1
|
Gniadek T. Production of Components by Apheresis. Transfus Med 2021. [DOI: 10.1002/9781119599586.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
2
|
LU D, Yin H, Wang S, Tang F, Huang W, Wang P. Chemical Synthesis of the Homogeneous Granulocyte-Macrophage Colony-Stimulating Factor Through Se-Auxiliary-Mediated Ligation. J Org Chem 2019; 85:1652-1660. [DOI: 10.1021/acs.joc.9b02232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Dan LU
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hongli Yin
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Siyao Wang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Feng Tang
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Center for Biotherapeutics Discovery Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Wei Huang
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Center for Biotherapeutics Discovery Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Ping Wang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
3
|
Zhan Y, Lew AM, Chopin M. The Pleiotropic Effects of the GM-CSF Rheostat on Myeloid Cell Differentiation and Function: More Than a Numbers Game. Front Immunol 2019; 10:2679. [PMID: 31803190 PMCID: PMC6873328 DOI: 10.3389/fimmu.2019.02679] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 10/30/2019] [Indexed: 12/27/2022] Open
Abstract
Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF) is a myelopoietic growth factor that has pleiotropic effects not only in promoting the differentiation of immature precursors into polymorphonuclear neutrophils (PMNs), monocytes/macrophages (MØs) and dendritic cells (DCs), but also in controlling the function of fully mature myeloid cells. This broad spectrum of GM-CSF action may elicit paradoxical outcomes-both immunostimulation and immunosuppression-in infection, inflammation, and cancer. The complexity of GM-CSF action remains to be fully unraveled. Several aspects of GM-CSF action could contribute to its diverse biological consequences. Firstly, GM-CSF as a single cytokine affects development of most myeloid cells from progenitors to mature immune cells. Secondly, GM-CSF activates JAK2/STAT5 and also activate multiple signaling modules and transcriptional factors that direct different biological processes. Thirdly, GM-CSF can be produced by different cell types including tumor cells in response to different environmental cues; thus, GM-CSF quantity can vary greatly under different pathophysiological settings. Finally, GM-CSF signaling is also fine-tuned by other less defined feedback mechanisms. In this review, we will discuss the role of GM-CSF in orchestrating the differentiation, survival, and proliferation during the generation of multiple lineages of myeloid cells (PMNs, MØs, and DCs). We will also discuss the role of GM-CSF in regulating the function of DCs and the functional polarization of MØs. We highlight how the dose of GM-CSF and corresponding signal strength acts as a rheostat to fine-tune cell fate, and thus the way GM-CSF may best be targeted for immuno-intervention in infection, inflammation and cancer.
Collapse
Affiliation(s)
- Yifan Zhan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Lew
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC, Australia
| | - Michael Chopin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
4
|
Abdelhakim H, Shune L, Bhatti S, Cantilena AR, Baran A, Lin TL, Ganguly S, Singh AK, Abhyankar S, Divine C, Lipe B, McGuirk J, Allin D, Aljitawi OS. Results of the First Clinical Study in Humans That Combines Hyperbaric Oxygen Pretreatment with Autologous Peripheral Blood Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1713-1719. [PMID: 31170519 DOI: 10.1016/j.bbmt.2019.05.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 12/26/2022]
Abstract
Patients undergoing high-dose chemotherapy and autologous hematopoietic cell transplantation (auto-HCT) are at risk for multiple morbidities, including mucosal inflammation and neutropenic fever, both related to neutropenia. Evidence from our preclinical work in an umbilical cord blood (UCB) transplantation murine model suggests that treatment with hyperbaric oxygen (HBO) before UCB infusion improves UCB CD34+ cell engraftment by reducing erythropoietin levels. A pilot clinical trial using HBO in patients undergoing UCB transplantation showed improvement in kinetics of blood count recovery. In this study, we evaluated HBO in combination with auto-HCT. Our primary aim was to determine the safety of HBO in this setting and secondarily to determine its efficacy in reducing time to neutrophil and platelet engraftment compared with matched historic controls. Patients with multiple myeloma, non-Hodgkin lymphoma, and Hodgkin disease eligible for auto-HCT were included. On day 0, patients received HBO treatment consisting of exposure to 2.5 atmosphere absolutes for a total of 90 minutes, in a monoplace hyperbaric chamber, breathing 100% oxygen. Six hours after the start of HBO, peripherally mobilized stem/progenitor cells were infused and patients were followed daily for toxicity and blood count recovery. All patients received daily granulocyte colony-stimulating factor starting on day +5 and until absolute neutrophil count (ANC) of ≥1500 or ANC of 500 for 3 consecutive days. A matched historic cohort of 225 patients who received auto-HCT between January 2008 and December 2012 was chosen for comparison and matched on sex, age, conditioning regimen, and disease type. We screened 26 patients for this study; 20 were treated and included in the primary analysis, and 19 completed the HBO therapy and were included in the secondary analysis. Although the median time to neutrophil count recovery was 11 days in both the HBO and control cohorts, the Kaplan-Meier estimates of the full distributions indicate that the time to neutrophil recovery was generally about 1 day sooner for HBO versus historical controls (log-rank P = .005; range, 9 to 13 for HBO patients and 7 to 18 for controls). The median time to platelet count recovery was 16 days (range, 14 to 21) for HBO versus 18 days (range, 11 to 86) for controls (log-rank P < .0001). In the secondary analysis comparing the HBO cohort who completed HBO therapy (n = 19) with our historical cohort, we evaluated neutropenic fever, growth factor use, mucositis, day +100 disease responses, and blood product use. HBO was associated with less growth factor use (median 6 days in HBO cohort versus median 8 days in controls, P < .0001). Packed RBC and platelet transfusion requirements were not statistically different between the 2 cohorts. Mucositis incidence was significantly lower in the HBO cohort (26.3% in HBO cohort versus 64.2% in controls, P = .002). HBO therapy appears to be well tolerated in the setting of high-dose therapy and auto-HCT. Prospective studies are needed to confirm potential benefits of HBO with respect to earlier blood count recovery, reduced mucositis, and growth factor use, and a cost-benefit analysis is warranted. © 2019 American Society for Blood and Marrow Transplantation.
Collapse
Affiliation(s)
- Haitham Abdelhakim
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Leyla Shune
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Sajjad Bhatti
- Department of Internal Medicine, University of Kansas School of Medicine, Kansas City, KS
| | - Amy Rose Cantilena
- University of Kansas School of Medicine, Kansas city, KS; Cardiovascular Research Institute, University of Kansas School of Medicine, Kansas City, KS
| | - Andrea Baran
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY
| | - Tara L Lin
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Siddhartha Ganguly
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Anurag K Singh
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Sunil Abhyankar
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Clint Divine
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Brea Lipe
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Joseph McGuirk
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS
| | - Dennis Allin
- Emergency Department, University of Kansas Medical Center, Kansas City, KS
| | - Omar S Aljitawi
- Division of Hematologic Malignancies and Cellular Therapy, Internal Medicine Department, University of Kansas Medical Center, Kansas city, KS; Division of Hematology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY.
| |
Collapse
|
5
|
Sun L, Rautela J, Delconte RB, Souza-Fonseca-Guimaraes F, Carrington EM, Schenk RL, Herold MJ, Huntington ND, Lew AM, Xu Y, Zhan Y. GM-CSF Quantity Has a Selective Effect on Granulocytic vs. Monocytic Myeloid Development and Function. Front Immunol 2018; 9:1922. [PMID: 30210491 PMCID: PMC6120981 DOI: 10.3389/fimmu.2018.01922] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/06/2018] [Indexed: 01/14/2023] Open
Abstract
GM-CSF promotes myeloid differentiation of cultured bone marrow cells into cells of the granulocytic and monocytic lineage; the latter can further differentiate into monocytes/macrophages and dendritic cells. How GM-CSF selects for these different myeloid fates is unresolved. GM-CSF levels can change either iatrogenically (e.g., augmenting leukopoiesis after radiotherapy) or naturally (e.g., during infection or inflammation) resulting in different immunological outcomes. Therefore, we asked whether the dose of GM-CSF may regulate the development of three types of myeloid cells. Here, we showed that GM-CSF acted as a molecular rheostat where the quantity determined which cell type was favored; moreover, the cellular process by which this was achieved was different for each cell type. Thus, low quantities of GM-CSF promoted the granulocytic lineage, mainly through survival. High quantities promoted the monocytic lineage, mainly through proliferation, whereas moderate quantities promoted moDCs, mainly through differentiation. Finally, we demonstrated that monocytes/macrophages generated with different doses of GM-CSF differed in function. We contend that this selective effect of GM-CSF dose on myeloid differentiation and function should be taken into consideration during pathophysiological states that may alter GM-CSF levels and during GM-CSF agonistic or antagonistic therapy.
