1
|
Shi X, Zhou Y, Du B, Yao X, Du X. Wound healing after surgical therapy for multiple myeloma: a case-control study. J Wound Care 2024; 33:866-874. [PMID: 39480735 DOI: 10.12968/jowc.2024.0324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
OBJECTIVE The aim of this study was to observe the surgical wound healing process in patients with multiple myeloma who had undergone surgery. METHOD We collected clinical data on patients with multiple myeloma and observed wound healing following surgical therapy. Additionally, we compared the expression of angiogenesis markers in patients with and without multiple myeloma (undergoing surgical excision of other tumour tissues). In patients who had multiple myeloma bone disease, we examined several clinical features: haemoglobin levels; albumin levels; blood glucose levels; and surgery programme. We then compared expression levels of the angiogenesis markers CD31, CD34 and vascular endothelial growth factor (VEGF) in samples scraped from the skin margin of the surgical incision in 12 patients without multiple myeloma (control) and nine patients with multiple myeloma. RESULTS All 61 patients with multiple myeloma observed showed no disunion, no delayed union and no infection in their wound healing. CD31 and VEGF expression was higher in the nine patients with multiple myeloma compared with the 12 control patients without. We observed no difference in CD34 expression between control and experimental groups. CONCLUSION The results of this study suggest that patients with multiple myeloma who have undergone surgery recover well and produce higher quantities of new vessels compared with patients without multiple myeloma. This occurs through increased expression of CD31 and VEGF, angiogenic factors which promote wound healing. We did not observe higher expression of these factors contributing to increased incisional implantation metastasis.
Collapse
Affiliation(s)
- Xiangjun Shi
- Department of Hematology, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100043, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yanyou Zhou
- Department of Orthopaedics, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100043, China
| | - Boran Du
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100084, China
| | - Xingchen Yao
- Department of Orthopaedics, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100043, China
| | - Xinru Du
- Department of Orthopaedics, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100043, China
| |
Collapse
|
2
|
Costa BA, Mouhieddine TH, Richter J. What's Old is New: The Past, Present and Future Role of Thalidomide in the Modern-Day Management of Multiple Myeloma. Target Oncol 2022; 17:383-405. [PMID: 35771402 DOI: 10.1007/s11523-022-00897-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 10/17/2022]
Abstract
Immunomodulatory drugs (IMiDs) have become an integral part of therapy for both newly diagnosed and relapsed/refractory multiple myeloma (RRMM). IMiDs bind to cereblon, leading to the degradation of proteins involved in B-cell survival and proliferation. Thalidomide, a first-generation IMiD, has little to no myelosuppressive potential, negligible renal clearance, and long-proven anti-myeloma activity. However, thalidomide's adverse effects (e.g., somnolence, constipation, and peripheral neuropathy) and the advent of more potent therapeutic options has led to the drug being less frequently used in many countries, including the US and Canada. Newer-generation IMiDs, such as lenalidomide and pomalidomide, are utilized far more frequently. In numerous previous trials, salvage therapy with thalidomide (50-200 mg/day) plus corticosteroids (with or without selected cytotoxic or targeted agents) has been shown to be effective and well-tolerated in the RRMM setting. Hence, thalidomide-based regimens remain important alternatives for heavily pretreated patients, especially for those who have no access to novel therapies and/or are not eligible for their use (due to renal failure, high-grade myelosuppression, or significant comorbidities). Ongoing and future trials may provide further insights into the current role of thalidomide, especially by comparing thalidomide-containing regimens with protocols based on newer-generation IMiDs and by investigating thalidomide's association with novel therapies (e.g., antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T cells).
Collapse
Affiliation(s)
- Bruno Almeida Costa
- Department of Medicine, Mount Sinai Morningside and West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tarek H Mouhieddine
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA.
| |
Collapse
|
3
|
Charliński G, Vesole DH, Jurczyszyn A. Rapid Progress in the Use of Immunomodulatory Drugs and Cereblon E3 Ligase Modulators in the Treatment of Multiple Myeloma. Cancers (Basel) 2021; 13:4666. [PMID: 34572892 PMCID: PMC8468542 DOI: 10.3390/cancers13184666] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades, the improvement in our understanding of the biology of MM and the introduction of new drug classes, including immunomodulatory drugs (IMiDs), proteasome inhibitors (PI), and monoclonal antibodies (MoAb), have significantly improved outcomes. The first IMiD introduced to treat MM was thalidomide. The side effects observed during treatment with thalidomide initiated work on the synthesis of IMiD analogs. Subsequently, lenalidomide and pomalidomide were developed, both with different safety profiles, and they have better tolerability than thalidomide. In 2010, the cereblon (CRBN) protein was discovered as a direct target of IMiDs. By binding to CRBN, IMiDs change the substrate specificity of the CRBN E3 ubiquitin ligase complex, which results in the breakdown of internal Ikaros and Aiolos proteins. Most clinical trials conducted, both in newly diagnosed, post-transplant maintenance and relapsed/refractory MM, report a beneficial effect of IMiDs on the extension of progression-free survival and overall survival in patients with MM. Due to side effects, thalidomide is used less frequently. Currently, lenalidomide is used at every phase of MM treatment. Lenalidomide is used in conjunction with other agents such as PIs and MoAb as induction and relapsed therapy. Pomalidomide is currently used to treat relapsed/refractory MM, also with PIs and monoclonal antibodies. Current clinical trials are evaluating the efficacy of IMiD derivatives, the CRBN E3 ligase modulators (CELMoDs). This review focuses on the impact of IMiDs for the treatment of MM.
Collapse
Affiliation(s)
- Grzegorz Charliński
- Department of Hematology, Warmian-Masurian Cancer Center of The Ministry of The Interior and Administration’s Hospital, 10-228 Olsztyn, Poland;
| | - David H. Vesole
- John Theurer Cancer Center at Hackensack Meridian School of Medicine, Hackensack, NJ 07601, USA;
| | - Artur Jurczyszyn
- Plasma Cell Dyscrasia Center, Department of Hematology, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Kraków, Poland
| |
Collapse
|
4
|
Watson N, Al-Samkari H. Thrombotic and bleeding risk of angiogenesis inhibitors in patients with and without malignancy. J Thromb Haemost 2021; 19:1852-1863. [PMID: 33928747 DOI: 10.1111/jth.15354] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023]
Abstract
Over the past two decades, therapies targeting angiogenesis have developed into a major class of cancer therapeutics. The vascular endothelial growth factor (VEGF) family of signaling proteins, a group of potent angiogenic growth factors, and their receptors represent the main targets of this therapeutic class. To date, 16 antiangiogenic agents have been approved in the United States for the treatment of cancer and several more are in development. An important consideration with antiangiogenic therapy is toxicity, in particular thrombotic and bleeding risks. These complications have emerged as a major clinical concern that may affect the use of these agents in patients both with and without cancer who may already have an elevated risk of thrombosis and bleeding. Although these agents are frequently considered together as a class when contemplating their bleeding and thrombotic risks, in fact the risks for venous thromboembolism, arterial thrombosis, and bleeding vary significantly between different classes of antiangiogenic agents and even among different agents within a class. In this narrative review, we describe the literature investigating the venous and arterial thrombotic and bleeding risks associated with the currently available antiangiogenic drugs. In addition, we discuss these specific complications in the context of both cancer therapy as well as the management of nonmalignant disorders now managed with antiangiogenic agents, including hereditary hemorrhagic telangiectasia and neovascular age-related macular degeneration.
Collapse
Affiliation(s)
| | - Hanny Al-Samkari
- Harvard Medical School, Boston, MA, USA
- Division of Hematology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
5
|
Barbarossa A, Iacopetta D, Sinicropi MS, Franchini C, Carocci A. Recent Advances in the Development of Thalidomide-Related Compounds as Anticancer Drugs. Curr Med Chem 2021; 29:19-40. [PMID: 34165402 DOI: 10.2174/0929867328666210623143526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Thalidomide is an old well-known drug that was first used as morning sickness relief in pregnant women before being withdrawn from the market due to its severe side effects on normal fetal development, However, over the last few decades, the interest in this old drug has been renewed because of its efficacy in several important disorders for instance, multiple myeloma, breast cancer, and HIV-related diseases due to its antiangiogenic and immunomodulatory properties. Unfortunately, even in these cases, many aftereffects as deep vein thrombosis, peripheral neuropathy, constipation, somnolence, pyrexia, pain, and teratogenicity have been reported, showing the requirement of careful and monitored use. For this reason, research efforts are geared toward the synthesis and optimization of new thalidomide analogues lacking in toxic effects to erase these limits and improve the pharmacological profile. AIMS This review aims to examine the state-of-the-art concerning the current studies on thalidomide and its analogues towards cancer diseases (with few hints regarding the antimicrobial activity), focusing the attention on the possible mechanisms of action involved and the lack of toxicity. CONCLUSION In the light of the collected data, thalidomide analogues and their ongoing optimization could lead, in the future, to the realization of a promising therapeutic alternative for cancer-fighting.
Collapse
Affiliation(s)
- Alexia Barbarossa
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Carlo Franchini
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Alessia Carocci
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy
| |
Collapse
|
6
|
Hill CS, Devesa SC, Ince W, Borg A, Aquilina K. A systematic review of ongoing clinical trials in optic pathway gliomas. Childs Nerv Syst 2020; 36:1869-1886. [PMID: 32556546 PMCID: PMC7434789 DOI: 10.1007/s00381-020-04724-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Optic pathway gliomas (OPGs), also known as Visual Pathway Gliomas, are insidious, debilitating tumours. They are most commonly WHO grade 1 pilocytic astrocytomas and frequently occur in patients with neurofibromatosis type 1. The location of OPGs within the optic pathway typically precludes complete resection or optimal radiation dosing, hence outcomes remain poor compared to many other low-grade gliomas. The aim of this systematic review was to formulate a comprehensive list of all current ongoing clinical trials that are specifically looking at clinical care of OPGs in order to identify trends in current research and provide an overview to guide future research efforts. METHODS This systematic review was conducted in line with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. The Cochrane Controlled Register of Trials (CENTRAL) and ClinicalTrials.gov were searched. Inclusion and exclusion criteria were applied and final results were reviewed. RESULTS 501 clinical trials were identified with the search strategy. All were screened and eligible studies extracted and reviewed. This yielded 36 ongoing clinical trials, 27 of which were pharmacological agents in phase I-III. The remaining trials were a mixture of biological agents, radiation optimisation, diagnostic imaging, surgical intervention, and a social function analysis. CONCLUSION OPG is a complex multifaceted disease, and advances in care require ongoing research efforts across a spectrum of different research fields. This review provides an update on the current state of research in OPG and summarises ongoing trials.
