1
|
Akter R, Rahman MR, Ahmed ZS, Afrose A. Plausibility of natural immunomodulators in the treatment of COVID-19-A comprehensive analysis and future recommendations. Heliyon 2023; 9:e17478. [PMID: 37366526 PMCID: PMC10284624 DOI: 10.1016/j.heliyon.2023.e17478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 06/28/2023] Open
Abstract
The COVID-19 pandemic has inflicted millions of deaths worldwide. Despite the availability of several vaccines and some special drugs approved for emergency use to prevent or treat this disease still, there is a huge concern regarding their effectiveness, adverse effects, and most importantly, their efficacy against the new variants. A cascade of immune-inflammatory responses is involved with the pathogenesis and severe complications with COVID-19. People with dysfunctional and compromised immune systems display severe complications, including acute respiratory distress syndrome, sepsis, multiple organ failure etc., when they get infected with the SARS-CoV-2 virus. Plant-derived natural immune-suppressant compounds, such as resveratrol, quercetin, curcumin, berberine, luteolin, etc., have been reported to inhibit pro-inflammatory cytokines and chemokines. Therefore, natural products with immunomodulatory and anti-inflammatory potential could be plausible targets to treat this contagious disease. This review aims to delineate the clinical trials status and outcomes of natural compounds with immunomodulatory potential in COVID-19 patients along with the outcomes of their in-vivo studies. In clinical trials several natural immunomodulators resulted in significant improvement of COVID-19 patients by diminishing COVID-19 symptoms such as fever, cough, sore throat, and breathlessness. Most importantly, they reduced the duration of hospitalization and the need for supplemental oxygen therapy, improved clinical outcomes in patients with COVID-19, especially weakness, and eliminated acute lung injury and acute respiratory distress syndrome. This paper also discusses many potent natural immunomodulators yet to undergo clinical trials. In-vivo studies with natural immunomodulators demonstrated reduction of a wide range of proinflammatory cytokines. Natural immunomodulators that were found effective, safe, and well tolerated in small-scale clinical trials are warranted to undergo large-scale trials to be used as drugs to treat COVID-19 infections. Alongside, compounds yet to test clinically must undergo clinical trials to find their effectiveness and safety in the treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Raushanara Akter
- School of Pharmacy, Brac University, 66 Mohakhali, Dhaka, Bangladesh
| | - Md. Rashidur Rahman
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Zainab Syed Ahmed
- School of Pharmacy, Brac University, 66 Mohakhali, Dhaka, Bangladesh
| | - Afrina Afrose
- School of Pharmacy, Brac University, 66 Mohakhali, Dhaka, Bangladesh
| |
Collapse
|
2
|
Induction of multiple myeloma bone marrow stromal cell apoptosis by inhibiting extracellular vesicle miR-10a secretion. Blood Adv 2020; 3:3228-3240. [PMID: 31698453 DOI: 10.1182/bloodadvances.2019000403] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/22/2019] [Indexed: 02/08/2023] Open
Abstract
Bone marrow stromal cells (BMSCs) interact with multiple myeloma (MM) cells in the bone marrow and create a permissive microenvironment for MM cell proliferation and survival. In this study, we investigated the role of extracellular vesicles (EVs) from BMSCs derived from patients with MM (MM-BMSCs). EV-encapsulated miR-10a expression was high while intracellular miR-10a was low in MM-BMSCs. We therefore hypothesized that miR-10a was packaged into EVs that were actively released into the extracellular space. Inhibition of EV release resulted in accumulation of intracellular miR-10a, inhibition of cell proliferation, and induction of apoptosis in MM-BMSCs. In contrast, proliferation and apoptosis of BMSCs derived from healthy individuals were unaffected by inhibition of EV release. Furthermore, miR-10a derived from MM-BMSCs was transferred into MM cells via EVs and enhanced their proliferation. These results suggest that inhibition of EV release induced apoptosis in MM-BMSCs and inhibited MM cell proliferation, indicating a possible role for MM-BMSC-targeted therapy.
Collapse
|
3
|
Biologic Implications of t(11;14) in Multiple Myeloma Explained With a Case of Refractory Disease Sensitive to Venetoclax. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e556-e559. [PMID: 32653454 DOI: 10.1016/j.clml.2020.05.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
|
4
|
Guerra-García P, Marshall LV, Cockle JV, Ramachandran PV, Saran FH, Jones C, Carceller F. Challenging the indiscriminate use of temozolomide in pediatric high-grade gliomas: A review of past, current, and emerging therapies. Pediatr Blood Cancer 2020; 67:e28011. [PMID: 31617673 DOI: 10.1002/pbc.28011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/31/2019] [Accepted: 09/11/2019] [Indexed: 01/26/2023]
Abstract
Pediatric high-grade gliomas (pHGG) constitute 8% to 12% of primary brain tumors in childhood. The most widely utilized treatment encompasses surgical resection followed by focal radiotherapy and temozolomide. However, experiences over past decades have not demonstrated improved outcomes. pHGG have been classified into different molecular subgroups defined by mutations in histone 3, IDH gene, MAPK pathway, and others, thereby providing a rationale for various targeted therapies. Additionally, immunotherapy and drug repurposing have also become attractive adjunctive treatments. This review focuses on past, present, and emerging treatments for pHGG integrating molecular research with the mainstream pediatric drug development in Europe and the United States to sketch a way forward in the development of novel therapeutic approaches. The implementation of randomized clinical trials with adaptive designs, underpinned by a robust biological rationale, and harnessing collaboration between the pharmaceutical industry, academia, regulators and patients/parents organizations will be essential to improve the outcomes for these children.
Collapse
Affiliation(s)
- Pilar Guerra-García
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.,Paediatric Oncology Department, 12 de Octubre University Hospital, Madrid, Spain
| | - Lynley V Marshall
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.,Division of Clinical Studies, Institute of Cancer Research, London, United Kingdom
| | - Julia V Cockle
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | | | - Frank H Saran
- Department of Radiation Oncology, The Royal Marsden NHS Foundation Trust, London, United Kingdom.,Department of Radiation Oncology, Auckland District Health Board, Auckland, New Zealand
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Fernando Carceller
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.,Division of Clinical Studies, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
5
|
Yang X, Zhao X, Wang X. Compound kushen injection for multiple myeloma: A protocol of systematic review and meta-analysis. Medicine (Baltimore) 2019; 98:e18257. [PMID: 31852094 PMCID: PMC6922587 DOI: 10.1097/md.0000000000018257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Compound kushen injection is increasingly used to treat various cancers. However, its role in the management of multiple myeloma (MM) is still controversial and requires further clarification. The aim of this study is to evaluate efficacy and safety of compound kushen injection for MM through systematic review and meta-analysis. METHODS We are going to search the 6 electronic databases: PubMed, Embase, Cochrane Library, China National Knowledge Infrastructure, Wangfang and Chinese Biomedical Literature Database. General characteristics and Specific trial characteristics will be collected from the included studies. The outcomes included overall response rate, complete response rate, 3-year progression-free survival rate, 3-year overall survival rate, and different types of treatment-related adverse events. We calculated the risk ratios as well as their 95% confidence intervals of these outcomes and pooled the results through RevMan 5.2 software. DISCUSSION The results of the study will be submitted to a peer-reviewed journal for publication. ETHICS AND DISSEMINATION This study is not a clinical study, ethical approval is not required.
