1
|
Bazarbachi AH, Mapara MY. Cytokines in hematopoietic cell transplantation and related cellular therapies. Best Pract Res Clin Haematol 2024; 37:101600. [PMID: 40074514 DOI: 10.1016/j.beha.2025.101600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025]
Abstract
Cytokines are pleiotropic molecules involved in hematopoiesis, immune responses, infections, and inflammation. They play critical roles in hematopoietic cell transplantation (HCT) and immune effector cell (IEC) therapies, mediating both therapeutic and adverse effects. Thus, cytokines contribute to the immunopathology of graft-versus-host disease (GVHD), cytokine release syndrome (CRS), and immune effector cell-associated neurotoxicity syndrome (ICANS). This review examines cytokine functions in these contexts, their influence on engraftment and immune recovery post-transplantation, and their role in mediating toxicities. We focus on current and potential uses of cytokines to enhance engraftment and potentiate IEC therapies, as well as strategies to mitigate cytokine-mediated complications using cytokine blockers (e.g., tocilizumab, anakinra) and JAK inhibitors (e.g., ruxolitinib). We discuss new insights into GVHD physiology that have led to novel treatments, such as CSF1R blockade, which is effective in refractory chronic GVHD.
Collapse
Affiliation(s)
- Abdul-Hamid Bazarbachi
- Division of Hematology/Oncology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA
| | - Markus Y Mapara
- Division of Hematology/Oncology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA; Columbia Center for Translational Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, USA.
| |
Collapse
|
2
|
Bordbar M, Hosseini-Bensenjan M, Sayadi M, Zekavat O, Bordbar S, Nozari F, Haghpanah S. The Impact of Prophylactic Post-Chemotherapy G-CSF on the Relapse Rates in Patients with Acute Myeloid Leukemia: A Meta-Analysis. Cancer Invest 2024; 42:452-468. [PMID: 38922312 DOI: 10.1080/07357907.2024.2352454] [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] [Received: 07/18/2023] [Accepted: 05/03/2024] [Indexed: 06/27/2024]
Abstract
This meta-analysis evaluated the impact of prophylactic post-chemotherapy granulocyte colony-stimulating factor (G-CSF) in patients with acute myeloid leukemia (AML). Overall, the relapse rate, overall survival, event-free survival, and mortality rate were similar in G-CSF (+) compared to G-CSF (-) patients. However, the relative risk (RR) of relapse was higher in children and in secondary AML patients who were treated with G-CSF compared to the G-CSF (-) group [RR, 95% confidence interval: 1.26, 1.04-1.52, and 1.12 (1.02-1.24)]. Treatment with post-chemotherapy G-CSF should be prescribed with caution in pediatric patients with AML and secondary AML as possibly increasing the relapse risk.
Collapse
Affiliation(s)
| | | | - Mehrab Sayadi
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Zekavat
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayan Bordbar
- Student Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnoosh Nozari
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sezaneh Haghpanah
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Karimdadi Sariani O, Eghbalpour S, Kazemi E, Rafiei Buzhani K, Zaker F. Pathogenic and therapeutic roles of cytokines in acute myeloid leukemia. Cytokine 2021; 142:155508. [PMID: 33810945 DOI: 10.1016/j.cyto.2021.155508] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with high mortality that accounts for the most common acute leukemia in adults. Despite all progress in the therapeutic strategies and increased rate of complete remission, many patients will eventually relapse and die from the disease. Cytokines as molecular messengers play a pivotal role in the immune system. The imbalance release of cytokine has been shown to exert a significant influence on the progression of hematopoietic malignancies including acute myeloid leukemia. This article aimed to summarize current knowledge about cytokines and their critical roles in the pathogenesis, treatment, and survival of AML patients.