Collapse
Affiliation(s)
- Li Sun
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Jai Rautela
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Rebecca B Delconte
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Emma M Carrington
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Robyn L Schenk
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Marco J Herold
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Nicholas D Huntington
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Lew
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC, Australia
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Yifan Zhan
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Guangzhou Women and Children's Medical Centre, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Wendel S, Dayawansa T, Das P, Smit Sibinga CT. A Simplified Method for Purification, Concentration and Freezing of Bone Marrow Cells for Autologous Transplantation. Int J Artif Organs 2018. [DOI: 10.1177/039139889001300410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Autologous bone marrow (BM) transplantation is being increasingly applied in hematological and oncological patients. However, because of the need to purify and preserve BM requiring high technology, such treatments are virtually concentrated in the “developed” countries. This paper examines methods of BM purification and freezing that could make the technique potentially applicable in developing countries. Hemapheresis is routinely applied for BM purification in our Dutch Centre, where the buffy coats obtained from routine blood donations were utilised in experimental settings. Using DMSO as cryoprotectant, semi-purified white cells were frozen in liquid N2 (LN2), by mechanical freezer or snap-frozen at -55° C. Different types of containers were compared including plastic tubes and ordinary blood bags. After thawing the results show that snap-freezing had a deleterious effect but the cell yields and viability were similar in LN2 or the mechanical freezer where the tubes and the bag were equally effective as containers (86% cell recovery with 90% viability). In the purification/concentration stage, reduction of the volume of the material by extra centrifugation, thus requiring less DMSO, produced better results — 96% cell yield and 90% viability after thawing. This simplified method was applied in a general hospital in Sao Paulo where four oncology patients underwent BM collection. BM was purified and concentrated within a blood bank facility. Hydroxyethyl starch sedimentation and centrifugation of the material in plastic blood bags gave 80% BM cell harvest. After thawing from the mechanical freezer 1 x 108 BM cells/kg were available for reinfusion to patients. There was no immediate untoward reaction. Three of the patients showed signs of bone marrow regeneration by three weeks, but one patient died 16 days after transplantation, of septicemia. We conclude that certain high-technology procedures for ABMT can be adapted for existing facilities in developing countries.
Collapse
Affiliation(s)
- S. Wendel
- Blood Bank, Hospital Sirio Libanes, Sao Paulo - Brasil
| | - T. Dayawansa
- Blood Bank, Cancer Institute, Maharagama - Sri Lanka
| | - P.C. Das
- Red Cross Blood Bank Groningen-Drenthe, Groningen - The Netherlands
| | | |
Collapse
|
7
|
Wu F, Dai L, Geng L, Zhu H, Jin T. Practically feasible production of sustained-release microspheres of granulocyte-macrophage colony-stimulating factor (rhGM-CSF). J Control Release 2017; 259:195-202. [PMID: 28389408 DOI: 10.1016/j.jconrel.2017.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/25/2017] [Accepted: 04/03/2017] [Indexed: 10/19/2022]
Abstract
Using recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) as a model drug, the present study demonstrated a practically feasible process to produce polymeric microspheres for sustained-release delivery of protein drugs with preserved integrity. This process is featured with pre-loading proteins into polysaccharide fine particles via a self-standing aqueous-aqueous "emulsion", prior to microencapsulation into the microspheres. The protein drug, rhGM-CSF, was partitioned thermodynamically into a dextran dispersed phase of the aqueous-aqueous emulsion, followed by lyophilization and removal of the polyethylene glycol (PEG) continuous phase (using an organic solvent not penetrating into dextran matrix). The harvested dextran particles were then suspended in a dichloromethane solution of polylatic-co-glyclic acids (PLGA) and emulsified in a polyvinyl alcohol (PVA) and NaCl solution of small volume to form embryonic microspheres. The emulsion was then transferred into a NaCl solution of large volume to extract the organic solvent and harden the embryonic microspheres. The obtained rhGM-CSF microspheres showed a satisfied release profile with the day-to-day variation within 9 folds over the multi-weeks long release period. At the same time, the integrity (defined freedom of aggregates measured by SEC-HPLC) and bioactivity (defined by TF-1 cell proliferation) of the proteins were well preserved. The present formulation process ensured good reproducibility and over 89% protein encapsulation efficiency, and practically feasible to adapt to scaled productions.
Collapse
Affiliation(s)
- Fei Wu
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 00240, China
| | - Lili Dai
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 00240, China
| | - Lee Geng
- New Jersey Medical School, Rutgers University, 185 S. Orange Ave., Newark 07103, USA
| | - Hua Zhu
- New Jersey Medical School, Rutgers University, 185 S. Orange Ave., Newark 07103, USA
| | - Tuo Jin
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 00240, China.
| |
Collapse
|
8
|
Production of Components by Apheresis. Transfus Med 2016. [DOI: 10.1002/9781119236504.ch7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
9
|
Klimowski LL, Rotstein C, Cummings KM. Incidence of Nosocomial Aspergillosis in Patients with Leukemia Over a Twenty-Year Period. Infect Control Hosp Epidemiol 2016. [DOI: 10.2307/30146471] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
AbstractThe incidence of invasive nosocomial aspergillosis was studied in leukemia patients at an oncology center from 1964 to 1983. A total of 97 cases of aspergillosis occurred in 1,866 patients, yielding an overall case rate of 5.2 cases per 100 patients and an incidence rate of 9.1 per 10,000 patient days. The highest incidence rate was in patients with chronic myelogenous leukemia (13.7 cases per 10,000 patient days), followed by patients with acute myelogenous leukemia (10.6 cases per 10,000 patient days). Subdividing patients after 1978 into those receiving bone marrow transplantation and those who did not demonstrated the predisposition of transplant recipients to aspergillosis. The rates of aspergillosis among those patients who did not receive a bone marrow transplant were highest for patients with acute myelogenous leukemia. Increases in the annual rates of aspergillosis over time coincided with the level of internal renovation activity and major construction projects upwind of patient care facilities.
Collapse
|
10
|
|
11
|
The potential role of recombinant hematopoietic colony-stimulating factors in preventing infections in the immunocompromised host. Can J Infect Dis 2012; 2:74-88. [PMID: 22529714 DOI: 10.1155/1991/782768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/1990] [Accepted: 10/15/1990] [Indexed: 11/17/2022] Open
Abstract
Hematopoietic colony-stimulating factors coordinate the proliferation and maturation of bone marrow and peripheral blood cells during normal hematopoiesis. Most of these factors are now available as recombinant human colony-stimulating factors, and preclinical and clinical testing is proceeding rapidly. Granulocyte and granulocyte/macrophage colony-stimulating factors have been the most extensively studied to date. In human clinical trials, granulocyte colony-stimulating factor improves neutrophil counts and function, reduces episodes of febrile neutropenia, improves neutrophil recovery after disease- or treatment-induced myelosuppression, and reduces the number of serious infections in several neutropenic disease states. Granulocyte/macrophage colony-stimulating factor has similar biological properties but may also improve eosinophil proliferation and function, and platelet cell recovery after myelotoxic bone marrow injury, Interleukin-1 boosts the effects of granulocyte colony-stimulating factor and granulocyte/macrophage colony-stimulating factor, but also may promote the resolution of established infections in conjunction with antibiotics. The therapeutic realities and future therapeutic implications of these agents for the therapy of infections, cancer and hemopoietic disorders are discussed.