Collapse
Affiliation(s)
- Xinwei Yang
- Department of Hematology, Gansu Provincial Hospital of Traditional Chinese Medicine
| | - Xiyun Zhao
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine
| | | |
Collapse
|
6
|
New generation drugs for treatment of multiple myeloma. Drug Discov Today 2019; 25:367-379. [PMID: 31765717 DOI: 10.1016/j.drudis.2019.11.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/08/2019] [Accepted: 11/15/2019] [Indexed: 12/28/2022]
Abstract
Multiple myeloma (MM), a plasma cell malignancy, is characterised by lesions in multiple bones involving transformed, matured post-follicular B cells. The course of the disease involves an initial development of monoclonal gammopathy of undetermined significance (MGUS), followed by smouldering MM, before the full MM disease emerges. Despite novel therapies, MM remains incurable, managed by combination therapies, including proteasome inhibitors (PIs), immunomodulators (IMiDs) and anti-human CD38 (daratumumab). MM patients have an increased risk of thromboembolic events due to combination treatments with IMiDs, PIs and anti-human CD38 antibody, and steroids. This review will examine the efficacy and pro-thrombotic effects of MM therapies.
Collapse
|
7
|
Jantan I, Haque MA, Ilangkovan M, Arshad L. An Insight Into the Modulatory Effects and Mechanisms of Action of Phyllanthus Species and Their Bioactive Metabolites on the Immune System. Front Pharmacol 2019; 10:878. [PMID: 31440162 PMCID: PMC6693410 DOI: 10.3389/fphar.2019.00878] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 07/10/2019] [Indexed: 11/25/2022] Open
Abstract
Phyllanthus species (family; Euphorbiaceae) have been intensively studied for their immunomodulating effects due to their wide-ranging uses to treat immune-related diseases in indigenous medicine, which are primarily lack of scientific basis. The focuses of this review are on the significance of Phyllanthus species and their bioactive metabolites particularly corilagin (1), geraniin (2), gallic acid (3), phyllanthin (4), hypophyllanthin (5), ellagic acid (6), phyltetralin (7), niranthin (8), catechin (9), quercetin (10), astragalin (11), and chebulagic acid (12) in the modulation of both innate and adaptive immune systems through various mechanisms and their possible therapeutic benefits for treatment of immune-related diseases. We have compiled all significant findings published in the literature, and the data were analyzed critically to provide perspectives and directions for future research for the plants as a prospective source of novel immunomodulating agents. Various Phyllanthus species particularly Phyllanthus amarus, Phyllanthus emblica, Phyllanthus niruri, and Phyllanthus urinaria have been documented to possess significant immunomodulatory effects. However, the possible challenges encountered by the application of extracts of various Phyllanthus species and their bioactive constituents as immunomodulators need to be addressed. Most reports on the biological and pharmacological studies of the plants were based on crude extracts. The extracts were not chemically characterized, and the contributions of their chemical constituents to the bioactivities were not identified. The underlying mechanisms involved in the immunomodulatory effects of the Phyllanthus species were not indepthly studied due to limitations in terms of design, conduct, and interpretation. Extensive experimental and preclinical studies on the immunomodulating potential of Phyllanthus species should be carried out to provide sufficient data to prove that their traditional uses are inherently effective and safe and will allow clinical trials to be pursued for their further development as therapeutic agents to treat immune-related disorders.
Collapse
Affiliation(s)
- Ibrahim Jantan
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Lakeside Campus, Subang Jaya, Malaysia
| | - Md. Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | | | - Laiba Arshad
- Department of Pharmacy, Forman Christian College (A Chartered University), Lahore, Pakistan
| |
Collapse
|
8
|
Xia X, Liu Y, Liao Y, Guo Z, Huang C, Zhang F, Jiang L, Wang X, Liu J, Huang H. Synergistic effects of gefitinib and thalidomide treatment on EGFR-TKI-sensitive and -resistant NSCLC. Eur J Pharmacol 2019; 856:172409. [DOI: 10.1016/j.ejphar.2019.172409] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/19/2019] [Accepted: 05/22/2019] [Indexed: 02/05/2023]
|
9
|
The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma. Blood Rev 2019; 34:56-66. [DOI: 10.1016/j.blre.2018.11.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/14/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022]
|
10
|
Sun S, Zhang Q, Wang Q, Wu Q, Xu G, Chang P, Hu H, Bai F. Local delivery of thalidomide to inhibit neointima formation in rat model with artery injury. Pathol Res Pract 2018; 214:1303-1308. [PMID: 30029933 DOI: 10.1016/j.prp.2018.02.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/03/2018] [Accepted: 02/18/2018] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To observe the effect of local administration of thalidomide on neointimal formation after balloon-induced carotid artery injury in rats. METHODS Forty-eight male Sprague-Dawley rats were randomly divided into 3 groups (n = 16): Sham operation group (group A), alone operation group (group B) and Thalidomide group (group C). The carotid arteries of group B and group C were injured by a conventional percutaneous transluminal coronary angioplasty (PTCA) balloon catheter. Group C was treated by local delivery of thalidomide, and group B did not receive thalidomide. The arteries of group A were not injured. Seven and 14 days after balloon injury, rats were sacrificed. Serum concentrations of vascular endothelial growth factor (VEGF) and tumor necrosis factor-α (TNF-α) were measured using enzyme-linked immunosorbent assay (ELISA). Neointima area, lumen area, macrophage infiltration and local expression of VEGF were measured using morphometric and immunohistochemical analyses. Real-time reverse transcriptase polymerase chain reaction (RT-PCR) was used to examine VEGF mRNA expression. RESULTS The VEGF levels were significantly increased in group B than in group C at 7 days (4.82 ± 0.17 pg/mL vs 0.98 ± 0.1 pg/mL, P < 0.01) and 14 days (6.3 ± 0.16 pg/mL vs 1.03 ± 0.09 pg/mL, P < 0.01). The TNF-α levels were also significantly increased in group B than in group C at 7 days (83 ± 1.01 pg/mL vs 76.37 ± 0.75 pg/mL, P < 0.01) and 14 days (84.06 ± 1.11 pg/mL vs 78.46 ± 0.94 pg/mL, P < 0.01). However, the area of neointimal formation was significantly reduced in group C than in group B at 14 days (0.07± 0.01 mm2 vs 0.12± 0.04 mm2, P < 0.01). Macrophage infiltration and local expression of VEGF in the injured arteries were significantly reduced in group C than in group B at 14 days. VEGF mRNA expression was significantly reduced in Group C than in group B at 14 days (6.3 ± 0.16 vs 1.02 ± 0.1, P < 0.01). CONCLUSIONS Thalidomide, which is a specific VEGF inhibitor, significantly inhibited neointimal hyperplasia and vascular restenosis after balloon injury to the carotid artery in rats, thus potentially providing a novel method for the prevention and treatment of restenosis, especially in-stent restenosis.