Collapse
Affiliation(s)
- Ciaran Scott Hill
- Department of Neurosurgery, Great Ormond Street Hospital, London, UK.
- UCL Cancer Institute, University College London, London, UK.
| | | | - William Ince
- Ipswich Hospital, East Suffolk and North Essex NHS Trust, Health Road, Ipswich, IP45PD, UK
| | - Anouk Borg
- Department of Neurosurgery, Oxford University Hospital, Oxford, UK
| | - Kristian Aquilina
- Department of Neurosurgery, Great Ormond Street Hospital, London, UK
| |
Collapse
|
7
|
Li Y, Shan NN, Sui XH. Research Progress on Artemisinin and Its Derivatives against Hematological Malignancies. Chin J Integr Med 2020; 26:947-955. [PMID: 32048169 DOI: 10.1007/s11655-019-3207-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
Although current therapeutic methods against hematological malignancies are effective in the early stage, they usually lose their effectiveness because of the development of drug resistances. Seeking new drugs with significant therapeutic effects is one of the current research hotspots. Artemisinin, an extract from the plant Artemisia annua Linne, and its derivatives have excellent antimalarial effects in clinical applications as well as excellent safety. Recent studies have documented that artemisinin and its derivatives (ARTs) also have significant effects against multiple types of tumours, including hematological malignancies. This review focuses on the latest research achievements of ARTs in the treatment of hematological malignancies as well as its mechanisms and future applications. The mechanisms of ARTs against different types of hematological malignancies mainly include cell cycle arrest, induction autophagy and apoptosis, inhibition of angiogenesis, production of reactive oxygen species, and induction of differentiation. Additionally, the review also summarizes the anticancer effects of ARTs in many drug-resistant hematological malignancies and its synergistic effects with other drugs.
Collapse
Affiliation(s)
- Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Ning-Ning Shan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Xiao-Hui Sui
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| |
Collapse
|
8
|
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: 1.3] [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
|
9
|
Thalidomide combined with chemotherapy in treating elderly patients with advanced gastric cancer. Aging Clin Exp Res 2018; 30:499-505. [PMID: 28660594 DOI: 10.1007/s40520-017-0790-z] [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: 04/04/2017] [Accepted: 06/17/2017] [Indexed: 10/19/2022]
Abstract
AIM The current systemic chemotherapy brings toxicity to human body, which elder patients suffer more than young people. The effective and well-tolerated treatment methods are of great importance for elderly advanced GC patients. This paper proposed an effective way of combining thalidomide with chemotherapy to treat elderly advanced GC patients, on the purpose of improving life quality and the treatment efficacy. METHODS In the control group, capecitabine was given with 2000 mg/m2 daily in a manner of 2 weeks on and 1 week off for elderly advanced GC patients. In the study group, thalidomide was given with 100 mg per day concurrently with chemotherapy additionally administered. RESULTS No significant differences were observed in the major prognostic factors among 64 eligible patients between the study and control groups. The ORRs and DCRs of the treatment and control groups showed no significant difference (P > 0.05). PFS of the study and control groups were 5.3 months (95% CI 4.5-6.2) and 4.2 months (95% CI 3.4-5.1), respectively. PFS exhibited a significant difference between the two group (P = 0.03), while the overall survivals of the patients between the two groups (10.4 months vs. 9.7 months) resulted as statistically non-significant (P = 0.47). Adverse effects were minimal in the study group, only a few patients suffered the grade 3 toxicity. The rate of drowsiness, fatigue, constipation of the study group was higher than that of the control group, and the rate of anorexia was lower (P < 0.05). CONCLUSIONS Our results demonstrated that thalidomide combined with capecitabine was mildly effective and safe for treating elderly patients with advanced GC.
Collapse
|
10
|
Abstract
Multiple myeloma (MM) is an incurable hematopoietic cancer that is characterized by malignant plasma cell infiltration of the bone marrow and/or extramedullary sites. Multi-modality approaches including "novel agents," traditional chemotherapy, and/or stem cell transplantation are used in MM therapy. Drug resistance, however, ultimately develops and the disease remains incurable for the vast majority of patients. In this chapter, we review both tumor cell-autonomous and non-autonomous (microenvironment-dependent) mechanisms of drug resistance. MM provides an attractive paradigm highlighting a number of current concepts and challenges in oncology. Firstly, identification of MM cancer stem cells and their unique drug resistance attributes may provide rational avenues towards MM eradication and cure. Secondly, the oligoclonal evolution of MM and alternation of "clonal tides" upon therapy challenge our current understanding of treatment responses. Thirdly, the success of MM "novel agents" provides exemplary evidence for the impact of therapies that target the immune and non-immune microenvironment. Fourthly, the rapid pace of drug approvals for MM creates an impetus for development of precision medicine strategies and biomarkers that promote efficacy and mitigate toxicity and cost. While routine cure of the disease remains the ultimate and yet unattainable prize, MM advances in the last 10-15 years have provided an astounding paradigm for the treatment of blood cancers in the modern era and have radically transformed patient outcomes.
Collapse
Affiliation(s)
- Athanasios Papadas
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- UW Carbone Cancer Center, Madison, WI, 53705, USA.
| | - Fotis Asimakopoulos
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
- UW Carbone Cancer Center, Madison, WI, 53705, USA
| |
Collapse
|
11
|
Ríos-Tamayo R, Martín-García A, Alarcón-Payer C, Sánchez-Rodríguez D, de la Guardia AMDVD, García Collado CG, Jiménez Morales A, Jurado Chacón M, Cabeza Barrera J. Pomalidomide in the treatment of multiple myeloma: design, development and place in therapy. Drug Des Devel Ther 2017; 11:2399-2408. [PMID: 28860711 PMCID: PMC5574598 DOI: 10.2147/dddt.s115456] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Multiple myeloma is a very heterogeneous disease with variable survival. Despite recent progress and the widespread use of new agents, patients with relapsed and refractory disease have a poor outcome. Immunomodulatory drugs play a key role in both the front-line and the relapsed/refractory setting. The combination of pomalidomide (POM) and dexamethasone is safe and effective in relapsed and refractory patients, even in those with high-risk cytogenetic features. Furthermore, it can be used in most patients without the need to adjust according to the degree of renal failure. In order to further improve the results, POM-based triplet therapies are currently used. This article highlights the most relevant issues of POM and POM-based combinations in the relapsed/refractory multiple myeloma setting, from a pharmacological and clinical point of view.
Collapse
Affiliation(s)
- Rafael Ríos-Tamayo
- Monoclonal Gammopathies Unit
- Department of Hematology, University Hospital Virgen de las Nieves, Granada, Spain
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| | - Agustín Martín-García
- Department of Pharmacy
- Clinical Trials Unit, University Hospital Virgen de las Nieves, Granada, Spain
| | | | | | | | | | | | - Manuel Jurado Chacón
- Monoclonal Gammopathies Unit
- Department of Hematology, University Hospital Virgen de las Nieves, Granada, Spain
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| | - José Cabeza Barrera
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- Department of Pharmacy
| |
Collapse
|
12
|
The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017. [PMID: 28635679 DOI: 10.3390/biomedicines5020034]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A number of anti-angiogenesis drugs have been FDA-approved and are being used in cancer treatment, and a number of other agents are in different stages of clinical development or in preclinical evaluation. However, pharmacologic anti-angiogenesis strategies that arrest tumor progression might not be enough to eradicate tumors. Decreased anti-angiogenesis activity in single mechanism-based anti-angiogenic strategies is due to the redundancy, multiplicity, and development of compensatory mechanism by which blood vessels are remodeled. Improving anti-angiogenesis drug efficacy will require identification of broad-spectrum anti-angiogenesis targets. These strategies may have novel features, such as increased porosity, and are the result of complex interactions among endothelial cells, extracellular matrix proteins, growth factors, pericyte, and smooth muscle cells. Thus, combinations of anti-angiogenic drugs and other anticancer strategies such as chemotherapy appear essential for optimal outcome in cancer patients. This review will focus on the role of anti-angiogenesis strategies in cancer treatment.