Collapse
|
12
|
Gamaletsou MN, Sipsas NV, Roilides E, Walsh TJ. Rhino-Orbital-Cerebral Mucormycosis. Curr Infect Dis Rep 2012; 14:423-34. [DOI: 10.1007/s11908-012-0272-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
Production of Components by Apheresis. Transfus Med 2011. [DOI: 10.1002/9781444398748.ch7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
14
|
Cheong SL, Federico S, Venkatesan G, Mandel AL, Shao YM, Moro S, Spalluto G, Pastorin G. The A3 adenosine receptor as multifaceted therapeutic target: pharmacology, medicinal chemistry, and in silico approaches. Med Res Rev 2011; 33:235-335. [PMID: 22095687 DOI: 10.1002/med.20254] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adenosine is an ubiquitous local modulator that regulates various physiological and pathological functions by stimulating four membrane receptors, namely A(1), A(2A), A(2B), and A(3). Among these G protein-coupled receptors, the A(3) subtype is found mainly in the lung, liver, heart, eyes, and brain in our body. It has been associated with cerebroprotection and cardioprotection, as well as modulation of cellular growth upon its selective activation. On the other hand, its inhibition by selective antagonists has been reported to be potentially useful in the treatment of pathological conditions including glaucoma, inflammatory diseases, and cancer. In this review, we focused on the pharmacology and the therapeutic implications of the human (h)A(3) adenosine receptor (AR), together with an overview on the progress of hA(3) AR agonists, antagonists, allosteric modulators, and radioligands, as well as on the recent advances pertaining to the computational approaches (e.g., quantitative structure-activity relationships, homology modeling, molecular docking, and molecular dynamics simulations) applied to the modeling of hA(3) AR and drug design.
Collapse
Affiliation(s)
- Siew Lee Cheong
- Department of Pharmacy, National University of Singapore, 3 Science Drive 2, Singapore 117543, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Hovgaard DJ, Nissen NI. Effect of Granulocyte-Macrophage Colony-Stimulating Factor on Chemotherapy-Related Neutropenia in Patients with Non-Hodgkin's Lymphomas—A Phase I/II Study of Dose and Mode of Administration. Leuk Lymphoma 2009; 6:39-48. [DOI: 10.3109/10428199109064877] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
Tsakona CP, Khwaja A, Goldstone AH. Growth Factor Administration in Lymphoma Transplants: Use of Flow Cytochemistry via the H*1 in Predictin Engraftment. Leuk Lymphoma 2009. [DOI: 10.3109/10428199209053569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- C. P. Tsakona
- University College Hospital and Middlesex School of Medicine, London, UK
| | - A. Khwaja
- University College Hospital and Middlesex School of Medicine, London, UK
| | - A. H. Goldstone
- University College Hospital and Middlesex School of Medicine, London, UK
| |
Collapse
|
18
|
Jones L, Newland AC. The Management of Relapsed and Refractory Acute Myeloid Leukaemia in Adults. Leuk Lymphoma 2009; 4:93-8. [DOI: 10.3109/10428199109068050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
Golidstone A, Gribben J, Mcmillan A. The Role of Growth Factors in Bone-Marrow Transplantation. Leuk Lymphoma 2009; 1:87-93. [DOI: 10.3109/10428199009042464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Marley SB, Russell NH, Reilly IAG. GM-CSF Stimulates Blast Cell Proliferation in Long-Term Culture of Bone Marrow from Patients with Myelodysplasia. Leuk Lymphoma 2009; 2:291-9. [DOI: 10.3109/10428199009106464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
21
|
Abstract
Patients with acute leukaemia and malignant lymphomas often are severely affected by fungal infections. There is in particular growing concern about disseminated candidosis. Oral, gastrointestinal and systemic candidosis seem to be closely linked. Predisposing factors are damaged mucosal barriers due to chemotherapy, protracted periods of neutropenia, and prolonged use of antibiotics and steroids. Oropharyngeal candidosis is very frequent. This can be prevented or controlled by the application of topical antifungals such as nystatin. The systemic application of antifungals is an alternative for patients who do not respond. Both oral ketoconazole and intravenous amphotericin B have been proven effective. Candida oesophagitis is also an important problem. Oral nystatin suspension can be helpful in mild cases. In others oral ketoconazole and intravenous amphotericin B have to be used. A whole range of measures has to be taken to prevent spread of the disease, i.e. H2-antagonists should be used only if definitely needed. Specific antifungal prophylaxis has also been discussed. Oral amphotericin B seems to be helpful. The azole itraconazole might be especially promising.
Collapse
|
22
|
Mortensen BT, Knudtzon S, Hovgaard D, Jensen AN. Bone marrow culture and haemopoietic growth factors: recent developments and current status. Eur J Haematol Suppl 2009; 48:77-84. [PMID: 3073963 DOI: 10.1111/j.1600-0609.1989.tb01241.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
23
|
Harmenberg J, Höglund M, Hellström-Lindberg E. G- and GM-CSF in oncology and oncological haematology. Eur J Haematol Suppl 2009; 55:1-28. [PMID: 7518779 DOI: 10.1111/j.1600-0609.1994.tb01617.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Administration of G- and GM-CSF increases the neutrophil counts in a number of clinical situations. GM-CSF shows the additional effect of increasing the number of monocytes and eosinophil granulocytes. Both G- and GM-CSF affect of neutrophil functions, in the case of GM-CSF there are some potentially negative effects on neutrophil migration and adhesiveness. The clinical relevance of the various effects on mature haematopoietic cells is not fully understood. Clinical data with G-CSF treatment indicate that increased levels of neutrophil granulocytes following cytotoxic chemotherapy may translate into clinical benefit such as a decreased rate of neutropenic infection and an increased cytotoxic chemotherapy dose even though the data are conflicting and the risk of "laboratory cosmetics" is apparent. Regarding treatment with GM-CSF following chemotherapy, the clinical benefit is unclear. The clinical benefit of GM-CSF-induced monocytes and eosinophils is unknown. G- and GM-CSF accelerates neutrophil recovery following autologous or allogeneic BMT. The influence on neutropenic infections is, however, less impressive. Pretreatment with G- or GM-CSF increases the yield of peripheral stem cell harvest, thereby reducing the number of leukaphereses needed. Transplantation of G- and GM-CSF primed autologous peripheral stem cells tends to reduce the period of post-transplant cytopenia, particularly thrombocytopenia, in comparison with traditional ABMT. In patients with MDS, G- and GM-CSF appear to increase the number of neutrophil granulocytes and there is some evidence that patients with severe infectious problems will benefit from this treatment. However, little influence was seen on the main clinical problems with these patients, which are anaemia and thrombocytopenia. In conclusion, G- and GM-CSF are two different proteins with different properties in vivo and in vitro. GM-CSF has, compared with G-CSF, more complex pharmacological effects and a more trouble-some side-effect profile. Early clinical development indicates that both compounds have a substantial influence on the levels of certain blood cells. Whether the increases in different blood cells translate into long-term clinical benefit for greater patient groups is the focus of ongoing research. The effects of G- and GM-CSF may be potentiated by other cytokines, an area which is presently being explored.
Collapse
Affiliation(s)
- J Harmenberg
- Department of Virology, Swedish Institute for Infectious Disease Control, Stockholm
| | | | | |
Collapse
|
24
|
Fishman P, Bar-Yehuda S, Synowitz M, Powell JD, Klotz KN, Gessi S, Borea PA. Adenosine receptors and cancer. Handb Exp Pharmacol 2009:399-441. [PMID: 19639290 PMCID: PMC3598010 DOI: 10.1007/978-3-540-89615-9_14] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The A(1), A(2A), A(2B) and A(3) G-protein-coupled cell surface adenosine receptors (ARs) are found to be upregulated in various tumor cells. Activation of the receptors by specific ligands, agonists or antagonists, modulates tumor growth via a range of signaling pathways. The A(1)AR was found to play a role in preventing the development of glioblastomas. This antitumor effect of the A(1)AR is mediated via tumor-associated microglial cells. Activation of the A(2A)AR results in inhibition of the immune response to tumors via suppression of T regulatory cell function and inhibition of natural killer cell cytotoxicity and tumor-specific CD4+/CD8+ activity. Therefore, it is suggested that pharmacological inhibition of A(2A)AR activation by specific antagonists may enhance immunotherapeutics in cancer therapy. Activation of the A(2B)AR plays a role in the development of tumors via upregulation of the expression levels of angiogenic factors in microvascular endothelial cells. In contrast, it was evident that activation of A(2B)AR results in inhibition of ERK1/2 phosphorylation and MAP kinase activity, which are involved in tumor cell growth signals. Finally, A(3)AR was found to be highly expressed in tumor cells and tissues while low expression levels were noted in normal cells or adjacent tissue. Receptor expression in the tumor tissues was directly correlated to disease severity. The high receptor expression in the tumors was attributed to overexpression of NF-kappaB, known to act as an A(3)AR transcription factor. Interestingly, high A(3)AR expression levels were found in peripheral blood mononuclear cells (PBMCs) derived from tumor-bearing animals and cancer patients, reflecting receptor status in the tumors. A(3)AR agonists were found to induce tumor growth inhibition, both in vitro and in vivo, via modulation of the Wnt and the NF-kappaB signaling pathways. Taken together, A(3)ARs that are abundantly expressed in tumor cells may be targeted by specific A(3)AR agonists, leading to tumor growth inhibition. The unique characteristics of these A(3)AR agonists make them attractive as drug candidates.