Collapse
Affiliation(s)
- Shougang Sun
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Quan Zhang
- Department of Cardiology, Pingliang People's Hospital, Pingliang, 744000, China
| | - Qiongying Wang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Qiang Wu
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Guangli Xu
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Peng Chang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Hao Hu
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Feng Bai
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| |
Collapse
|
11
|
Bruserud Ø, Hansen BA, Vetti N, Johansen S, Reikvam H. Successful eradication of leptomeningeal plasma cell disease. Oxf Med Case Reports 2018; 2018:omy038. [PMID: 29992033 PMCID: PMC6031028 DOI: 10.1093/omcr/omy038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/04/2018] [Accepted: 05/07/2018] [Indexed: 11/23/2022] Open
Abstract
Plasma cell leukaemia (PCL) is a rare and aggressive form of malignant monoclonal gammopathy characterized by the presence of high levels of plasma cells in peripheral blood. Central nervous system involvement of PCL has no established treatment and an extremely poor prognosis. We here present a 59-year-old male patient diagnosed with PCL, initially treated with induction chemotherapy followed by autologous peripheral blood hematopoietic stem cell transplantation. After achieving a partial response, he relapsed and presented with leptomeningeal disease. He was then successfully treated with dexamethasone, pomalidomide, and an intrathecal combination of methotrexate, methylprednisolone and cytarabine. This cleared his cerebrospinal fluid from plasma cells achieving a durable partial response.
Collapse
Affiliation(s)
- Øyvind Bruserud
- Section for Endocrinology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Nils Vetti
- Department of Radiology, Haukeland University Hospital, Bergen, Norway.,Section for Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Silje Johansen
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Section for Haematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
12
|
Shaulov A, Avivi I, Cohen Y, Duek A, Leiba M, Gatt ME. Gastrointestinal perforation in light chain amyloidosis in the era of novel agent therapy - a case series and review of the literature. Amyloid 2018; 25:11-17. [PMID: 29241368 DOI: 10.1080/13506129.2017.1416350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gastrointestinal (GI) perforation is remarkably rare in patients with light chain (AL) amyloidosis and has not yet been reported in patients with AL amyloidosis treated with novel agents. Only 24 cases of GI perforation have previously been reported in the setting of AL amyloidosis of which 15 had available information in English. All 15 did not receive novel agent therapy and six died early after experiencing GI perforation. This study reports the characteristics and outcome of AL patients that developed GI perforation in the era of novel agent treatment. Seven patients were reviewed. In two patients, GI perforation was the presenting symptom of AL amyloidosis, whereas five patients developed GI perforations following initiation of an anti-AL therapy (three after bortezomib-based, 1 after lenalidomide-based and 1 after thalidomide-based therapy). All patients underwent surgery and survived the perforation. Treatment was renewed following surgery in six of seven patients, with no further GI complications. In conclusion, GI perforation in AL amyloidosis is rare and mostly reported after treatment initiation. Urgent surgery appears to be lifesaving and renewal of the anti-AL novel therapy appears to be safe, with no significant risk for re-perforation or GI toxicity. Prognosis in these patients is related to severity of the disease and response to therapy rather than the development of GI perforation.
Collapse
Affiliation(s)
- Adir Shaulov
- a Department of Haematology , Hadassah-Hebrew University Medical Center , Jerusalem , Israel
| | - Irit Avivi
- b Department of Haematology and Bone Marrow Transplantation , Tel Aviv Sourasky Medical Center , Tel Aviv , Israel
| | - Yael Cohen
- b Department of Haematology and Bone Marrow Transplantation , Tel Aviv Sourasky Medical Center , Tel Aviv , Israel
| | - Adrian Duek
- c Department of Haematology , Sheba-Tel HaShomer Medical Center , Tel Aviv , Israel
| | - Merav Leiba
- c Department of Haematology , Sheba-Tel HaShomer Medical Center , Tel Aviv , Israel
| | - Moshe E Gatt
- a Department of Haematology , Hadassah-Hebrew University Medical Center , Jerusalem , Israel
| |
Collapse
|
13
|
Enhancement of pomalidomide anti-tumor response with ACY-241, a selective HDAC6 inhibitor. PLoS One 2017; 12:e0173507. [PMID: 28264055 PMCID: PMC5338861 DOI: 10.1371/journal.pone.0173507] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/21/2017] [Indexed: 11/22/2022] Open
Abstract
Thalidomide-based Immunomodulatory Drugs (IMiDs®), including lenalidomide and pomalidomide, are effective therapeutics for multiple myeloma. These agents have been approved with, or are under clinical development with, other targeted therapies including proteasome inhibitors, αCD38 monoclonal antibodies, as well as histone deacetylase (HDAC) inhibitors for combination therapy. HDAC inhibitors broadly targeting Class I and IIb HDACs have shown potent preclinical efficacy but have frequently demonstrated an undesirable safety profile in combination therapy approaches in clinical studies. Therefore, development of more selective HDAC inhibitors could provide enhanced efficacy with reduced side effects in combination with IMiDs® for the treatment of B-cell malignancies, including multiple myeloma. Here, the second generation selective HDAC6 inhibitor citarinostat (ACY-241), with a more favorable safety profile than non-selective pan-HDAC inhibitors, is shown to synergize with pomalidomide in in vitro assays through promoting greater apoptosis and cell cycle arrest. Furthermore, utilizing a multiple myeloma in vivo murine xenograft model, combination treatment with pomalidomide and ACY-241 leads to increased tumor growth inhibition. At the molecular level, combination treatment with ACY-241 and pomalidomide leads to greater suppression of the pro-survival factors survivin, Myc, and IRF4. The results presented here demonstrate synergy between pomalidomide and ACY-241 in both in vitro and in vivo preclinical models, providing further impetus for clinical development of ACY-241 for use in combination with IMiDs for patients with multiple myeloma and potentially other B-cell malignancies.
Collapse
|
14
|
Yamashita Y, Tamura S, Oiwa T, Kobata H, Kuriyama K, Mushino T, Murata S, Hosoi H, Nishikawa A, Hanaoka N, Sonoki T. Successful Intrathecal Chemotherapy Combined with Radiotherapy Followed by Pomalidomide and Low-Dose Dexamethasone Maintenance Therapy for a Primary Plasma Cell Leukemia Patient. Hematol Rep 2017; 9:6986. [PMID: 28286633 PMCID: PMC5337827 DOI: 10.4081/hr.2017.6986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/03/2017] [Accepted: 02/05/2017] [Indexed: 01/04/2023] Open
Abstract
Primary plasma cell leukemia (PPCL) is a rare aggressive variant of plasma cell disorder and frequently presents with extramedullary disease. Central nervous system (CNS) involvement with PPCL has an extremely poor prognosis. We describe a 46-year-old man with PPCL treated with a combination of lenalidomide, bortezomib, and dexamethasone as induction therapy following upfront allogeneic stem cell transplantation (allo-SCT). Despite achieving a very good partial response, the patient suffered from an isolated CNS relapse 12 months after allo-SCT. He was immediately started on concurrent intrathecal chemotherapy (IT) and cranial irradiation (RT). Subsequently, pomalidomide and low-dose dexamethasone (Pd) were given as maintenance therapy. He has been without CNS recurrence for more than 18 months. Our case suggests that concurrent IT and RT followed by Pd maintenance therapy may be an effective option to control CNS relapse of PPCL after allo-SCT.
