multibacillary (MB) leprosy, 12 months treatment seems insufficient. Thus, (A) For MB with bacterial index (BI) > 3 before treatment, 2 years treatment by rifampicin, dapsone and clofazimine (MDT/MB) is necessary. When BI becomes negative and active lesion is lost within 2 years, no maintenance therapy is necessary. When BI is still positive, one year of MDT/MB is added (3 years in total), followed by maintenance therapy by dapsone and clofazimine until BI negativity and loss of active lesions. (B) For MB with BI < 3 or fresh MB (less than 6 months after the onset of the disease) with BI > 3, 1 year treatment by MDT/MB is necessary. When BI becomes negative and active lesion is lost within one year, no maintenance therapy is necessary. When BI is still positive or active lesion is remaining, additional therapy with MDT/MB for one more year is recommended. Brief summary of diagnosis, purpose of therapy, character of drugs, and prevention of deformity is also described.
Collapse
Affiliation(s)
| | | | | | | | - Rie Yotsu
- National Center for Global Health and Medicine
| | | | | | - Masanori Kai
- Leprosy Research Center, National Institute of Infectious Diseases
| | - Norihisa Ishii
- Leprosy Research Center, National Institute of Infectious Diseases
| | | | | | | | | |
Collapse
|
108
|
Chang X, Zhu Y, Shi C, Stewart AK. Mechanism of immunomodulatory drugs' action in the treatment of multiple myeloma. Acta Biochim Biophys Sin (Shanghai) 2014; 46:240-53. [PMID: 24374776 DOI: 10.1093/abbs/gmt142] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although immunomodulatory drugs (IMiDs), such as thalidomide, lenalidomide, and pomalidomide, are widely used in the treatment of multiple myeloma (MM), the molecular mechanism of IMiDs' action is largely unknown. In this review, we will summarize recent advances in the application of IMiDs in MM cancer treatment as well as their effects on immunomodulatory activities, anti-angiogenic activities, intervention of cell surface adhesion molecules between myeloma cells and bone marrow stromal cells, anti-inflammatory activities, anti-proliferation, pro-apoptotic effects, cell cycle arrest, and inhibition of cell migration and metastasis. In addition, the potential IMiDs' target protein, IMiDs' target protein's functional role, and the potential molecular mechanisms of IMiDs resistance will be discussed. We wish, by presentation of our naive discussion, that this review article will facilitate further investigation in these fields.
Collapse
Affiliation(s)
- Xiubao Chang
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | | | | | | |
Collapse
|
109
|
Raje N, Hideshima T, Anderson KC. Therapeutic use of immunomodulatory drugs in the treatment of multiple myeloma. Expert Rev Anticancer Ther 2014; 6:1239-47. [PMID: 17020458 DOI: 10.1586/14737140.6.9.1239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Immunomodulatory drugs, such as thalidomide, lenalidomide (Revlimid, CC-5013) and actimid (CC-4047), have a broad spectrum of activity and have shown remarkable responses in patients with multiple myeloma and related hematological diseases, such as myelodysplastic syndrome. They are currently being tested in other cancer types. This review will focus on the preclinical and clinical activity of thalidomide and its more potent immunomodulatory derivatives that are used to treat multiple myeloma. They represent a new class of antitumor agents that not only target the tumor cell directly, but also have significant activity within the bone marrow milieu. These agents have shown high responses in all phases of multiple myeloma, including the upfront setting, relapsed refractory stage and also as maintenance therapy for the disease. They have been used in combination with dexamethasone, chemotherapy and, more recently, with other novel agents, such as proteasome inhibitors. Thalidomide and lenalidomide in combination with dexamethasone have recently been approved by the US FDA for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Noopur Raje
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| | | | | |
Collapse
|
110
|
Mentlik James A, Cohen AD, Campbell KS. Combination immune therapies to enhance anti-tumor responses by NK cells. Front Immunol 2013; 4:481. [PMID: 24391651 PMCID: PMC3870292 DOI: 10.3389/fimmu.2013.00481] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/09/2013] [Indexed: 01/22/2023] Open
Abstract
Natural killer (NK) cells are critical innate immune lymphocytes capable of destroying virally infected or cancerous cells through targeted cytotoxicity and further assisting in the immune response by releasing inflammatory cytokines. NK cells are thought to contribute to the process of tumor killing by certain therapeutic monoclonal antibodies (mAb) by directing antibody-dependent cellular cytotoxicity (ADCC) through FcγRIIIA (CD16). Numerous therapeutic mAb have been developed that target distinct cancer-specific cell markers and may direct NK cell-mediated ADCC. Recent therapeutic approaches have combined some of these cancer-specific mAb with additional strategies to optimize NK cell cytotoxicity. These include agonistic mAb targeting NK cell activating receptors and mAbs blocking NK cell inhibitory receptors to enhance NK cell functions. Furthermore, several drugs that can potentiate NK cell cytotoxicity through other mechanisms are being used in combination with therapeutic mAb. In this review, we examine the mechanisms employed by several promising agents used in combination therapies that enhance natural or Ab-dependent cytotoxicity of cancer cells by NK cells, with a focus on treatments for leukemia and multiple myeloma.
Collapse
Affiliation(s)
- Ashley Mentlik James
- Immune Cell Development and Host Defense Program, The Research Institute at Fox Chase Cancer Center , Philadelphia, PA , USA
| | - Adam D Cohen
- Abramson Cancer Center at the University of Pennsylvania , Philadelphia, PA , USA
| | - Kerry S Campbell
- Immune Cell Development and Host Defense Program, The Research Institute at Fox Chase Cancer Center , Philadelphia, PA , USA
| |
Collapse
|
111
|
Krönke J, Udeshi ND, Narla A, Grauman P, Hurst SN, McConkey M, Svinkina T, Heckl D, Comer E, Li X, Ciarlo C, Hartman E, Munshi N, Schenone M, Schreiber SL, Carr SA, Ebert BL. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 2013; 343:301-5. [PMID: 24292625 DOI: 10.1126/science.1244851] [Citation(s) in RCA: 1272] [Impact Index Per Article: 115.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lenalidomide is a drug with clinical efficacy in multiple myeloma and other B cell neoplasms, but its mechanism of action is unknown. Using quantitative proteomics, we found that lenalidomide causes selective ubiquitination and degradation of two lymphoid transcription factors, IKZF1 and IKZF3, by the CRBN-CRL4 ubiquitin ligase. IKZF1 and IKZF3 are essential transcription factors in multiple myeloma. A single amino acid substitution of IKZF3 conferred resistance to lenalidomide-induced degradation and rescued lenalidomide-induced inhibition of cell growth. Similarly, we found that lenalidomide-induced interleukin-2 production in T cells is due to depletion of IKZF1 and IKZF3. These findings reveal a previously unknown mechanism of action for a therapeutic agent: alteration of the activity of an E3 ubiquitin ligase, leading to selective degradation of specific targets.
