1
|
Saleh Al Ward MM, Abdallah AE, Zayed MF, Ayyad RR, Abdelghany TM, Bakhotmah DA, El-Zahabi MA. New immunomodulatory anticancer quinazolinone-based thalidomide analogs: design, synthesis and biological evaluation. Future Med Chem 2024:1-11. [PMID: 39530517 DOI: 10.1080/17568919.2024.2419361] [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: 07/13/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Aim: The current work is an extension to our previous work for the development of new thalidomide analogs.Materials & methods: Quinazolinone-based molecules carrying a glutarimide moiety have been designed, synthesized and biologically evaluated for immunomodulatory and anticancer activity.Results: Compounds 7d and 12 showed considerable immunomodulatory properties in comparison to thalidomide. 7d and 12 significantly reduced TNF-α levels in HepG-2 cells from 162.5 to 57.4 pg/ml and 49.2 pg/ml, respectively, compared with 53.1 pg/ml reported for thalidomide. Moreover, they caused 69.33 and 77.74% reduction in NF-κB P65, respectively, compared with 60.26% reduction for thalidomide. Similarly, they reduced VEGF from 432.5 to 161.3 pg/ml and 132.8 pg/ml, respectively, in comparison to 153.2 pg/ml reported for thalidomide. The two new derivatives, 7d and 12 also showed about eightfold increases in caspase-8 levels in cells treated with them. These results were slightly better than those of thalidomide. The obtained results revealed that Compound 12 had better immunomodulatory properties than thalidomide, with stronger effects on TNF-α, NF-κB P65, VEGF and caspase-8.Conclusion: This work indicates that compounds 7d and 12 have interesting biological properties that should be further evaluated and modified in order to develop clinically useful thalidomide analogs.
Collapse
Affiliation(s)
- Maged Mohammed Saleh Al Ward
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Abdallah E Abdallah
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Mohamed F Zayed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
- Pharmaceutical Sciences Department, Fakeeh College for Medical Sciences, Jeddah, 21461, Saudi Arabia
| | - Rezk R Ayyad
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
- College of Pharmacy, University of Hilla, Babylon, Iraq
| | - Tamer M Abdelghany
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, 11785, Egypt
- Pharmacology & Toxicology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | | | - Mohamed Ayman El-Zahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| |
Collapse
|
2
|
Tang B, Ma W, Lin Y. Emerging applications of anti-angiogenic nanomaterials in oncotherapy. J Control Release 2023; 364:61-78. [PMID: 37871753 DOI: 10.1016/j.jconrel.2023.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Angiogenesis is the process of generating new blood vessels from pre-existing vasculature. Under normal conditions, this process is delicately controlled by pro-angiogenic and anti-angiogenic factors. Tumor cells can produce plentiful pro-angiogenic molecules promoting pathological angiogenesis for uncontrollable growth. Therefore, anti-angiogenic therapy, which aims to inhibit tumor angiogenesis, has become an attractive approach for oncotherapy. However, classic anti-angiogenic agents have several limitations in clinical use, such as lack of specific targeting, low bioavailability, and poor therapeutic outcomes. Hence, alternative angiogenic inhibitors are highly desired. With the emergence of nanotechnology, various nanomaterials have been designed for anti-angiogenesis purposes, offering promising features like excellent targeting capabilities, reduced side effects, and enhanced therapeutic efficacy. In this review, we describe tumor vascular features, discuss current dilemma of traditional anti-angiogenic medicines in oncotherapy, and underline the potential of nanomaterials in tumor anti-angiogenic therapy. Moreover, we discuss the current challenges of anti-angiogenic cancer treatment. We expect that this summary of anti-angiogenic nanomaterials in oncotherapy will offer valuable insights, facilitating their extensive applications in the future.
Collapse
Affiliation(s)
- Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Forster S, Radpour R, Ochsenbein AF. Molecular and immunological mechanisms of clonal evolution in multiple myeloma. Front Immunol 2023; 14:1243997. [PMID: 37744361 PMCID: PMC10516567 DOI: 10.3389/fimmu.2023.1243997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by the proliferation of clonal plasma cells in the bone marrow (BM). It is known that early genetic mutations in post-germinal center B/plasma cells are the cause of myelomagenesis. The acquisition of additional chromosomal abnormalities and distinct mutations further promote the outgrowth of malignant plasma cell populations that are resistant to conventional treatments, finally resulting in relapsed and therapy-refractory terminal stages of MM. In addition, myeloma cells are supported by autocrine signaling pathways and the tumor microenvironment (TME), which consists of diverse cell types such as stromal cells, immune cells, and components of the extracellular matrix. The TME provides essential signals and stimuli that induce proliferation and/or prevent apoptosis. In particular, the molecular pathways by which MM cells interact with the TME are crucial for the development of MM. To generate successful therapies and prevent MM recurrence, a thorough understanding of the molecular mechanisms that drive MM progression and therapy resistance is essential. In this review, we summarize key mechanisms that promote myelomagenesis and drive the clonal expansion in the course of MM progression such as autocrine signaling cascades, as well as direct and indirect interactions between the TME and malignant plasma cells. In addition, we highlight drug-resistance mechanisms and emerging therapies that are currently tested in clinical trials to overcome therapy-refractory MM stages.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F. Ochsenbein
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Mabrouk RR, Abdallah AE, Mahdy HA, El-Kalyoubi SA, Kamal OJ, Abdelghany TM, Zayed MF, Alshaeri HK, Alasmari MM, El-Zahabi MA. Design, Synthesis, and Biological Evaluation of New Potential Unusual Modified Anticancer Immunomodulators for Possible Non-Teratogenic Quinazoline-Based Thalidomide Analogs. Int J Mol Sci 2023; 24:12416. [PMID: 37569792 PMCID: PMC10418715 DOI: 10.3390/ijms241512416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Sixteen new thalidomide analogs were synthesized. The new candidates showed potent in vitro antiproliferative activities against three human cancer cell lines, namely hepatocellular carcinoma (HepG-2), prostate cancer (PC3), and breast cancer (MCF-7). It was found that compounds XII, XIIIa, XIIIb, XIIIc, XIIId, XIVa, XIVb, and XIVc showed IC50 values ranging from 2.03 to 13.39 µg/mL, exhibiting higher activities than thalidomide against all tested cancer cell lines. Compound XIIIa was the most potent candidate, with an IC50 of 2.03 ± 0.11, 2.51 ± 0.2, and 0.82 ± 0.02 µg/mL compared to 11.26 ± 0.54, 14.58 ± 0.57, and 16.87 ± 0.7 µg/mL for thalidomide against HepG-2, PC3, and MCF-7 cells, respectively. Furthermore, compound XIVc reduced the expression of NFκB P65 levels in HepG-2 cells from 278.1 pg/mL to 63.1 pg/mL compared to 110.5 pg/mL for thalidomide. Moreover, compound XIVc induced an eightfold increase in caspase-8 levels with a simultaneous decrease in TNF-α and VEGF levels in HepG-2 cells. Additionally, compound XIVc induced apoptosis and cell cycle arrest. Our results reveal that the new candidates are potential anticancer candidates, particularly XIIIa and XIVc. Consequently, they should be considered for further evaluation for the development of new anticancer drugs.
Collapse
Affiliation(s)
- Reda R. Mabrouk
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt; (R.R.M.); (A.E.A.); (H.A.M.)
- Directorate of Health Affairs in Buhaira-Clinical Research Department, Ministry of Health and Population, Damanhour 22511, Egypt
| | - Abdallah E. Abdallah
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt; (R.R.M.); (A.E.A.); (H.A.M.)
| | - Hazem A. Mahdy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt; (R.R.M.); (A.E.A.); (H.A.M.)
| | - Samar A. El-Kalyoubi
- Department of Pharmaceutical Organic Chemistry, Port Said University, Port Said 42511, Egypt;
| | - Omar Jamal Kamal
- King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21461, Saudi Arabia;
| | - Tamer M. Abdelghany
- Pharmacology & Toxicology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt;
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo 11785, Egypt
| | - Mohamed F. Zayed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt; (R.R.M.); (A.E.A.); (H.A.M.)