Collapse
|
13
|
Abstract
A number of anti-angiogenesis drugs have been FDA-approved and are being used in cancer treatment, and a number of other agents are in different stages of clinical development or in preclinical evaluation. However, pharmacologic anti-angiogenesis strategies that arrest tumor progression might not be enough to eradicate tumors. Decreased anti-angiogenesis activity in single mechanism-based anti-angiogenic strategies is due to the redundancy, multiplicity, and development of compensatory mechanism by which blood vessels are remodeled. Improving anti-angiogenesis drug efficacy will require identification of broad-spectrum anti-angiogenesis targets. These strategies may have novel features, such as increased porosity, and are the result of complex interactions among endothelial cells, extracellular matrix proteins, growth factors, pericyte, and smooth muscle cells. Thus, combinations of anti-angiogenic drugs and other anticancer strategies such as chemotherapy appear essential for optimal outcome in cancer patients. This review will focus on the role of anti-angiogenesis strategies in cancer treatment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
14
|
Rajabi M, Mousa SA. The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017; 5:E34. [PMID: 28635679 PMCID: PMC5489820 DOI: 10.3390/biomedicines5020034] [Citation(s) in RCA: 401] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/09/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022] Open
Abstract
A number of anti-angiogenesis drugs have been FDA-approved and are being used in cancer treatment, and a number of other agents are in different stages of clinical development or in preclinical evaluation. However, pharmacologic anti-angiogenesis strategies that arrest tumor progression might not be enough to eradicate tumors. Decreased anti-angiogenesis activity in single mechanism-based anti-angiogenic strategies is due to the redundancy, multiplicity, and development of compensatory mechanism by which blood vessels are remodeled. Improving anti-angiogenesis drug efficacy will require identification of broad-spectrum anti-angiogenesis targets. These strategies may have novel features, such as increased porosity, and are the result of complex interactions among endothelial cells, extracellular matrix proteins, growth factors, pericyte, and smooth muscle cells. Thus, combinations of anti-angiogenic drugs and other anticancer strategies such as chemotherapy appear essential for optimal outcome in cancer patients. This review will focus on the role of anti-angiogenesis strategies in cancer treatment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
15
|
Shared mechanism of teratogenicity of anti-angiogenic drugs identified in the chicken embryo model. Sci Rep 2016; 6:30038. [PMID: 27443489 PMCID: PMC4957076 DOI: 10.1038/srep30038] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/29/2016] [Indexed: 12/24/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is essential for tumor growth, stabilization and progression. Angiogenesis inhibitors are now widely used in the clinic; however, there are relatively few published studies on the mechanism of their presumed teratogenic effects. To address this issue, we screened a variety of angiogenesis inhibitors in developing zebrafish and chicken embryo models to assess for developmental defects and potential teratogenic effects. We confirmed previous reports that sunitinib, sorafenib and TNP-470 are teratogenic and demonstrate that axitinib, pazopanib, vandetanib, and everolimus are also teratogens in these models. A dose response study identified the drugs inhibit HUVEC cell proliferation in vitro, and also target the developing blood vessels of embryos in vivo. This provides further evidence for the potential risk of fetal toxicity when using these drugs in a clinical setting, and emphasizes the importance of the development and maintenance of the vasculature in the embryo. We conclude that angiogenesis inhibitors, regardless of the molecular target, are teratogenic when exposed to chicken embryos.
Collapse
|
16
|
Smith SL, Singh P, Harding D, Lun D, Chambers JP. Thalidomide pharmacokinetics in sheep. N Z Vet J 2016; 64:238-42. [PMID: 26727254 DOI: 10.1080/00480169.2015.1130663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
AIM To determine the half life (T1/2), time taken to reach maximum plasma concentration (Tmax) and maximum plasma concentration (Cmax) of thalidomide in sheep following I/V, oral and topical treatment with a single dose of thalidomide. METHOD Three groups of 4-6-month-old ram lambs were treated with thalidomide dissolved in dimethylsulphoxide (DMSO). The first group (n=10) was treated I/V with 100 mg thalidomide in 2 mL DMSO; the second group (n=8) received 400 mg thalidomide in 2 mL DMSO orally, and the third group (n=8) had 400 mg thalidomide in 4 mL DMSO applied topically. Plasma samples were collected up to 36 hours after treatment, snap-frozen at -80°C and analysed for concentrations of thalidomide using high performance liquid chromatography. RESULTS Following I/V administration, T1/2 was 5.0 (SEM 0.4) hours, volume of distribution was 3,372.0 (SEM 244.3) mL/kg and clearance was 487.1 (SEM 46.1) mL/hour.kg. Topical application of 400 mg thalidomide did not increase plasma concentrations. Following oral administration, thalidomide bioavailability was 89%, with T1/2, Tmax, and Cmax being 7.2 (SEM 0.8) hours, 3.0 (SEM 0.4) hours and 1,767.3 (SEM 178.1) ng/mL, respectively. CONCLUSION Topical administration using DMSO as a solvent did not increase concentrations of thalidomide in plasma. The mean pharmacokinetic parameters determined following oral treatment with 400 mg of thalidomide were similar to those reported in humans receiving a single 400 mg oral dose (T1/2 7.3 hours; Tmax 4.3 hours and Cmax 2,820 ng/mL). There is potential for thalidomide to be used as a model for the treatment of chronic inflammatory conditions in sheep, such as Johne's disease, where tumour necrosis factor alpha plays a pathogenic role.
Collapse
Affiliation(s)
- S L Smith
- a Institute of Veterinary, Animal and Biological Sciences , Massey University , Tennent Drive, Palmerston North 4443 , New Zealand
| | - P Singh
- a Institute of Veterinary, Animal and Biological Sciences , Massey University , Tennent Drive, Palmerston North 4443 , New Zealand
| | - D Harding
- b Institute of Fundamental Sciences , Massey University , Tennent Drive, Palmerston North 4474 , New Zealand
| | - D Lun
- b Institute of Fundamental Sciences , Massey University , Tennent Drive, Palmerston North 4474 , New Zealand
| | - J P Chambers
- a Institute of Veterinary, Animal and Biological Sciences , Massey University , Tennent Drive, Palmerston North 4443 , New Zealand
| |
Collapse
|
17
|
Huang XE, Yan XC, Wang L, Ji ZQ, Li L, Liu MY, Qian T, Shen HL, Gu HG, Liu Y, Gu M, Deng LC. Thalidomide Combined with Chemotherapy in Treating Patients with Advanced Colorectal Cancer. Asian Pac J Cancer Prev 2015; 16:7867-9. [DOI: 10.7314/apjcp.2015.16.17.7867] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
18
|
Assis PA, De Figueiredo-Pontes LL, Lima ASG, Leão V, Cândido LA, Pintão CT, Garcia AB, Saggioro FP, Panepucci RA, Chahud F, Nagler A, Falcão RP, Rego EM. Halofuginone inhibits phosphorylation of SMAD-2 reducing angiogenesis and leukemia burden in an acute promyelocytic leukemia mouse model. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:65. [PMID: 26099922 PMCID: PMC4486128 DOI: 10.1186/s13046-015-0181-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 06/11/2015] [Indexed: 01/09/2023]
Abstract
Background Halofuginone (HF) is a low-molecular-weight alkaloid that has been demonstrated to interfere with Metalloproteinase-2 (MMP-2) and Tumor Growth Factor-β (TGF-β) function and, to present antiangiogenic, antiproliferative and proapoptotic properties in several solid tumor models. Based on the fact that high levels of Vascular Endothelial Growth Factor (VEGF) and increased angiogenesis have been described in acute myeloid leukemia and associated with disease progression, we studied the in vivo effects of HF using an Acute Promyelocytic Leukemia (APL) mouse model. Methods NOD/SCID mice were transplanted with leukemic cells from hCG-PML/RARA transgenic mice (TM) and treated with HF 150 μg/kg/day for 21 days. The leukemic infiltration and the percentage of VEGF+ cells were evaluated by morphology and flow cytometry. The effect of HF on the gene expression of several pro- and antiangiogenic factors, phosphorylation of SMAD2 and VEGF secretion was assessed in vitro using NB4 and HUVEC cells. Results HF treatment resulted in hematological remission with decreased accumulation of immature cell and lower amounts of VEGF in BM of leukemic mice. In vitro, HF modulated gene expression of several pro- and antiangiogenic factors, reduced VEGF secretion and phosphorylation of SMAD2, blocking TGF-β-signaling. Conclusion Taken together, our results demonstrate that HF inhibits SMAD2 signaling and reduces leukemia growth and angiogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0181-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patricia A Assis
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Lorena L De Figueiredo-Pontes
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Ana Silvia G Lima
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Vitor Leão
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Larissa A Cândido
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Carolina T Pintão
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Aglair B Garcia
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Fabiano P Saggioro
- Pathology Department, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Rodrigo A Panepucci
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Fernando Chahud
- Pathology Department, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Arnon Nagler
- Hematology Division and Cord Blood Bank, Chaim Sheba Medical Center, Tel Aviv University, Tel Hashomer, 6997801, Israel.
| | - Roberto P Falcão
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Eduardo M Rego
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| |
Collapse
|
19
|
Calcinotto A, Ponzoni M, Ria R, Grioni M, Cattaneo E, Villa I, Sabrina Bertilaccio MT, Chesi M, Rubinacci A, Tonon G, Bergsagel PL, Vacca A, Bellone M. Modifications of the mouse bone marrow microenvironment favor angiogenesis and correlate with disease progression from asymptomatic to symptomatic multiple myeloma. Oncoimmunology 2015; 4:e1008850. [PMID: 26155424 PMCID: PMC4485787 DOI: 10.1080/2162402x.2015.1008850] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/14/2015] [Indexed: 11/12/2022] Open
Abstract
While multiple myeloma (MM) is almost invariably preceded by asymptomatic monoclonal gammopathy of undetermined significance (MGUS) and/or smoldering MM (SMM), the alterations of the bone marrow (BM) microenvironment that establish progression to symptomatic disease are circumstantial. Here we show that in Vk*MYC mice harboring oncogene-driven plasma cell proliferative disorder, disease appearance associated with substantial modifications of the BM microenvironment, including a progressive accumulation of both CD8+ and CD4+ T cells with a dominant T helper type 1 (Th1) response. Progression from asymptomatic to symptomatic MM was characterized by further BM accrual of T cells with reduced Th1 and persistently increased Th2 cytokine production, which associated with accumulation of CD206+Tie2+ macrophages, and increased pro-angiogenic cytokines and microvessel density (MVD). Notably, MVD was also increased at diagnosis in the BM of MGUS and SMM patients that subsequently progressed to MM when compared with MGUS and SMM that remained quiescent. These findings suggest a multistep pathogenic process in MM, in which the immune system may contribute to angiogenesis and disease progression. They also suggest initiating a large multicenter study to investigate MVD in asymptomatic patients as prognostic factor for the progression and outcome of this disease.