Collapse
Affiliation(s)
- Jan Krönke
- Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Yang Y, Gao S, Fan H, Lin H, Li W, Wang J. Analysis of the efficacy of lenalidomide in patients with intermediate-1 risk myelodysplastic syndrome without 5q deletion. Exp Ther Med 2013; 6:803-807. [PMID: 24137269 PMCID: PMC3786804 DOI: 10.3892/etm.2013.1218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 06/18/2013] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to evaluate the efficacy and adverse effects of lenalidomide in the treatment of intermediate-1 risk non-5q deletion [non-del (5q)] myelodysplastic syndrome (MDS). A total of 30 patients with MDS were classified through G-banding chromosome karyotype analysis and fluorescence in situ hybridization (FISH). According to the International Prognostic Scoring System scores, among the 30 patients, 23 and seven cases had scores of 0.5 and 1.0, respectively. Lenalidomide (Revlimid®), 10 mg/day) was administered for 21 days every 28 days. All 30 cases were treated with lenalidomide for at least three cycles, including 20 cases with four cycles. The patients did not require erythropoietin, cyclosporine or iron chelation treatments. Statistical analysis was performed using SPSS statistical software version 13.0, and comparisons among groups were conducted using a t-test. The efficacy of lenalidomide was demonstrated in patients with intermediate-1 risk non-del (5q) MDS. Peripheral blood cell counts were improved following treatment, and absolute neutrophil, haemoglobin and platelet counts increased following 2–4 cycles of treatment. All patients became stable having undergone three cycles of treatment; however, 17 patients with chromosomal abnormalities had no cytogenetic response to the treatment, as confirmed through the FISH test. Patients with intermediate-1 risk non-del (5q) MDS treated with lenalidomide did not achieve complete haematological remission, although they demonstrated haematological improvement.
Collapse
Affiliation(s)
- Yan Yang
- Department of Haematology and Oncology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | | | | | | | | | | |
Collapse
|
113
|
Kröger N, Stübig T, Atanackovic D. Immune-modulating drugs and hypomethylating agents to prevent or treat relapse after allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2013; 20:168-72. [PMID: 24067503 DOI: 10.1016/j.bbmt.2013.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 09/15/2013] [Indexed: 02/05/2023]
Abstract
Allogeneic stem cell transplantation is a curative treatment option for many hematological diseases, and the numbers of transplantations are steadily increasing worldwide. Major progress has been made in lowering treatment-related mortality by reducing intensity of the conditioning regimen and by improving supportive care (eg, for infectious complications). Accordingly, relapse after allogeneic stem cell transplantation has become the major cause for treatment failure. Major efforts to prevent or treat relapse are focused on cellular- (T cell, natural killer cell), cytokine-, or antibody-based strategies to enhance the graft-versus-tumor effect or circumvent immunoescape. In the more recent years, new classes of agents have shown activity in several hematological malignancies, and besides their immediate antitumor activity, most of them also possess immune-modulatory qualities that may be useful alone or in combination with adoptive immunotherapy after allogeneic stem cell transplantation to enhance graft-versus-tumor effects. Here, we summarize the current knowledge and potential use of 2 of these compounds in preventing or treating relapse after allogeneic stem cell transplantation, namely immune-modulating drugs and hypomethylating agents.
Collapse
Affiliation(s)
- Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Thomas Stübig
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Djordje Atanackovic
- Department of Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
114
|
Bryant C, Suen H, Brown R, Yang S, Favaloro J, Aklilu E, Gibson J, Ho PJ, Iland H, Fromm P, Woodland N, Nassif N, Hart D, Joshua DE. Long-term survival in multiple myeloma is associated with a distinct immunological profile, which includes proliferative cytotoxic T-cell clones and a favourable Treg/Th17 balance. Blood Cancer J 2013; 3:e148. [PMID: 24036947 PMCID: PMC3789202 DOI: 10.1038/bcj.2013.34] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 07/15/2013] [Indexed: 12/31/2022] Open
Abstract
Despite improved outcomes in multiple myeloma (MM), a cure remains elusive. However, even before the current therapeutic era, 5% of patients survived >10 years and we propose that immune factors contribute to this longer survival. We identified patients attending our clinic, who had survived >10 years (n=20) and analysed their blood for the presence of T-cell clones, T-regulatory cells (Tregs) and T helper 17 (Th17) cells. These results were compared with MM patients with shorter follow-up and age-matched healthy control donors. The frequency of cytotoxic T-cell clonal expansions in patients with <10 years follow-up (MM patients) was 54% (n=144), whereas it was 100% (n=19/19) in the long-survivors (LTS-MM). T-cell clones from MM patients proliferated poorly in vitro, whereas those from LTS-MM patients proliferated readily (median proliferations 6.1% and 61.5%, respectively (P<0.0001)). In addition, we found significantly higher Th17 cells and lower Tregs in the LTS-MM group when compared with the MM group. These results indicate that long-term survival in MM is associated with a distinct immunological profile, which is consistent with decreased immune suppression.
Collapse
Affiliation(s)
- C Bryant
- 1] Institute of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia [2] Dendritic Cell Biology and Therapeutics, ANZAC Research Institute, Concord Hospital, Sydney, New South Wales, Australia [3] Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Santos ES, Goodman M, Byrnes JJ, Fernandez HF. Thalidomide Effects in the Post-transplantation Setting in Patients with Multiple Myeloma. Hematology 2013; 9:35-9. [PMID: 14965866 DOI: 10.1080/10245330310001652428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
UNLABELLED Thalidomide recently has been proven to have an impact on plasma cell dyscrasia through multiple mechanisms. Its effects on hematopoietic stem cells both in harvesting and in the immediate post-transplant setting are still unknown. We report on 12 cases (9 males and 3 females), median age 56 years old (range 41-65 years old) who underwent autologous peripheral stem cell transplantation for multiple myeloma and received thalidomide as maintenance therapy post-transplantation. Patients received various cytoreductive therapies prior to stem cell harvest. Eleven patients were in partial remission (PR) and one in complete remission (CR) on entry into the transplant phase of therapy. The median CD34+/kg harvested was 4.7 x 10(6) (range 1.9-55.4 x 10(6) CD34+/kg). All patients received intravenous melphalan 200 mg/m2 as their conditioning regimen. Six of twelve patients attained a CR post-transplant, and six a PR. Thalidomide was started after all patients engrafted post-transplant (absolute neutrophil count >0.5 x 10(9)/l and self-sustained platelet count >20 x 10(9)/l) and following satisfactory resolution of transplant toxicity including mucositis and diarrhea. Thalidomide was initiated at a median of 43 days post-transplant (range 23-138 days). The median leukocyte and platelet counts at the moment of thalidomide initiation were 5.8 x 10(9)/l (range 2.9-8.6 x 10(9)/l) and 196 x 10(9)/l (range 30-351 x 10(9)/l), respectively. Thalidomide was started at 100 mg daily, increasing 100 mg/day/month until reaching a dose of 400 mg/day. One patient failed to tolerate thalidomide due to CNS symptoms and stopped therapy at 12 days. Another patient stopped thalidomide therapy after 71 days, because of severe fatigue secondary to hypothyroidism. The most common adverse effects were constipation (5), rash (4), dry skin (3) and dizziness (3). No grade 3-4 adverse effects were documented. Neutropenia, previously reported as an adverse effect in this setting, was not seen to date in our cohort. All patients attained a CR or PR after transplant and thalidomide maintenance. We have had two relapses during a median follow-up of 68 weeks (range 42-172 weeks). CONCLUSION Thalidomide appears to be a safe drug in the post-transplant setting, perhaps adding to the response achieved post-transplant without major toxicity. Longer follow up and future randomized trials will be needed to validate the role of thalidomide and its long-term effect when used as maintenance therapy in the post-transplant setting.