- Pharmaceutical Sciences Department, Fakeeh College for Medical Sciences, Jeddah 21461, Saudi Arabia;
| | - Heba K. Alshaeri
- Pharmaceutical Sciences Department, Fakeeh College for Medical Sciences, Jeddah 21461, Saudi Arabia;
| | - Moudi M. Alasmari
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah 21461, Saudi Arabia;
- King Abdullah International Medical Research Center (KAIMRC), Jeddah 21423, Saudi Arabia
| | - Mohamed Ayman El-Zahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt; (R.R.M.); (A.E.A.); (H.A.M.)
| |
Collapse
|
5
|
Lin JY, Liu HJ, Wu Y, Jin JM, Zhou YD, Zhang H, Nagle DG, Chen HZ, Zhang WD, Luan X. Targeted Protein Degradation Technology and Nanomedicine: Powerful Allies against Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207778. [PMID: 36693784 DOI: 10.1002/smll.202207778] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/08/2023] [Indexed: 05/04/2023]
Abstract
Targeted protein degradation (TPD) is an emerging therapeutic strategy with the potential of targeting undruggable pathogenic proteins. After the first proof-of-concept proteolysis-targeting chimeric (PROTAC) molecule was reported, the TPD field has entered a new era. In addition to PROTAC, numerous novel TPD strategies have emerged to expand the degradation landscape. However, their physicochemical properties and uncontrolled off-target side effects have limited their therapeutic efficacy, raising concerns regarding TPD delivery system. The combination of TPD and nanotechnology offers great promise in improving safety and therapeutic efficacy. This review provides an overview of novel TPD technologies, discusses their clinical applications, and highlights the trends and perspectives in TPD nanomedicine.
Collapse
Affiliation(s)
- Jia-Yi Lin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hai-Jun Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ye Wu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jin-Mei Jin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu-Dong Zhou
- Department of Chemistry and Biochemistry, College of Liberal Arts, University of Mississippi, University-1848, Boston, MA, 38677, USA
| | - Hong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dale G Nagle
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University-1848, Boston, MA, 38677, USA
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
6
|
Abdallah AE, Eissa IH, Mehany AB, Sakr H, Atwa A, El-Adl K, El-Zahabi MA. Immunomodulatory quinazoline-based thalidomide analogs: Design, synthesis, apoptosis and anticancer evaluations. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
7
|
Lv X, Mao Y, Cao S, Feng Y. Animal models of chemotherapy-induced peripheral neuropathy for hematological malignancies: A review. IBRAIN 2022; 9:72-89. [PMID: 37786517 PMCID: PMC10529012 DOI: 10.1002/ibra.12086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 10/04/2023]
Abstract
Chemotherapy is one of the main treatments for hematologic malignancies. However, chemotherapy-induced peripheral neuropathy (CIPN) is one of the most common long-term toxic reactions in chemotherapy, and the occurrence of CIPN affects patients' quality of life and can cause interruption of chemotherapy in severe cases, thus reducing the efficacy of chemotherapy. We currently summarize the existing CIPN animal models, including the characteristics of several common animal models such as bortezomib-induced peripheral neuropathy, vincristine-induced peripheral neuropathy, and oxaliplatin-induced peripheral neuropathy. It was found that CIPN may lead to behavioral, histopathological, and neurophysiological changes inducing peripheral neuropathy. However, the mechanism of CIPN has not been fully elucidated, especially the prevention and treatment protocols need to be improved. Therefore, this review article summarizes the progress of research on CIPN animal models and the possible mechanisms and treatment of CIPN.
Collapse
Affiliation(s)
- Xiaoli Lv
- Department of HematologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yingwei Mao
- Department of BiologyPenn State UniversityUniversity ParkPennsylvaniaUSA
| | - Song Cao
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- Department of Pain MedicineAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Yonghuai Feng
- Department of HematologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
8
|
Minařík J, Ševčíková S. Immunomodulatory Agents for Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14235759. [PMID: 36497241 PMCID: PMC9739922 DOI: 10.3390/cancers14235759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
The treatment of multiple myeloma (MM) has undergone a significant paradigm shift in the last 20 years, from conventional chemotherapy to more tumor-specific treatments, based on the interference with pathogenesis of the malignant clone as well as the bone microenvironment [...].
Collapse
Affiliation(s)
- Jiří Minařík
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, 779 00 Olomouc, Czech Republic
- Correspondence:
| | - Sabina Ševčíková
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| |
Collapse
|
9
|
Kotb AR, Bakhotmah DA, Abdallah AE, Elkady H, Taghour MS, Eissa IH, El-Zahabi MA. Design, synthesis, and biological evaluation of novel bioactive thalidomide analogs as anticancer immunomodulatory agents. RSC Adv 2022; 12:33525-33539. [PMID: 36505721 PMCID: PMC9680624 DOI: 10.1039/d2ra06188k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022] Open
Abstract
Cancer is still a dangerous disease with a high mortality rate all over the world. In our attempt to develop potential anticancer candidates, new quinazoline and phthalazine based compounds were designed and synthesized. The new derivatives were built in line with the pharmacophoric features of thalidomide. The new derivatives as well as thalidomide were examined against three cancer cell lines, namely: hepatocellular carcinoma (HepG-2), breast cancer (MCF-7) and prostate cancer (PC3). Then the effects on the expression levels of caspase-8, VEGF, NF-κB P65, and TNF-α in HepG-2 cells were evaluated. The biological data revealed the high importance of phthalazine based compounds (24a-c), which were far better than thalidomide with regard to the antiproliferative activity. 24b showed IC50 of 2.51, 5.80 and 4.11 μg mL-1 compared to 11.26, 14.58, and 16.87 μg mL-1 for thalidomide against the three cell lines respectively. 24b raised caspase-8 level by about 7 folds, compared to 8 folds reported for thalidomide. Also, VEGF level in HepG-2 cells treated with 24b was 185.3 pg mL-1, compared to 432.5 pg mL-1 in control cells. Furthermore, the immunomodulatory properties were proven to 24b, which reduced TNF-α level by approximately half. At the same time, NF-κB P65 level in HepG-2 cells treated with 24b was 76.5 pg mL-1 compared to 278.1 and 110.5 pg mL-1 measured for control cells and thalidomide treated HepG-2 cells respectively. Moreover, an in vitro viability study against Vero non-cancerous cell line was investigated and the results reflected a high safety profile of all tested compounds. This work suggests 24b as a promising lead compound for development of new immunomodulatory anticancer agents.
Collapse
Affiliation(s)
- Anas Ramadan Kotb
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar UniversityCairo11884Egypt
| | - Dina A. Bakhotmah
- Department of Chemistry, Faculty of Science, King Abdulaziz UniversityJeddahSaudi Arabia
| | - Abdallah E. Abdallah
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar UniversityCairo11884Egypt
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar UniversityCairo11884Egypt
| | - Mohammed S. Taghour
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar UniversityCairo11884Egypt
| | - Ibrahim. H. Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar UniversityCairo11884Egypt
| | - Mohamed Ayman El-Zahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar UniversityCairo11884Egypt
| |
Collapse
|
10
|
Tao R, Peng X, Liu X, Su J, Lang G, Huang Y, Zhang Y, Zhu B. Lenalidomide Improves Cognitive Function and Reduces Immune Reconstitution Inflammatory Syndrome in HIV-1-Related Cryptococcal Meningitis. J Inflamm Res 2022; 15:2891-2899. [PMID: 35586751 PMCID: PMC9109900 DOI: 10.2147/jir.s353463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/27/2022] [Indexed: 11/26/2022] Open
Abstract
Cryptococcal meningitis (CM) is a common opportunistic infection in patients with acquired immune deficiency syndrome. Although there is a standardized treatment for CM, some patients still have CM-associated immune reconstitution inflammatory syndrome (IRIS) after anti-cryptococcal and antiretroviral therapy, which manifests as cognitive impairment. We report two cases of CM-associated IRIS in human immunodeficiency virus (HIV) patients that were treated with lenalidomide. The treatment yielded a rapid clinical remission and improved cognitive function in both patients; their Montreal Cognitive Assessment (MoCA) and International HIV Dementia Scale (IHDS) scores improved. Furthermore, we evaluated changes in 32 cytokines in the cerebrospinal fluid of two patients and found that both MoCA and IHDS were significantly negatively correlated with inflammation-related factors (growth-related oncogene, interleukin [IL]-10, IL-2, IL-8, macrophage inflammatory protein-1β, tumor necrosis factor [TNF]-α) and significantly positively correlated with dementia-related factors (αβ42 and total tau). Our study reveals the potential of lenalidomide in treating cognitive impairment caused by immune-mediated inflammation in patients with HIV-CM. Moreover, we speculate that lenalidomide improves cognitive function by regulating intracranial inflammation via multiple pathways, not only by TNF-α blocking.