Collapse
Affiliation(s)
- Arianna Calcinotto
- Cellular Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Università Vita-Salute San Raffaele ; Milan, Italy
| | - Maurilio Ponzoni
- Pathology and Myeloma Units; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology; Internal Medicine and Clinical Oncology Unit; University of Bari Medical School ; Bari, Italy
| | - Matteo Grioni
- Cellular Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Elena Cattaneo
- Cellular Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Isabella Villa
- Bone Metabolic Unit; Division of Cardiovascular Science; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | | | - Marta Chesi
- Comprehensive Cancer Center; Mayo Clinic Arizona ; Scottsdale, AZ, USA
| | - Alessandro Rubinacci
- Bone Metabolic Unit; Division of Cardiovascular Science; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Giovanni Tonon
- Division of Molecular Oncology; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - P Leif Bergsagel
- Comprehensive Cancer Center; Mayo Clinic Arizona ; Scottsdale, AZ, USA
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology; Internal Medicine and Clinical Oncology Unit; University of Bari Medical School ; Bari, Italy
| | - Matteo Bellone
- Cellular Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| |
Collapse
|
20
|
Hassler MR, Sax C, Flechl B, Ackerl M, Preusser M, Hainfellner JA, Woehrer A, Dieckmann KU, Rössler K, Prayer D, Marosi C. Thalidomide as palliative treatment in patients with advanced secondary glioblastoma. Oncology 2015; 88:173-9. [PMID: 25427949 DOI: 10.1159/000368903] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/05/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND For its numerous abilities including sedation, we have been using thalidomide (TH) as the 'last therapeutic option' in patients with advanced gliomas. We noticed that a small subgroup, i.e. patients with secondary glioblastoma (GBM, whose GBM has evolved over several months or years from a less malignant glioma), survived for prolonged periods. Therefore, we retrospectively evaluated the outcomes of patients with secondary GBM treated with TH at our centre. PATIENTS AND METHODS Starting in the year 2000, we have studied 23 patients (13 females, 10 males, with a median age of 31.5 years) with secondary GBM who have received palliative treatment with TH 100 mg at bedtime. All patients had previously undergone radiotherapy and received at least 1 and up to 5 regimens of chemotherapy. RESULTS The median duration of TH administration was 4.0 months (range 0.8-32). The median duration of overall survival after the start of TH therapy was 18.3 months (range 0.8-57). Eleven patients with secondary GBM survived longer than 1 year. Symptomatic improvement was most prominent in the restoration of a normal sleep pattern. CONCLUSION The palliative effects of TH, especially the normalization of a sleep pattern, were highly valued by patients and families. The prolongation of survival of patients with secondary GBM has not been reported previously.
Collapse
|
21
|
Abraham J, Salama NN, Azab AK. The role of P-glycoprotein in drug resistance in multiple myeloma. Leuk Lymphoma 2014; 56:26-33. [PMID: 24678978 DOI: 10.3109/10428194.2014.907890] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multiple myeloma (MM) is a malignant neoplastic cancer of the plasma cells that involves the bone marrow. The majority of patients with MM initially respond to chemotherapy, but they eventually become resistant to later drug therapy. One of the reasons for drug resistance in patients with MM is efflux transporters. P-glycoprotein (P-gp) is the most studied of the multidrug resistance proteins, and is up-regulated in response to many chemotherapeutic drugs. This up-regulation of P-gp causes a decrease in the intracellular accumulation of these drugs, limiting their therapeutic efficacy. In this review, we focus on the role of P-gp in drugs used for patients with MM. P-gp has been found to be an important factor with regard to drug resistance in many of the drug classes used in the treatment of MM (proteasome inhibitors, anthracyclines, alkylating agents and immunomodulators are examples). Thus, our further understanding of its mechanism and inhibitory effects will help us decrease drug resistance in patients with MM.
Collapse
Affiliation(s)
- Joseph Abraham
- Cancer Biology Division, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis , St. Louis, MO , USA
| | | | | |
Collapse
|
22
|
Lee H, Banskota S, Kim DG, Been JH, Jin YJ, Gautam J, Jang H, Nam TG, Kim JA, Jeong BS. Synthesis and antiangiogenic activity of 6-amido-2,4,5-trimethylpyridin-3-ols. Bioorg Med Chem Lett 2014; 24:3131-6. [PMID: 24894557 DOI: 10.1016/j.bmcl.2014.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/23/2014] [Accepted: 05/02/2014] [Indexed: 12/24/2022]
Abstract
We recently reported that 6-aminoalkyl-2,4,5-trimethylpyridin-3-ols, novel series of 6-aminopyridin-3-ol-based antioxidants, have high antiangiogenic activities. In pursuit of wider variety in the analogues, we here report the synthesis and antiangiogenic activities of 6-amidoalkyl-2,4,5-trimethylpyridin-3-ols, which would not be considered excellent antioxidants because of the poorer electron-donating effect of the C(6)-amido group than the corresponding C(6)-amino group. The selected 6-amido compounds showed up to several fold-higher antiangiogenic activities and up to an order of magnitude better antitumor activities in the chick embryo chorioallantoic membrane (CAM) assay than SU4312, a positive control. We also found that paracetamol, as a direct phenolic analogue of our simplest 6-amidopyridin-3-ol, showed a moderate level of antiangiogenic activity. We propose this study will offer a basis for a scaffold of novel angiogenesis inhibitors that can perturb angiogenesis-related pathologies.
Collapse
Affiliation(s)
- Hyunji Lee
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Suhrid Banskota
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Dong-Guk Kim
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Jae-Hui Been
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - You-Jin Jin
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Republic of Korea
| | - Jaya Gautam
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Hyeonjin Jang
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Tae-gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Republic of Korea.
| | - Jung-Ae Kim
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea.
| | - Byeong-Seon Jeong
- College of Pharmacy and Institute for Drug Research, Yeungnam University, Gyeongsan 712-749, Republic of Korea.
| |
Collapse
|
23
|
The evaluation of angiogenesis and matrix metalloproteinase-2 secretion in bone marrow of multiple myeloma patients before and after the treatment. Adv Med Sci 2014; 58:118-25. [PMID: 23333895 DOI: 10.2478/v10039-012-0048-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Angiogenesis appears to be a prominent feature of many hematological disorders, particularly in multiple myeloma (MM). Progression in MM also involves secretion of the metaloproteinases (MMPs). In this study, the expression of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF) and its receptor, in bone marrow trephine biopsy (TB) of thirty six MM patients before and after the treatment or during progression was examined. The MMP-2 secretion was assessed from the same patients. MATERIAL/METHODS Immunohistochemical staining of bone marrow specimens for angiogenic factors and microvessel density (MVD) and bone marrow aspirates for Western blot analysis of MMP-2 expression was performed. RESULTS In active, untreated MM patients, we found statistically significant differences in the expression of angiogenic factors according to the patients after the anti-angiogenic treatment. We found statistical differences of the expression of angiogenic factors between the group of patients with a response after the treatment and the patients who had progression during the treatment. The data showed statistically significant decreased MVD after the treatment. The results showed statistically significant differences between initial secretion of MMP-2 in active, untreated MM patients and patients with a response after the treatment and patients with progression during the treatment. CONCLUSIONS We showed that not only decreased expression of angiogenic cytokines is present after the anti-angiogenic treatment but also activity of MMP-2 in MM patients who responded to the treatment. Combination therapy with the inhibition of the activity of MMPs could represent an interesting therapeutical approach in MM.
Collapse
|
24
|
BAFF and APRIL as TNF superfamily molecules and angiogenesis parallel progression of human multiple myeloma. Ann Hematol 2013; 93:635-44. [PMID: 24141333 PMCID: PMC3945232 DOI: 10.1007/s00277-013-1924-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/02/2013] [Indexed: 02/03/2023]
Abstract
Tumour necrosis factor alpha (TNF-α) is an inflammatory cytokine with a wide spectrum of biological activity, including angiogenesis. B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) are members of the TNF-α family. Vascular endothelial growth factor (VEGF), on the other hand, is one of the most characteristic pro-angiogenic cytokines produced by multiple cell types in multiple myeloma (MM). We have analysed BAFF and APRIL concentrations in parallel with pro-angiogenic cytokines in serum and trephine biopsy, and the bone marrow microvascular density (MVD) in 50 patients with newly diagnosed IgG MM and 24 healthy volunteers. The study showed statistically higher concentrations of BAFF, APRIL and TNF-α, as well as VEGF and its receptor, in MM patients compared to healthy volunteers and patients in advanced stages of the disease. A statistically positive correlation between the concentration of TNF-α and the expression of VEGF was demonstrated, and so was a positive link between BAFF, APRIL, MVD and lactate dehydrogenase (LDH). Furthermore, we observed a significant decrease in all studied cytokines after anti-angiogenic therapy, with meaningful differences between responders (at least partial remission) and patients with stable disease. It was also established that APRIL, but not BAFF, correlated with pro-angiogenic cytokines such as VEGF with its receptor, MVD and syndecan-1. Finally, our results showed that serum BAFF and APRIL levels could be useful biomarkers of MM disease activity and its progression which suggests that APRIL could be a possible novel therapeutic target in MM.
Collapse
|
25
|
Bone marrow microenvironment in multiple myeloma progression. J Biomed Biotechnol 2012; 2012:157496. [PMID: 23093834 PMCID: PMC3471001 DOI: 10.1155/2012/157496] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 09/18/2012] [Indexed: 12/28/2022] Open
Abstract
Substantial advances have been made in understanding the biology of multiple myeloma (MM) through the study of the bone marrow (BM) microenvironment. Indeed, the BM niche appears to play an important role in differentiation, migration, proliferation, survival, and drug resistance of the malignant plasma cells. The BM niche is composed of a cellular compartment (stromal cells, osteoblasts, osteoclasts, endothelial cells, and immune cells) and a noncellular compartment including the extracellular matrix (ECM) and the liquid milieu (cytokines, growth factors, and chemokines). In this paper we discuss how the interaction between the malignant plasma cell and the BM microenvironment allowed myeloma progression through cell homing and the new concept of premetastatic niche.