Collapse
Affiliation(s)
- P Fishman
- Can-Fite BioPharma, Kiryat Matalon, Petach Tikva, 49170, Israel.
| | | | | | | | | | | | | |
Collapse
|
25
|
Thompson JM, Carlton P, Akard LP, Dugan MJ, Jansen J. Starting granulocyte-colony-stimulating factor (filgrastim) early after autologous peripheral blood progenitor cell transplantation leads to faster engraftment without increased resource utilization. Transfusion 2008; 49:548-54. [PMID: 19040494 DOI: 10.1111/j.1537-2995.2008.02006.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Hematopoietic growth factor support is routinely used after autologous stem cell transplantation. The optimal starting date of this growth factor support has not been established yet, but many engraftment studies now recommend the fifth day after stem cell infusion (Day 5). STUDY DESIGN AND METHODS After switching the start date of granulocyte-colony-stimulating factor (G-CSF) support from the day of transplant (Day 0 group), to Day 5 after stem cell infusion (Day 5 group), the impression arose that there was an associated delay in engraftment of both white blood cells and platelets (PLTs). A retrospective analysis of two cohorts was performed with attention to engraftment variables and resource utilization. RESULTS Patients in the Day 0 group recovered an absolute granulocyte count of more than 0.500 x 10(9) per L significantly earlier than patients in the Day 5 group (p < 0.001 in log-rank test; median difference, 1 day). Time to PLT recovery of more than 20 x 10(9) per L, without transfusion support, was not significantly different between the Day 0 and Day 5 groups (p = 0.16; medians of 10 and 12 days, respectively). Resource utilization, defined as number of red blood cell and PLT transfusions, days with fever or on intravenous antibiotics, days with mucositis, and length of hospital stay, were not significantly different between the two groups (p >or= 0.15 in each case). Total charges for the transplant episode were also not different between the two groups (p = 0.48). CONCLUSION Starting G-CSF support on the day of stem cell infusion, instead of on Day +5, leads to faster hematologic recovery without a significant impact on resource utilization.
Collapse
Affiliation(s)
- James M Thompson
- Indiana Blood and Marrow Transplantation, Beech Grove, Indiana 46107, USA.
| | | | | | | | | |
Collapse
|
26
|
Metcalf D. THE CONSEQUENCES OF EXCESS LEVELS OF HAEMOPOIETIC GROWTH FACTORS. Br J Haematol 2008. [DOI: 10.1111/j.1365-2141.1990.00001.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Sachs L, Lotem J, Shabo Y. The molecular regulators of macrophage and granulocyte development. Role of MGI-2/IL-6. Ann N Y Acad Sci 2008; 557:417-35, discussion 435-7. [PMID: 2660699 DOI: 10.1111/j.1749-6632.1989.tb24035.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The development of a cell culture system for the in vitro cloning and clonal differentiation of normal hematopoietic cells made it possible to identify the proteins that regulate growth and differentiation of different hematopoietic cell lineages and the change in normal controls that produce leukemia. A model system with myeloid cells has identified different myeloid cell colony-inducing proteins, which we called MGI-1 (= CSF, including IL-3). There is another protein that we first described in 1976 and called MGI-2 in 1980 that induces differentiation of myeloid cells to macrophages or granulocytes without inducing the clonal growth of myeloid cells. The four CSF proteins and IL-1 induce the production of MGI-2 in myeloid cells and MGI-2 induces the production of GM-CSF. This shows the participation of MGI-2 in the network of interactions with different myeloid regulatory proteins. Using a monoclonal antibody to MGI-2, amino acid sequencing, and recombinant protein, we have shown in collaboration with the Genetics Institute that the major form of MGI-2 (MGI-2A) is IL-6. This shows that IL-6 is a myeloid cell differentiation inducing protein. The results also suggest new clinical potentials for MGI-2/IL-6.
Collapse
Affiliation(s)
- L Sachs
- Department of Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
28
|
Sneller MC, Lane HC. Infections in the immunocompromised host. Clin Immunol 2008. [DOI: 10.1016/b978-0-323-04404-2.10031-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Crowther D, Scarffe JH, Howell A, Thatcher N, Bronchud M, Steward WP, Testa N, Dexter M. Growth factor-assisted chemotherapy--the Manchester experience. CIBA FOUNDATION SYMPOSIUM 2007; 148:201-10; discussion 211-4. [PMID: 1690624 DOI: 10.1002/9780470513880.ch14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Stimulation of red cell production by erythropoietin and of granulocyte production by granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage-CSF (GM-CSF) has been demonstrated in several clinical studies. The first study to show that a human CSF could be used to shorten the period of neutropenia and reduce the risk of serious infection following intensive combination chemotherapy was carried out in Manchester using G-CSF. The period of neutropenia was significantly shortenened (by a median of 80%) and the neutrophil count levels were restored and above normal by 14 days after chemotherapy. In view of these results a further study was undertaken to examine the possibility of using intensive two weekly chemotherapy under cover of G-CSF. Treatment with Doxorubicin at doses of 75, 100, 125 and 150 mg/m2 was followed by infusion of G-CSF for 11 days. The neutrophil counts returned to normal within 12-14 days, allowing the delivery of up to three cycles of high dose chemotherapy at 14 day intervals. These studies demonstrated that intensive chemotherapy with dose-limiting myelodepression can be given with increased frequency under cover of G-CSF. Our studies using GM-CSF have also shown that administration by continuous i.v. infusion can reduce the period of life-threatening neutropenia following high dose Melphalan (120 mg/m2) without resort to autologous bone marrow transplantation (ABMT). In this study the period of granulocytopenia following Melphalan (less than 500 g x 10(9)/m2) was less than 15 days. This compares favourably with other series using high dose Melphalan followed by ABMT without CSF, where the duration of severe neutropenia was prolonged beyond three weeks. Although it appears that G-CSF and GM-CSF should be given either by continuous i.v. infusion or s.c. injection at doses between 3-10 micrograms/kg/day to obtain maximum biological effect, a great deal more work is required to determine optimum schedules and investigate the possibility of using more than one bioregulator.
Collapse
Affiliation(s)
- D Crowther
- Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Jansen EM, Hanks SG, Terry C, Akard LP, Thompson JM, Dugan MJ, Jansen J. Prediction of engraftment after autologous peripheral blood progenitor cell transplantation: CD34, colony-forming unit?granulocyte-macrophage, or both? Transfusion 2007; 47:817-23. [PMID: 17465946 DOI: 10.1111/j.1537-2995.2007.01195.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The rate of hematologic recovery after peripheral blood progenitor cell (PBPC) transplantation is influenced by the dose of progenitor cells. Enumeration of cells that express CD34+ on their surface is the most frequently used method to determine progenitor cell dose. In vitro growth of myeloid progenitor cells (colony-forming unit-granulocyte-macrophage [CFU-GM]) requires more time and resources, but may add predictive information. STUDY DESIGN AND METHODS A series of 323 patients, who underwent autologous PBPC transplantation for multiple myeloma, malignant lymphoma, or locally advanced breast cancer, were studied for the effect of CD34+ dose and CFU-GM dose on hematologic recovery. Measures for engraftment were days to absolute granulocyte and platelet (PLT) counts to greater than 500 per muL and than 20 x 10(9) per L, respectively, and number of PLT transfusions and red cell units required. RESULTS The CD34+ dose had a median of 8.4 x 10(6) per kg, and the CFU-GM dose a median of 84.9 x 10(4) per kg. The CD34+ and CFU-GM doses showed significant correlation (R = 0.63; p < 0.0001) but a wide variation in the ratio of CD34+ and CFU-GM. Both CD34+ and CFU-GM doses had significant correlation with the measures of engraftment, but for all measures the relationship of CD34+ was stronger. Multivariate analysis and subgroup analysis of patients receiving CD34+ doses of less than 5 x 10(6) per kg also did not reveal an independent predictive value for CFU-GM. CONCLUSION For prediction of hematologic recovery after autologous PBPC transplantation, determination of CFU-GM dose does not add to the predictive value of the CD34+ dose.