Collapse
Affiliation(s)
- Ran Tao
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaorong Peng
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiang Liu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Junwei Su
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Guanjing Lang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Ying Huang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yafei Zhang
- PET Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Biao Zhu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- Correspondence: Biao Zhu, Tel +0086-571-87236417, Fax +0086-571-87236416, Email
| |
Collapse
|
11
|
Singh R, Adhya P, Sharma SS. Redox-sensitive TRP channels: a promising pharmacological target in chemotherapy-induced peripheral neuropathy. Expert Opin Ther Targets 2021; 25:529-545. [PMID: 34289785 DOI: 10.1080/14728222.2021.1956464] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chemotherapy-induced peripheral neuropathy (CIPN) and its related pain is a major side effect of certain chemotherapeutic agents used in cancer treatment. Available analgesics are mostly symptomatic, and on prolonged treatment, patients become refractive to them. Hence, the development of improved therapeutics that act on novel therapeutic targets is necessary. Potential targets include the redox-sensitive TRP channels [e.g. TRPA1, TRPC5, TRPC6, TRPM2, TRPM8, TRPV1, TRPV2, and TRPV4] which are activated under oxidative stress associated with CIPN. AREAS COVERED We have examined numerous neuropathy-inducing cancer chemotherapeutics and their pathophysiological mechanisms. Oxidative stress and its downstream targets, the redox-sensitive TRP channels, together with their potential pharmacological modulators, are discussed. Finally, we reflect upon the barriers to getting new therapeutic approaches into the clinic. The literature search was conducted in PubMed upto and including April 2021. EXPERT OPINION Redox-sensitive TRP channels are a promising target in CIPN. Pharmacological modulators of these channels have reduced pain in preclinical models and in clinical studies. Clinical scrutiny suggests that TRPA1, TRPM8, and TRPV1 are the most promising targets because of their pain-relieving potential. In addition to the analgesic effect, TRPV1 agonist-Capsaicin possesses a disease-modifying effect in CIPN through its restorative property in damaged sensory nerves.
Collapse
Affiliation(s)
- Ramandeep Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Pratik Adhya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| |
Collapse
|
12
|
Lu J, Chen W. Cost-effectiveness of lenalidomide plus low-dose dexamethasone for newly diagnosed multiple myeloma patients ineligible for stem cell transplantation in China. J Comp Eff Res 2019; 8:979-992. [PMID: 31232089 DOI: 10.2217/cer-2019-0052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To assess the cost-effectiveness of lenalidomide plus low dose dexamethasone (Rd) relative to bortezomib-contained therapy (BCT) for newly diagnosed multiple myeloma patients ineligible for stem cell transplantation (ndMM) in China. Materials & methods: A literature review was conducted to identify appropriate evidence for developing a cost-effectiveness model comparing Rd with BCT for lifetime health outcomes and direct medical costs in Chinese ndMM patients. Results: The estimated incremental cost-effectiveness ratio per gained quality-adjusted life years for Rd versus BCT was ¥49,793. The chance for Rd to be cost effective, under the cost-effectiveness thresholds of three-times the 2018 Chinese gross domestic goods per capita, was 90.8%. Conclusion: The cost-effectiveness of Rd relative to BCT for ndMM in Chinese patients is highly attractive.
Collapse
Affiliation(s)
- Jin Lu
- Department of Hematology, Peking University People's Hospital, Beijing 100032, China
| | - Wendong Chen
- Toronto Health Economics & Technology Assessment Collaborative, University of Toronto, ON M5G 2C4, Canada
| |
Collapse
|
13
|
Wang B, Li PK, Ma JX, Chen D. Therapeutic Effects of a Novel Phenylphthalimide Analog for Corneal Neovascularization and Retinal Vascular Leakage. Invest Ophthalmol Vis Sci 2019; 59:3630-3642. [PMID: 30029250 PMCID: PMC6054429 DOI: 10.1167/iovs.18-24015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose Neovascularization (NV) and retinal vascular leakage are major causes of impaired vision in ocular diseases. The purpose of this study was to identify novel phenylphthalimide analogs with therapeutic effects on NV and vascular leakage and to explore the mechanism of action. Methods Antiangiogenic activities of novel phenylphthalimide analogs were assessed in vitro by using VEGF ELISA and endothelial cell proliferation assay. Their efficacies on retinal vascular leakage were evaluated using rat models of oxygen-induced retinopathy (OIR) and streptozotocin (STZ)-induced diabetes. The in vivo antiangiogenic activity was evaluated using topical administration in the alkali burn-induced corneal NV model. The expression of VEGF and intercellular adhesion molecule-1 (ICAM-1) were measured using ELISA. Results Thalidomide and three novel analogs all showed inhibitory effects on endothelial cell proliferation and VEGF expression in vitro. Through intravitreal injection, all of the compounds reduced retinal vascular leakage in the OIR and STZ-induced diabetic models. Among these compounds, (2,6-diisopropylphenyl)-5-amino-1H-isoindole-1,3-dione (DAID) displayed the most potent efficacy and reduced retinal vascular leakage in a dose-dependent manner in both the OIR and STZ-diabetes models. Topical administration of DAID also inhibited alkali burn-induced corneal NV. Furthermore, DAID attenuated the overexpression of VEGF and ICAM-1 in the retina of the OIR model. Intravitreal injection of DAID did not result in any detectable side effects, as shown by electroretinogram and retinal histological analysis. Conclusions DAID is a novel phenylphthalimide analog with potent effects on NV and retinal vascular leakage through downregulation of VEGF and inflammatory factors and has therapeutic potential.
Collapse
Affiliation(s)
- Bing Wang
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States.,Department of Ophthalmology, Fujian Medical University Union Hospital, Fujian Province, China
| | - Pui-Kai Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Ohio State University, Columbus, Ohio, United States
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Danyang Chen
- Charlesson, LLC, Oklahoma City, Oklahoma, United States
| |
Collapse
|
14
|
Duan X, Mu M, Yan J, Bai L, Zhong L, Zhu Y, Pan H, Zhang M, Shi J. Co-delivery of Aurora-A inhibitor XY-4 and Bcl-xl siRNA enhances antitumor efficacy for melanoma therapy. Int J Nanomedicine 2018; 13:1443-1456. [PMID: 29563798 PMCID: PMC5849942 DOI: 10.2147/ijn.s147759] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background The newly synthesized Aurora-A kinase inhibitor XY-4 is a potential anti-cancer agent, but its hydrophobicity and limited efficiency restrict further application. Nanotechnology based combined therapy provides an optimized strategy for solving these issues. Methods In this study, the newly synthesized Aurora-A kinase inhibitor XY-4 and Bcl-xl targeted siRNA were co-delivered by cationic liposomes, creating an injectable co-delivery formulation. The anti-cancer ability and mechanisms of XY-4/Bcl-xl siRNA co-loaded cationic liposomes were studied both in vitro and in vivo. Results The prepared liposomes had a mean particle size of 91.3±4.5 nm with a zeta potential of 38.5±0.5 mV and were monodispersed (Polydispersity index =0.183) in water solution, with high drug loading capacity and stability. Intriguingly, the positive charges of co-delivery liposomes not only facilitated gene delivery, but also obviously enhanced drug uptake. The XY-4/Bcl-xl siRNA co-loaded cationic liposomes demonstrated enhanced anti-cancer effects on B16 melanoma cells in vitro by activation mitochondrial apoptosis pathway. Moreover, intratumoral injection of this co-delivery formulation efficiently inhibited the growth of a B16 melanoma xenograft model in vivo. Conclusion By co-delivering Aurora-A kinase inhibitor XY-4 and Bcl-xl targeting siRNA in a nanoformulation, our study supplied a potential combination strategy for melanoma therapy.