Collapse
|
26
|
Chen F, Feng Y, Zheng K, De Keyzer F, Li J, Feng Y, Cona MM, Wang H, Jiang Y, Yu J, Marchal G, Verfaillie C, De Geest B, Oyen R, Ni Y. Enhanced antitumor efficacy of a vascular disrupting agent combined with an antiangiogenic in a rat liver tumor model evaluated by multiparametric MRI. PLoS One 2012; 7:e41140. [PMID: 22815943 PMCID: PMC3399789 DOI: 10.1371/journal.pone.0041140] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 06/17/2012] [Indexed: 12/16/2022] Open
Abstract
A key problem in solid tumor therapy is tumor regrowth from a residual viable rim after treatment with a vascular disrupting agent (VDA). As a potential solution, we studied a combined treatment of a VDA and antiangiogenic. This study was approved by the institutional ethical committee for the use and care of laboratory animals. Rats with implanted liver tumors were randomized into four treatment groups: 1) Zd6126 (Zd); 2) Thalidomide (Tha); 3) Zd in combination with Tha (ZdTha); and 4) controls. Multiparametric MRIs were performed and quantified before and after treatment. Circulating endothelial progenitor cells (EPCs) and plasma stromal cell-derived factor-1α (SDF-1α) were monitored. Tumor apoptosis, necrosis, and microvessels were verified by histopathology. A single use of Zd or Tha did not significantly delay tumor growth. The combined ZdTha showed enhanced antitumor efficacy due to synergistic effects; it induced a cumulative tumor apoptosis or necrosis, which resulted in significant delay in tumor growth and reduction in the viable tumor rim; it also reduced tumor vessel permeability; and it improved tumor hemodynamic indexes, most likely via a transient normalization of tumor vasculature induced by Tha. A stepwise linear regression analysis showed that the apparent diffusion coefficient was an independent predictor of tumor growth. We found no significant increases in Zd-induced circulating EPCs or plasma SDF-1α. ZdTha showed improved therapeutic efficacy in solid tumors compared to either agent alone. The therapeutic effects were successfully tracked in vivo with multiparametric MRI.
Collapse
Affiliation(s)
- Feng Chen
- Theragnostic Laboratory, Department of Imaging and Pathology, University Hospital, University of Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chauhan D, Bandi M, Singh AV, Ray A, Raje N, Richardson P, Anderson KC. Preclinical evaluation of a novel SIRT1 modulator SRT1720 in multiple myeloma cells. Br J Haematol 2011; 155:588-98. [PMID: 21950728 DOI: 10.1111/j.1365-2141.2011.08888.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIRT1 belongs to the silent information regulator 2 (Sir2) protein family of enzymes and functions as a NAD(+) -dependent class III histone deacetylase. Here, we examined the anti-multiple myeloma (MM) activity of a novel oral agent, SRT1720, which targets SIRT1. Treatment of MM cells with SRT1720 inhibited growth and induced apoptosis in MM cells resistant to conventional and bortezomib therapies without significantly affecting the viability of normal cells. Mechanistic studies showed that anti-MM activity of SRT1720 is associated with: (i) activation of caspase-8, caspase-9, caspase-3, poly(ADP) ribose polymerase; (ii) increase in reactive oxygen species; (iii) induction of phosphorylated ataxia telangiectasia mutated/checkpoint kinase 2 signalling; (iv) decrease in vascular endothelial growth factor-induced migration of MM cells and associated angiogenesis; and (v) inhibition of nuclear factor-κB. Blockade of ATM attenuated SRT1720-induced MM cell death. In animal tumour model studies, SRT1720 inhibited MM tumour growth. Finally, SRT1720 enhanced the cytotoxic activity of bortezomib or dexamethasone. Our preclinical studies provide the rationale for novel therapeutics targeting SIRT1 in MM.
Collapse
Affiliation(s)
- Dharminder Chauhan
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Cheng J, Gu YJ, Wang Y, Cheng SH, Wong WT. Nanotherapeutics in angiogenesis: synthesis and in vivo assessment of drug efficacy and biocompatibility in zebrafish embryos. Int J Nanomedicine 2011; 6:2007-21. [PMID: 21976976 PMCID: PMC3181060 DOI: 10.2147/ijn.s20145] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Carbon nanotubes have shown broad potential in biomedical applications, given their unique mechanical, optical, and chemical properties. In this pilot study, carbon nanotubes have been explored as multimodal drug delivery vectors that facilitate antiangiogenic therapy in zebrafish embryos. METHODS Three different agents, ie, an antiangiogenic binding site (cyclic arginine-glycin-easpartic acid), an antiangiogenic drug (thalidomide), and a tracking dye (rhodamine), were conjugated onto single-walled carbon nanotubes (SWCNT). The biodistribution, efficacy, and biocompatibility of these triple functionalized SWCNT were tested in mammalian cells and validated in transparent zebrafish embryos. RESULTS Accumulation of SWCNT-associated nanoconjugates in blastoderm cells facilitated drug delivery applications. Mammalian cell xenograft assays demonstrated that these antiangiogenic SWCNT nanoconjugates specifically inhibited ectopic angiogenesis in the engrafted zebrafish embryos. CONCLUSION This study highlights the potential of using SWCNT for generating efficient nanotherapeutics.
Collapse
Affiliation(s)
- Jinping Cheng
- Department of Biology and Chemistry, The City University of Hong Kong, Kowloon
| | | | | | | | | |
Collapse
|
29
|
Mirandola L, Yu Y, Chui K, Jenkins MR, Cobos E, John CM, Chiriva-Internati M. Galectin-3C inhibits tumor growth and increases the anticancer activity of bortezomib in a murine model of human multiple myeloma. PLoS One 2011; 6:e21811. [PMID: 21765917 PMCID: PMC3135605 DOI: 10.1371/journal.pone.0021811] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/08/2011] [Indexed: 02/07/2023] Open
Abstract
Galectin-3 is a human lectin involved in many cellular processes including differentiation, apoptosis, angiogenesis, neoplastic transformation, and metastasis. We evaluated galectin-3C, an N-terminally truncated form of galectin-3 that is thought to act as a dominant negative inhibitor, as a potential treatment for multiple myeloma (MM). Galectin-3 was expressed at varying levels by all 9 human MM cell lines tested. In vitro galectin-3C exhibited modest anti-proliferative effects on MM cells and inhibited chemotaxis and invasion of U266 MM cells induced by stromal cell-derived factor (SDF)-1α. Galectin-3C facilitated the anticancer activity of bortezomib, a proteasome inhibitor approved by the FDA for MM treatment. Galectin-3C and bortezomib also synergistically inhibited MM-induced angiogenesis activity in vitro. Delivery of galectin-3C intravenously via an osmotic pump in a subcutaneous U266 cell NOD/SCID mouse model of MM significantly inhibited tumor growth. The average tumor volume of bortezomib-treated animals was 19.6% and of galectin-3C treated animals was 13.5% of the average volume of the untreated controls at day 35. The maximal effect was obtained with the combination of galectin-3C with bortezomib that afforded a reduction of 94% in the mean tumor volume compared to the untreated controls at day 35. In conclusion, this is the first study to show that inhibition of galectin-3 is efficacious in a murine model of human MM. Our results demonstrated that galectin-3C alone was efficacious in a xenograft mouse model of human MM, and that it enhanced the anti-tumor activity of bortezomib in vitro and in vivo. These data provide the rationale for continued testing of galectin-3C towards initiation of clinical trials for treatment of MM.
Collapse
Affiliation(s)
- Leonardo Mirandola
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, Texas, United States of America
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Yuefei Yu
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, Texas, United States of America
| | - Kitty Chui
- MandalMed, San Francisco, California, United States of America
| | - Marjorie R. Jenkins
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, Texas, United States of America
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Everardo Cobos
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, Texas, United States of America
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | | | - Maurizio Chiriva-Internati
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, Texas, United States of America
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
- * E-mail:
| |
Collapse
|
30
|
Tsukamoto I, Sakakibara N, Maruyama T, Igarashi J, Kosaka H, Kubota Y, Tokuda M, Ashino H, Hattori K, Tanaka S, Kawata M, Konishi R. A novel nucleic acid analogue shows strong angiogenic activity. Biochem Biophys Res Commun 2010; 399:699-704. [DOI: 10.1016/j.bbrc.2010.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 08/02/2010] [Indexed: 11/25/2022]
|
31
|
Yang DH, Kim YK, Sohn SK, Chung JS, Joo YD, Lee JH, Lee JL, Ahn JS, Moon JH, Shin HJ, Choi YJ, Lee WS, Kim HJ, Lee JJ. Induction treatment with cyclophosphamide, thalidomide, and dexamethasone in newly diagnosed multiple myeloma: a phase II study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2010; 10:62-7. [PMID: 20223731 DOI: 10.3816/clml.2010.n.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Thalidomide has alternative mechanisms of action; it can be combined with dexamethasone or alkylating agents for the treatment of multiple myeloma (MM); however, the optimal doses and appropriate intervals of thalidomide continue to be debated. PATIENTS AND METHODS We assessed the clinical efficacy and toxicity of thalidomide in patients with newly diagnosed MM; 68 patients were treated with pulsed cyclophosphamide, thalidomide, and dexamethasone (CTD) chemotherapy for induction treatment. RESULTS After a median of 28 months' follow-up, the overall response rate was 79.4%, with a 42.6% complete response (CR) or very good partial response (VGPR). Patients with cytogenetically high-risk disease had poor CR/VGPR rates (27.3%) at a median of 11.5 months of time to progression (TTP) compared with patients with standard-risk disease who achieved CR/VGPR rates (50%) at a median of 20.3 months of TTP. The major adverse events included peripheral sensory neuropathy (14.3%), infection (10.2%), and thromboembolic complications (5.9%). Thirty-two patients who achieved more than a PR proceeded to peripheral blood stem cell collection with a median number of 5.0 x 106 CD34+ cells/kg collected. CONCLUSION CTD resulted in a favorable response with tolerable toxicity in patients with MM and did not affect the yield of the stem cell collection.