Collapse
Affiliation(s)
- Eva M Jansen
- Indiana Blood and Marrow Transplantation, Beech Grove, Indiana, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Gómez Raposo C, Pinto Marín A, González Barón M. Colony-stimulating factors: clinical evidence for treatment and prophylaxis of chemotherapy-induced febrile neutropenia. Clin Transl Oncol 2006; 8:729-34. [PMID: 17074671 DOI: 10.1007/s12094-006-0119-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The hematopoietic growth factors (HGFs) are a family of glycoproteins which plays a major role in the proliferation, differentiation, and survival of primitive hematopoietic stem and progenitor cells, and in the functions of some mature cells. More than 20 different molecules of HGF have been identified. Among them, granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) have been demostrated to be effective in reducing the incidence of febrile neutropenia when administered inmediately after chemotherapy and as supportive therapy in patients undergoing bone marrow transplantation. Chemotherapy used for treatment of cancer often causes neutropenia, which may be profound, requiring hospitalization, and leading to potentially fatal infection. The uses of the recombinant human hematopoietic colony-stimulating factors G-CSF and GM-CSF for treatment and prophylaxis of chemotherapy-induced febrile neutropenia will be reviewed here.
Collapse
Affiliation(s)
- César Gómez Raposo
- Servicio de Oncología Médica, Hospital Universitario La Paz, Madrid, Spain.
| | | | | |
Collapse
|
32
|
Abstract
GVAX is a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine. Original work with GM-CSF as a recombinant DNA protein (Leukine) involved proliferative stimulation of macrophages and neutrophils for the purpose of reducing hematopoietic toxicity related to dose-intensive chemotherapy. Following US Food and Drug Administration approval of Leukine several years ago, extensive preclinical results have demonstrated an immunostimulatory effect related to GM-CSF gene when transfected into tumor cells and used as a vaccine (GVAX). Tumor regression and prolonged survival was demonstrated in animal models. Toxicology with GVAX indicated no adverse effects, which enabled further testing in cancer patients. A small number of responses were demonstrated in Phase I trials in immunosensitive cancer patients (renal cell carcinoma and melanoma). However, a series of dramatic complete and durable responses in advanced non-small cell lung cancer patients, demonstrated in recent clinical trials, have generated interest in further development of this vaccine in nontraditional cancer disease types. The rationale of GVAX development and a summary of clinical results are reviewed.
Collapse
|
33
|
Kipar A, Meli ML, Failing K, Euler T, Gomes-Keller MA, Schwartz D, Lutz H, Reinacher M. Natural feline coronavirus infection: differences in cytokine patterns in association with the outcome of infection. Vet Immunol Immunopathol 2006; 112:141-55. [PMID: 16621029 PMCID: PMC7112699 DOI: 10.1016/j.vetimm.2006.02.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 01/05/2006] [Accepted: 02/15/2006] [Indexed: 11/22/2022]
Abstract
Natural and experimental feline coronavirus (FCoV) infection leads to systemic viral spread via monocyte-associated viraemia and induces systemic proliferation of monocytes/macrophages. In the majority of naturally infected animals, FCoV infection remains subclinical and is associated with generalised B and T cell hyperplasia, but no other pathological findings. A minority of cats, however, develop feline infectious peritonitis (FIP), a fatal systemic granulomatous disease. This is generally accompanied by B and T cell depletion. The obvious functional differences of lymphatic tissues in FCoV-infected cats with and without FIP suggest that they contribute to the outcome of FCoV infection. This study attempted to evaluate the functional changes in haemolymphatic tissues after natural FCoV infection, with special emphasis on the magnitude, phenotype and function of the monocyte/macrophage population. The spleen, mesenteric lymph nodes and bone marrow from naturally FCoV-infected cats with and without FIP and specific pathogen-free (SPF) control cats were examined for the quantity and activation state of monocytes/macrophages both by immunohistology and by quantitative real time PCR for the transcription of interleukin (IL)-1β, IL-6, IL-10, IL-12 p40, tumour necrosis factor (TNF), granulocyte colony stimulating factor (G-CSF), macrophage-CSF (M-CSF) and GM-CSF. Compared to cats with FIP, FCoV-infected cats without FIP exhibited significantly higher IL-10 levels in the spleen and significantly lower levels of IL-6, G- and M-CSF in mesenteric lymph nodes. In cats with FIP, however, IL-12 p40 levels were significantly lower in lymphatic tissues in comparison to both SPF cats and FCoV-infected cats without FIP. In comparison to SPF cats, FIP cats had significantly higher IL-1β levels and lower TNF levels in mesenteric lymph nodes and lower M-CSF levels in the spleen. Findings indicate that FCoV-infected cats which do not develop FIP are able to mount an effective FCoV-specific immune response and can avoid excessive macrophage activation and FIP, possibly by upregulation of IL-10 production. Development of FIP, however, might be due to a lack of IL-12 which inhibits an effective cellular immune response and allows for monocyte/macrophage activation and the development of FIP.
Collapse
Affiliation(s)
- Anja Kipar
- Institut für Veterinär-Pathologie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 96, 35392 Giessen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Burroughs L, Mielcarek M, Little MT, Bridger G, Macfarland R, Fricker S, Labrecque J, Sandmaier BM, Storb R. Durable engraftment of AMD3100-mobilized autologous and allogeneic peripheral-blood mononuclear cells in a canine transplantation model. Blood 2005; 106:4002-8. [PMID: 16105977 PMCID: PMC1895108 DOI: 10.1182/blood-2005-05-1937] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Accepted: 07/28/2005] [Indexed: 11/20/2022] Open
Abstract
Peripheral-blood mononuclear cells (PBMCs) mobilized with AMD3100, a CXCR4 antagonist, combined with granulocyte colony-stimulating factor (G-CSF) have reconstituted autologous hematopoiesis in cancer patients following myeloablative conditioning. The engraftment potential of PBMCs mobilized with AMD3100 alone, however, has remained unproven. We therefore studied AMD3100-mobilized PBMCs in a canine model. Four dogs received 920 cGy total body irradiation (TBI) before infusion of autologous AMD3100-mobilized PBMCs (median CD34 cell count, 3.9 x 10(6)/kg). Neutrophil (> 0.5 x 10(9)/L [500/microL]) and platelet (> 20 x 10(9)/L [> 20 000/microL]) recoveries occurred at medians of 9 (range, 7-10) days and 25 (range, 23-38) days, respectively, after TBI, and all dogs had normal marrow function at 1 year after transplantation. To evaluate the long-term engraftment potential of AMD3100-mobilized PBMCs, 5 dogs were given 920 cGy TBI followed by infusion of AMD3100-mobilized PBMCs (median CD34 cell dose, 4.7 x 10(6)/kg) from their dog leukocyte antigen (DLA)-identical littermates. Neutrophil and platelet recoveries occurred at medians of 8 (range, 8-10) days and 26 (range, 26-37) days, respectively, after TBI. With a median follow-up of 53 (range, 33-61) weeks, recipients' marrow function was normal, and blood-donor chimerism levels were 97% to 100%. In summary, both autologous and allogeneic AMD3100-mobilized PBMCs led to prompt and durable engraftment in dogs after 920 cGy TBI.