Collapse
Affiliation(s)
- Xingmei Duan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu
| | - Minjie Mu
- Key Laboratory Standardization of Chinese Herbal Medicines of Ministry of Education, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Medicine (TCM), Chengdu, People's Republic of China
| | - Junfeng Yan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Lei Zhong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Yuxuan Zhu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Haixia Pan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Mei Zhang
- Key Laboratory Standardization of Chinese Herbal Medicines of Ministry of Education, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Medicine (TCM), Chengdu, People's Republic of China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu.,Key Laboratory Standardization of Chinese Herbal Medicines of Ministry of Education, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Medicine (TCM), Chengdu, People's Republic of China
| |
Collapse
|
15
|
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: 4] [Impact Index Per Article: 0.7] [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
|
16
|
Ogino Y, Tanaka M, Shimozawa T, Asahi T. LC-MS/MS and chiroptical spectroscopic analyses of multidimensional metabolic systems of chiral thalidomide and its derivatives. Chirality 2017; 29:282-293. [DOI: 10.1002/chir.22683] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/12/2016] [Accepted: 01/04/2017] [Indexed: 02/02/2023]
Affiliation(s)
- Yoshiyuki Ogino
- Waseda University; Department of Life Science and Medical Bioscience; Tokyo Japan
| | - Masahito Tanaka
- National Institute of Advanced Industrial Science and Technology (AIST); Research Institute of Instrumentation Frontier; Tsukuba Japan
| | - Togo Shimozawa
- Waseda University; Department of Life Science and Medical Bioscience; Tokyo Japan
| | - Toru Asahi
- Waseda University; Department of Life Science and Medical Bioscience; Tokyo Japan
| |
Collapse
|
17
|
|
18
|
Pourcher V, Desnoyer A, Assoumou L, Lebbe C, Curjol A, Marcelin AG, Cardon F, Gibowski S, Salmon D, Chennebault JM, Poizot-Martin I, Peytavin G, Boué F, Costagliola D. Phase II Trial of Lenalidomide in HIV-Infected Patients with Previously Treated Kaposi's Sarcoma: Results of the ANRS 154 Lenakap Trial. AIDS Res Hum Retroviruses 2017; 33:1-10. [PMID: 27405442 DOI: 10.1089/aid.2016.0069] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lenalidomide, an oral immunomodulating agent, has shown promising activity in HIV-infected individuals with Kaposi's sarcoma (KS). This single-arm, multicenter, open-label, Gehan's two-stage phase II trial evaluated the efficacy and safety of lenalidomide in HIV-infected patients with progressive KS despite previous chemotherapy (NCT01282047, ANRS 154 Lenakap trial). The primary endpoint was the rate of partial response (PR) or complete response (CR) at week 24, evaluated by both the study investigators and the patients using the Physical Global Assessment (PGA). AIDS Clinical Trials Group (ACTG) criteria for KS treatment evaluation were used as a secondary endpoint. The data and safety monitoring board recommended that enrollments be halted on April 24, 2013, because of lack of responses. We enrolled 12 antiretroviral-treated HIV-infected men with progressive KS despite previous chemotherapy. Their HIV plasma viral load was <50 copies/ml and their median CD4 cell count 444/mm3. One patient stopped taking lenalidomide because of hives at week 1 and a second patient died at week 7. The remaining 10 patients were assessable at week 24, when none had PGA-defined CR or PR and one had ACTG-defined PR. There were no additional PGA responses at week 48, but an additional three patients had ACTG responses, for a total of four patients with ACTG PR at week 48 (40%; 95% confidence interval: 12.2-73.8). Fourteen grade 3-4 adverse events were considered at least possibly related to lenalidomide during a total of 101 cycles. Lenalidomide was well tolerated in antiretroviral experienced patients with progressive KS previously treated with chemotherapy. The ACTG-defined response rate at week 48 was 40%, while it was 0% using PGA criteria.
Collapse
Affiliation(s)
- Valérie Pourcher
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Service de Maladies Infectieuses et Tropicales, Paris, France
- Sorbonne Universités, UPMC Paris Univ 06, Paris, France
- INSERM UMR-S996, Clamart, France
| | - Aude Desnoyer
- INSERM UMR-S996, Clamart, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Département de Pharmaco-Toxicologie Clinique, Paris, France
| | - Lambert Assoumou
- INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMR 1136), Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Céleste Lebbe
- Assistance Publique Hôpitaux de Paris, Hôpital Saint Louis, Service de Dermatologie, Paris, France
- INSERM U976, CIC et Dermatologie, Univ Paris Diderot, Paris, France
| | - Angélique Curjol
- INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMR 1136), Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Anne-Geneviève Marcelin
- INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMR 1136), Sorbonne Universités, UPMC Univ Paris 06, Paris, France
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Virologie, Paris, France
| | - Fanny Cardon
- ANRS (France REcherche Nord&Sud Sida-hiv Hépatites), Clinical and Therapeutic Research on HIV/AIDS Office, Paris, France
| | - Séverine Gibowski
- ANRS (France REcherche Nord&Sud Sida-hiv Hépatites), Clinical Research Safety Office, Paris, France
| | - Dominique Salmon
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Cochin, Service des Maladies Infectieuses, Paris, France
| | | | - Isabelle Poizot-Martin
- APHM Sainte-Marguerite, Service d'Immunohématologie Clinique, Marseille, France
- INSERM U912 (SESSTIM), Université Aix Marseille, Marseille, France
| | - Gilles Peytavin
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Département de Pharmaco-Toxicologie Clinique, Paris, France
- INSERM, IAME UMR 1137, Université Paris Diderot, Paris, France
| | - François Boué
- INSERM UMR-S996, Clamart, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Antoine Béclère, Service de Médecine Interne et Immunologie Clinique, Clamart, France
| | - Dominique Costagliola
- INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMR 1136), Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | | |
Collapse
|
19
|
Li M, Wang Y, Zhang J, Xie S, Wang C, Wu Y. Synthesis and Biological Evaluation of Novel Aromatic Imide-Polyamine Conjugates. Molecules 2016; 21:molecules21121637. [PMID: 27916902 PMCID: PMC6273765 DOI: 10.3390/molecules21121637] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/17/2016] [Accepted: 11/23/2016] [Indexed: 11/16/2022] Open
Abstract
Three types of conjugates in which aromatic imide scaffolds were coupled to diverse amine/polyamine motifs were synthesized, and their antitumor activities were evaluated in vitro and in vivo. Results showed that the conjugate 11e of 1,8-naphthilimide with spermine had pronounced effects on inhibiting tumor cell proliferation and inducing tumor cell apoptosis via ROS-mediated mitochondrial pathway. The in vivo assays on three H22 tumor transplant models revealed that compound 11e exerted potent ability in preventing lung cancer metastasis and extending lifespan. Furthermore, the efficacy of 11e in inhibiting tumor growth and improving body weight index were better than that of positive control, amonafide. Our study demonstrates that compound 11e is a valuable lead compound for further investigation.
Collapse
Affiliation(s)
- Ming Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China.
- Pharmaceutical College, Henan University, Kaifeng 475001, China.
| | - Yuxia Wang
- College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475001, China.
| | - Jianying Zhang
- Pharmaceutical College, Henan University, Kaifeng 475001, China.
| | - Songqiang Xie
- Pharmaceutical College, Henan University, Kaifeng 475001, China.
| | - Chaojie Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering, Kaifeng 475001, China.
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
20
|
Li Y, Yang T, Li J, Hao HL, Wang SY, Yang J, Luo JM. Inhibition of multiple myeloma cell proliferation by ginsenoside Rg3 via reduction in the secretion of IGF-1. Mol Med Rep 2016; 14:2222-30. [PMID: 27430248 DOI: 10.3892/mmr.2016.5475] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 05/31/2016] [Indexed: 11/05/2022] Open
Abstract
Ginsenoside Rg3 (Rg3) is one of the primary constituents isolated from ginseng, and has been found to exhibit cytotoxic effects against cancer cells. The present study aimed to investigate the effects of Rg3 on human multiple myeloma cell proliferation and apoptosis, and to examine its underlying molecular mechanisms. Cell viability was detected using a Cell Counting kit‑8 assay, and cell cycle arrest and cell apoptosis were analyzed using flow cytometry. In addition, the expression levels of cell cycle‑associated markers and apoptosis‑associated proteins, and the release of cytochrome C were determined using western blot analysis. The effects of Rg3 on the insulin‑like growth factor (IGF)-1/AKT/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase signaling pathways were also investigated using western blot analysis. The results showed that Rg3 inhibited cell viability in U266, RPMI8226 and SKO‑007 cells in a time‑ and dose‑dependent manner, and caused cell cycle arrest in the G1 phase by regulating the cyclin‑dependent kinase pathway. Furthermore, Rg3 induced multiple myeloma cell apoptosis, and was involved in B cell lymphoma-2 (Bcl2)/Bcl2-associated X protein imbalance, caspase activation and the release of cytochrome C from the mitochondria into the cytoplasm. Mechanistically, it was found that the inhibitory effects of Rg3 on multiple myeloma cell proliferation were essential for secretion of IGF‑1 and inactivation of the Akt/mTOR pathway. Collectively, these findings demonstrated that Rg3 effectively inhibited cell proliferation and induced apoptosis of multiple myeloma cells. These data broaden the clinical investigation of Rg3 in the treatment of multiple myeloma, associated with the inactivation of IGF-1/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Yan Li
- Department of Hematology, The Second Hospital, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Tao Yang
- Department of Urinary Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Jing Li
- Department of Hematology, Hebei Province Chinese Medicine Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Hong-Ling Hao
- Department of Hematology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Su-Yun Wang
- Department of Hematology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Jie Yang
- Department of Hematology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Jian-Min Luo
- Department of Hematology, The Second Hospital, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
21
|
Shen Z, Lv C, Zeng S. Significance and challenges of stereoselectivity assessing methods in drug metabolism. J Pharm Anal 2016; 6:1-10. [PMID: 29403956 PMCID: PMC5762452 DOI: 10.1016/j.jpha.2015.12.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 12/21/2015] [Accepted: 12/21/2015] [Indexed: 12/14/2022] Open
Abstract
Stereoselectivity in drug metabolism can not only influence the pharmacological activities, tolerability, safety, and bioavailability of drugs directly, but also cause different kinds of drug-drug interactions. Thus, assessing stereoselectivity in drug metabolism is of great significance for pharmaceutical research and development (R&D) and rational use in clinic. Although there are various methods available for assessing stereoselectivity in drug metabolism, many of them have shortcomings. The indirect method of chromatographic methods can only be applicable to specific samples with functional groups to be derivatized or form complex with a chiral selector, while the direct method achieved by chiral stationary phases (CSPs) is expensive. As a detector of chromatographic methods, mass spectrometry (MS) is highly sensitive and specific, whereas the matrix interference is still a challenge to overcome. In addition, the use of nuclear magnetic resonance (NMR) and immunoassay in chiral analysis are worth noting. This review presents several typical examples of drug stereoselective metabolism and provides a literature-based evaluation on current chiral analytical techniques to show the significance and challenges of stereoselectivity assessing methods in drug metabolism.