Collapse
Affiliation(s)
- Deok-Hwan Yang
- Chonnam National University Hwasun Hospital, Republic of Korea, Jeollanam-do, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Despite the recent discovery that thalidomide causes limb defects by targeting highly angiogenic, immature blood vessels, several challenges still remain and new ones have arisen. These include understanding the drug's species specificity, determining molecular target(s) in the endothelial cell, shedding light on the molecular basis of phocomelia and producing a form of the drug that is clinically effective without having side effects. Now that the trigger of thalidomide-induced teratogenesis has been uncovered, a framework is proposed, incorporating and uniting previous models of thalidomide action, explaining how thalidomide causes not just limb defects, but also all the other defects it induces.
Collapse
Affiliation(s)
- Neil Vargesson
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK.
| |
Collapse
|
33
|
Segarra M, Lozano E, Corbera-Bellalta M, Vilardell C, Cibeira MT, Esparza J, Izco N, Bladé J, Cid MC. Thalidomide decreases gelatinase production by malignant B lymphoid cell lines through disruption of multiple integrin-mediated signaling pathways. Haematologica 2009; 95:456-63. [PMID: 19815837 DOI: 10.3324/haematol.2009.006395] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Thalidomide and its analogs are effective agents in the treatment of multiple myeloma. Since gelatinases (matrix metalloproteinases-2 and -9) play a crucial role in tumor progression, we explored the effect of thalidomide on gelatinase production by malignant B lymphoid cell lines. DESIGN AND METHODS We investigated the effect of therapeutic doses of thalidomide on integrin-mediated production of gelatinases by malignant B lymphoid cell lines by gelatin zymography, western-blot, reverse transcriptase polymerase chain reaction and invasive capacity through Matrigel-coated Boyden chambers. We also explored the effect of thalidomide on the activation status of the main signaling pathways involved in this process. RESULTS Thalidomide strongly inhibited gelatinase production by B-cell lines and primary myeloma cells in response to fibronectin, the most efficient gelatinase inducer identified in lymphoid cells. Thalidomide disrupted integrin-mediated signaling pathways involved in gelatinase induction and release, such as Src and MAP-kinase ERK activation, resulting in decreased cell motility and invasiveness. Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. CONCLUSIONS Disruption of integrin-mediated signaling may be an important mechanism through which thalidomide and its analogs impair tumor cell interactions with the microenvironment. The unexpected effects of thalidomide on Akt activation indicate the need for further studies to elucidate whether the interference with Akt downstream effects would synergize with the anti-tumor activity of thalidomide.
Collapse
Affiliation(s)
- Marta Segarra
- Department of Systemic Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ryu JK, Jantaratnotai N, McLarnon JG. Thalidomide inhibition of vascular remodeling and inflammatory reactivity in the quinolinic acid-injected rat striatum. Neuroscience 2009; 163:601-8. [PMID: 19591904 DOI: 10.1016/j.neuroscience.2009.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 06/30/2009] [Accepted: 07/05/2009] [Indexed: 11/25/2022]
Abstract
Effects of thalidomide administration on vascular remodeling, gliosis and neuronal viability have been studied in excitotoxin-injected rat striatum. Intrastriatal injection of quinolinic acid (QUIN) caused time-dependent changes (durations of 6 h, 1 and 7 d post-injection) in vascular remodeling. QUIN excitotoxic insult was associated with increased numbers of vessels (laminin or collagen IV markers) demonstrating considerable abnormalities in morphology, including short fragments and vascular loops. Non-lesioned striatum, with injection of phosphate buffer solution (PBS) as a vehicle, showed no evidence for vascular remodeling. A maximal extent of vascular remodeling was measured at 1 d post-QUIN and was correlated with marked increases in microgliosis (ED1 marker) and astrogliosis (glial fibrillary acidic protein [GFAP] marker) relative to control PBS injection. Double staining of laminin with ED1 and GFAP demonstrated areas of close association of glial cells with blood vessels. Treatment of QUIN-injected animals with the anti-inflammatory compound, thalidomide significantly inhibited vascular remodeling (by 43%) and reduced microgliosis (by 33%) but was ineffective in modifying extents of astrogliosis. Intrastriatal QUIN injection was associated with a marked loss of striatal neurons relative to non-lesioned control with thalidomide treatment exhibiting a significant degree of neuroprotection (24% recovery) against QUIN-induced neurotoxicity. These results suggest close links between microglial-mediated inflammatory responses and vascular remodeling, with inflammatory reactivity associated with, and contributing to, neuronal damage in excitotoxically-lesioned striatum.
Collapse
Affiliation(s)
- J K Ryu
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, 2176 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
35
|
|
36
|
Fadul CE, Kingman LS, Meyer LP, Cole BF, Eskey CJ, Rhodes CH, Roberts DW, Newton HB, Pipas JM. A phase II study of thalidomide and irinotecan for treatment of glioblastoma multiforme. J Neurooncol 2008; 90:229-35. [PMID: 18661102 PMCID: PMC3885231 DOI: 10.1007/s11060-008-9655-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Accepted: 07/07/2008] [Indexed: 01/24/2023]
Abstract
PURPOSE Irinotecan is a cytotoxic agent with activity against gliomas. Thalidomide, an antiangiogenic agent, may play a role in the treatment of glioblastoma multiforme (GBM). To evaluate the combination of thalidomide and irinotecan, we conducted a phase II trial in adults with newly-diagnosed or recurrent GBM. PATIENTS AND METHODS Thalidomide was given at a dose of 100 mg/day, followed by dose escalation every 2 weeks by 100 mg/day to a target of 400 mg/day. Irinotecan was administered on day 1 of each 3 week cycle. Irinotecan dose was 700 mg/m(2) for patients taking enzyme-inducing anticonvulsants and 350 mg/m(2) for all others. The primary endpoint was tumor response, assessed by MRI. Secondary endpoints were toxicity, progression-free survival, and overall survival. RESULTS Twenty-six patients with a median age of 55 years were enrolled, with fourteen evaluable for the primary outcome, although all patients were included for secondary endpoints. One patient (7%) exhibited a partial response after twelve cycles, and eleven patients (79%) had stable disease. The intention to treat group with recurrent disease included 16 patients who had a 6-month PFS of 19% (95% CI: 4-46%) and with newly-diagnosed disease included 10 patients who had a 6-month PFS of 40% (95% CI: 12-74%). Gastrointestinal (GI) toxicity was mild, but six patients (23%) experienced a venous thromboembolic complication. Two patients had Grade 4 treatment-related serious adverse events that required hospitalization. There were no treatment-related deaths. CONCLUSION The combination of irinotecan and thalidomide has limited activity against GBM. Mild GI toxicity was observed, but venous thromboembolic complications were common.
Collapse
Affiliation(s)
- Camilo E Fadul
- Department of Medicine, Section of Hematology/Oncology, Norris Cotton Cancer Center, Dartmouth Hitchcock Medical Center Drive, One Medical Center Drive, Lebanon, NH 03756, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Robinson AA, Watson WJ, Leslie KK. Targeted treatment using monoclonal antibodies and tyrosine-kinase inhibitors in pregnancy. Lancet Oncol 2007; 8:738-43. [PMID: 17679084 DOI: 10.1016/s1470-2045(07)70242-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An expanding knowledge of the signalling pathways involved in the cell cycle has led to great improvements in the understanding of the molecular events involved in carcinogenesis. The past decade has seen substantial advances with the introduction of several classes of targeted therapeutics for the treatment of various cancers and autoimmune disorders. However, the question arises as to whether pregnant women can take advantage of these new treatments in view of the potential risks to the fetus. Published work suggests that biological agents, like traditional treatments, have the potential to affect the fetus, and should, therefore, be used with caution during pregnancy. However, when targeted treatment is clearly indicated the magnitude of the risk to the fetus might not reach that of standard chemotherapy. In circumstances where better alternative treatments do not exist, or where failure to use targeted treatments would result in suboptimum patient care or survival, the risk-benefit analysis might favour the use of potentially effective molecular treatment during pregnancy.
Collapse
Affiliation(s)
- Alice A Robinson
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
38
|
Varker KA, Campbell J, Shah MH. Phase II study of thalidomide in patients with metastatic carcinoid and islet cell tumors. Cancer Chemother Pharmacol 2007; 61:661-8. [PMID: 17589846 DOI: 10.1007/s00280-007-0521-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Accepted: 05/07/2007] [Indexed: 10/23/2022]
Abstract
PURPOSE Carcinoid and islet cell tumors are known to be highly vascular. There is no effective systemic therapy currently available for metastatic disease. We conducted a phase II trial to evaluate the efficacy of the anti-antiangiogenic agent thalidomide in metastatic neuroendocrine tumors. PATIENTS AND METHODS Eighteen patients with measurable, histologically proven metastatic carcinoid neuroendocrine carcinomas (well-differentiated, n = 13; moderately-differentiated, n = 5) were enrolled on this study. The majority of the patients had gastrointestinal primaries (small bowel, 8; pancreas, 5; colon, 1). All but one patient had hepatic metastases, and 12 patients (67%) had carcinoid syndrome. All patients had Eastern Cooperative Oncology Group performance status of zero or one. Eight patients (44%) had received previous hepatic artery chemoembolization and 11 (61%) had undergone surgical resection. Patients were started on oral thalidomide at a daily dose of 200 mg that was escalated to the target dose of 400 mg daily after 2 weeks. Tumor response was assessed at 12-week intervals using RECIST criteria. Planned treatment duration was 24 weeks unless unacceptable toxicity or disease progression was observed. RESULTS No patient achieved a partial remission or a complete remission. Best response was stable disease (SD) in 11 of 16 response-evaluable patients (69%). Serum pancreastatin results did not correlate with clinical response. Grade 3 toxicities included dizziness with orthostatic hypotension (n = 5), sensory neuropathy (n = 2), fatigue (n = 2), hemorrhagic cystitis (n = 1), and deep venous thrombosis (n = 1). Frequent Grade 1-2 toxicities were: fatigue (n = 13), constipation (n = 13), dry mouth (n = 12), somnolence (n = 12), dizziness/syncope (n = 10), weight gain (n = 5), and peripheral neuropathy (n = 5). CONCLUSIONS Thalidomide was fairly well tolerated in patients with metastatic carcinoid/islet cell tumors, but failed to reveal any objective responses. The single stage design of the trial makes it difficult to determine whether observed SD in a subset of patients was attributable to the indolent nature of these tumors, or to beneficial effect of thalidomide.