Collapse
Affiliation(s)
- Lauri Burroughs
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D1-100, PO Box 19024, Seattle, WA 98109-1024, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Eager R, Nemunaitis J. GM-CSF Gene-Transduced Tumor Vaccines. Mol Ther 2005; 12:18-27. [PMID: 15963916 DOI: 10.1016/j.ymthe.2005.02.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Revised: 02/04/2005] [Accepted: 02/05/2005] [Indexed: 10/25/2022] Open
Abstract
GVAX is a GM-CSF gene-transduced tumor vaccine. Expression of the GM-CSF gene within either autologous or allogeneic tumor cell populations has demonstrated evidence of immune stimulation in patients and evidence of antitumor activity particularly in prostate cancer and non-small-cell lung cancer. Results of preclinical studies justify clinical investigation. A summary of clinical results is presented.
Collapse
Affiliation(s)
- Robert Eager
- Mary Crowley Medical Research Center/Texas Oncology PA, Dallas, TX 75201, USA
| | | |
Collapse
|
36
|
Dubois RW, Pinto LA, Bernal M, Badamgarav E, Lyman GH. Benefits of GM-CSF Versus Placebo or G-CSF in Reducing Chemotherapy-Induced Complications: A Systematic Review of the Literature. ACTA ACUST UNITED AC 2004; 2:34-41. [DOI: 10.3816/sct.2004.n.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Chiarini R, Moran O, Revoltella RP. Identification of an Antigenic Domain Near the C Terminus of Human Granulocyte-Macrophage Colony-stimulating Factor and Its Spatial Localization. J Biol Chem 2004; 279:37908-17. [PMID: 15201279 DOI: 10.1074/jbc.m404663200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The goal of this study was to map an epitope on the human granulocyte-macrophage colony-stimulating factor (hGM-CSF) at its C terminus, a region whose integrity is fundamental in maintaining the normal function of this molecule. Residues including the fourth alpha-helix (D, 103-116) were analyzed for their role in the interaction with antibodies (Abs) raised against the protein. Five peptides homologous to different segments of the C terminus of hGM-CSF were synthesized. Peptide-(102-121) included the same residues of the alpha-helix D and the next five amino acids toward the C terminus; peptide-[E108A]-(102-121) introduced the mutation E108A in order to verify the role of acidic residues; peptide-[C96A](93-110) encompassed the beta-sheet 2 and half of the alpha-helix D; peptide-[C121A]-(110-127) included the second half of the alpha-helix D and the C terminus of hGMCSF; peptide-(13-31)-Gly-Pro-Gly-(103-116) included both the alpha-helices A and D connected by the tripeptide Gly-Pro-Gly, which allows the original antiparallel orientation of the two alpha-helices to be maintained. Both anti-protein and anti-peptide-(102-121) antibodies, capable of neutralizing the stimulatory activity of hGMCSF in the bone marrow colony-forming assays, recognized a specific epitope in the C terminus of hGM-CSF. Molecular modeling estimated the surface accessibility of hGM-CSF and the stability of the synthetic peptides in aqueous solution. Altogether, our results showed that the immunogenic region includes part of the alpha-helix D and the residues 116-120, which are external to this helix and particularly exposed on the protein surface, confirming the feasible participation of this region in antibody binding.
Collapse
Affiliation(s)
- Riccardo Chiarini
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Via G. Moruzzi, 1-56100 Pisa, Italy
| | | | | |
Collapse
|
38
|
Nemunaitis J, Nemunaitis J. Granulocyte-macrophage colony-stimulating factor gene-transfected autologous tumor cell vaccine: focus[correction to fcous] on non-small-cell lung cancer. Clin Lung Cancer 2004; 5:148-57. [PMID: 14667270 DOI: 10.3816/clc.2003.n.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Traditionally, non-small-cell lung cancer (NSCLC) is not thought of as an immunosensitive malignancy. However, recent clinical results with GVAX, a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transduced autologous tumor vaccine, may suggest otherwise. This review summarizes immune-induced activity caused by GM-CSF protein and GM-CSF gene-transfected vaccines. Initial indication of use for GM-CSF protein (sargramostim) was to improve neutrophil recovery following cytotoxic chemotherapy. However, several trials involving patients with hematologic malignancy demonstrated improvement in survival related to delayed disease progression in patients receiving sargramostim in combination with chemotherapy. Subsequently, others explored potential antitumor activity with sargramostim in a variety of trials. Results did not consistently demonstrate sufficient antitumor activity to justify routine use of sargramostim as an anticancer agent. Preclinical work with GM-CSF gene-transfected vaccines, however, did demonstrate significant activity, thereby justifying clinical investigation. Patients with metastatic NSCLC who had previously failed chemotherapy demonstrated response to GVAX (3 of 33 complete responses) and dose-related improvement in survival (471 days vs. 174 days).
Collapse
|
39
|
Zumberg MS, Leather HL, Nejame C, Meyer C, Wingard JR. GM-CSF versus G-CSF: engraftment characteristics, resource utilization, and cost following autologous PBSC transplantation. Cytotherapy 2003; 4:531-8. [PMID: 12568989 DOI: 10.1080/146532402761624692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND G-CSF and GM-CSF have both been shown to decrease the time to hematopoietic recovery when administered after autologous BM or peripheral stem cell re-infusion. However, few studies have compared G-CSF and GM-CSF to determine which is the preferred myeloid growth factor. METHODS This study compares a prospectively accrued cohort of 22 patients receiving GM-CSF with a historical cohort of patients who received G-CSF commencing Day + 6 after autologous PBSC transplantation. Patients were matched based on disease type and stage, CD34(+) cell dose/kg, conditioning regimen, and prior treatment. Time to myeloid engraftment, growth factor utilization, antibiotic utilization, fever incidence, and cost were compared. RESULTS The median time to neutrophil and platelet engraftment was similar in the two groups (ANC > 500 /mm(3), GM-CSF 12 versus G-CSF 11, P = 0.69). There was a trend towards more days of temperature > 38.0 masculine C (six versus three, P = 0.05) and febrile neutropenia (three versus two, P = 0.06) in the GM-CSF arm. There was a trend towards increased use of i.v. antibiotics in the GM-CSF cohort (7.6 days versus 5.5 days, P = 0.06). More chest X-rays (1.5 versus 1.0, P = 0.03) were ordered, and more blood cultures drawn (4.2 versus 2.7, P = 0.05) as part of fever evaluation in the group treated with GM-CSF. Resource utilization based on actual wholesale pricing (AWP) favored the G-CSF cohort. Applying a sensitivity analysis, GM-CSF became cost-effective when priced below $94 per 250 micro g, despite greater resource utilization. DISCUSSION This study suggests that engraftment characteristics are similar with GM-CSF and G-CSF following PBSC transplantation. Resource utilization for fever treatment and evaluation may be greater with GM-CSF. Determination of which agent is more cost-effective depends on institutional acquisition costs.
Collapse
Affiliation(s)
- M S Zumberg
- University of Florida College of Medicine, Department of Medicine and the Bone Marrow Transplant Program, Shands Hospital at the University of Florida, Gainesville, USA
| | | | | | | | | |
Collapse
|
40
|
Merimsky O, Bar-Yehuda S, Madi L, Fishman P. Modulation of the A3 adenosine receptor by low agonist concentration induces antitumor and myelostimulatory effects. Drug Dev Res 2003. [DOI: 10.1002/ddr.10182] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
41
|
Ito A, Tanaka K, Kondo K, Shinkai M, Honda H, Matsumoto K, Saida T, Kobayashi T. Tumor regression by combined immunotherapy and hyperthermia using magnetic nanoparticles in an experimental subcutaneous murine melanoma. Cancer Sci 2003; 94:308-13. [PMID: 12824927 PMCID: PMC11160160 DOI: 10.1111/j.1349-7006.2003.tb01438.x] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Immunotherapy (IT) has become an accepted therapeutic modality. We previously reported that intracellular hyperthermia (IH) using magnetic nanoparticles induces antitumor immunity. We undertook these studies in order to study the combined effects of IT and IH on melanoma. Magnetite cationic liposomes (MCLs) have a positive surface charge and generate heat in an alternating magnetic field (AMF) due to hysteresis loss. MCLs were injected into a B16 melanoma nodule in C57BL/6 mice, which were subjected to AMF for 30 min. The temperature at the tumor reached 43 degrees C and was maintained by controlling the magnetic field intensity. At 24 h after IH, interleukin-2 (IL-2) or granulocyte macrophage-colony stimulating factor (GM-CSF) was injected directly into the melanoma. Mice were divided into six groups: group I (control), group II (IH), group III (IL-2), group IV (GM-CSF), group V (IH + IL-2), and group VI (IH + GM-CSF). Complete regression of tumors was observed in mice of groups V and VI (75% (6/8) and 40% (4/10) of the mice, respectively), while no tumor regression was observed in mice of the other groups. This study supports the combined use of IT and IH using MCLs in patients with advanced malignancies.