Collapse
Affiliation(s)
- Zhuowei Shen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chuang Lv
- Biogen Idec, Cambridge, MA 02142, USA
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
22
|
Sriskandarajah P, Scott F, Muthalali S, Mercieca J. Life-threatening bowel perforation while on thalidomide-based triplet regimen for multiple myeloma: a retrospective case series. Br J Haematol 2015; 173:786-8. [DOI: 10.1111/bjh.13630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/03/2015] [Indexed: 12/31/2022]
Affiliation(s)
| | - Francis Scott
- Department of Haematology; Epsom and St Helier Hospital; Carshalton Surrey UK
| | - Sneha Muthalali
- Department of Haematology; Epsom and St Helier Hospital; Carshalton Surrey UK
| | - Jane Mercieca
- Department of Haematology; Epsom and St Helier Hospital; Carshalton Surrey UK
| |
Collapse
|
23
|
Ghosh N, Grunwald MR, Fasan O, Bhutani M. Expanding role of lenalidomide in hematologic malignancies. Cancer Manag Res 2015; 7:105-19. [PMID: 25999761 PMCID: PMC4427066 DOI: 10.2147/cmar.s81310] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Lenalidomide is an immunomodulatory agent that has been approved by the US Food and Drug Administration for treatment of multiple myeloma, deletion 5q myelodysplastic syndrome, and mantle cell lymphoma. In addition, it has clinical activity in lymphoproliferative disorders and acute myeloid leukemia. The mode of action includes immunomodulatory, anti-inflammatory, antiangiogenic, and antiproliferative mechanisms. The antitumor effect is a result of direct interference of key pathways in tumor cells and indirect modulation of the tumor microenvironment. There has been no recent collective review on lenalidomide in multiple myeloma, myelodysplastic syndrome/acute myeloid leukemia, and lymphoma. This review summarizes the results of current clinical studies of lenalidomide, alone and in combination with other agents, as a therapeutic option for various hematologic malignancies.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - Michael R Grunwald
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - Omotayo Fasan
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - Manisha Bhutani
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| |
Collapse
|
24
|
Maher HM, Alzoman NZ, Alshehri MM, Aljohar HI, Shehata S, Alossaimi M, Abanmy NO. Simultaneous determination of dexamethasone and lenalidomide in rat plasma by solid phase extraction and ultra-performance liquid chromatography-tandem mass spectrometry: application to pharmacokinetic studies. RSC Adv 2015. [DOI: 10.1039/c5ra22339c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Determination of dexamethasone and lenalidomide in plasma by SPE and UPLC-MS/MS for the purpose of TDM: application to pharmacokinetic studies.
Collapse
Affiliation(s)
- Hadir M. Maher
- College of Pharmacy
- Department of Pharmaceutical Chemistry
- King Saud University
- Riyadh 11495
- Saudi Arabia
| | - Nourah Z. Alzoman
- College of Pharmacy
- Department of Pharmaceutical Chemistry
- King Saud University
- Riyadh 11495
- Saudi Arabia
| | - Mona M. Alshehri
- College of Pharmacy
- Department of Pharmaceutical Chemistry
- King Saud University
- Riyadh 11495
- Saudi Arabia
| | - Haya I. Aljohar
- College of Pharmacy
- Department of Pharmaceutical Chemistry
- King Saud University
- Riyadh 11495
- Saudi Arabia
| | - Shereen Shehata
- College of Pharmacy
- Department of Pharmaceutical Chemistry
- King Saud University
- Riyadh 11495
- Saudi Arabia
| | - Manal Alossaimi
- Pharmaceutical Analysis Department
- University of Strathclyde
- UK
| | - Norah O. Abanmy
- College of Pharmacy
- Department of Clinical Pharmacy
- King Saud University
- Riyadh 11495
- Saudi Arabia
| |
Collapse
|
25
|
Azimirad A, Alborzi S, Kumar PV, Zolghadri J, Zarei A, Tavana Z, Azimirad M. Thalidomide affects experimental endometriosis: A randomized controlled study in the rat. J Obstet Gynaecol Res 2014; 40:1989-97. [DOI: 10.1111/jog.12434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 02/20/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Afshin Azimirad
- Laparoscopy Research Center; Department of Obstetrics and Gynecology; Shiraz University of Medical Sciences; Shiraz Iran
- Student Research Committee; Shiraz University of Medical Sciences; Shiraz Iran
- Infertility Research Center; Shiraz University of Medical Sciences; Shiraz Iran
| | - Saeed Alborzi
- Laparoscopy Research Center; Department of Obstetrics and Gynecology; Shiraz University of Medical Sciences; Shiraz Iran
- Infertility Research Center; Shiraz University of Medical Sciences; Shiraz Iran
| | | | - Jaleh Zolghadri
- Infertility Research Center; Shiraz University of Medical Sciences; Shiraz Iran
| | - Afsoon Zarei
- Laparoscopy Research Center; Department of Obstetrics and Gynecology; Shiraz University of Medical Sciences; Shiraz Iran
| | - Zohreh Tavana
- Laparoscopy Research Center; Department of Obstetrics and Gynecology; Shiraz University of Medical Sciences; Shiraz Iran
| | - Mina Azimirad
- Shariati Hospital; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
26
|
Mucosal healing with thalidomide in refractory Crohn's disease patients intolerant of anti-TNF-α drugs: report of 3 cases and literature review. J Clin Gastroenterol 2014; 48:530-3. [PMID: 24667589 DOI: 10.1097/mcg.0000000000000122] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thalidomide is an oral immunomodulatory and anti-inflammatory drug with antitumor necrosis factor-α (TNF-α) activity. Several case reports and some clinical trials have demonstrated its efficacy in the treatment of refractory Crohn's disease (CD). We report the effect and tolerability of thalidomide in 3 patients with moderate-to-severe CD who were not responsive to anti-TNF-α therapies, and review the relevant literature. The first case is of a 28-year-old female affected by Crohn's colitis complicated by a severe fistulizing perianal disease; she was treated with infliximab, adalimumab, and certolizumab pegol, which were stopped because of intolerance. The second case is of a 39-year-old female with fistulizing ileocolitis complicated by severe arthralgias and perianal disease with loss of response to infliximab and intolerance of certolizumab pegol. The third case is of a 39-year-old male with gastric and ileocolonic CD refractory to immunosuppressors and intolerant of infliximab. All the 3 cases achieved complete clinical remission and endoscopic healing of mucosal lesions at a low dose of thalidomide (50 to 150 mg/d). In our CD patients who experienced loss of response or were unable to tolerate anti-TNF-α drugs, thalidomide was an effective and well-tolerated therapy for inducing and maintaining long-term remission.