Collapse
Affiliation(s)
- Kimberly A Varker
- Division of Hematology-Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, A438 Starling-Loving Hall, 320 West 10th Avenue, Columbus, OH 43210, USA
| | | | | |
Collapse
|
39
|
Dürk HA. Maintenance therapy for multiple myeloma with particular emphasis on thalidomide. Oncol Res Treat 2006; 29:582-90. [PMID: 17202830 DOI: 10.1159/000096262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Treatment standards are changing as a result of new findings in the therapy of multiple myeloma. So far, prednisone, dexamethasone and interferon-a have mainly been used as maintenance therapy after achieving remission or stable disease. At present, thalidomide is being considered as a new therapeutic option in several studies investigating maintenance therapy. As a result of the dose dependence of adverse effects such as neuropathy, constipation, sedation/vertigo and bradycardia, individual adjustment of the thalidomide dose is recommended. Only isolated cases of thrombosis occurred in the maintenance phase of therapy, and discontinuation of therapy is generally not necessary. While important study results on the efficacy of thalidomide following conventional chemotherapy are still awaited, it is the best documented drug so far for maintenance therapy following autologous stem cell transplantation. An upgrade of the response was seen in 22-73% of patients, as well as a significant prolongation of progression-free survival. For the first time, maintenance therapy with thalidomide showed a significant improvement in overall survival in a phase III study published recently. The tolerability of thalidomide could be further improved by including the option of intermittent administration of the drug.
Collapse
Affiliation(s)
- Heinz Albert Dürk
- Klinik für Hämatologie/Onkologie, St. Marien-Hospital Hamm, Germany.
| |
Collapse
|
40
|
Mirshahi P, Toprak SK, Faussat AM, Dubrulle S, Marie JP, Soria C, Soria J, Mirshahi M. Malignant hematopoietic cells induce an increased expression of VEGFR-1 and VEGFR-3 on bone marrow endothelial cells via AKT and mTOR signalling pathways. Biochem Biophys Res Commun 2006; 349:1003-1010. [PMID: 16959214 DOI: 10.1016/j.bbrc.2006.08.132] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Accepted: 08/21/2006] [Indexed: 11/26/2022]
Abstract
Angiogenesis plays a significant role in a variety of malignant hematologic diseases, and it is recognized that it has prognostic value. However, the cellular mechanisms by which malignant hematologic cells induce angiogenesis are not well understood. In order to investigate the role of cells from B-cell chronic lymphocytic leukemia (B-CLL) and multiple myeloma (MM) in angiogenesis on human bone marrow endothelial cells (HBMEC), we analyzed the impact of factors secreted by B-CLL cells and by MM cells on HBMEC capillary tube formation on matrigel. It was found that, in addition to the secretion of angiogenic factors VEGF and b-FGF by B-CLL and MM cells, MM cells (but not B-CLL cells) induced a dramatic increase in expression of VEGFR-1 and VEGFR-3 on human bone marrow endothelial cells (HBMEC). It would seem that this increase in VEGFR-3 occurred via the ERK and mTOR pathways, since their respective inhibitors U0126, LY294002 or rapamycin were responsible for a decrease of VEGFR-3. In response to MM cells-increased VEGF receptors on HBMEC, endothelial cell migration was enhanced in a wound artificially produced in a semi-confluent HBMEC culture, a phenomenon which was also down-regulated by the same inhibitors that reversed the increase in VEGF receptors. The present study suggests that, in addition to the classic angiogenic pathway, another mechanism related to an increased expression of VEGFRs on HBMEC might exist in malignant hematopoietic angiogenesis.
Collapse
Affiliation(s)
- P Mirshahi
- INSERM, UMR736, IFR 58, Université Pierre et Marie Curie, 75006 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Chanan-Khan A, Miller KC, Takeshita K, Koryzna A, Donohue K, Bernstein ZP, Mohr A, Klippenstein D, Wallace P, Zeldis JB, Berger C, Czuczman MS. Results of a phase 1 clinical trial of thalidomide in combination with fludarabine as initial therapy for patients with treatment-requiring chronic lymphocytic leukemia (CLL). Blood 2005; 106:3348-52. [PMID: 16051743 DOI: 10.1182/blood-2005-02-0669] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractTumor necrosis factor α (TNF-α) and vascular endothelial growth factor (VEGF) play an important role in the biology of chronic lymphocytic leukemia (CLL) cells. Thalidomide is a first-generation immuno-modulating agent that down-regulates TNF-α and VEGF. We initiated a phase 1/2 clinical trial to determine the safety and efficacy of combining thalidomide with fludarabine in patients with treatment-naïve CLL. Patients received 6 months of continuous daily thalidomide with standard monthly doses of fludarabine. Three dose levels of thalidomide (100, 200, and 300 mg) were studied. Results from the phase 1 part of this study are reported here. Thirteen patients were enrolled in the phase 1 component of the study. Dose-limiting toxicity was not reached. The most common toxicities noted were fatigue, constipation, and peripheral sensory neuropathy. Overall response rate was 100% with 55% of patients achieving complete remissions. At a median follow-up of 15+ months none of the patients have had a relapse and the median time to disease progression has not yet been reached. Responses were noted at all dose levels. Thalidomide given up to 300 mg/day concurrently with fludarabine in patients with previously untreated CLL shows encouraging clinical efficacy and acceptable toxicity. An ongoing phase 2 part of this study will help validate the clinical efficacy of this regimen.
Collapse
Affiliation(s)
- Asher Chanan-Khan
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14051, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Daruwalla J, Nikfarjam M, Malcontenti-Wilson C, Muralidharan V, Christophi C. Effect of thalidomide on colorectal cancer liver metastases in CBA mice. J Surg Oncol 2005; 91:134-40. [PMID: 16028287 DOI: 10.1002/jso.20289] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND AIMS Thalidomide has undergone resurgence in the treatment of specific malignancies. One of the possible actions of thalidomide may be an antiangiogenic effect. This study investigates the effects of thalidomide on tumor growth and long-term survival in a murine model of colorectal liver metastases. METHODS Liver metastases were produced in male CBA mice by intrasplenic injection of a dimethyl hydrazine induced MoCR colon cancer murine cell line. Thalidomide was administered daily at doses ranging from 50 to 300 mg/kg by intraperitoneal injection. Tumor growth was assessed using quantitative stereological analysis. The effect on long-term survival was determined at the maximum tolerated dose using Kaplan-Meier analysis. The microvascular effects of thalidomide were assessed by laser Doppler flowmetry (LDF) and microvascular resin casting. Immunohistochemistry was used to determine vascular endothelial growth factor (VGEF) and basic fibroblast growth factor (bFGF) expression. RESULTS Thalidomide, (50-300 mg/kg) caused no significant reduction in tumor growth by day 21 following induction of liver metastases and caused systemic toxicity at a dose of 300 mg/kg. At a dose of 200 mg/kg given beyond 35 days, thalidomide significantly reduced tumor growth compared to control, (P = 0.029). No significant impact on survival was however observed (P = 0.93). LDF and microvascular resin casting showed no differences in blood flow or tumor microvascular architecture. VGEF and FGF were expressed in tumors, but remained unaltered by thalidomide administration compared to matched controls. CONCLUSIONS Thalidomide caused a significant reduction in the volume of colorectal liver metastases during the late phase of tumor growth. There was however no improvement in survival. Tumor growth reduction in this model did not appear to be due to microvascular changes or altered expression of VGEF or basic FGF. Further investigation into potential mechanisms of action of thalidomide and its synergistic use with other therapies is required.
Collapse
Affiliation(s)
- Jurstine Daruwalla
- The University of Melbourne, Department of Surgery, Austin Health Hospital, Studley Road, Heidelberg, Australia.
| | | | | | | | | |
Collapse
|
43
|
Lee KW, Yun T, Song EK, Na II, Shin H, Bang SM, Lee JH, Lee ST, Kim JH, Yoon SS, Lee JS, Park S, Kim BK, Kim NK. A pilot study of bortezomib in Korean patients with relapsed or refractory myeloma. J Korean Med Sci 2005; 20:598-62. [PMID: 16100450 PMCID: PMC2782154 DOI: 10.3346/jkms.2005.20.4.598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent clinical trials showed that bortezomib, a novel proteasome inhibitor, had therapeutic activity in multiple myeloma. However, there was no data about the feasibility of bortezomib in Korean patients. We performed a pilot study of bortezomib in patients with relapsed or refractory myeloma (1.3 mg/m2 twice weekly for 2 week in a 3-week cycle). Seven patients were enrolled. The median age of patients was 59 yr. All patients previously received VAD (vincristine, doxorubicin and dexamethasone) and thalidomide chemotherapy. Three patients previously received alkylator-containing chemotherapy and 4 patients, autologous stem cell transplantation. Bortezomib monotherapy resulted in 3 partial remissions (43%), 3 no changes (43%) and 1 progressive disease (14%). One patient who had no response to bortezomib monotherapy experienced partial remission after addition of dexamethasone to bortezomib. The most common serious toxicity was thrombocytopenia (grade 3/4, 10 of 20 cycles (50%)) and grade 3 peripheral neuropathy was developed in 2 of 20 cycles (10%). Drug-related adverse event led to discontinuation of bortezomib in 1 patient. There was no treatment related mortality. Overall, bortezomib seems to be effective and feasible. Conduction of larger clinical studies on Korean patients is necessary to characterize clinical efficacy and safety of bortezomib more precisely.