Collapse
Affiliation(s)
- Akira Ito
- Department of Biotechnology, School of Engineering, Nagoya University
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Nelson RP, Ballow M. 26. Immunomodulation and immunotherapy: drugs, cytokines, cytokine receptors, and antibodies. J Allergy Clin Immunol 2003; 111:S720-43. [PMID: 12592317 DOI: 10.1067/mai.2003.146] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The preceding chapters in this primer have provided an overview of the immune response that serves as a background for understanding potential sites for immune modulation and immunotherapy. A number of soluble growth and activation factors are released from various cell populations involved in the immune response. They play vital roles in the initiation, propagation, and regulation of immunologic responses. Pharmacologic immunomodulators include suppressive and stimulatory agents. Immunosuppressive therapies include antimetabolites, cytotoxic drugs, radiation, adrenocortical glucocorticosteroids, immunophilins, and therapeutic antibodies. The field of clinical immunostimulation is emerging as an important therapeutic modality for a number of immunodeficiency diseases, chronic viral infections, and cancer. These compounds will be discussed in terms of general principles, molecular targets, major indications, and adverse effects.
Collapse
Affiliation(s)
- Robert P Nelson
- Division of Hematology/Oncology, Hematological Malignancy Program/Immunology, Indiana University School of Medicine, 55 Barnhill Drive, Ste 473, Indianapolis, IN 46202, USA
| | | |
Collapse
|
43
|
Bar-Yehuda S, Madi L, Barak D, Mittelman M, Ardon E, Ochaion A, Cohn S, Fishman P. Agonists to the A3 adenosine receptor induce G-CSF production via NF-kappaB activation: a new class of myeloprotective agents. Exp Hematol 2002; 30:1390-8. [PMID: 12482500 DOI: 10.1016/s0301-472x(02)00962-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the effect of CF101, a synthetic agonist to the A3 adenosine receptor (A3AR), on the production of granulocyte colony-stimulating factor (G-CSF). The ability of CF101 to act as a myeloprotective agent in chemotherapy-treated mice was tested. METHODS CF101 was administered orally to naïve mice and its effect was studied on blood cell counts (coulter counter), serum G-CSF level (ELISA), bone marrow colony-forming cells (soft agar culture), and splenocytes' ability to produce ex vivo G-CSF. Protein extract was prepared from splenocytes and Western blot analysis was carried out to evaluate expression level of key proteins. In an additional set of experiments, CF101 was administered to mice 48 hours after cyclophosphamide treatment and blood cell counts as well as serum G-CSF levels were monitored. RESULTS Oral administration of CF101 to naïve mice led to the elevation of serum G-CSF levels, an increase in absolute neutrophil counts (ANC), and bone marrow colony-forming cells. Splenocytes derived from these mice produced higher G-CSF level than controls. The molecular mechanisms underlying the events prior to G-CSF production included the upregulation of NF-kappaB and the upstream kinases phosphoinositide 3-kinase (PI3K), protein kinase B/Akt (PKB/Akt), and IKK. Accelerated recovery of white blood cells and neutrophil counts were observed in cyclophosphamide-treated mice following CF101 administration. CONCLUSION CF101 induced upregulation of the PI3K/NF-kappaB pathway leading to G-CSF production, resulting in myeloprotective effect in cyclophosphamide-treated mice.
Collapse
Affiliation(s)
- Sara Bar-Yehuda
- Laboratory of Clinical and Tumor Immunology, The Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Rabin Medical Center, Petach-Tikva, 49100 Israel
| | | | | | | | | | | | | | | |
Collapse
|
44
|
de Lima Mota PJ. Iatrogenia a fármacos no contexto do Aparelho Respiratório. REVISTA PORTUGUESA DE PNEUMOLOGIA 2002. [DOI: 10.1016/s0873-2159(15)30803-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
45
|
Estrov Z, Huh YO, Ginsberg CF, Harris D, Van Q, Mirza NQ, Talpaz M, Korbling M. Ex vivo expansion of apheresis-derived peripheral blood hematopoietic progenitors. J Clin Apher 2002; 17:7-16. [PMID: 11948700 DOI: 10.1002/jca.10012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Because the administration of hematopoietic growth factors and the use of stem cell support often fails to alleviate the neutropenic phase induced by cytotoxic drugs, several investigators have attempted to expand ex vivo hematopoietic progenitors for clinical use. These attempts have clearly shown that the cultured cells are functional and can be safely administered to patients, but that the in vivo performance is disappointing and the concept as a whole is not yet clinically useful. The major reasons for these unsuccessful attempts are thought to be cumbersome cell fractionation techniques, contamination, prolonged incubation, and the use of less than ideal cytokine combinations. In response, we have developed a simple procedure for ex vivo expansion of myeloid progenitor cells. In this assay, unfractionated mononuclear cells from apheresis donors are incubated in nonpyrogenic plastic bags for 7 days in the presence of culture medium either containing fetal calf serum or human plasma, granulocyte colony-stimulating factor, and stem cell factor. We have demonstrated that under these conditions the number of colony-forming units (CFU) granulocyte-macrophage (CFU-GM) and of CFU-granulocyte-macrophage-erythroid-megakaryocyte (CFU-GEMM) increased 7- and 9-fold, respectively, by day 7 and the number of burst-forming units-erythroid (BFU-E) increased 2.7-fold by day 5 of culture. Significant increases in the numbers of cells expressing CD34+, CD34+/CD38+, CD34+/CD33+, CD34+/CD15+, and CD34+/CD90+ and significant declines in the numbers of cells expressing CD34+/CD38- and CD19 surface antigens were also observed. The relative numbers of cells expressing T-cell markers and CD56 surface antigen did not change. By using different concentrations of various hematopoietic growth factor combinations, we can increase the number of mature and immature cells of different hematopoietic lineages.
Collapse
Affiliation(s)
- Zeev Estrov
- Department of Bioimmunotherapy, Univ. of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Tsimberidou AM, O'Brien SM, Cortes JE, Faderl S, Andreeff M, Kantarjian HM, Keating MJ, Giles FJ. Phase II study of fludarabine, cytarabine (Ara-C), cyclophosphamide, cisplatin and GM-CSF (FACPGM) in patients with Richter's syndrome or refractory lymphoproliferative disorders. Leuk Lymphoma 2002; 43:767-72. [PMID: 12153163 DOI: 10.1080/10428190290016872] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
A phase II study was conducted to evaluate the safety and efficacy of fludarabine, cytarabine (ara-C), cyclophosphamide, cisplatin and GM-CSF (FACPGM) treatment in patients with Richter's syndrome (RS), refractory prolymphocytic leukemia (PLL) or refractory non-Hodgkin's lymphoma (NHL). Twenty-two patients with RS, refractory PLL, or refractory NHL were entered into this trial between March 1997 and February 2001. Median age was 62 years (42-74); 77% were over 60 years of age. Histologic diagnosis was large cell NHL transformation in 15 patients with CLL, immunoblastic transformation of CLL in one, refractory PLL in three, and refractory NHL in three patients. Treatment consisted of fludarabine 30mg/m2 (days 1-3), ara-C 0.5g/m2 (days 3-4), cyclophosphamide 250 mg/m2 (days 2-4), cisplatin 15 mg/m2 IV CI (days 1-4) with GM-CSF 250 microg/m2 from day 5 to recovery of neutrophils and antibiotic prophylaxis. Patients with response were to receive a maximum of six cycles of therapy. Eighteen patients were evaluable for response; one patient achieved a complete remission (5%), 12 stable disease/no response (67%) and five patients had progressive disease (28%). The median survival was 2.2 months (range, 1-19); the median failure-free survival was 1.5 months (range, 0.5-18.6). Grade III/IV toxicities were as follows: anemia in 62% of cycles; leucopoenia in 66%; granulocytopenia in 90%; thrombocytopenia in 83%; hyperbilirubinemia in 14%; hyperuricemia in 17%; hyponatremia in 17%; hypokalemia in 14%; hypophosphatemia in 10%; hypoalbulinemia in 14%; hypocalcemia in 7%; and hypercalcemia in 3%. One (3%) patient developed cardiac failure. Forty-one percent of the cycles were complicated with fever, 34% with non-neutropenic fever, and 55% cycles with infections (fungal 31%; bacterial 57%; HSV 6%; VZV 6%). FACPGM had very limited activity and significant toxicity in a cohort of patients with heavily pretreated refractory lymphoproliferative disorders.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- Department of Leukemia, University of Texas, M. D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Stasko J, Drouet L, Soria C, Mazoyer E, Caen J, Kubisz P. Erythropoietin and granulocyte colony-stimulating factor increase plasminogen activator inhibitor-1 release in HUVEC culture. Thromb Res 2002; 105:161-4. [PMID: 11958807 DOI: 10.1016/s0049-3848(02)00011-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
UNLABELLED Recombinant human erythropoietin (rHuEPO) is considered to be a mitogenic and chemotactic agent in cultured endothelial cells (ECs). The effect of recombinant granulocyte-macrophage (rGM-CSF) and granulocyte (rG-CSF) colony-stimulating factors on the proliferation of human umbilical vein endothelial cells (HUVECs) has been controversial. METHODS HUVEC proliferation and the release of endothelial markers in HUVEC culture stimulated by rHuEPO, rGM-CSF and G-CSF were measured. RESULTS We found the dose-dependent stimulating effect of rHuEPO and rGM-CSF on HUVEC proliferation, but we did not achieve this with rG-CSF. rHuEPO like rG-CSF was an effective agent in stimulating the plasminogen activator inhibitor (PAI)-1 release from HUVECs to a cultured medium, while rGM-CSF failed. CONCLUSION We suggest that rHuEPO showed prothrombotic changes in HUVEC culture. Our results support the idea of suspected rHuEPO direct prothrombotic role in haemodialysed patients treated with rHuEPO.