Collapse
Affiliation(s)
- Yusuke Yamashita
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Shinobu Tamura
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Takehiro Oiwa
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Hiroshi Kobata
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Kodai Kuriyama
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Toshiki Mushino
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Shogo Murata
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Hiroki Hosoi
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Akinori Nishikawa
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Nobuyoshi Hanaoka
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| | - Takashi Sonoki
- Department of Hematology/Oncology, Wakayama Medical University , Wakayama, Japan
| |
Collapse
|
15
|
Li Y, Du Z, Wang X, Wang G, Li W. Association of IL-6 Promoter and Receptor Polymorphisms with Multiple Myeloma Risk: A Systematic Review and Meta-Analysis. Genet Test Mol Biomarkers 2016; 20:587-596. [PMID: 27525545 DOI: 10.1089/gtmb.2015.0169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A number of studies show that the pleiotropic cytokine interleukin-6 (IL-6) plays an important role in the pathogenesis of multiple myeloma (MM). However, whether MM risk is associated with IL-6 genetic variability remains uncertain. OBJECTIVE The aim of our study was to evaluate the association between two different IL-6 polymorphisms (located in the IL-6 promoter and receptor, respectively) and the risk of developing MM using a meta-analytic approach. MATERIALS AND METHODS A systematic search for studies on the association of IL-6/IL-6R single-nucleotide polymorphisms with susceptibility to MM was conducted in PubMed, Cochrane Library, Embase, CNKI (Chinese) and Wanfang (Chinese) Digital Dissertations Databases from inception through November 2014. A meta-analysis was performed and results were presented as odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS A total of eight case-control studies on the IL-6 promoter polymorphism and three studies on the IL-6 receptor (IL-6R) polymorphism were included. No significant association was found between the IL-6 promoter rs1800795 (G>C) polymorphism and MM susceptibility. A significantly increased risk of MM was observed with the IL-6R rs8192284 (A>C) polymorphism. In subgroup analyses, grouped by ethnicity, region, quality of studies, and Hardy-Weinberg equilibrium of control group, similar results were found. CONCLUSION Unlike the IL-6 promoter rs1800795 (G>C) polymorphism, the IL-6R rs8192284 (A>C) polymorphism may be associated with MM risk. However, large-scale studies are needed to validate our findings since they are based on a relatively small number of studies.
Collapse
Affiliation(s)
- Yuying Li
- Cancer Center, First Hospital of Jilin University , Changchun, China
| | - Zhonghua Du
- Cancer Center, First Hospital of Jilin University , Changchun, China
| | - Xu Wang
- Cancer Center, First Hospital of Jilin University , Changchun, China
| | - Guanjun Wang
- Cancer Center, First Hospital of Jilin University , Changchun, China
| | - Wei Li
- Cancer Center, First Hospital of Jilin University , Changchun, China
| |
Collapse
|
16
|
Krishnan SR, Jaiswal R, Brown RD, Luk F, Bebawy M. Multiple myeloma and persistence of drug resistance in the age of novel drugs (Review). Int J Oncol 2016; 49:33-50. [PMID: 27175906 DOI: 10.3892/ijo.2016.3516] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/17/2015] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is a mature B cell neoplasm that results in multi-organ failure. The median age of onset, diverse clinical manifestations, heterogeneous survival rate, clonal evolution, intrinsic and acquired drug resistance have impact on the therapeutic management of the disease. Specifically, the emergence of multidrug resistance (MDR) during the course of treatment contributes significantly to treatment failure. The introduction of the immunomodulatory agents and proteasome inhibitors has seen an increase in overall patient survival, however, for the majority of patients, relapse remains inevitable with evidence that these agents, like the conventional chemotherapeutics are also subject to the development of MDR. Clinical management of patients with MM is currently compromised by lack of a suitable procedure to monitor the development of clinical drug resistance in individual patients. The current MM prognostic measures fail to pick the clonotypic tumor cells overexpressing drug efflux pumps, and invasive biopsy is insufficient in detecting sporadic tumors in the skeletal system. This review summarizes the challenges associated with treating the complex disease spectrum of myeloma, with an emphasis on the role of deleterious multidrug resistant clones orchestrating relapse.
Collapse
Affiliation(s)
- Sabna Rajeev Krishnan
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ritu Jaiswal
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ross D Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Frederick Luk
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Mary Bebawy
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| |
Collapse
|
17
|
Park J, Park E, Jung CK, Kang SW, Kim BG, Jung Y, Kim TH, Lim JY, Lee SE, Min CK, Won KA. Oral proteasome inhibitor with strong preclinical efficacy in myeloma models. BMC Cancer 2016; 16:247. [PMID: 27012957 PMCID: PMC4806471 DOI: 10.1186/s12885-016-2285-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/17/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The proteasome is a validated anti-cancer target and various small-molecule inhibitors are currently in clinical development or on the market. However, adverse events and resistance associated with those proteasome inhibitors indicate the need for a new generation of drugs. Therefore, we focused on developing an oral proteasome inhibitor with improved efficacy and safety profiles. METHOD The in vitro inhibition of the 20S proteasome catalytic activities was determined in human multiple myeloma (MM) cellular lysates with fluorogenic peptide substrates specific for each catalytic subunit. Cell cytotoxicity was assessed with the ATP bioluminescence assay using human cell samples from tumor cell lines, MM patients or normal healthy donors. In mice bearing human MM xenografts, a single dose of LC53-0110 was administered orally, and concentration-time profiles of LC53-0110 and the 20S proteasome catalytic activities in plasma, blood, and tumor were determined. The efficacy of repeat-dose compound with regard to tumor growth inhibition in vivo was also evaluated in the same MM xenograft models. RESULTS LC53-0110 is far more specific for the chymotrypsin-like proteolytic (β5) site of the 20S proteasome as compared to bortezomib, carfilzomib, or ixazomib. LC53-0110 treatment showed accumulation of ubiquitinated proteins, inhibited cell viability with a low nM range potency in various tumor cell lines, and showed potent activity on CD138(+) cells isolated from MM patients who are resistant/refractory to current FDA-approved drug treatment. When a single dose was administered orally to tumor-bearing mice, LC53-0110 showed both greater maximum and sustained tumor proteasome inhibition as compared with ixazomib in MM xenograft models. The robust pharmacodynamic responses in tumor correlated with tumor growth regression. In addition, LC53-0151, an analog of LC53-0110, in combination with pomalidomide, a third-generation immunomodulatory drug, showed synergistic inhibition of tumor growth both in vitro and in the xenograft mouse model. CONCLUSIONS In view of the in vitro, in vivo, and ex vivo profiles, further investigation of additional LC compounds in preclinical studies is warranted for the nomination of a clinical development candidate.