Collapse
Affiliation(s)
- Keun-Wook Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Tak Yun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Kee Song
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Im il Na
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyunchoon Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Soo-Mee Bang
- Department of Internal Medicine, Gachon Medical School, Incheon, Korea
| | - Jae Hoon Lee
- Department of Internal Medicine, Gachon Medical School, Incheon, Korea
| | - Seung Tae Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Clinical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Jong Seok Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seonyang Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Clinical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Byoung Kook Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Clinical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Noe Kyeong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
Hus I, Dmoszynska A, Manko J, Hus M, Jawniak D, Soroka-Wojtaszko M, Hellmann A, Ciepluch H, Skotnicki A, Wolska-Smolen T, Sulek K, Robak T, Konopka L, Kloczko J. An evaluation of factors predicting long-term response to thalidomide in 234 patients with relapsed or resistant multiple myeloma. Br J Cancer 2005; 91:1873-9. [PMID: 15520820 PMCID: PMC2409770 DOI: 10.1038/sj.bjc.6602225] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The aim of this study was to assess the prognostic value of pretreatment clinical and laboratory parameters in refractory or relapsed multiple myeloma (MM) patients who have a long-term response to thalidomide (THAL), lasting at least 18 months. The study was carried out on 234 patients who received THAL for relapsed/refractory myeloma. Out of the 234 patients, 129 patients (55.1%) responded to THAL with a mean response duration of 11.9 months (ranging from 1 to 48) and an overall survival rate of 20.3 months (ranging 1–55 months). In 64 patients (27.4% of the whole group), the response to THAL lasted ⩾18 months with a mean response lasting 24 months. Statistical analysis of the group of nonresponders and patients with long-term response to THAL showed a significantly higher serum albumin level (P=0.0003) and haemoglobin level (P=0.05), as well as a lower β2 microglobulin (β2M) (P=0.022), LDH (P=0.045) serum level in patients with long-term response. In this study, the LDH and serum albumin level were predictors for response to THAL therapy. The β2M serum level was not a predictor for response to THAL. The albumin serum level was the best parameter distinguishing the group of patients with long-term response to THAL from the entire responding group (P=0.02).
Collapse
Affiliation(s)
- I Hus
- Department of Haematooncology, Medical University of Lublin, Poland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Brennen WN, Cooper CR, Capitosti S, Brown ML, Sikes RA. Thalidomide and analogues: current proposed mechanisms and therapeutic usage. ACTA ACUST UNITED AC 2004; 3:54-61. [PMID: 15279692 DOI: 10.3816/cgc.2004.n.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microvessel density is a prognostic factor for many cancers, including prostate. For this reason, several studies and therapeutic approaches that target the tumor microvasculature have been attempted. Thalidomide has long been recognized as an antiangiogenic molecule. Recently, this drug has regained favor as an anticancer agent and is in clinical trial for multiple myeloma and prostate cancer, among others. This article will briefly review the proposed mechanisms of action for thalidomide, discuss why these activities are of therapeutic value in diseases currently undergoing clinical trials, and summarize the current status of clinical trials for prostate cancer. The focus will be predominantly on the relationship of thalidomide to angiogenesis, as well as on the future and potential value of thalidomide-inspired structural derivatives.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Laboratory for Cancer Ontogeny and Therapeutics, University of Delaware, Newark 19716, USA
| | | | | | | | | |
Collapse
|
46
|
Du W, Hattori Y, Hashiguchi A, Kondoh K, Hozumi N, Ikeda Y, Sakamoto M, Hata JI, Yamada T. Tumor angiogenesis in the bone marrow of multiple myeloma patients and its alteration by thalidomide treatment. Pathol Int 2004; 54:285-94. [PMID: 15086832 DOI: 10.1111/j.1440-1827.2004.01622.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Angiogenesis in solid tumors is important to tumor growth, invasion and metastasis. Recently, it has been suggested that angiogenesis plays a certain role in the development of hematopoietic malignancies, including leukemia and multiple myeloma. We evaluated tumor angiogenesis in the bone marrow (BM) of multiple myeloma (MM) patients by calculating microvessel density (MVD) in needle-biopsy specimens obtained from 51 cases of untreated MM or monoclonal gammopathy of undetermined significance (MGUS). The MVD in the BM of donors for transplantation and patients with non-hematological diseases was calculated as a control. There was an obvious increase in MVD in the BM of MM patients, and the MVD correlated with the grade of myeloma cell invasion of the BM in the untreated MM cases. It was recently reported that thalidomide might be effective for the treatment of MM. We assessed the effect of thalidomide on angiogenesis in BM treatment of 11 patients with refractory MM. The concentration of M-protein in the serum or urine of seven of the 11 patients was reduced by at least 30% after thalidomide treatment, and MVD in the BM decreased in three of these seven cases in response to thalidomide. Increased plasma concentrations of basic fibroblast growth factor (FGF-2) and vascular endothelial growth factor (VEGF) were observed in all 11 cases before thalidomide administration and both levels were reduced after treatment with thalidomide. Augmented angiogenesis in the bone marrow of MM patients was confirmed in the present study. It seems that thalidomide is effective in the treatment of MM through the impairment of angiogenesis by decreasing FGF-2 and VEGF production. This is the first report on pathological evidence in the bone marrow of MM before and after thalidomide treatment, in Japan.
Collapse
Affiliation(s)
- Wenlin Du
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sherman AC, Simonton S, Latif U, Spohn R, Tricot G. Psychosocial adjustment and quality of life among multiple myeloma patients undergoing evaluation for autologous stem cell transplantation. Bone Marrow Transplant 2004; 33:955-62. [PMID: 15034542 DOI: 10.1038/sj.bmt.1704465] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Stem cell transplantation has assumed a prominent place in the treatment of multiple myeloma, but relative to patients with other malignancies there is surprisingly little information about the adjustment difficulties and quality-of-life changes that these patients experience. This study examined psychosocial and functional deficits among myeloma patients assessed at a uniform period during their initial diagnostic evaluation, prior to beginning protocols at a transplant center. Validated self-report measures and clinician rating scales were used to assess 213 patients. Outcomes evaluated included emotional distress (Hospital Anxiety and Depression Scale, Brief Symptom Inventory), depression (Hamilton Depression Rating Scale), physical functioning, pain, and energy (SF-12). A significant proportion of patients experienced compromised psychosocial and physical functioning. Roughly one-third reported clinically elevated levels of distress, anxiety, and depression. In all, 59% scored below age-adjusted norms for daily physical functioning, 58% reported at least moderate levels of pain, and over 80% noted at least moderate fatigue. Clinical and demographic correlates of these outcomes were examined. These findings are among the first to characterize quality-of-life outcomes among myeloma patients in the transplant setting, and indicate that many patients experience considerable supportive care needs even prior to beginning aggressive regimens. Results highlight the importance of early screening.
Collapse
Affiliation(s)
- A C Sherman
- Behavioral Medicine, Arkansas Cancer Research Center, University of Arkansas for Medical Sciences, Little Rock 72205, USA.
| | | | | | | | | |
Collapse
|
48
|
Zangari M, Anaissie E, Stopeck A, Morimoto A, Tan N, Lancet J, Cooper M, Hannah A, Garcia-Manero G, Faderl S, Kantarjian H, Cherrington J, Albitar M, Giles FJ. Phase II study of SU5416, a small molecule vascular endothelial growth factor tyrosine kinase receptor inhibitor, in patients with refractory multiple myeloma. Clin Cancer Res 2004; 10:88-95. [PMID: 14734456 DOI: 10.1158/1078-0432.ccr-0221-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Increased bone marrow angiogenesis and vascular endothelial growth factor (VEGF) levels are of adverse prognostic significance in patients with multiple myeloma (MM). VEGF, a soluble circulating angiogenic molecule, acts via receptor tyrosine kinases, including VEGF receptor 2. SU5416 is a small molecule VEGF receptor 2 inhibitor. EXPERIMENTAL DESIGN Adult patients with advanced MM were entered on a multicenter phase II study. RESULTS Twenty-seven patients (median age 69, range 39-79), median 4 (0-10) lines of prior therapy, 14 with prior thalidomide therapy, received SU5416 at 145 mg/m(2) twice weekly i.v. for a median of two 4-week cycles (range 0.2-9). Grade 3/4 toxicities were rarely observed; the most frequent was thrombocytopenia (12%). Mild-to-moderate toxicities included nausea (63%), headache (56%), diarrhea, vomiting (both 37%), and fatigue (33%). There were three thromboembolic episodes and five cases of new onset hypertension. Two (7%) patients did not complete the first 4-week cycle of therapy because of adverse events (pneumonia and headache). There were no objective responses. Four patients had disease stabilization for >/==" BORDER="0">4 months. A decrease in median VEGF plasma levels was observed in patients with stable disease (n = 7) compared with patients with progressive disease (n = 5). Overall median survival was 42 weeks (range 3-92+). CONCLUSIONS Although SU5416 had minimal clinical activity, signs of biological activity (decrease in plasma VEGF levels) suggest that angiogenic modulation may be of value in patients with MM.
Collapse
Affiliation(s)
- Maurizio Zangari
- University of Arkansas for Medical Sciences, The Multiple Myeloma Institute for Research and Therapy, Little Rock, Arkansas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- S Vincent Rajkumar
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, Minn 55905, USA.
| |
Collapse
|
50
|
Abstract
OBJECTIVE To review the pharmacology, pharmacokinetics, and place of bortezomib in the therapy of multiple myeloma (MM). DATA SOURCES A MEDLINE search was conducted (1985-May 2003). Meeting abstracts, bibliographies from identified articles, and the package insert were also used. Search terms were bortezomib, multiple myeloma, Velcade, PS-341, LDP-341, MLNM341, and proteasome inhibitor. STUDY SELECTION AND DATA EXTRACTION All published information relevant to the clinical activity of bortezomib in MM was considered. All human clinical studies, with an emphasis on Phase II trials, were selected. DATA SYNTHESIS Current therapy for MM yields significant, although temporary, responses. Bortezomib is a novel anticancer agent with significant activity in relapsed and refractory MM. CONCLUSIONS Although the clinical trial data are incomplete, bortezomib offers a novel therapeutic modality for patients with MM who would otherwise have few options.
Collapse
Affiliation(s)
- Brad L Stanford
- School of Pharmacy, Texas Tech Health Sciences Center, Lubbock, TX, USA.
| | | |
Collapse
|