Collapse
|
27
|
NF-kappa B modulation is involved in celastrol induced human multiple myeloma cell apoptosis. PLoS One 2014; 9:e95846. [PMID: 24755677 PMCID: PMC3995890 DOI: 10.1371/journal.pone.0095846] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 03/31/2014] [Indexed: 11/19/2022] Open
Abstract
Celastrol is an active compound extracted from the root bark of the traditional Chinese medicine Tripterygium wilfordii Hook F. To investigate the effect of celastrol on human multiple myeloma cell cycle arrest and apoptosis and explore its molecular mechanism of action. The activity of celastrol on LP-1 cell proliferation was detected by WST-8 assay. The celastrol-induced cell cycle arrest was analyzed by flow cytometry after propidium iodide staining. Nuclear translocation of the nuclear factor kappa B (NF-κB) was observed by fluorescence microscope. Celastrol inhibited cell proliferation of LP-1 myeloma cell in a dose-dependent manner with IC50 values of 0.8817 µM, which was mediated through G1 cell cycle arrest and p27 induction. Celastrol induced apoptosis in LP-1 and RPMI 8226 myeloma cells in a time and dose dependent manner, and it involved Caspase-3 activation and NF-κB pathway. Celastrol down-modulated antiapoptotic proteins including Bcl-2 and survivin expression. The expression of NF-κB and IKKa were decreased after celastrol treatment. Celastrol effectively blocked the nuclear translocation of the p65 subunit and induced human multiple myeloma cell cycle arrest and apoptosis by p27 upregulation and NF-kB modulation. It has been demonstrated that the effect of celastrol on NF-kB was HO-1-independent by using zinc protoporphyrin-9 (ZnPPIX), a selective heme oxygenase inhibitor. From the results, it could be inferred that celastrol may be used as a NF-kB inhibitor to inhibit myeloma cell proliferation.
Collapse
|
28
|
Gomathi T, Govindarajan C, Rose H R MH, Sudha PN, Imran PKM, Venkatesan J, Kim SK. Studies on drug-polymer interaction, in vitro release and cytotoxicity from chitosan particles excipient. Int J Pharm 2014; 468:214-22. [PMID: 24742716 DOI: 10.1016/j.ijpharm.2014.04.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/24/2014] [Accepted: 04/10/2014] [Indexed: 11/24/2022]
Abstract
Nonobvious controlled polymeric pharmaceutical excipient, chitosan nanoparticles (CS-NPs) for lenalidomide encapsulation were geared up by the simple ionic cross linking method to get better bioavailability and to reduce under as well as overloading of hydrophobic and sparingly soluble drug lenalidomide towards cancer cells. Lenalidomide loaded chitosan nanoparticles (LND-CS-NPs) were in the size range of 220-295 nm and characterized by DLS, TEM, FT-IR, TGA and XRD. Encapsulation of lenalidomide over chitosan nanoparticles was observed about 99.35% using UV spectrophotometry method. In vitro release and the cytotoxic studies were performed using LND-CS-NPs. This study implies the new drug delivery route for lenalidomide and illustrates that the CS-NPs serves as the effective pharmaceutical carrier for sustained delivery of lenalidomide.
Collapse
Affiliation(s)
- Thandapani Gomathi
- Department of Chemistry, D.K.M. College for Women, Vellore, Tamil Nadu, India
| | - C Govindarajan
- Indian Institute of Chromatography and Mass Spectrometry, Chennai, Tamil Nadu, India
| | | | - P N Sudha
- Department of Chemistry, D.K.M. College for Women, Vellore, Tamil Nadu, India.
| | - P K Mohamed Imran
- Department of Chemistry, Islamiah College, Vaniyambadi, Tamil Nadu, India
| | - Jayachandran Venkatesan
- Department of Marine Bio Convergence Science and Marine Bioprocess Research Center, Pukyong National University, Busan 608-737, South Korea.
| | - Se-Kwon Kim
- Department of Marine Bio Convergence Science and Marine Bioprocess Research Center, Pukyong National University, Busan 608-737, South Korea.
| |
Collapse
|
29
|
Abstract
The drugs used to treat hematologic disorders constitute an extraordinary array of agents and include therapy for anemias, bleeding diatheses, thromboembolism and hematologic malignancies. Within many of these categories, hematologic therapies represent molecular understanding of the disease pathophysiology and a treatment targeted precisely at the known aberration. In this chapter we discuss these treatments, with greater emphasis on the most commonly used agents and the latest advances in the specific field. The chapter includes discussion on old (cyclooxygenase inhibitors) and new antiplatelet agents (ADP receptor P2Y12 inhibitors), newly described oral thrombin inhibitors such as dabigatran, treatment targeted at the bcr-abl tyrosine kinase in chronic myeloid leukemia and demethylating agents in myelodysplasia. The discussion is also oriented towards neurologic implications, both therapeutic and toxic, for each therapy or therapeutic group.
Collapse
|
30
|
Huang X, Wang Y, Nan X, He S, Xu X, Zhu X, Tang J, Yang X, Yao L, Wang X, Cheng C. The role of the orphan G protein-coupled receptor 37 (GPR37) in multiple myeloma cells. Leuk Res 2013; 38:225-35. [PMID: 24290813 DOI: 10.1016/j.leukres.2013.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 11/04/2013] [Accepted: 11/07/2013] [Indexed: 01/14/2023]
Abstract
The orphan G protein-coupled receptor 37 (GPR37) is homologous to endothelin (ETB-R) and bombesin (GRP-R, NMB-R) receptors. The present study was undertaken to determine the expression and functional significance of GPR37 in human multiple myeloma (MM). We found that GPR37 was lowly expressed in MM cell adhesion model and highly expressed in proliferating cells. In vitro, meddling with the expression of GPR37 affected the CAM-DR by regulating the ability of cell adhesion and the activity of Akt and ERK in MM cells. Further studies indicated the positive role of GPR37 in the proliferation of MM cells.
Collapse
Affiliation(s)
- Xianting Huang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Yuchan Wang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Xun Nan
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Song He
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu 226001, People's Republic of China
| | - Xiaohong Xu
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu 226001, People's Republic of China
| | - Xinghua Zhu
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu 226001, People's Republic of China
| | - Jie Tang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Xiaojing Yang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Li Yao
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Xinxiu Wang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Chun Cheng
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
31
|
Olivier-Abbal P, Teisseyre AC, Montastruc JL. Comparison of serious adverse reactions between thalidomide and lenalidomide: analysis in the French Pharmacovigilance database. Med Oncol 2013; 30:733. [DOI: 10.1007/s12032-013-0733-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/21/2013] [Indexed: 01/18/2023]
|
32
|
Silbermann R, Roodman GD. Myeloma bone disease: Pathophysiology and management. J Bone Oncol 2013; 2:59-69. [PMID: 26909272 PMCID: PMC4723362 DOI: 10.1016/j.jbo.2013.04.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/29/2013] [Accepted: 04/02/2013] [Indexed: 10/26/2022] Open
Abstract
Multiple myeloma bone disease is marked by severe dysfunction of both bone formation and resorption and serves as a model for understanding the regulation of osteoblasts (OBL) and osteoclasts (OCL) in cancer. Myeloma bone lesions are purely osteolytic and are associated with severe and debilitating bone pain, pathologic fractures, hypercalcemia, and spinal cord compression, as well as increased mortality. Interactions within the bone marrow microenvironment in myeloma are responsible for the abnormal bone remodeling in myeloma bone disease. Myeloma cells drive bone destruction that increases tumor growth, directly stimulates the OCL formation, and induces cells in the marrow microenvironment to produce factors that drive OCL formation and suppress OBL formation. Factors produced by marrow stromal cells and OCL promote tumor growth through direct action on myeloma cells and by increasing angiogenesis. Current therapies targeting MMBD focus on preventing osteoclastic bone destruction; however regulators of OBL inhibition in MMBD have also been identified, and targeted agents with a potential anabolic effect in MMBD are under investigation. This review will discuss the mechanisms responsible for MMBD and therapeutic approaches currently in use and in development for the management of MMBD.
Collapse
Affiliation(s)
- Rebecca Silbermann
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
33
|
Srivastava G, Rana V, Lacy MQ, Buadi FK, Hayman SR, Dispenzieri A, Gertz MA, Dingli D, Zeldenrust S, Russell S, McCurdy A, Kapoor P, Kyle R, Rajkumar SV, Kumar S. Long-term outcome with lenalidomide and dexamethasone therapy for newly diagnosed multiple myeloma. Leukemia 2013; 27:2062-6. [PMID: 23648667 DOI: 10.1038/leu.2013.143] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 05/01/2013] [Indexed: 01/25/2023]
Abstract
The combination of lenalidomide and dexamethasone (Len-Dex) is a commonly used initial therapy for newly diagnosed multiple myeloma (MM). Although the initial response rates and toxicity are well known, long-term outcome is not well described. We studied 286 consecutive patients with newly diagnosed MM initially treated with Len-Dex. The median (range) age at diagnosis was 63 (28-92) years, 166 (58%) patients ≤ 65 years and 175 (61%) male. The median estimated duration on Len-Dex was 5.3 months with overall response (≥ partial response) of 72%, including 26% with very good partial response or better. The median overall survival (OS) from the diagnosis was not reached (NR) and the estimated 5-year survival was 71%. The median time to first disease progression, irrespective of transplant status, was 30.2 months. Overall, 143 (50%) patients underwent stem cell transplant. The median OS was NR for patients ≤ 70 years and 5.8 years for the older patients (P=0.01). The 5-year OS estimate for patients in International Staging System stage 1, 2 and 3 were 82, 65, and 44% respectively. There were 21 new second malignancies after MM diagnosis (6.6%). The median survival exceeding 7 years reflects the efficacy of novel agents. The risk of second malignancies doesn't appear to be excessive in this population.