Collapse
Affiliation(s)
- Jonghoon Park
- R&D Center, LG Life Sciences, Ltd, Daejeon, South Korea
| | - Eok Park
- R&D Center, LG Life Sciences, Ltd, Daejeon, South Korea
| | | | | | - Byung Gyu Kim
- R&D Center, LG Life Sciences, Ltd, Daejeon, South Korea
| | - Youngjoo Jung
- R&D Center, LG Life Sciences, Ltd, Daejeon, South Korea
| | - Tae Hun Kim
- R&D Center, LG Life Sciences, Ltd, Daejeon, South Korea
| | - Ji-Young Lim
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Eun Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Kwang-Ai Won
- R&D Center, LG Life Sciences, Ltd, Daejeon, South Korea.
| |
Collapse
|
18
|
Efficacy and Safety of Novel Agent-Based Therapies for Multiple Myeloma: A Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6848902. [PMID: 26949704 PMCID: PMC4753325 DOI: 10.1155/2016/6848902] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/15/2015] [Accepted: 12/22/2015] [Indexed: 12/26/2022]
Abstract
This study aimed at comparing bortezomib, thalidomide, and lenalidomide in patients with multiple myeloma (MM) for safety and efficacy using meta-analysis. This meta-analysis identified 17 randomized controlled trials (RCTs) including 6742 patients. These RCTs were separated according to the different agent-based regimens and to autologous stem-cell transplantation (ASCT). Complete response (CR), progression-free survival (PFS), overall survival (OS), and adverse events (AE) were combined. The total weighted risk ratio (RR) of CR was 3.29 [95% confidence interval (95% CI): 2.22–4.88] (P < 0.0001) for the novel agent-based regimens. These novel agent-based regimens showed greater benefit in terms of PFS of all subgroups irrespective of whether the patient received ASCT or not. The hazard ratio (HR) for PFS was 0.64 [95% CI: 0.60–0.69] (P < 0.00001). Improvements of OS could be found only in the bortezomib- and thalidomide-based regimens without ASCT. The pooled HRs were 0.74 [95% CI: 0.65–0.86] (P < 0.0001) and 0.80 [95% CI: 0.70–0.90] (P = 0.0004), respectively. Several AEs were shown more frequently in the novel agent-based regimens compared with controls such as hematologic events (neutropenia, anemia, and thrombocytopenia), gastrointestinal infection, peripheral neuropathy, thrombosis, and embolism events. In conclusion, in spite of the AEs, novel agent-based regimens are safe and effective for the treatment of MM.
Collapse
|
19
|
McBride A, Klaus JO, Stockerl-Goldstein K. Carfilzomib: a second-generation proteasome inhibitor for the treatment of multiple myeloma. Am J Health Syst Pharm 2015; 72:353-60. [PMID: 25694410 DOI: 10.2146/ajhp130281] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
PURPOSE The pharmacology, clinical efficacy, safety, cost, dosage and administration, and place in therapy of carfilzomib for the treatment of multiple myeloma (MM) are reviewed. SUMMARY Proteasome inhibition in MM has become a cornerstone in treatment regimens. Carfilzomib, a second-generation proteasome inhibitor, has demonstrated efficacy in patients with relapsed or refractory disease who have received at least two prior therapies including bortezomib and an immunomodulatory agent. Carfilzomib is an irreversible inhibitor and binds to a different site than bortezomib on the proteasome. A Phase II study evaluated 266 heavily pretreated patients with relapsed or refractory MM who had received at least two prior therapies, including bortezomib and either thalidomide or lenalidomide. The overall response rate was 23.7%, with a median duration of response of 7.8 months. The median overall survival time was 15.6 months. Carfilzomib has a similar adverse-effect profile to bortezomib, including anemia, thrombocytopenia, fatigue, dyspnea, and nausea; however, it does not result in the development or worsening of peripheral neuropathy. Carfilzomib is infused intravenously over 2-10 minutes for 2 consecutive days every week for three out of four weeks, with a 12-day rest period. Dosing is based on the patient's actual body surface area. Carfilzomib is available in 60-mg vials for single infusion. The total cost for a year of therapy is approximately $155,852. CONCLUSION Carfilzomib, a second-generation proteasome inhibitor that irreversibly inhibits the 26S proteasome, has shown efficacy in clinical studies of patients with relapsed or refractory MM, though the drug's role in the management of MM is not yet clear.
Collapse
Affiliation(s)
- Ali McBride
- Ali McBride, Pharm.D., M.S., BCPS, is Clinical Coordinator Hematology/Oncology, Department of Pharmacy, University of Arizona Cancer Center, Tucson. Jeff O. Klaus, Pharm.D., is Clinical Pharmacist, Hematologic Malignancies/Stem Cell Transplant, Department of Pharmacy, Barnes-Jewish Hospital, St. Louis, MO. Keith Stockerl-Goldstein, M.D.,is Associate Professor, Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis.
| | - Jeff O Klaus
- Ali McBride, Pharm.D., M.S., BCPS, is Clinical Coordinator Hematology/Oncology, Department of Pharmacy, University of Arizona Cancer Center, Tucson. Jeff O. Klaus, Pharm.D., is Clinical Pharmacist, Hematologic Malignancies/Stem Cell Transplant, Department of Pharmacy, Barnes-Jewish Hospital, St. Louis, MO. Keith Stockerl-Goldstein, M.D.,is Associate Professor, Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis
| | - Keith Stockerl-Goldstein
- Ali McBride, Pharm.D., M.S., BCPS, is Clinical Coordinator Hematology/Oncology, Department of Pharmacy, University of Arizona Cancer Center, Tucson. Jeff O. Klaus, Pharm.D., is Clinical Pharmacist, Hematologic Malignancies/Stem Cell Transplant, Department of Pharmacy, Barnes-Jewish Hospital, St. Louis, MO. Keith Stockerl-Goldstein, M.D.,is Associate Professor, Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis
| |
Collapse
|
20
|
Wang J, Qin T, Xu Z, Zhang Y, Zhang H, Fang L, Pan L, Hu N, Qu S, Li B, Xiao Z. [Long- term outcome of thalidomide and cyclosporine in patients with IPSS low/intermediate- 1 myelodysplastic syndromes]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:942-6. [PMID: 26632468 PMCID: PMC7342423 DOI: 10.3760/cma.j.issn.0253-2727.2015.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To investigate the long- term outcome of cyclosporin A (CsA) combined with thalidomide regime for Chinese patients with IPSS low/intermediate- 1 myelodysplastic syndromes (MDS) without del(5q)and the predictive variables which could impact the response to the therapy. METHODS Seventy-six MDS patients who were treated with these drugs at a single institute in China were retrospectively analyzed. The polymorphism of cereblon gene, rs1672753, was detected in patients of this cohort by PCR and direct sequencing. RESULTS A total of 53% of patients showed hematological improvement(HI)to the therapy. Thirty-one patients(31/73, 43%)achieved erythrocyte response(HI-E); 15 patients(15/50, 30%)achieved neutrophil response(HI-N); 18 patients(18/58, 31%)achieved platelet response(HI-P). Twenty-seven of the 50 patients(46%)who were dependent on red blood cell transfusion achieved HI- E and became independent of transfusion. The median duration of response among the responders was 22 months (range, 1- 131 + months). Bone marrow blasts ≤2% was the only factor associated with longer response duration in univariate analysis (P=0.010). There was no significant difference between the two groups of celeblon gene rs1672753 polymorphism either on the response rate or the response duration. The median survival of 67 patients without stem cell transplantation was 82 months. In multivariate analyses, factors significantly correlated with survival were IPSS-R(HR=3.461, 95%CI 1.126-10.639, P=0.030), age ≥ 60 y(HR=4.120, 95%CI 1.070-15.867, P=0.040)and HI-N(HR=7.733, 95%CI 1.007-59.396, P=0.049). CONCLUSION CsA combined with thalidomide regime could improve the anemia symptom in low/int-1 risk MDS patients without del(5q). The predictive value of cereblon gene polymorphism, rs1672753, could not be verified in this study.