Collapse
Affiliation(s)
- Omid Karimdadi Sariani
- Department of Genetics, College of Science, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Sara Eghbalpour
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Elahe Kazemi
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Farhad Zaker
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Xia T, Zhang B, Li Y, Fang B, Zhu X, Xu B, Zhang J, Wang M, Fang J. New insight into 20(S)-ginsenoside Rh2 against T-cell acute lymphoblastic leukemia associated with the gut microbiota and the immune system. Eur J Med Chem 2020; 203:112582. [PMID: 32682197 DOI: 10.1016/j.ejmech.2020.112582] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/13/2020] [Accepted: 06/14/2020] [Indexed: 02/07/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a hematopoietic malignancy associated with unfavorable factors including male gender and over nine years of age. Chemotherapy toxicity continues to present a major challenge. There is a need to develop novel natural agents to improve survival and quality of life in patients with T-ALL. 20(S)-ginsenoside Rh2 (GRh2) exhibits immune regulation and anti-tumor effects in both cellular and murine xenograft models. In the present study, the anti-cancer mechanisms of 20(S)-GRh2 involved in the immune system and intestinal microbiota were investigated in T-ALL mice. We revealed that 20(S)-Rh2 suppressed T-ALL by blocking the PI3K/Akt/mTOR signaling pathway, and enhanced immunity in the spleen by regulating immune factors. In addition, 20(S)-GRh2 altered the composition of the gut microbiota, and promoted intestinal homeostasis by elevating the levels of tight junction proteins, antimicrobial peptides and IgA. 20(S)-GRh2 ameliorated the LPS-induced inflammatory response in the intestine of T-ALL mice. Furthermore, Bacteroidetes, Verrucomicrobia, Akkermansia, Lactobacillus, and Lachnospiraceae_NK4A136_group were positively correlated with anti-tumor immune factors, intestinal barrier-related factors, and the anti-inflammatory response. Conversely, Firmicutes, Proteobacteria, Parabacteroides and Alistipes had the opposite correlation. Collectively, these results suggest that 20(S)-GRh2 is a safe and effective natural product, that shows promise for the prevention and treatment of T-ALL.
Collapse
Affiliation(s)
- Ting Xia
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China.
| | - Bo Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Yu Li
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Bin Fang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Xiaoxuan Zhu
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Bicheng Xu
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Jin Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Min Wang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China.
| | - Jianpei Fang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guang Dong, 510120, PR China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, Guang Dong, 510120, PR China.
| |
Collapse
|
5
|
Abstract
Acute myeloid leukemia (AML) is one of the best studied malignancies, and significant progress has been made in understanding the clinical implications of its disease biology. Unfortunately, drug development has not kept pace, as the '7+3' induction regimen remains the standard of care for patients fit for intensive therapy 40 years after its first use. Temporal improvements in overall survival were mostly confined to younger patients and driven by improvements in supportive care and use of hematopoietic stem cell transplantation. Multiple forms of novel therapy are currently in clinical trials and are attempting to bring bench discoveries to the bedside to benefit patients. These novel therapies include improved chemotherapeutic agents, targeted molecular inhibitors, cell cycle regulators, pro-apoptotic agents, epigenetic modifiers, and metabolic therapies. Immunotherapies in the form of vaccines; naked, conjugated and bispecific monoclonal antibodies; cell-based therapy; and immune checkpoint inhibitors are also being evaluated in an effort to replicate the success seen in other malignancies. Herein, we review the scientific basis of these novel therapeutic approaches, summarize the currently available evidence, and look into the future of AML therapy by highlighting key clinical studies and the challenges the field continues to face.
Collapse
|
6
|
Stiehl T, Ho AD, Marciniak-Czochra A. Mathematical modeling of the impact of cytokine response of acute myeloid leukemia cells on patient prognosis. Sci Rep 2018; 8:2809. [PMID: 29434256 PMCID: PMC5809606 DOI: 10.1038/s41598-018-21115-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/30/2018] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease. One reason for the heterogeneity may originate from inter-individual differences in the responses of leukemic cells to endogenous cytokines. On the basis of mathematical modeling, computer simulations and patient data, we have provided evidence that cytokine-independent leukemic cell proliferation may be linked to early relapses and poor overall survival. Depending whether the model of cytokine-dependent or cytokine-independent leukemic cell proliferation fits to the clinical data, patients can be assigned to two groups that differ significantly with respect to overall survival. The modeling approach further enables us to identify parameter constellations that can explain unexpected responses of some patients to external cytokines such as blast crisis or remission without chemotherapy.
Collapse
Affiliation(s)
- Thomas Stiehl
- Institute of Applied Mathematics, Interdisciplinary Center of Scientific Computing and BIOQUANT Center, Heidelberg University, Im Neuenheimer Feld 205, 69120, Heidelberg, Germany.