Collapse
Affiliation(s)
- Ján Stasko
- Department of Hematology and Transfusiology, University Hospital, Kollárova 2, 036 59 Martin, Slovakia.
| | | | | | | | | | | |
Collapse
|
48
|
Biesma B, van Kralingen KW, van Leen RW, Koster MC, Postmus PE. Recombinant human interleukin-3 administered concomitantly with chemotherapy in patients with relapsed small cell lung cancer. JOURNAL OF EXPERIMENTAL THERAPEUTICS AND ONCOLOGY 2002; 2:47-52. [PMID: 12415620 DOI: 10.1046/j.1359-4117.2002.01006.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although recombinant human interleukin-3 (rhIL-3) shortens both the duration of chemotherapy-induced neutropenia and thrombocytopenia, its effect on nadir counts is limited. Concurrent administration of rhIL-3 and chemotherapy may enhance this effect. However, simultaneous administration of other hematopoietic growth factors and chemotherapy has resulted in enhanced myelosuppression. We investigated whether concomitant administration of rhIL-3 and chemotherapy would result in enhanced myelosuppression. Twelve patients with relapsed small cell lung cancer received vincristine, ifosfamide, mesna, and carboplatin on day 1 every four weeks. RhIL-3 was administered subcutaneously on days 1-14 during cycle 1 at doses of 4 (three patients) or 8 micrograms/kg/day (nine patients). During cycle 2 patients received only chemotherapy. No significant difference in leukocyte (1.4 +/- 1.0 vs. 0.9 +/- 0.4 x 10(9)/l (mean +/- SD), neutrophil (0.5 +/- 0.6 vs. 0.2 +/- 0.2 x 10(9)/l), and platelet (64 +/- 60 vs. 38 +/- 58 x 10(9)/l) nadir counts were demonstrated. The hemoglobin nadir level was significantly higher during cycle 1 (6.5 +/- 1.1 vs. 5.5 +/- 0.9 mmol/l, P = 0.05). Both leukocyte and platelet recovery were significantly enhanced in the rhIL-3 cycle. There was no significant difference in chemotherapy postponement or platelet transfusions. As a result of severe headaches, rhIL-3 administration was discontinued in one patient at 8 micrograms. RhIL-3 during this chemotherapy regimen for relapsed small cell lung cancer did not enhance myelotoxicity but did improve bone marrow recovery. This observation may increase the application of rhIL-3, for instance in combination with other hematopoietic growth factors.
Collapse
Affiliation(s)
- Bonne Biesma
- Department of Pulmonology, University Hospital, Free University of Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
49
|
Zuberbier T, Welker P, Grabbe J, Henz BM. Effect of granulocyte macrophage colony-stimulating factor in a patient with benign systemic mastocytosis. Br J Dermatol 2001; 145:661-6. [PMID: 11703299 DOI: 10.1046/j.1365-2133.2001.04435.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We report the in vitro and in vivo effects of granulocyte macrophage colony stimulating factor (GM-CSF), a known inhibitor of in vitro mast cell differentiation, in a patient with benign, adult-onset systemic mastocytosis. In vitro effects of GM-CSF on bone marrow cultures before the start of treatment showed a marked inhibition of mast cell marker expression [tryptase, Kit, and high-affinity IgE receptor (FcepsilonRIalpha)] at both protein and mRNA levels. Therefore, the patient was treated with daily injections of GM-CSF for 10 weeks. After an initial improvement, increasing worsening of clinical symptoms was noted, and the patient refused further treatment. Lesional skin biopsies showed an increase of toluidine blue-positive mast cells, compared with uninvolved skin, with further significant increase after treatment. Similar results were obtained on staining for mast cell-specific tryptase and Kit, as well as for CD1a and FcepsilonRIalpha. These findings show that GM-CSF inhibits human bone marrow mast cell differentiation in vitro, and also in mastocytosis. However, GM-CSF apparently enhances recruitment of mast cell as well as dendritic cell precursors into the tissue during systemic treatment. These findings and the observed adverse clinical effects in the present patient make it unlikely that GM-CSF monotherapy will be beneficial for the treatment of mastocytosis.
Collapse
Affiliation(s)
- T Zuberbier
- Department of Dermatology and Allergy, Charité, Humboldt University, 13344 Berlin, Germany.
| | | | | | | |
Collapse
|
50
|
Khatami S, Brummer E, Stevens DA. Effects of granulocyte-macrophage colony stimulating factor (GM-CSF) in vivo on cytokine production and proliferation by spleen cells. Clin Exp Immunol 2001; 125:198-201. [PMID: 11529909 PMCID: PMC1906115 DOI: 10.1046/j.1365-2249.2001.01600.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
GM-CSF is a potent stimulator of haematopoietic cells as well as some functions of granulocytes and macrophages. GM-CSF has many clinical uses; however, little is known about the effects of GM-CSF treatment in vivo on the responses of tissue lymphocytes in terms of secretion of Th-1 and Th-2 cytokines. We investigated this issue by measuring the responses of spleen cells from mice 24 h after treatment i.p. with saline or rmGM-CSF. GM-CSF at 16.7-50.0 microg/kg significantly increased (P < 0.01) spleen cellularity 2-2.5-fold and enhanced proliferative responses of non-stimulated (no mitogen) as well as concanavalin A (Con A)-stimulated spleen cells. Secretion of IFN-gamma by Con A (2.5 microg/ml)-stimulated spleen cells was significantly (P < 0.01) increased from 1.8 microg/ml by control spleen cells to 5.2 microg/ml by GM-CSF spleen cells. IL-10 production was greater (0.25 microg/ml, P < 0.05) by Con A-stimulated spleen cells from GM-CSF-treated mice compared to control spleen cells (0.06 microg/ml). By contrast, there were no significant differences in IL-4 production by Con A-stimulated spleen cells from the different groups. These results show that GM-CSF treatment increases spleen cellularity and primes lymphocytes for enhanced responses. The enhanced production of Th-1 cytokines by primed lymphocytes may partially explain the beneficial role of in vivo administration of GM-CSF in several clinical situations.
Collapse
Affiliation(s)
- S Khatami
- California Institute for Medical Research, Santa Clara Valley Medical Center, San Jose, CA, USA
| | | | | |
Collapse
|