Collapse
Affiliation(s)
- Jingya Wang
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Tiejun Qin
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Zefeng Xu
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Yue Zhang
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Hongli Zhang
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Liwei Fang
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Lijuan Pan
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Naibo Hu
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Shiqiang Qu
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Bing Li
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| | - Zhijian Xiao
- Institute of Hematology and Blood Disease Hospital, CAMS & PUMC, State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| |
Collapse
|
21
|
Huang YT, Cheng CC, Chiu TH, Lai PC. Therapeutic potential of thalidomide for gemcitabine-resistant bladder cancer. Int J Oncol 2015; 47:1711-24. [PMID: 26398114 DOI: 10.3892/ijo.2015.3155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/23/2015] [Indexed: 11/05/2022] Open
Abstract
Controversial effects of thalidomide for solid malignancies have been reported. In the present study, we evaluate the effects of thalidomide for transitional cell carcinoma (TCC), the most common type of bladder cancer. Thalidomide precipitates were observed when its DMSO solution was added to the culture medium. No precipitation was found when thalidomide was dissolved in 45% γ-cyclodextrin, and this concentration of γ-cyclodextrin elicited slight cytotoxicity on TCC BFTC905 and primary human urothelial cells. Thalidomide-γ-cyclodextrin complex exerted a concentration-dependent cytotoxicity in TCC cells, but was relatively less cytotoxic (with IC50 of 200 µM) in BFTC905 cells than the other 3 TCC cell lines, possibly due to upregulation of Bcl-xL and HIF-1α mediated carbonic anhydrase IX, and promotion of quiescence. Gemcitabine-resistant BFTC905 cells were chosen for additional experiments. Thalidomide induced apoptosis through downregulation of survivin and securin. The secretion of VEGF and TNF-α was ameliorated by thalidomide, but they did not affect cell proliferation. Immune-modulating lenalidomide and pomalidomide did not elicit cytotoxicity. In addition, cereblon did not play a role in the thalidomide effect. Oxidative DNA damage was triggered by thalidomide, and anti-oxidants reversed the effect. Thalidomide also inhibited TNF-α induced invasion through inhibition of NF-κB, and downregulation of effectors, ICAM-1 and MMP-9. Thalidomide inhibited the growth of BFTC905 xenograft tumors in SCID mice via induction of DNA damage and suppression of angiogenesis. Higher average body weight, indicating less chachexia, was observed in thalidomide treated group. Sedative effect was observed within one-week of treatment. These pre-clinical results suggest therapeutic potential of thalidomide for gemcitabine-resistant bladder cancer.
Collapse
Affiliation(s)
- Yen Ta Huang
- Department of Medicine, Tzu Chi University, Hualien, Taiwan, R.O.C
| | - Chuan Chu Cheng
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, R.O.C
| | - Ted H Chiu
- Department of Pharmacology, College of Medicine, Tzu Chi University, Hualien, Taiwan, R.O.C
| | - Pei Chun Lai
- Department of Medicine, Tzu Chi University, Hualien, Taiwan, R.O.C
| |
Collapse
|
22
|
Ullenhag GJ, Rossmann E, Liljefors M. A phase I dose-escalation study of lenalidomide in combination with gemcitabine in patients with advanced pancreatic cancer. PLoS One 2015; 10:e0121197. [PMID: 25837499 PMCID: PMC4383423 DOI: 10.1371/journal.pone.0121197] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/19/2015] [Indexed: 01/05/2023] Open
Abstract
Purpose Lenalidomide have both immunomodulatory and anti-angiogenic properties which could confer anti-cancer effects. The aim of this study was to assess the feasibility of combining lenalidomide with the standard treatment gemcitabine in pancreatic cancer patients with advanced disease. Patients and Methods Eligible patients had locally advanced or metastatic adenocarcinoma of the pancreas. Patients received lenalidomide days 1–21 orally and gemcitabine 1000 mg/m2 intravenously (days 1, 8 and 15), each 28 day cycle. Three cohorts of lenalidomide were examined (Cohort I = 15 mg, Cohort II = 20 mg and Cohort III = 25 mg daily). The maximum tolerated dose (MTD) of lenalidomide given in combination with gemcitabine was defined as the highest dose level at which no more than one out of four (25%) subjects experiences a dose-limiting toxicity (DLT). Patients should also be able to receive daily low molecular weight heparin (LMWH) (e.g. dalteparin 5000 IU s.c. daily) as a prophylactic anticoagulant for venous thromboembolic events (VTEs). Twelve patients (n = 4, n = 3 and n = 5 in cohort I, II and III, respectively) were enrolled in this study. Results Median duration of treatment was 11 weeks (range 1–66), and median number of treatment cycles were three (range 1–14). The only DLT was a cardiac failure grade 3 in cohort III. Frequent treatment-related adverse events (AEs) (all grades) included neutropenia, leucopenia and fatigue (83% each, but there was no febrile neutropenia); thrombocytopenia (75%); dermatological toxicity (75%); diarrhea and nausea (42% each); and neuropathy (42%). Discussion This phase I study demonstrates the feasibility of the combination of lenalidomide and gemcitabine as first-line treatment in patients with advanced pancreatic cancer. The tolerability profile demonstrated in the dose escalation schedule of lenalidomide suggests the dosing of lenalidomide to be 25 mg daily on days 1–21 with standard dosing of gemcitabine and merits further evaluation in a phase II trial. Trial Registration ClinicalTrials.gov NCT01547260
Collapse
Affiliation(s)
- Gustav J. Ullenhag
- Department of Radiology, Oncology and Radiation Science, Section of Oncology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Entrance 78, 751 85 Uppsala, Sweden
| | - Eva Rossmann
- Department of Oncology and Pathology (Radiumhemmet), Cancer Centre Karolinska, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Maria Liljefors
- Department of Oncology and Pathology (Radiumhemmet), Cancer Centre Karolinska, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
23
|
Obrist F, Manic G, Kroemer G, Vitale I, Galluzzi L. Trial Watch: Proteasomal inhibitors for anticancer therapy. Mol Cell Oncol 2015; 2:e974463. [PMID: 27308423 PMCID: PMC4904962 DOI: 10.4161/23723556.2014.974463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/12/2023]
Abstract
The so-called "ubiquitin-proteasome system" (UPS) is a multicomponent molecular apparatus that catalyzes the covalent attachment of several copies of the small protein ubiquitin to other proteins that are generally (but not always) destined to proteasomal degradation. This enzymatic cascade is crucial for the maintenance of intracellular protein homeostasis (both in physiological conditions and in the course of adaptive stress responses), and regulates a wide array of signaling pathways. In line with this notion, defects in the UPS have been associated with aging as well as with several pathological conditions including cardiac, neurodegenerative, and neoplastic disorders. As transformed cells often experience a constant state of stress (as a result of the hyperactivation of oncogenic signaling pathways and/or adverse microenvironmental conditions), their survival and proliferation are highly dependent on the integrity of the UPS. This rationale has driven an intense wave of preclinical and clinical investigation culminating in 2003 with the approval of the proteasomal inhibitor bortezomib by the US Food and Drug Administration for use in multiple myeloma patients. Another proteasomal inhibitor, carfilzomib, is now licensed by international regulatory agencies for use in multiple myeloma patients, and the approved indications for bortezomib have been extended to mantle cell lymphoma. This said, the clinical activity of bortezomib and carfilzomib is often limited by off-target effects, innate/acquired resistance, and the absence of validated predictive biomarkers. Moreover, the antineoplastic activity of proteasome inhibitors against solid tumors is poor. In this Trial Watch we discuss the contribution of the UPS to oncogenesis and tumor progression and summarize the design and/or results of recent clinical studies evaluating the therapeutic profile of proteasome inhibitors in cancer patients.