Collapse
Affiliation(s)
- Edgardo S Santos
- Division of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami, FL 33136, USA
| | | | | | | |
Collapse
|
116
|
Kuruvilla J, Song K, Mollee P, Panzarella T, McCrae J, Nagy T, Crump M, Keating A. A phase II study of thalidomide and vinblastine for palliative patients with Hodgkin's lymphoma. Hematology 2013; 11:25-9. [PMID: 16522545 DOI: 10.1080/10245330500276592] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
INTRODUCTION Patients with Hodgkin's Lymphoma (HL) who relapse or progress after primary therapy and subsequent high dose chemotherapy with autologous stem cell transplantation (ASCT) cannot be cured with conventional treatment. We combined thalidomide (THAL), an agent with anti-angiogenic and immunomodulatory properties, with vinblastine, which is active after ASCT, to determine the objective response rate, improvement in B symptoms and toxicity in patients with refractory HD. METHODS Patients were eligible if they HD that progressed after chemotherapy and ASCT or had declined or were ineligible for curative therapy. Treatment consisted of THAL 200 mg orally given daily. After 2 weeks, VBL 6 mg IV was given weekly x 6 doses on an eight-week cycle. Response and toxicity assessment occurred following each cycle. RESULTS Eleven patients were enrolled, 1 progressed within 6 days of study enrollment and was subsequently treated with alternative palliative therapy and thus 11 patients are response evaluable and 10 are evaluable for toxicity. PATIENT CHARACTERISTICS relapsed after ASCT: 7; median number of prior chemotherapy regimens: 3 (range 1-5); median time to progression post-ASCT: 7 months (range 2-29). Four patients had a partial response to treatment (response rate 36%); two patients had stable disease. B symptoms were present at enrollment in four patients and resolved completely on treatment in two patients. Five had disease progression within 3 months of starting treatment. The median duration of response was 9 months (range 0-22 months). Toxicity was mild and limited to grade 2 neuropathy in 6 patients and grade 2 or 3 neutropenia in 4 patients. CONCLUSIONS In this small study in chemotherapy- refractory HL, THAL and VBL demonstrated encouraging activity with some durable responses and acceptable toxicity. These results suggest that chronic low dose chemotherapy combined with less toxic immunomodulatory or anti-angiogenic drugs warrants further study.
Collapse
Affiliation(s)
- John Kuruvilla
- The University of Toronto Autologous Blood and Marrow Transplant Program, Princess Margaret Hospital, Toronto, Ont., Canada.
| | | | | | | | | | | | | | | |
Collapse
|
117
|
Abstract
This spotlight review focuses on the second-generation immunomodulatory drug pomalidomide, which was recently approved by the US Food and Drug Administration. This drug was approved for patients with multiple myeloma who have received at least 2 prior therapies, including lenalidomide and bortezomib, and have demonstrated disease progression on or within 60 days of completion of the last therapy. This review focuses on the clinical trial data that led to approval and provides advice for treating physicians who are now prescribing this drug for patients.
Collapse
|
118
|
Morgan GJ, Davies FE. Role of thalidomide in the treatment of patients with multiple myeloma. Crit Rev Oncol Hematol 2013; 88 Suppl 1:S14-22. [PMID: 23827438 DOI: 10.1016/j.critrevonc.2013.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 05/23/2013] [Accepted: 05/30/2013] [Indexed: 01/06/2023] Open
Abstract
The first of the so-called "novel agents" (thalidomide, lenalidomide, and bortezomib), thalidomide has demonstrated activity as a single agent and in combination with other agents in patients with relapsed and/or refractory MM. The combination of melphalan, prednisone, and thalidomide (MPT) has become a standard treatment option for newly diagnosed patients who are ineligible for high-dose chemotherapy with autologous stem cell transplantation (ASCT). For patients intending to undergo ASCT, the combination of thalidomide, dexamethasone and cyclophosphamide can be used as a non-myelosuppressive induction regimen. Treatment with thalidomide is associated with an increased risk of developing peripheral neuropathy, which can be managed with dose reductions and discontinuation, and venous thromboembolism, which warrants thromboprophylaxis. While its adverse event profile may preclude prolonged use as maintenance therapy, thalidomide is an effective and well-tolerated salvage therapy option. Ongoing trials continue to evaluate novel thalidomide-based regimens to further optimize the use of thalidomide in the management of MM.
Collapse
Affiliation(s)
- Gareth J Morgan
- Institute of Cancer Research, Royal Marsden Hospital, London, UK.
| | | |
Collapse
|
119
|
Immunomodulation as a therapeutic strategy in the treatment of multiple myeloma. Crit Rev Oncol Hematol 2013; 88 Suppl 1:S5-13. [PMID: 23806982 DOI: 10.1016/j.critrevonc.2013.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 05/24/2013] [Accepted: 05/30/2013] [Indexed: 12/27/2022] Open
Abstract
Growth and survival of multiple myeloma (MM) cells depend on intrinsic, cell-autonomous parameters, such as the genetic lesions harboured by the MM cells, as well as extracellular, cell-non-autonomous factors, including the interaction between MM cells and bone-marrow stromal cells and the suppression of the host's anticancer immune responses. Thalidomide and the immunomodulatory agents lenalidomide and pomalidomide have pleiotropic effects on MM cells and their microenvironment, including promotion of direct mechanisms of MM-cell apoptosis, as well as indirect mechanisms mediated by perturbation of cell adhesion, modulation of cytokine production, and inhibition of tumor-associated angiogenesis. The immunomodulatory properties of these agents are mediated by effects on T-cell proliferation and function, stimulation of natural killer cells, and inhibition of regulatory T cells. Thalidomide and lenalidomide have established roles in the treatment of patients with newly diagnosed MM and those with relapsed/refractory disease. Pomalidomide is currently being evaluated in clinical trials, and preliminary clinical data suggest that it is active in patients with MM that is refractory to lenalidomide and bortezomib treatment. This article provides an overview of the current and potential future roles of immunomodulation in the management of MM, and how improved anticancer immune responses may improve treatment outcomes.