| | - Anthony D Ho
- Department of Medicine V, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Anna Marciniak-Czochra
- Institute of Applied Mathematics, Interdisciplinary Center of Scientific Computing and BIOQUANT Center, Heidelberg University, Im Neuenheimer Feld 205, 69120, Heidelberg, Germany
| |
Collapse
|
7
|
Anderson G, Mackay N, Gilroy K, Hay J, Borland G, McDonald A, Bell M, Hassanudin SA, Cameron E, Neil JC, Kilbey A. RUNX-mediated growth arrest and senescence are attenuated by diverse mechanisms in cells expressing RUNX1 fusion oncoproteins. J Cell Biochem 2017; 119:2750-2762. [PMID: 29052866 PMCID: PMC5813226 DOI: 10.1002/jcb.26443] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 01/27/2023]
Abstract
RUNX gene over‐expression inhibits growth of primary cells but transforms cells with tumor suppressor defects, consistent with reported associations with tumor progression. In contrast, chromosomal translocations involving RUNX1 are detectable in utero, suggesting an initiating role in leukemias. How do cells expressing RUNX1 fusion oncoproteins evade RUNX‐mediated growth suppression? Previous studies showed that the TEL‐RUNX1 fusion from t(12;21) B‐ALLs is unable to induce senescence‐like growth arrest (SLGA) in primary fibroblasts while potent activity is displayed by the RUNX1‐ETO fusion found in t(8;21) AMLs. We now show that SLGA potential is suppressed in TEL‐RUNX1 but reactivated by deletion of the TEL HLH domain or mutation of a key residue (K99R). Attenuation of SLGA activity is also a feature of RUNX1‐ETO9a, a minor product of t(8;21) translocations with increased leukemogenicity. Finally, while RUNX1‐ETO induces SLGA it also drives a potent senescence‐associated secretory phenotype (SASP), and promotes the immortalization of rare cells that escape SLGA. Moreover, the RUNX1‐ETO SASP is not strictly linked to growth arrest as it is largely suppressed by RUNX1 and partially activated by RUNX1‐ETO9a. These findings underline the heterogeneous nature of premature senescence and the multiple mechanisms by which this failsafe process is subverted in cells expressing RUNX1 oncoproteins.
Collapse
Affiliation(s)
- Gail Anderson
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Nancy Mackay
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Kathryn Gilroy
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Jodie Hay
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Gillian Borland
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Alma McDonald
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Margaret Bell
- School of Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Siti Ayuni Hassanudin
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Ewan Cameron
- School of Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - James C Neil
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Anna Kilbey
- Molecular Oncology Laboratory, Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
8
|
Weng S, Matsuura S, Mowery CT, Stoner SA, Lam K, Ran D, Davis AG, Lo MC, Zhang DE. Restoration of MYC-repressed targets mediates the negative effects of GM-CSF on RUNX1-ETO leukemogenicity. Leukemia 2016; 31:159-169. [PMID: 27389055 PMCID: PMC5214981 DOI: 10.1038/leu.2016.167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023]
Abstract
GM-CSF signaling regulates hematopoiesis and immune responses. CSF2RA, the gene encoding the α subunit for GM-CSF, is significantly downregulated in t(8;21) (RUNX1-ETO or RE) leukemia patients, suggesting that it may serve as a tumor suppressor. We previously reported that GM-CSF signaling is inhibitory to RE leukemogenesis. Here we conducted gene expression profiling of primary RE hematopoietic stem/progenitor cells (HSPCs) treated with GM-CSF to elucidate the mechanisms mediating the negative effects of GM on RE leukemogenicity. We observed that GM treatment of RE HSPCs resulted in a unique gene expression profile that resembles primary human cells undergoing myelopoiesis, which was not observed in control HSPCs. Additionally we discovered that GM-CSF signaling attenuates MYC-associated gene signatures in RE HSPCs. In agreement with this, a functional screen of a subset of GM-CSF-responsive genes demonstrated that a MYC inhibitor, MXI1, reduced the leukemic potential of RE HSPCs and t(8;21) AML cells. Furthermore, MYC knockdown and treatment with the BET inhibitor JQ1 reduced the leukemic potential of t(8;21) cell lines. Altogether, we discovered a novel molecular mechanism mediating the GM-CSF-induced reduction in leukemic potential of RE cells, and our findings support MYC inhibition as an effective strategy for reducing the leukemogenicity of t(8;21) AML.