Collapse
Affiliation(s)
- Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “Tor Vergata”
| | - Lorenzo Galluzzi
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
24
|
Patel UH, Mir MA, Sivik JK, Raheja D, Pandey MK, Talamo G. Central neurotoxicity of immunomodulatory drugs in multiple myeloma. Hematol Rep 2015; 7:5704. [PMID: 25852850 PMCID: PMC4378207 DOI: 10.4081/hr.2015.5704] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 01/12/2023] Open
Abstract
Immunomodulatory drugs (IMiDs) currently used in the treatment of multiple myeloma, are thalidomide, lenalidomide and pomalidomide. One of the most common side effects of thalidomide is neurotoxicity, predominantly in the form of peripheral neuropathy. We report 6 cases of significant central neurotoxicity associated with IMiD therapy. Treatment with thalidomide (1 patient), lenalidomide (4 patients), and pomalidomide (1 patient) was associated with various clinical manifestations of central neurotoxicity, including reversible coma, amnesia, expressive aphasia, and dysarthria. Central neurotoxicity should be recognized as an important side effect of IMiD therapy.
Collapse
|
25
|
Deng S, Xu Y, An G, Sui W, Zou D, Zhao Y, Qi J, Li F, Hao M, Qiu L. Features of extramedullary disease of multiple myeloma: high frequency of p53 deletion and poor survival: a retrospective single-center study of 834 cases. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2015; 15:286-91. [PMID: 25640025 DOI: 10.1016/j.clml.2014.12.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/24/2014] [Accepted: 12/29/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Multiple myeloma (MM) is a heterogeneous disease in which most patients have myeloma restricted to the bone marrow, and some patients develop extramedullary disease (EMD) at the time of diagnosis or during follow-up, and show different clinical characteristics and a dismal prognosis. PATIENTS AND METHODS We studied 834 consecutive MM patients in a single center in China and compared clinical features of patients with and without EMD. RESULTS In general, the prevalence of EMD was 4.8% at the time of diagnosis and 3.4% during follow-up, with a significant increase in recent years. MM patients with EMD at the time of diagnosis had remarkably greater prevalence of P53 deletion determined using fluorescence in situ hybridization (FISH) analysis (34.5% vs. 11.9%; P = .037) and higher level of lactate dehydrogenase (LDH) (P = .003) compared with patients without EMD. EMD relapse/progression during follow-up was correlated with EMD presentation at diagnosis, immunoglobulin (Ig)D subtype and P53 deletion in FISH analysis, but not previous treatment (thalidomide, bortizomib, or transplantation). With respect to prognosis, multivariate analysis showed that EMD was an independent adverse prognostic factor. The overall survival of patients with and without EMD at diagnosis were 16.5 and 40 months, respectively (P < .001), and the time to disease progression of the 2 groups was 11.5 and 25 months, respectively (P < .001). CONCLUSION MM patients with EMD at the time of diagnosis showed remarkably greater prevalence of P53 deletion in FISH analysis and higher LDH levels. EMD relapse/progression was correlated with EMD presentation at diagnosis, IgD subtype, and P53 deletion in FISH analysis, but not previous exposure to new drugs or transplantation. The presence of EMD involvement negatively affected survival.
Collapse
Affiliation(s)
- Shuhui Deng
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Gang An
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Yaozhong Zhao
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Junyuan Qi
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Fei Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College.
| |
Collapse
|
26
|
Teh BW, Teng JC, Urbancic K, Grigg A, Harrison SJ, Worth LJ, Slavin MA, Thursky KA. Invasive fungal infections in patients with multiple myeloma: a multi-center study in the era of novel myeloma therapies. Haematologica 2014; 100:e28-31. [PMID: 25304609 DOI: 10.3324/haematol.2014.114025] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Benjamin W Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne Sir Peter MacCallum Department of Oncology, University of Melbourne
| | - Jasmine C Teng
- Department of Infectious Diseases, St Vincent's Hospital Melbourne, Fitzroy
| | | | - Andrew Grigg
- Department of Haematology, Austin Hospital, Heidelberg
| | - Simon J Harrison
- Sir Peter MacCallum Department of Oncology, University of Melbourne Department of Haematology, Peter MacCallum Cancer Centre, East Melbourne
| | - Leon J Worth
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne Department of Medicine, University of Melbourne
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne Department of Medicine, University of Melbourne Victorian Infectious Diseases Service, Peter Doherty Institute for Infection and Immunity, Parkville, Australia
| | - Karin A Thursky
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne Department of Medicine, University of Melbourne Victorian Infectious Diseases Service, Peter Doherty Institute for Infection and Immunity, Parkville, Australia
| |
Collapse
|
27
|
Abstract
Pomalidomide (Pomalyst(®)) is a synthetic compound derived by modifying the chemical structure of thalidomide to improve its potency and reduce its side effects and third drug in the class of immunomodulatory drugs. Pomalidomide is under global development with Celgene Corporation, was approved by the U.S. Food and Drug Administration on February 8, 2013 to treat patients with relapsed and refractory multiple myeloma (MM) who have received at least two prior therapies including bortezomib and lenalidomide. In October 2009, it has found orphan designation for the treatment of relapsed and refractory MM by the EMA, and on August 2013, marketing authorization has issued in Europe by gaining a positive response. It inhibits myeloma cell growth and angiogenesis directly. Pomalidomide is the latest myeloma cell growth inhibitor to be approved in both USA and EU. The predominant side effects are thrombocytopenia, neuropathy, and deep vein thrombosis. Pomalidomide is also being investigated in patients with amyloidosis, prostate cancer, small cell lung cancer, pancreatic cancer, graft-versus-host disease, and Waldenstrom's macroglobulinemia. This article reviews the available information on pomalidomide with respect to its clinical pharmacology, mechanism of action, pharmacokinetics, pharmacodynamics, metabolism, pre-clinical studies, and clinical trials.