Collapse
|
120
|
McCurdy AR, Lacy MQ. Pomalidomide and its clinical potential for relapsed or refractory multiple myeloma: an update for the hematologist. Ther Adv Hematol 2013; 4:211-6. [PMID: 23730498 DOI: 10.1177/2040620713480155] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Multiple myeloma is a common plasma cell neoplasm that is incurable with conventional therapy. The treatment paradigm of multiple myeloma is not standardized and is evolving. The advent of novel drugs, including the proteasome inhibitor bortezomib and the immunomodulatory agents, has resulted in increased median survival. Unfortunately, all patients eventually relapse and require further therapy. Pomalidomide is the newest immunomodulatory drug, created by chemical modification of thalidomide with the intention of increasing therapeutic activity while limiting toxicity. Its mechanism of action is incompletely understood but involves anti-angiogenic effects, immunomodulation, an effect on the myeloma tumor microenvironment, and the protein cereblon. It is more potent than thalidomide and lenalidomide. In phase II studies, it has shown significant activity in patients with relapsed and refractory multiple myeloma, including patients who are heavily pretreated, have disease refractory to lenalidomide and bortezomib, and those with high-risk cytogenetic or molecular markers. It is generally well tolerated, with adverse effects including fatigue, neutropenia, neuropathy, and thromboembolic disease. Pomalidomide is a promising new agent in the expanding arsenal of antimyeloma drugs. In this review, we discuss the clinical experience to date with pomalidomide in multiple myeloma.
Collapse
Affiliation(s)
- Arleigh R McCurdy
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | |
Collapse
|
121
|
Slawinska-Brych A, Zdzisinska B, Mizerska-Dudka M, Kandefer-Szerszen M. Induction of apoptosis in multiple myeloma cells by a statin–thalidomide combination can be enhanced by p38 MAPK inhibition. Leuk Res 2013; 37:586-94. [DOI: 10.1016/j.leukres.2013.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 01/06/2013] [Accepted: 01/29/2013] [Indexed: 01/03/2023]
|
122
|
Feyler S, Selby PJ, Cook G. Regulating the regulators in cancer-immunosuppression in multiple myeloma (MM). Blood Rev 2013; 27:155-64. [PMID: 23623928 DOI: 10.1016/j.blre.2013.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An effective immune response requires a prompt but measured action against the pathological insult, to prevent over-zealous inflammatory-mediated tissue destruction. In cancer, defective or incompetent immune responses may paradoxically result in disease progression despite an immune attempt at elimination. Tumour-induced immunosuppression may not only result from soluble factors and altered antigenicity, but also from cellular-mediated tumour-induced immune evasion. Multiple myeloma (MM) is associated with both cellular and humoral immune deficiencies and increased T(Reg) cells. In vitro modelling has indicated that the tumour cells directly induce functional T(Reg) cells. In light of this recent evidence, it now seems that the most promising and synergistic approaches for cancer immunotherapy would involve specific anti-tumour immunity and simultaneous reduction of tumour-induced immune-regulation. This review sets out the basic understanding of the human immune response, its dysregulation in cancer and proposes how this knowledge may influence future treatment strategies to maximise the anti-tumour immune response.
Collapse
Affiliation(s)
- Sylvia Feyler
- Transplant Immunology Group, Academic Department of Haematology & Oncology, University of Leeds, UK
| | | | | |
Collapse
|
123
|
Abstract
One of the greatest challenges in all of medicine is to improve on the life of an asymptomatic patient; however, in this issue of Blood, Richter and colleagues share provocative new data taking steps toward accomplishing this goal.
Collapse
|
124
|
Abstract
SUMMARY Although there have been advances in the field, multiple myeloma, the second most common hematological malignancy, remains an incurable disease characterized by ever-shortening cycles of treatment and relapse. Myriad experimental and observational studies over the last few decades have comprehensively documented a state of profound immune dysfunction, which is progressive and correlated with disease stage. Nonetheless, immune responses against the tumor have demonstrated efficacy ex vivo, in animal models and in human disease. In this review we examine the immune defects in multiple myeloma and consider current and future approaches toward correction and manipulation of immune responses to affect clinically useful antitumor effects.
Collapse
Affiliation(s)
- Christopher Parrish
- Transplant Immunology Group, Leeds Institute of Molecular Medicine, University of Leeds, Beckett Street, Leeds, LS9 7TF, UK
| | - Gina B Scott
- Transplant Immunology Group, Leeds Institute of Molecular Medicine, University of Leeds, Beckett Street, Leeds, LS9 7TF, UK
| | - Gordon Cook
- Transplant Immunology Group, Leeds Institute of Molecular Medicine, University of Leeds, Beckett Street, Leeds, LS9 7TF, UK
| |
Collapse
|
125
|
Leitch HA, Buckstein R, Shamy A, Storring JM. The immunomodulatory agents lenalidomide and thalidomide for treatment of the myelodysplastic syndromes: A clinical practice guideline. Crit Rev Oncol Hematol 2013; 85:162-92. [DOI: 10.1016/j.critrevonc.2012.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 07/08/2012] [Accepted: 07/10/2012] [Indexed: 12/27/2022] Open
|
126
|
Lymphocytopenia is associated with an increased risk of severe infections in patients with multiple myeloma treated with bortezomib-based regimens. Int J Hematol 2013; 97:382-7. [DOI: 10.1007/s12185-013-1270-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/07/2013] [Accepted: 01/15/2013] [Indexed: 10/27/2022]
|
127
|
Thalidomide-analogue biology: immunological, molecular and epigenetic targets in cancer therapy. Oncogene 2013; 32:4191-202. [PMID: 23318436 DOI: 10.1038/onc.2012.599] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/04/2012] [Accepted: 11/05/2012] [Indexed: 11/08/2022]
|
128
|
Cives M, Simone V, Brunetti O, Longo V, Silvestris F. Novel lenalidomide-based combinations for treatment of multiple myeloma. Crit Rev Oncol Hematol 2013; 85:9-20. [DOI: 10.1016/j.critrevonc.2012.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 06/04/2012] [Accepted: 06/27/2012] [Indexed: 01/08/2023] Open
|
129
|
Quach H, Kalff A, Spencer A. Lenalidomide in multiple myeloma: Current status and future potential. Am J Hematol 2012; 87:1089-95. [PMID: 22641420 DOI: 10.1002/ajh.23234] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/06/2012] [Accepted: 04/11/2012] [Indexed: 11/05/2022]
Abstract
The clinical development of lenalidomide (Revlimid™), then pomalidomide (Actimid™) as members of immunomodulatory drugs (IMiDs) for the treatment of multiple myeloma (MM), exemplifies how insight into disease biology can lead to design of effective therapeutic agents. Increased experience and understanding of IMiD's diverse biological effects has lead to rational design of lenalidomide-based treatment-regimens over recent years. However, much about lenalidomide is yet to be understood and fully exploited. Here, we review what is known of lenalidomide's biological effects, clinical certainties and uncertainties in the treatment of MM, and explore its future potential with other synergistic therapeutic agents.