Collapse
Affiliation(s)
- S Weng
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - S Matsuura
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - C T Mowery
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - S A Stoner
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - K Lam
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - D Ran
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - A G Davis
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - M-C Lo
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - D-E Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.,Department of Pathology and Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
9
|
Ravandi F, Ritchie EK, Sayar H, Lancet JE, Craig MD, Vey N, Strickland SA, Schiller GJ, Jabbour E, Erba HP, Pigneux A, Horst HA, Recher C, Klimek VM, Cortes J, Roboz GJ, Odenike O, Thomas X, Havelange V, Maertens J, Derigs HG, Heuser M, Damon L, Powell BL, Gaidano G, Carella AM, Wei A, Hogge D, Craig AR, Fox JA, Ward R, Smith JA, Acton G, Mehta C, Stuart RK, Kantarjian HM. Vosaroxin plus cytarabine versus placebo plus cytarabine in patients with first relapsed or refractory acute myeloid leukaemia (VALOR): a randomised, controlled, double-blind, multinational, phase 3 study. Lancet Oncol 2015; 16:1025-1036. [PMID: 26234174 DOI: 10.1016/s1470-2045(15)00201-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/20/2015] [Accepted: 05/20/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Safe and effective treatments are urgently needed for patients with relapsed or refractory acute myeloid leukaemia. We investigated the efficacy and safety of vosaroxin, a first-in-class anticancer quinolone derivative, plus cytarabine in patients with relapsed or refractory acute myeloid leukaemia. METHODS This phase 3, double-blind, placebo-controlled trial was undertaken at 101 international sites. Eligible patients with acute myeloid leukaemia were aged 18 years of age or older and had refractory disease or were in first relapse after one or two cycles of previous induction chemotherapy, including at least one cycle of anthracycline (or anthracenedione) plus cytarabine. Patients were randomly assigned 1:1 to vosaroxin (90 mg/m(2) intravenously on days 1 and 4 in a first cycle; 70 mg/m(2) in subsequent cycles) plus cytarabine (1 g/m(2) intravenously on days 1-5) or placebo plus cytarabine through a central interactive voice system with a permuted block procedure stratified by disease status, age, and geographical location. All participants were masked to treatment assignment. The primary efficacy endpoint was overall survival and the primary safety endpoint was 30-day and 60-day all-cause mortality. Efficacy analyses were done by intention to treat; safety analyses included all treated patients. This study is registered with ClinicalTrials.gov, number NCT01191801. FINDINGS Between Dec 17, 2010, and Sept 25, 2013, 711 patients were randomly assigned to vosaroxin plus cytarabine (n=356) or placebo plus cytarabine (n=355). At the final analysis, median overall survival was 7·5 months (95% CI 6·4-8·5) in the vosaroxin plus cytarabine group and 6·1 months (5·2-7·1) in the placebo plus cytarabine group (hazard ratio 0·87, 95% CI 0·73-1·02; unstratified log-rank p=0·061; stratified p=0·024). A higher proportion of patients achieved complete remission in the vosaroxin plus cytarabine group than in the placebo plus cytarabine group (107 [30%] of 356 patients vs 58 [16%] of 355 patients, p<0·0001). Early mortality was similar between treatment groups (30-day: 28 [8%] of 355 patients in the vosaroxin plus cytarabine group vs 23 [7%] of 350 in the placebo plus cytarabine group; 60-day: 70 [20%] vs 68 [19%]). Treatment-related deaths occurred at any time in 20 (6%) of 355 patients given vosaroxin plus cytarabine and in eight (2%) of 350 patients given placebo plus cytarabine. Treatment-related serious adverse events occurred in 116 (33%) and 58 (17%) patients in each group, respectively. Grade 3 or worse adverse events that were more frequent in the vosaroxin plus cytarabine group than in the placebo plus cytarabine group included febrile neutropenia (167 [47%] vs 117 [33%]), neutropenia (66 [19%] vs 49 [14%]), stomatitis (54 [15%] vs 10 [3%]), hypokalaemia (52 [15%] vs 21 [6%]), bacteraemia (43 [12%] vs 16 [5%]), sepsis (42 [12%] vs 18 [5%]), and pneumonia (39 [11%] vs 26 [7%]). INTERPRETATION Although there was no significant difference in the primary endpoint between groups, the prespecified secondary analysis stratified by randomisation factors suggests that the addition of vosaroxin to cytarabine might be of clinical benefit to some patients with relapsed or refractory acute myeloid leukaemia. FUNDING Sunesis Pharmaceuticals.