Collapse
Affiliation(s)
- G Srivastava
- Division of Hematology and Blood and Marrow Transplant, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sondhi SM, Arya S, Rani R, Kumar N, Roy P. Synthesis, anti-inflammatory and anticancer activity evaluation of some mono- and bis-Schiff’s bases. Med Chem Res 2012; 21:3620-3628. [DOI: 10.1007/s00044-011-9899-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
35
|
Abstract
Background Combination of different agents is widely used in clinic to combat complex diseases with improved therapy and reduced side effects. However, the identification of effective drug combinations remains a challenging task due to the huge number of possible combinations among candidate drugs that makes it impractical to screen putative combinations. Results In this work, we construct a 'drug cocktail network' using all the known effective drug combinations extracted from the Drug Combination Database (DCDB), and propose a network-based approach to investigate drug combinations. Our results show that the agents in an effective combination tend to have more similar therapeutic effects and share more interaction partners. Based on our observations, we further develop a statistical approach termed as DCPred (Drug Combination Predictor) to predict possible drug combinations by exploiting the topological features of the drug cocktail network. Validating on the known drug combinations, DCPred achieves the overall AUC (Area Under the receiver operating characteristic Curve) score of 0.92, indicating the predictive power of our proposed approach. Conclusions The drug cocktail network constructed in this work provides useful insights into the underlying rules of effective drug combinations and offer important clues to accelerate the future discovery of new drug combinations.
Collapse
Affiliation(s)
- Ke-Jia Xu
- Institute of Systems Biology, Shanghai University, Shanghai, China.
| | | | | |
Collapse
|
36
|
Gaballa MR, Laubach JP, Schlossman RL, Redman K, Noonan K, Mitsiades CS, Ghobrial IM, Munshi N, Anderson KC, Richardson PG. Management of myeloma-associated renal dysfunction in the era of novel therapies. Expert Rev Hematol 2012; 5:51-66; quiz 67-8. [PMID: 22272706 DOI: 10.1586/ehm.11.72] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm often associated with renal impairment (RI), with myeloma cast nephropathy recognized as the most common cause. While RI is present in over 50% of MM patients at some point in their disease course, it is associated with higher tumor burden, more aggressive disease, diminished quality of life, development of complications and increased mortality. The introduction of novel therapies, including bortezomib, lenalidomide and thalidomide, has revolutionized the management of MM. They are now considered first-line therapies in induction, maintenance and salvage therapy for MM. In addition to their anti-MM effect, they can improve outcome in patients with RI, especially when combined, and bortezomib with dexamethasone may have a renal protective effect. This review focuses on the use of these agents in patients with MM and RI, and evaluates their efficacy, safety, need for dose adjustment and impact on RI.
Collapse
Affiliation(s)
- Mahmoud R Gaballa
- Department of Internal Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kannaiyan R, Hay HS, Rajendran P, Li F, Shanmugam MK, Vali S, Abbasi T, Kapoor S, Sharma A, Kumar AP, Chng WJ, Sethi G. Celastrol inhibits proliferation and induces chemosensitization through down-regulation of NF-κB and STAT3 regulated gene products in multiple myeloma cells. Br J Pharmacol 2012; 164:1506-21. [PMID: 21506956 DOI: 10.1111/j.1476-5381.2011.01449.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Activation of pro-inflammatory transcription factors NF-κB and signal transducer and activator of transcription 3 (STAT3) is one of the major contributors to both pathogenesis and chemoresistance in multiple myeloma (MM), which results in high mortality rate. Thus, in the present study, we investigated whether celastrol could suppress the proliferation and induce chemosensitization of MM cells by interfering with NF-κB and STAT3 activation pathways. EXPERIMENTAL APPROACH The effects of celastrol were investigated using both a virtual predictive tumour cell system and different MM cell lines resistant to doxorubicin, melphalan and bortezomib. KEY RESULTS Celastrol inhibited the proliferation of MM cell lines regardless of whether they were sensitive or resistant to bortezomib and other conventional chemotherapeutic drugs. It also synergistically enhanced the apoptotic effects of thalidomide and bortezomib. This correlated with the down-regulation of various proliferative and anti-apoptotic gene products including cyclin D1, Bcl-2, Bcl-xL, survivin, XIAP and Mcl-1. These effects of celastrol were mediated through suppression of constitutively active NF-κB induced by inhibition of IκBα kinase activation; and the phosphorylation of IκBα and of p65. Celastrol also inhibited both the constitutive and IL6-induced activation of STAT3, which induced apoptosis as indicated by an increase in the accumulation of cells in the sub-G1 phase, an increase in the expression of pro-apoptotic proteins and activation of caspase-3. CONCLUSIONS AND IMPLICATIONS Thus, based on our experimental findings, we conclude that celastrol may have great potential as a treatment for MM and other haematological malignancies.
Collapse
Affiliation(s)
- Radhamani Kannaiyan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Roccaro AM, Ghobrial IM, Blotta S, Treon SP, Malagola M, Anderson KC, Richardson PG, Russo D. Advances in the treatment of monoclonal gammopaties: The emerging role of targeted therapy in plasma cell dyscrasias. Biologics 2011; 2:419-31. [PMID: 19707373 PMCID: PMC2721375 DOI: 10.2147/btt.s3088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The paradigm for the treatment of monoclonal gammopaties has dramatically changed: therapeutic options in multiple myeloma (MM) have evolved from the introduction of melphalan and prednisone in the 1960s, high-dose chemotherapy and stem cell transplantation in the late 1980s and 1990s, to the rapid introduction of small novel molecules within the last seven years. Based on the understanding of the complex interaction of the MM cells with the bone marrow microenvironment and the signaling pathways that are dysregulated in this process, a number of novel therapeutic agents are now available. Specifically, three novel agents with a specific-targeted anti-MM activity, have been FDA-approved for the treatment of this disease, namely Bortezomib, thalidomide, and lenalidomide which are now all playing a key role in the treatment of MM. The success of targeted therapy in MM has since led to the development and investigation of more than 30 new compounds in this disease and in other plasma cell dyscrasias such as Waldenström’s macroglobulinemia and primary amyloidosis, both in the preclinical settings and as part of clinical trials.
Collapse
Affiliation(s)
- Aldo M Roccaro
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Yang Y, Zhu YQ, Jiang L, Li LF, Ge JP. Thalidomide induces apoptosis in human oral squamous cell carcinoma cell line with altered expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Oral Oncol 2011; 47:927-8. [PMID: 21724449 DOI: 10.1016/j.oraloncology.2011.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 05/31/2011] [Accepted: 06/07/2011] [Indexed: 11/30/2022]
|
40
|
Madan S, Kumar S. Current treatment options for elderly patients with multiple myeloma: clinical impact of novel agents. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/thy.11.39] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
|
42
|
Guglielmelli T, Bringhen S, Rrodhe S, Gay F, Cavallo F, Berruti A, Montefusco V, Piro E, Benevolo G, Petrucci MT, Caravita T, Offidani M, Corradini P, Boccadoro M, Saglio G, Palumbo A. Previous thalidomide therapy may not affect lenalidomide response and outcome in relapse or refractory multiple myeloma patients. Eur J Cancer 2011; 47:814-8. [PMID: 21334196 DOI: 10.1016/j.ejca.2010.12.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 12/23/2010] [Accepted: 12/23/2010] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Lenalidomide is a thalidomide analogue, designed to have improved efficacy and tolerability over the parent drug. The aim of this retrospective analysis is to evaluate the impact of thalidomide therapy on lenalidomide response and outcome in relapse or refractory multiple myeloma patients. PATIENTS AND METHODS A total of 106 relapsed or refractory multiple myeloma patients received lenalidomide 25mg plus dexamethasone as salvage therapy; 80 patients progressed on thalidomide treatment (thalidomide-resistant) and 26 patients discontinued thalidomide in at least partial remission (thalidomide-sensitive). Median time from diagnosis to lenalidomide treatment was 57 months. Median prior lines of therapies were 3, range 1-6. 62% of patients were previously treated with autologous stem cell transplantation, and 71% with bortezomib-based regimens. RESULTS In the thalidomide-resistant and -sensitive groups, the at least partial response rates were 56.2% and 61.5% (P = .45), including at least VGPR rates of 16.2% and 11.5%; the median progression free survival was 10 and 12 months (P=.12) and the median overall survival was 17 and 18.5 months (P = .50), respectively. CONCLUSION Lenalidomide may be equally effective in heavily pre-treated multiple myeloma patients who are thalidomide-resistant or thalidomide-sensitive to a previous therapy.