Collapse
Affiliation(s)
- Hang Quach
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University, Clayton, Victoria, Australia.
| | | | | |
Collapse
|
130
|
Bolzoni M, Storti P, Bonomini S, Todoerti K, Guasco D, Toscani D, Agnelli L, Neri A, Rizzoli V, Giuliani N. Immunomodulatory drugs lenalidomide and pomalidomide inhibit multiple myeloma-induced osteoclast formation and the RANKL/OPG ratio in the myeloma microenvironment targeting the expression of adhesion molecules. Exp Hematol 2012. [PMID: 23178378 DOI: 10.1016/j.exphem.2012.11.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Multiple myeloma (MM)-induced osteoclast (OC) formation is mainly due to an imbalance of the receptor activator NF-κB ligand (RANKL)-osteoprotegerin (OPG) ratio in favor of RANKL in the bone microenvironment and to the CCL3 production by MM cells. The purpose of the study was to investigate the effect of the immunomodulatory drugs on RANKL/OPG ratio, the production of pro-osteoclastogenic cytokines, and MM-induced OC formation. We found that in vivo concentrations of both lenalidomide (LEN) and pomalidomide (POM) significantly blunted RANKL upregulation normalizing the RANKL/OPG ratio in human osteoprogenitor cells (PreOBs) when co-cultured with MM cells and also inhibited CCL3 production by MM cells. A reduction in CD49d expression, a molecule critically involved in RANKL upregulation in the MM microenvironment, accompanied this effect. Consistently, the pro-osteoclastogenic property of MM cells co-cultured with PreOBs was reduced by both LEN and POM. We further investigated the effect of these drugs on the transcriptional profile of both MM cells and PreOBs by microarray analysis, which showed that adhesion molecules, such as ITGA8 and ICAM2, are significantly downregulated in MM cells. Our data suggest that LEN and POM inhibit MM-induced OC formation through normalization of the RANKL/OPG ratio targeting the expression of adhesion molecules by MM cells.
Collapse
Affiliation(s)
- Marina Bolzoni
- Hematology and BMT Center, University of Parma, Parma, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Herth I, Witzens-Harig M, Beckhove P, Hose D, Meissner T, Neuber B, Engelhardt M, Haas J, Neben K, Ho AD, Klein B, Goldschmidt H, Hundemer M. Thalidomide maintenance therapy maturates the T cell compartment and compromises antigen-specific antitumor immunity in patients with multiple myeloma. Exp Hematol 2012; 41:231-40. [PMID: 23142220 DOI: 10.1016/j.exphem.2012.10.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 09/25/2012] [Accepted: 10/23/2012] [Indexed: 01/08/2023]
Abstract
Interferon (INF)-α was the maintenance treatment of choice after autologous stem cell transplantation in multiple myeloma in the past, but currently Thalidomide is commonly used. In this prospective study, the implications of the various types of maintenance therapy on the patients T cell pattern and activation status were assessed. T cells were analyzed for expression of surface molecules, cytokine secretion, the presence of regulatory T cells, and the specific activation against the multiple myeloma antigen HM1.24. T cells from 69 multiple myeloma patients were analyzed: 19 patients were treated with IFN-α; 26 were treated with Thalidomide; and 24 patients received no maintenance therapy. Specific T cell activation with an immunogenic HLA-A2(+)-restricted peptide from the myeloma-associated antigen HM1.24 was impaired in the Thalidomide group. In accordance with this observation, there was a trend toward a higher amount of regulatory T cells in the Thalidomide group. Furthermore, patients treated with IFN-α showed high rates of naive T cells, whereas a high rate of effector memory T cells was observed in the Thalidomide group. Importantly, after cessation of Thalidomide therapy, this effect was reversible in the CD8 compartment. In conclusion, Thalidomide maintenance therapy has profound implications on T cell pattern and activation status, which compromise antigen specific antitumor immunity.
Collapse
Affiliation(s)
- Isabelle Herth
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Abstract
Despite improvements in disease management, multiple myeloma (MM) remains incurable. Conventional treatment methods are unsatisfactory, leading to a pattern of regression and remission, and ultimately failure. This pattern suggests that one of the possible strategies for improving outcomes is continuous therapy to maintain suppression of the surviving tumor cells. Optimal management of MM requires potent agents and modalities with direct tumoricidal activity, which can also provide continuous suppression of the residual tumor to prevent disease relapse. Immunomodulatory agents exert immunomodulatory and tumoricidal effects, and cause disruption of stromal cell support from the bone marrow microenvironment. Therefore continuous therapy with immunomodulatory agents may be able to provide both tumor reduction and tumor suppression, enabling physicians to consider the possibility of incorporating continuous therapy into the treatment paradigm of patients with MM.
Collapse
Affiliation(s)
- Ivan Borrello
- The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
133
|
Ruchelman AL, Man HW, Zhang W, Chen R, Capone L, Kang J, Parton A, Corral L, Schafer PH, Babusis D, Moghaddam MF, Tang Y, Shirley MA, Muller GW. Isosteric analogs of lenalidomide and pomalidomide: synthesis and biological activity. Bioorg Med Chem Lett 2012; 23:360-5. [PMID: 23168019 DOI: 10.1016/j.bmcl.2012.10.071] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/12/2012] [Accepted: 10/15/2012] [Indexed: 11/26/2022]
Abstract
A series of analogs of the immunomodulary drugs lenalidomide (1) and pomalidomide (2), in which the amino group is replaced with various isosteres, was prepared and assayed for immunomodulatory activity and activity against cancer cell lines. The 4-methyl and 4-chloro analogs 4 and 15, respectively, displayed potent inhibition of tumor necrosis factor-α (TNF-α) in LPS-stimulated hPBMC, potent stimulation of IL-2 in a human T cell co-stimulation assay, and anti-proliferative activity against the Namalwa lymphoma cell line. Both of these analogs displayed oral bioavailability in rat.
Collapse
Affiliation(s)
- Alexander L Ruchelman
- Drug Discovery Department, Celgene Corporation, 86 Morris Avenue, Summit, NJ 07901, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Clinical regressions and broad immune activation following combination therapy targeting human NKT cells in myeloma. Blood 2012; 121:423-30. [PMID: 23100308 DOI: 10.1182/blood-2012-06-435503] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Natural killer T (iNKT) cells can help mediate immune surveillance against tumors in mice. Prior studies targeting human iNKT cells were limited to therapy of advanced cancer and led to only modest activation of innate immunity. Clinical myeloma is preceded by an asymptomatic precursor phase. Lenalidomide was shown to mediate antigen-specific costimulation of human iNKT cells. We treated 6 patients with asymptomatic myeloma with 3 cycles of combination of α-galactosylceramide-loaded monocyte-derived dendritic cells and low-dose lenalidomide. Therapy was well tolerated and led to reduction in tumor-associated monoclonal immunoglobulin in 3 of 4 patients with measurable disease. Combination therapy led to activation-induced decline in measurable iNKT cells and activation of NK cells with an increase in NKG2D and CD56 expression. Treatment also led to activation of monocytes with an increase in CD16 expression. Each cycle of therapy was associated with induction of eosinophilia as well as an increase in serum soluble IL2 receptor. Clinical responses correlated with pre-existing or treatment-induced antitumor T-cell immunity. These data demonstrate synergistic activation of several innate immune cells by this combination and the capacity to mediate tumor regression. Combination therapies targeting iNKT cells may be of benefit toward prevention of cancer in humans.