Collapse
Affiliation(s)
- Farhad Ravandi
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | - Hamid Sayar
- Indiana University Cancer Center, Indianapolis, IN, USA
| | | | | | - Norbert Vey
- Institut Paoli-Calmettes and Aix-Marseille University, Marseille, France
| | | | | | - Elias Jabbour
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Harry P Erba
- Division of Hematology and Oncology, University of Alabama, Birmingham, AL, USA
| | - Arnaud Pigneux
- Université de Bordeaux, CHU de Bordeaux, Bordeaux, France
| | - Heinz-August Horst
- Medizinische Klinik und Poliklinik, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christian Recher
- Institut Universitaire du Cancer de Toulouse Oncopole, Université de Toulouse III, CHU de Toulouse, Toulouse, France
| | | | - Jorge Cortes
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | - Lloyd Damon
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Bayard L Powell
- Wake Forest University Baptist Medical Center-Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Gianluca Gaidano
- Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | | | - Andrew Wei
- The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Donna Hogge
- Vancouver General Hospital, Vancouver, BC, Canada
| | - Adam R Craig
- Sunesis Pharmaceuticals, South San Francisco, CA, USA
| | - Judith A Fox
- Sunesis Pharmaceuticals, South San Francisco, CA, USA
| | - Renee Ward
- Sunesis Pharmaceuticals, South San Francisco, CA, USA
| | | | - Gary Acton
- Sunesis Pharmaceuticals, South San Francisco, CA, USA
| | - Cyrus Mehta
- Cytel, Cambridge, MA, USA; Harvard School of Public Health, Cambridge, MA, USA
| | | | | |
Collapse
|
10
|
Nipp RD, Rao AV. Performance status in elderly patients with acute myeloid leukemia: exploring gene expression signatures of cytokines and chemokines. J Gerontol A Biol Sci Med Sci 2013; 70:714-21. [PMID: 23783402 DOI: 10.1093/gerona/glt039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/21/2013] [Indexed: 01/30/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive disease that predominantly affects elderly patients. Cytokines and chemokines are major players in the pathogenesis of AML. They regulate the disease course and play a deleterious role in the progression of AML. The geriatric population is particularly vulnerable to these mediators as these cytokines and chemokines are also implicated in the development of frailty, fatigue, and declining cognitive function. It is the combination of these adverse effects of cytokines and chemokines that affect performance status and, in turn, the poor prognosis in this age group. Cytokines and chemokines are emerging as therapeutic targets in AML. Future endeavors to treat AML will likely involve cytokines and chemokines as attempts are made to disrupt the bone marrow environment. By modulating the bone marrow stroma, the goal is to create an environment less favorable to AML cells and more favorable to the effects of chemotherapy against AML.
Collapse
Affiliation(s)
- Ryan D Nipp
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Arati V Rao
- Department of Medicine, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
11
|
|
12
|
Anti-leukemic properties of IL-12, IL-23 and IL-27: Differences and similarities in the control of pediatric B acute lymphoblastic leukemia. Crit Rev Oncol Hematol 2012; 83:310-8. [DOI: 10.1016/j.critrevonc.2011.11.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 11/11/2011] [Accepted: 11/15/2011] [Indexed: 12/19/2022] Open
|
13
|
Ferretti E, Cocco C, Airoldi I, Pistoia V. Targeting acute myeloid leukemia cells with cytokines. J Leukoc Biol 2012; 92:567-575. [DOI: 10.1189/jlb.0112036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Abstract
Review of data identifying IL-12 and IL-27 as potential therapeutic agents for pediatric AML by targeting leukemia initiating cells and/or blasts.
AML is a hematologic malignancy that represents 15–20% of all childhood acute leukemias and is responsible for more than one-half of pediatric leukemic deaths. The bulk tumor is continuously regenerated and sustained by rare leukemic ICs that proliferate slowly, thus resulting refractory to chemotherapeutic agents targeting highly proliferating cells within the tumor. Therefore, a complete eradication of the bulk tumor may depend on efficacy of therapies that target IC. In spite of the improvements in the treatment of AML, the difficulty to eradicate completely the disease incites research for innovative therapeutic approaches. In this regard, the role of cytokines in the treatment of AML has been investigated for many years, and some of them have been tested in clinical trials as a result of their immunomodulatory properties. Furthermore, recent preclinical studies highlighted the ability of the IL-12 superfamily cytokines as potent antileukemic agents that act directly on tumor cells and on leukemic IC, thus opening new perspectives for leukemic patient treatment. Here, we review the current knowledge about the antileukemic effects of cytokines, documented in preclinical and clinical studies, discussing their potential clinical application.