Collapse
|
28
|
Bae J, Prabhala R, Voskertchian A, Brown A, Maguire C, Richardson P, Dranoff G, Anderson KC, Munshi NC. A multiepitope of XBP1, CD138 and CS1 peptides induces myeloma-specific cytotoxic T lymphocytes in T cells of smoldering myeloma patients. Leukemia 2014; 29:218-29. [PMID: 24935722 PMCID: PMC4237716 DOI: 10.1038/leu.2014.159] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/24/2014] [Accepted: 04/02/2014] [Indexed: 02/07/2023]
Abstract
We evaluated a cocktail of HLA-A2-specific peptides including heteroclitic XBP1 US184-192 (YISPWILAV), heteroclitic XBP1 SP367-375 (YLFPQLISV), native CD138260-268 (GLVGLIFAV) and native CS1239-247 (SLFVLGLFL), for their ability to elicit multipeptide-specific cytotoxic T lymphocytes (MP-CTLs) using T cells from smoldering multiple myeloma (SMM) patients. Our results demonstrate that MP-CTLs generated from SMM patients' T cells show effective anti-MM responses including CD137 (4-1BB) upregulation, CTL proliferation, interferon-γ production and degranulation (CD107a) in an HLA-A2-restricted and peptide-specific manner. Phenotypically, we observed increased total CD3(+)CD8(+) T cells (>80%) and cellular activation (CD69(+)) within the memory SMM MP-CTL (CD45RO(+)/CD3(+)CD8(+)) subset after repeated multipeptide stimulation. Importantly, SMM patients could be categorized into distinct groups by their level of MP-CTL expansion and antitumor activity. In high responders, the effector memory (CCR7(-)CD45RO(+)/CD3(+)CD8(+)) T-cell subset was enriched, whereas the remaining responders' CTL contained a higher frequency of the terminal effector (CCR7(-)CD45RO(-)/CD3(+)CD8(+)) subset. These results suggest that this multipeptide cocktail has the potential to induce effective and durable memory MP-CTL in SMM patients. Therefore, our findings provide the rationale for clinical evaluation of a therapeutic vaccine to prevent or delay progression of SMM to active disease.
Collapse
Affiliation(s)
- J Bae
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - R Prabhala
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA [3] VA Boston Healthcare System, Boston, MA, USA
| | - A Voskertchian
- Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - A Brown
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - C Maguire
- Tufts University School of Medicine, Boston, MA, USA
| | - P Richardson
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - G Dranoff
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - K C Anderson
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - N C Munshi
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA [3] VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
29
|
|
30
|
Heparin-binding epidermal growth factor-like growth factor/diphtheria toxin receptor in normal and neoplastic hematopoiesis. Toxins (Basel) 2013; 5:1180-1201. [PMID: 23888518 PMCID: PMC3717776 DOI: 10.3390/toxins5061180] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Heparin-binding EGF-like growth factor (HB-EGF) belongs to the EGF family of growth factors. It is biologically active either as a molecule anchored to the membrane or as a soluble form released by proteolytic cleavage of the extracellular domain. HB-EGF is involved in relevant physiological and pathological processes spanning from proliferation and apoptosis to morphogenesis. We outline here the main activities of HB-EGF in connection with normal or neoplastic differentiative or proliferative events taking place primitively in the hematopoietic microenvironment.
Collapse
|
31
|
Roeven MWH, Hobo W, Schaap N, Dolstra H. Immunotherapeutic approaches to treat multiple myeloma. Hum Vaccin Immunother 2013; 10:896-910. [PMID: 24335570 PMCID: PMC4896532 DOI: 10.4161/hv.27380] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 11/21/2013] [Accepted: 11/29/2013] [Indexed: 11/19/2022] Open
Abstract
Cellular immunotherapy can be an effective adjuvant treatment for multiple myeloma (MM), as demonstrated by induction of durable remissions after allogeneic stem cell transplantation. However, anti-myeloma immunity is often hampered by suppressive mechanisms in the tumor micro-environment resulting in relapse or disease progression. To overcome this immunosuppression, new cellular immunotherapies have been developed, based on the important effector cells in anti-myeloma immunity, namely T cells and natural killer cells. These effectors can be modulated to improve their functionality, activated by dendritic cell vaccines, or combined with immune stimulating antibodies or immunomodulatory drugs to enhance their efficacy. In this review, we discuss promising pre-clinical and clinical data in the field of cellular immunotherapy in MM. In addition, we address the potential of combining these strategies with other therapies to maximize clinical effects without increasing toxicity. The reviewed therapies might pave the way to effective personalized treatments for MM patients.
Collapse
Affiliation(s)
- Mieke WH Roeven
- Department of Hematology; Radboud University Medical Center; Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine—Laboratory of Hematology Radboud; University Medical Center; Nijmegen, The Netherlands
| | - Nicolaas Schaap
- Department of Hematology; Radboud University Medical Center; Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine—Laboratory of Hematology Radboud; University Medical Center; Nijmegen, The Netherlands
| |
Collapse
|
32
|
Abstract
The incorporation of novel agents such as bortezomib and lenalidomide into initial therapy for multiple myeloma has improved the response rate of induction regimens. Also, these drugs are being increasingly used in the peri-transplant setting for transplant-eligible patients, and as part of consolidation and/or maintenance after front-line treatment, including in transplant-ineligible patients. Together, these and other strategies have contributed to a prolongation of progression-free survival (PFS) and overall survival (OS) in myeloma patients, and an increasing proportion are able to sustain a remission for many years. Despite these improvements, however, the vast majority of patients continue to suffer relapses, which suggests a prominent role for either primary, innate drug resistance, or secondary, acquired drug resistance. As a result, there remains a strong need to develop new proteasome inhibitors and immunomodulatory agents, as well as new drug classes, which would be effective in the relapsed and/or refractory setting, and overcome drug resistance. This review will focus on novel drugs that have reached phase III trials, including carfilzomib and pomalidomide, which have recently garnered regulatory approvals. In addition, agents that are in phase II or III, potentially registration-enabling trials will be described as well, to provide an overview of the possible landscape in the relapsed and/or refractory arena over the next 5 years.
Collapse
Affiliation(s)
- Robert Z Orlowski
- Department of Lymphoma/Myeloma, and Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
33
|
Pomalidomide is nonteratogenic in chicken and zebrafish embryos and nonneurotoxic in vitro. Proc Natl Acad Sci U S A 2013; 110:12703-8. [PMID: 23858438 DOI: 10.1073/pnas.1307684110] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Thalidomide and its analog, Lenalidomide, are in current use clinically for treatment of multiple myeloma, complications of leprosy and cancers. An additional analog, Pomalidomide, has recently been licensed for treatment of multiple myeloma, and is purported to be clinically more potent than either Thalidomide or Lenalidomide. Using a combination of zebrafish and chicken embryos together with in vitro assays we have determined the relative anti-inflammatory activity of each compound. We demonstrate that in vivo embryonic assays Pomalidomide is a significantly more potent anti-inflammatory agent than either Thalidomide or Lenalidomide. We tested the effect of Pomalidomide and Lenalidomide on angiogenesis, teratogenesis, and neurite outgrowth, known detrimental effects of Thalidomide. We found that Pomalidomide, displays a high degree of cell specificity, and has no detectable teratogenic, antiangiogenic or neurotoxic effects at potent anti-inflammatory concentrations. This is in marked contrast to Thalidomide and Lenalidomide, which had detrimental effects on blood vessels, nerves, and embryonic development at anti-inflammatory concentrations. This work has implications for Pomalidomide as a treatment for conditions Thalidomide and Lenalidomide treat currently.
Collapse
|
34
|
|
35
|
Sardnal V, Rouquette A, Kaltenbach S, Bally C, Chesnais V, Leschi C, Ades L, Santini V, Park S, Toma A, Fenaux P, Dreyfus F, Fontenay M, Kosmider O. A G polymorphism in the CRBN gene acts as a biomarker of response to treatment with lenalidomide in low/int-1 risk MDS without del(5q). Leukemia 2013; 27:1610-3. [DOI: 10.1038/leu.2013.59] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|