Collapse
|
135
|
Zhu YX, Kortuem KM, Stewart AK. Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk Lymphoma 2012; 54:683-7. [PMID: 22966948 DOI: 10.3109/10428194.2012.728597] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Although several mechanisms have been proposed to explain the activity of thalidomide, lenalidomide and pomalidomide in multiple myeloma (MM), including demonstrable anti-angiogenic, anti-proliferative and immunomodulatory effects, the precise cellular targets and molecular mechanisms have only recently become clear. A landmark study recently identified cereblon (CRBN) as a primary target of thalidomide teratogenicity. Subsequently it was demonstrated that CRBN is also required for the anti-myeloma activity of thalidomide and related drugs, the so-called immune-modulatory drugs (IMiDs). Low CRBN expression was found to correlate with drug resistance in MM cell lines and primary MM cells. One of the downstream targets of CRBN identified is interferon regulatory factor 4 (IRF4), which is critical for myeloma cell survival and is down-regulated by IMiD treatment. CRBN is also implicated in several effects of IMiDs, such as down-regulation of tumor necrosis factor-α (TNF-α) and T cell immunomodulatory activity, demonstrating that the pleotropic actions of the IMiDs are initiated by binding to CRBN. Future dissection of CRBN downstream signaling will help to delineate the underlying mechanisms for IMiD action and eventually lead to development of new drugs with more specific anti-myeloma activities. It may also provide a biomarker to predict IMiD response and resistance.
Collapse
Affiliation(s)
- Yuan Xiao Zhu
- Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | | | | |
Collapse
|
136
|
Jin X, Lu S, Xing X, Wang L, Mu D, He M, Huang H, Zeng X, Chen Q. Thalidomide: features and potential significance in oral precancerous conditions and oral cancer. J Oral Pathol Med 2012; 42:355-62. [PMID: 22978368 DOI: 10.1111/jop.12004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2012] [Indexed: 02/05/2023]
Affiliation(s)
- Xin Jin
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Shujing Lu
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Xue Xing
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Lili Wang
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Dongliang Mu
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Mingjing He
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Haiyun Huang
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| |
Collapse
|
137
|
Molecular action of lenalidomide in lymphocytes and hematologic malignancies. Adv Hematol 2012; 2012:513702. [PMID: 22888354 PMCID: PMC3409527 DOI: 10.1155/2012/513702] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 05/12/2012] [Accepted: 06/18/2012] [Indexed: 01/10/2023] Open
Abstract
The immunomodulatory agent, lenalidomide, is a structural analogue of thalidomide approved by the US Food and Drug Administration for the treatment of myelodysplastic syndrome (MDS) and multiple myeloma (MM). This agent is also currently under active investigation for the treatment of chronic lymphocytic leukemia (CLL) and non-Hodgkin's lymphoma (NHL), as well as in drug combinations for some solid tumors and mantle cell lymphoma (MCL). Although treatment with lenalidomide has translated into a significant extension in overall survival in MM and MDS and has superior safety and efficacy relative to thalidomide, the mechanism of action as it relates to immune modulation remains elusive. Based on preclinical models and clinical trials, lenalidomide, as well as other structural thalidomide derivatives, enhances the proliferative and functional capacity of T-lymphocytes and amplifies costimulatory signaling pathways that activate effector responses and suppress inflammation. This paper summarizes our current understanding of T- and natural killer (NK) cell pathways that are modified by lenalidomide in hematopoietic neoplasms to inform future decisions about potential combination therapies.
Collapse
|
138
|
Mechanism of immunomodulatory drugs in multiple myeloma. Leuk Res 2012; 36:1218-24. [PMID: 22727252 DOI: 10.1016/j.leukres.2012.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 03/18/2012] [Accepted: 05/25/2012] [Indexed: 11/24/2022]
Abstract
Multiple myeloma is the second most common hematological cancer in the world. It is characterized by accumulation of malignant plasma cells in the bone marrow, osteolytic lesions and monoclonal immunoglobulins in blood/urine. With the introduction of immunomodulatory drugs into the treatment protocol, the outcome of multiple myeloma patients has dramatically improved with more than 30% of patients surviving for 10 years thus shifting multiple myeloma to a treatable condition.
Collapse
|
139
|
Puzik A, Thiel A, Faust K, Härtel C. Thalidomide has anti-inflammatory properties in neonatal immune cells. Innate Immun 2012; 19:42-52. [PMID: 22710762 DOI: 10.1177/1753425912449881] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Neonates demonstrate functional immaturity and dysregulation of immune responses leading to systemic inflammation and enhanced apoptosis of immune cells. Thalidomide has already been proven to differentially regulate immune responses and support anti-apoptosis in immunodeficiency syndromes. Thus, it was the aim of this study to evaluate the effects of thalidomide on the cytokine response and apoptosis of neonatal immune cells. After whole blood culture and stimulation of cord and adult blood samples, the intracytoplasmic expression and the secreted amounts of IL-2, TNF-α, IFN-γ, IL-6, IL-10 and IL-8 were assessed by flow cytometry and Cytokine Bead Array. Apoptosis was detected using Annexin-V staining. Bcl-2 expression was analysed using the Cytokine Bead Array Apoptosis Kit. Exposure to thalidomide (100 µg/ml) reduced the intracytoplasmic pro-inflammatory cytokine production of neonatal monocytes and the IFN-γ production of neonatal lymphocytes. In supernatants, the addition of thalidomide resulted in reduction of TNF-α, IL-6, IL-10 and, by trend, IFN-γ. While stimulated neonatal lymphocytes exhibited susceptibility to apoptosis, thalidomide tended to diminish apoptotic cells. Bcl-2 expression tended to be increased after addition of thalidomide. The potent anti-inflammatory effects of thalidomide and its anti-apoptotic properties in cord blood immune cells provide the basis for future strategies to optimise treatment of neonatal infections and immunodeficiency syndromes.
Collapse
Affiliation(s)
- Alexander Puzik
- Department of Pediatrics, Albert-Ludwigs-University of Freiburg, Germany.
| | | | | | | |
Collapse
|
140
|
Segler A, Tsimberidou AM. Lenalidomide in solid tumors. Cancer Chemother Pharmacol 2012; 69:1393-406. [PMID: 22584909 DOI: 10.1007/s00280-012-1874-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 04/22/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Lenalidomide is a thalidomide analogue with immunomodulatory and anti-angiogenic properties that include altering cytokine production, activating T cells, and augmenting natural killer cell function. Lenalidomide is approved by the U.S. Food and Drug Administration (FDA) for single-agent treatment of myelodysplastic syndromes associated with a 5q deletion and as a combination therapy with dexamethasone for the treatment of multiple myeloma. METHODS All prospective phase I-III clinical trials and preclinical data published until October 2011 and relevant literature were reviewed. RESULTS In phase I and/or II studies of single-agent lenalidomide in patients with advanced cancer, responses were reported in patients with prostate, thyroid, hepatocellular, pancreatic, and renal cancer and melanoma. The most common toxicities were hematologic, and in the first clinical trials, thrombotic events were noted. When anticoagulation prophylaxis and exclusion of patients with a history of thrombosis were implemented, thrombotic complications became uncommon. CONCLUSION Monitoring of blood counts and for evidence of thromboembolic events is essential for patients treated with lenalidomide. Ongoing trials of lenalidomide combination therapy offer a treatment option for patients with advanced cancer and will better define the role of lenalidomide in solid tumors.