Collapse
Affiliation(s)
- T Guglielmelli
- Department of Clinical and Biological Sciences, University of Turin and San Luigi Hospital, Orbassano, Turin, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Valsami S, Ruf W, Leikauf MS, Madon J, Kaech A, Asmis LM. Immunomodulatory drugs increase endothelial tissue factor expression in vitro. Thromb Res 2010; 127:264-71. [PMID: 21159364 DOI: 10.1016/j.thromres.2010.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 11/17/2010] [Accepted: 11/17/2010] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Immunomodulatory compounds such as thalidomide (THL) and lenalidomide (LEN) represent treatment options for multiple myeloma. Venous thromboembolism is a potential complication of immunomodulatory treatment in myeloma patients. The optimal thromboprophylactic strategy to prevent this drug-induced hypercoagulable state is debated. It is the aim of this study to investigate i) the effect of immunomodulatory treatment on endothelial cell tissue factor (TF) expression and function, ii) the regulation of the observed TF activity, and iii) the modulating effect of low molecular weight heparin and aspirin on TF activity in vitro. MATERIALS AND METHODS These aims were addressed in an in vitro culture model, human umbilical vein endothelial cells, using TF activity and antigen assays as well flow cytometry, real time PCR and electron microscopy. RESULTS At THL and LEN concentrations resembling those observed in myeloma patients in vivo and in the presence of tumor necrosis factor-α (TNFα) we observed significantly increased TF activity in human umbilical vein endothelial cells in vitro. Concordant changes were detected for tissue factor mRNA and TF whole cell antigen. Dalteparin and a mixture of monoclonal anti-TF antibodies inhibited TF activity by 100% and more than 80% respectively, while aspirin's inhibitory effect was only approximately 30%. In the presence of TNFα we detected the generation of endothelial cell-derived microparticles which expressed TF activity. CONCLUSIONS Our in vitro data support the hypothesis that THL and LEN induce a hypercoagulable state through increased endothelial TF expression.
Collapse
Affiliation(s)
- Serena Valsami
- Division of Hematology, University Hospital of Zurich, CH 8091 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
44
|
Richardson P, Mitsiades C, Laubach J, Schlossman R, Ghobrial I, Hideshima T, Munshi N, Anderson K. Lenalidomide in multiple myeloma: an evidence-based review of its role in therapy. CORE EVIDENCE 2010; 4:215-45. [PMID: 20694078 PMCID: PMC2899783 DOI: 10.2147/ce.s6002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is a relatively common and incurable hematological malignancy. Currently, there is no single standard therapy, with choice of treatment dependent on individual patient factors. Lenalidomide is an immunomodulatory drug with potent antitumor, antiangiogenic, immunomodulatory, and proapoptotic activity in MM. AIMS To evaluate the evidence for the use of lenalidomide in its current indication in relapsed or refractory MM, and additionally its investigational use for the treatment of newly diagnosed MM. EVIDENCE REVIEW In patients with relapsed and refractory MM, adding lenalidomide to high-dose dexamethasone significantly improves response rates and time-to-progression, relative to high-dose dexamethasone alone. This translates into a significant extension of overall survival (with a median extension of 9.1 months in a pivotal phase III study). Outcome is independent of patient age, number of previous therapies, type of previous therapy (including thalidomide or autologous stem cell transplantation), renal impairment, and beta(2)-microglobulin level. Evidence suggests that combining lenalidomide with low-dose dexamethasone improves outcomes in patients with newly diagnosed disease and is superior to lenalidomide combined with high-dose dexamethasone. Myelosuppression is the predominant toxicity observed, although some studies have shown high incidences of venous thromboembolism in the absence of prophylactic antithrombotic anticoagulation therapy. There is currently only limited evidence regarding the health economics of lenalidomide. ROLE IN THERAPY: The encouraging results obtained with lenalidomide alone and in combination with dexamethasone in patients with relapsed or refractory MM have led to its adoption as a recommended therapy in patients who have received at least one prior treatment. Emerging evidence supports the ongoing investigation of lenalidomide in combination with low-dose dexamethasone, and in other combinations including bortezomib, for use both in relapsed, refractory, and newly diagnosed MM.
Collapse
Affiliation(s)
- Paul Richardson
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Constantine Mitsiades
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jacob Laubach
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Robert Schlossman
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Irene Ghobrial
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Teru Hideshima
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth Anderson
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Struga M, Miroslaw B, Pakosinska-Parys M, Drzewiecka A, Borowski P, Kossakowski J, Koziol AE. Synthesis, characterization and supramolecular synthons in crystals of new derivatives of 10-oxa-4-azatricyclo[5.2.1.02,6]dec-8-ene-3,5-dione. J Mol Struct 2010. [DOI: 10.1016/j.molstruc.2009.11.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Antimicrobial activity of 10-(diphenylmethylene)-4-azatricyclo[5.2.1.02,6]dec-8-ene-3,5-dione derivatives. ANN MICROBIOL 2010. [DOI: 10.1007/s13213-009-0013-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
47
|
Christoffersen T, Guren TK, Spindler KLG, Dahl O, Lønning PE, Gjertsen BT. Cancer therapy targeted at cellular signal transduction mechanisms: Strategies, clinical results, and unresolved issues. Eur J Pharmacol 2009; 625:6-22. [DOI: 10.1016/j.ejphar.2009.10.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 09/30/2009] [Accepted: 10/08/2009] [Indexed: 12/19/2022]
|
48
|
Abstract
After decades of disuse because of its teratogenic effects, thalidomide has had a resurgence of use as a promising therapeutic agent for multiple myeloma. Its mechanism of action involves activation of the immune system, antiangiogenic effects, and inhibition of cytokines. Thalidomide does not interact with the cytochrome oxidase system. It is not significantly metabolized, but it does undergo nonenzymatic hydrolysis in plasma. The resulting products are inactive. Despite the potential adverse effects of peripheral neuropathy, constipation, deep vein thrombosis, somnolence, rash, and orthostatic hypotension, thalidomide is an effective first-line agent for multiple myeloma in combination with dexamethasone or melphalan and prednisone. It has also been studied in the palliative care of patients with cytokine-based syndromes such as anorexia-cachexia syndrome. This review describes its use in oncology, hematology, and palliative care.
Collapse
Affiliation(s)
- Eric E. Prommer
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, Arizona,
| |
Collapse
|
49
|
Monocytes promote tumor cell survival in T-cell lymphoproliferative disorders and are impaired in their ability to differentiate into mature dendritic cells. Blood 2009; 114:2936-44. [PMID: 19671921 DOI: 10.1182/blood-2009-05-220111] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A variety of nonmalignant cells present in the tumor microenvironment promotes tumorigenesis by stimulating tumor cell growth and metastasis or suppressing host immunity. The role of such stromal cells in T-cell lymphoproliferative disorders is incompletely understood. Monocyte-derived cells (MDCs), including professional antigen-presenting cells such as dendritic cells (DCs), play a central role in T-cell biology. Here, we provide evidence that monocytes promote the survival of malignant T cells and demonstrate that MDCs are abundant within the tumor microenvironment of T cell-derived lymphomas. Malignant T cells were observed to remain viable during in vitro culture with autologous monocytes, but cell death was significantly increased after monocyte depletion. Furthermore, monocytes prevent the induction of cell death in T-cell lymphoma lines in response to either serum starvation or doxorubicin, and promote the engraftment of these cells in nonobese diabetic/severe combined immunodeficient mice. Monocytes are actively recruited to the tumor microenvironment by CCL5 (RANTES), where their differentiation into mature DCs is impaired by tumor-derived interleukin-10. Collectively, the data presented demonstrate a previously undescribed role for monocytes in T-cell lymphoproliferative disorders.
Collapse
|
50
|
Cardiovascular toxicities: clues to optimal administration of vascular endothelial growth factor signaling pathway inhibitors. Target Oncol 2009; 4:67-76. [PMID: 19373440 DOI: 10.1007/s11523-009-0106-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Accepted: 03/25/2009] [Indexed: 01/07/2023]
Abstract
Several angiogenesis inhibitors have been approved for commercial use and many additional agents are under development for the treatment of various malignancies. Cardiovascular toxicities have been increasingly recognized as effects of this entire class of new anticancer therapeutics. There is a limited but growing understanding of the mechanism of action of these drugs in the human cancer patient and the factors affecting the therapeutic index. In addition to reviewing current concepts for the cardiovascular toxicities of angiogenesis inhibitors, we discuss how better understanding the pharmacologic basis for these effects could optimize their use for individual patients.
Collapse
|