Collapse
Affiliation(s)
- Angela Segler
- Department of Investigational Cancer Therapeutics, Phase I Clinical Trials Program, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 455, Houston, TX 77030, USA
| | | |
Collapse
|
141
|
Martiniuk F, Giovinazzo J, Tan AU, Shahidullah R, Haslett P, Kaplan G, Levis WR. Lessons of leprosy: the emergence of TH17 cytokines during type II reactions (ENL) is teaching us about T-cell plasticity. J Drugs Dermatol 2012; 11:626-630. [PMID: 22527432 PMCID: PMC3412264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
BACKGROUND Leprosy was the first disease classified according to the thymus derived T-cell in the 1960s and the first disease classified by the cytokine profile as intact interferon-γ (IFN-γ) and interleukin-2 (IL2) or TH1 (tuberculoid) and deficient IFN-γ and IL2 or TH2 (lepromatous), in the 1980s. OBJECTIVE In the present study, we set out to explore the T helper 17 (TH17) lymphocyte subset, the hallmark of T-cell plasticity, in skin biopsies from patients with erythema nodosum leprosum (ENL) who were treated with thalidomide. METHOD RNA was extracted from paraffin embedded tissue before and after thalidomide treatment of ENL and RT-PCR was performed. RESULTS IL17A, the hallmark of TH17, was consistently seen before and after thalidomide treatment, confirming the TH17 subset to be involved in ENL and potentially up-regulated by thalidomide. CONCLUSION A reduction in CD70, GARP, IDO, IL17B (IL-20), and IL17E (IL-25), coupled with increases in RORγT, ARNT, FoxP3, and IL17C (IL-21) following thalidomide treatment, opens the door to understanding the complexity of the immunomodulatory drug thalidomide, which can operate as an anti-inflammatory while simultaneously stimulating cell-mediated immunity (CMI). We conclude that TH17 is involved in the immunopathogenesis of ENL and that thalidomide suppresses inflammatory components of TH17, while enhancing other components of TH17 that are potentially involved in CMI.
Collapse
Affiliation(s)
- Frank Martiniuk
- Pulmonary Division, New York University Department of Medicine, New York, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
142
|
Pruritus in cutaneous T-cell lymphoma: a review. J Am Acad Dermatol 2012; 67:760-8. [PMID: 22285672 DOI: 10.1016/j.jaad.2011.12.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 12/12/2011] [Accepted: 12/20/2011] [Indexed: 12/21/2022]
Abstract
BACKGROUND Pruritus can be a distressing and even debilitating symptom for patients with cutaneous T-cell lymphoma (CTCL). To date, few studies have evaluated the pathophysiology of this symptom. Because of this, therapy for pruritus in CTCL has mainly relied on those therapies that target and treat the lymphoma. For patients living with CTCL that relapses or becomes refractory to treatment, and who continue to experience severe itch, this lymphoma-targeted treatment may not be enough to combat their pruritus. Therefore, other itch-targeted therapies are needed for use in this disease. OBJECTIVE We sought to evaluate the current evidence regarding the mechanism of action and treatments for pruritus associated with CTCL. METHODS An explicit and thorough search was restricted to all peer-reviewed literature available through MEDLINE (1950 to September 2011) and PubMed. Search terms used were "pruritus," "cutaneous T-cell lymphoma," "CTCL," "mycosis fungoides," "MF," and "Sézary syndrome." All studies that involved pruritus in CTCL, mycosis fungoides, or Sézary syndrome were evaluated by all 3 authors. RESULTS The current literature helps to identify therapies and possible mechanisms for treating patients with CTCL-associated pruritus. LIMITATION Most studies were preclinical. Only studies involving mechanisms of action or treatment were included. CONCLUSION A guideline is necessary to assist in the treatment of pruritus in CTCL and additional studies are necessary to uncover the exact mechanism or mechanisms of action.
Collapse
|
143
|
|
144
|
Sibon D, Cannas G, Baracco F, Prebet T, Vey N, Banos A, Besson C, Corm S, Blanc M, Slama B, Perrier H, Fenaux P, Wattel E. Lenalidomide in lower-risk myelodysplastic syndromes with karyotypes other than deletion 5q and refractory to erythropoiesis-stimulating agents. Br J Haematol 2011; 156:619-25. [DOI: 10.1111/j.1365-2141.2011.08979.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
145
|
Abstract
PURPOSE OF REVIEW To alert physician to timely recognition and current treatment of recurrent hypopyon iridocyclitis or panuveitis in ocular Behçet disease (OBD). RECENT FINDINGS Interferon-α, rituximab, intravitreal triamcinolone, and biological response modifiers by tumor necrosis factor inhibitors such as infliximab and adalimumab are being used increasingly for the treatment of severe sight-threatening ocular inflammation including retinal vasculitis and cystoid macular edema (CME). SUMMARY Biological agents offer tremendous potential in the treatment of OBD. Given that OBD predominantly afflicts the younger adults in their most productive years, dermatologist, rheumatologist, internist, or general practitioners supervising patients with oculo-articulo-oromucocutaneous syndromes should be aware of systemic Behçet disease. Early recognition of ocular involvement is important and such patients should strongly be instructed to visit immediately an ophthalmologist, as uveitis management differs from extraocular involvements with high ocular morbidity from sight-threatening complications due to relapsing inflammatory attacks in the posterior segment of the eye. A single infliximab infusion should be considered for the control of acute panuveitis, whereas repeated long-term infliximab infusions were proved to be more effective in reducing the number of episodes in refractory uveoretinitis with faster regression and complete remission of CME.
Collapse
Affiliation(s)
- Cem Evereklioglu
- Department of Ophthalmology, Erciyes University Medical Faculty, Kayseri, Turkey
| |
Collapse
|
146
|
Leite MR, Santos SS, Lyra AC, Mota J, Santana GO. Thalidomide induces mucosal healing in Crohn's disease: case report. World J Gastroenterol 2011; 17:5028-31. [PMID: 22174554 PMCID: PMC3236580 DOI: 10.3748/wjg.v17.i45.5028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Revised: 05/02/2011] [Accepted: 05/09/2011] [Indexed: 02/06/2023] Open
Abstract
Crohn's disease is a chronic inflammatory disorder of the gastrointestinal tract that is defined by relapsing and remitting episodes. Tumor necrosis factor alpha (TNF-α) appears to play a central role in the pathophysiology of the disease. Standard therapies for inflammatory bowel disease fail to induce remission in about 30% of patients. Biological therapies have been associated with an increased incidence of infections, especially infection by Mycobacterium tuberculosis (Mtb). Thalidomide is an oral immunomodulatory agent with anti-TNF-α properties. Recent studies have suggested that thalidomide is effective in refractory luminal and fistulizing Crohn's disease. Thalidomide costimulates T lymphocytes, with greater effect on CD8+ than on CD4+ T cells, which contributes to the protective immune response to Mtb infection. We present a case of Crohn's disease with gastric, ileal, colon and rectum involvement as well as steroid dependency, which progressed with loss of response to infliximab after three years of therapy. The thorax computed tomography scan demonstrated a pulmonary nodule suspected to be Mtb infection. The patient was started on thalidomide therapy and exhibited an excellent response.
Collapse
|
147
|
Germano G, Mantovani A, Allavena P. Targeting of the innate immunity/inflammation as complementary anti-tumor therapies. Ann Med 2011; 43:581-93. [PMID: 21756064 DOI: 10.3109/07853890.2011.595732] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Different types of cancer take advantage of inflammatory components to improve their life-span in the organs. A sustenance of growth factors and cytokines (e.g. interleukin (IL)-1, tumor necrosis factor, IL-6, vascular endothelial growth factor) supports malignant cell progression and contributes to suppress the body immune defense. Strategies to modulate the host micro-environment offer new approaches for anti-cancer therapies. For these reasons new molecules with anti-tumor and anti-inflammatory features (e.g. trabectedin) are looked at with new eyes in the light of the crucial link between inflammation and cancer.
Collapse
Affiliation(s)
- Giovanni Germano
- Department of Immunology and Inflammation, IRCCS Humanitas Clinical Institute, Rozzano, Milan, Italy
| | | | | |
Collapse
|
148
|
Muscal JA, Sun Y, Nuchtern JG, Dauser RC, McGuffey LH, Gibson BW, Berg SL. Plasma and cerebrospinal fluid pharmacokinetics of thalidomide and lenalidomide in nonhuman primates. Cancer Chemother Pharmacol 2011; 69:943-7. [PMID: 22109830 DOI: 10.1007/s00280-011-1781-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 11/07/2011] [Indexed: 11/28/2022]
Abstract
PURPOSE Thalidomide, originally developed as a sedative, was subsequently identified to have antiangiogenic properties. Lenalidomide is an antiangiogenic and immunomodulatory agent that has been utilized in the treatment of patients with brain tumors. We studied the pharmacokinetics and cerebrospinal fluid (CSF) penetration of thalidomide and lenalidomide in a nonhuman primate model. METHODS A dose of 50 mg of thalidomide or 20 mg of lenalidomide was administered once orally to each of three rhesus monkeys. Plasma and CSF samples were obtained at specified intervals, and the thalidomide or lenalidomide concentrations were determined by high-performance liquid chromatography with tandem mass spectrometry. Pharmacokinetic parameters were estimated using noncompartmental methods. CSF penetration was calculated as area under the concentration-time curve (AUC) CSF/AUC plasma. RESULTS For thalidomide, the median apparent clearance (Cl/F) was 2.9 mL/min/kg, the median plasma AUC was 80 μM h, and the median terminal half-life (t(½)) was 13.3 h. For lenalidomide, the median Cl/F was 8.7 mL/min/kg, the median AUC was 9 μM h, and the median t(½) was 5.6 h. Thalidomide was detected in the CSF of all animals, with a median penetration of 42%. Lenalidomide was detected in the CSF of 2 of 3 animals, with a CSF penetration of 11% in each. CONCLUSION Thalidomide and lenalidomide penetrate into the CSF after oral administration of clinically relevant doses. Plasma exposure to lenalidomide was similar in our model to that observed in studies involving children who have brain tumors. These results support further development of lenalidomide for the treatment of central nervous system malignancies.
Collapse
Affiliation(s)
- Jodi A Muscal
- Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates Street, Suite 1220, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
149
|
Bruno B, Gay F, Boccadoro M, Palumbo A. Management of myeloma: an Italian perspective. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2011; 11 Suppl 1:S82-6. [PMID: 22035755 DOI: 10.1016/j.clml.2011.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 03/23/2011] [Accepted: 03/25/2011] [Indexed: 11/26/2022]
Abstract
Multiple myeloma remains a fatal plasma cell malignancy. However, new insights into the disease biology and immunology have identified molecular mechanisms, underling functional interactions between plasma cells and the bone marrow microenvironment that have become molecular targets of so-called "new drugs" such as thalidomide, lenalidomide, and bortezomib. Recently, the combinations of new drugs with melphalan and prednisone in elderly patients, and with autologous stem cell transplantation in induction and/or maintenance schedules in younger patients have significantly prolonged overall survival. Optimal combinations and timing are a matter of debate. Moreover, management of side effects is a key clinical target to improve long-term quality of life. Many randomized phase III studies are currently in progress to address these issues. Whether these new advancements in myeloma treatment will eventually translate into a long chronic phase or a monoclonal gammopathy of undetermined significance-like status for the majority of patients remains, however, still unanswered.
Collapse
Affiliation(s)
- Benedetto Bruno
- Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliero-Universitaria San Giovanni Battista di Torino, Torino, Italy.
| | | | | | | |
Collapse
|
150
|
Rehman W, Arfons LM, Lazarus HM. The rise, fall and subsequent triumph of thalidomide: lessons learned in drug development. Ther Adv Hematol 2011; 2:291-308. [PMID: 23556097 PMCID: PMC3573415 DOI: 10.1177/2040620711413165] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Perhaps no other drug in modern medicine rivals the dramatic revitalization of thalidomide. Originally marketed as a sedative, thalidomide gained immense popularity worldwide among pregnant women because of its effective anti-emetic properties in morning sickness. Mounting evidence of human teratogenicity marked a dramatic fall from grace and led to widespread social, legal and economic ramifications. Despite its tragic past thalidomide emerged several decades later as a novel and highly effective agent in the treatment of various inflammatory and malignant diseases. In 2006 thalidomide completed its remarkable renaissance becoming the first new agent in over a decade to gain approval for the treatment of plasma cell myeloma. The catastrophic collapse yet subsequent revival of thalidomide provides important lessons in drug development. Never entirely abandoned by the medical community, thalidomide resurfaced as an important drug once the mechanisms of action were further studied and better understood. Ongoing research and development of related drugs such as lenalidomide now represent a class of irreplaceable drugs in hematological malignancies. Further, the tragedies associated with this agent stimulated the legislation which revamped the FDA regulatory process, expanded patient informed consent procedures and mandated more transparency from drug manufacturers. Finally, we review recent clinical trials summarizing selected medical indications for thalidomide with an emphasis on hematologic malignancies. Herein, we provide a historic perspective regarding the up-and-down development of thalidomide. Using PubMed databases we conducted searches using thalidomide and associated keywords highlighting pharmacology, mechanisms of action, and clinical uses.
Collapse
Affiliation(s)
- Waqas Rehman
- Department of Medicine, Division of Hematology-Oncology, Case Comprehensive Cancer Center, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Lisa M. Arfons
- Department of Medicine, Division of Hematology/Oncology, Louis Stokes Cleveland VAMC, Cleveland, OH, USA
| | - Hillard M. Lazarus
- Department of Medicine, University Hospitals Case Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|