Collapse
Affiliation(s)
- Elisa Ferretti
- Laboratory of Oncology, Istituto Giannina Gaslini , Genova, Italy
| | - Claudia Cocco
- Associazione Italiana Ricerca sul Cancro Laboratory of Immunology and Tumors, Department of Experimental and Laboratory Medicine, Istituto Giannina Gaslini , Genova, Italy
| | - Irma Airoldi
- Associazione Italiana Ricerca sul Cancro Laboratory of Immunology and Tumors, Department of Experimental and Laboratory Medicine, Istituto Giannina Gaslini , Genova, Italy
| | - Vito Pistoia
- Laboratory of Oncology, Istituto Giannina Gaslini , Genova, Italy
| |
Collapse
|
14
|
Zorzoli A, Di Carlo E, Cocco C, Ognio E, Ribatti D, Ferretti E, Dufour C, Locatelli F, Montagna D, Airoldi I. Interleukin-27 inhibits the growth of pediatric acute myeloid leukemia in NOD/SCID/Il2rg-/- mice. Clin Cancer Res 2012; 18:1630-40. [PMID: 22383738 DOI: 10.1158/1078-0432.ccr-11-2432] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Acute myeloid leukemia (AML) accounts for more than half of fatal cases in all pediatric leukemia patients; this observation highlights the need of more effective therapies. Thus, we investigated whether interleukin (IL)-27, an immunomodulatory cytokine, functions as an antitumor agent against pediatric AML cells. EXPERIMENTAL DESIGN Expression of WSX-1 and gp130 on AML cells from 16 pediatric patients was studied by flow cytometry. Modulation of leukemia cell proliferation or apoptosis upon IL-27 treatment in vitro was tested by bromodeoxyuridine/propidium iodide (PI) and Ki67, or Annexin V/PI staining and flow cytometric analysis. The angiogenic potential of AML cells treated or not with IL-27 was studied by chorioallantoic membrane assay and PCR array. In vivo studies were carried out using nonobese diabetic/severe combined immunodeficient (NOD/SCID)/Il2rg(-/-) mice injected intravenously with five pediatric AML cell samples. Leukemic cells engrafted in PBS and IL-27-treated animals were studied by immunohistochemical/morphologic analysis and by PCR array for expression angiogenic/dissemination-related genes. RESULTS We provided the first demonstration that (i) AML cells injected into NOD/SCID/Il2rg(-/-) mice gave rise to leukemia dissemination that was severely hampered by IL-27, (ii) compared with controls, leukemia cells harvested from IL-27-treated mice showed significant reduction of their angiogenic and spreading related genes, and (iii) similarly to what was observed in vivo, IL-27 reduced in vitro AML cell proliferation and modulated the expression of different genes involved in the angiogenic/spreading process. CONCLUSION These results provide an experimental rationale for the development of future clinical trials aimed at evaluating the toxicity and efficacy of IL-27.
Collapse
Affiliation(s)
- Alessia Zorzoli
- Department of Experimental and Laboratory Medicine, IRCCS G. Gaslini Institute, Genoa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Bijou F, Ivanovic Z, Boiron JM, Nicolini F. [Hematopoietic stem cells mobilization: state of the art in 2011 and perspectives]. Transfus Clin Biol 2011; 18:503-15. [PMID: 22019608 DOI: 10.1016/j.tracli.2011.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 02/19/2011] [Indexed: 10/16/2022]
Abstract
High-dose chemotherapy with stem cells support has largely improved in terms of hematopoietic stem and progenitor cells harvest procedures as well as in those, which target or manipulate the cellular composition of autologous graft. Optimal preparative regimens and supportive care had lead to better use of autologous transplantation procedure. For other patients assigned to hematopoietic transplantation, availability of allogeneic donors appears to be an interesting alternative source of hematopoietic stem cells. Since three decades, hematopoietic growth factors development has allowed mobilization optimization and collection of peripheral hematopoietic stem cells leading to reduced days of hospitalization and less blood products requirements, being more cost-effective for patients in autologous transplantation settings and for stem cell collection facilities in allogeneic ones. New perspectives include, besides ex vivo manipulation of graft, development of mobilizing drugs in order to perform transplantation even in poor mobilizers patients. An important goal is achieved with the description of genetic polymorphisms related to optimal mobilization of stem cells. New approach using more promising and selective agents called chemokines, such as plerixafor the main leader among these agents are now available and appear complementary for alternative approach using cytokines alone (G-CSF, GM-CSF, SCF). The aim of this review is to assess the evolution of theses biotechnologies and their role in different steps of autologous transplantation and allogeneic stem cells collection.
Collapse
Affiliation(s)
- F Bijou
- Établissement français du sang Aquitaine-Limousin, place Amélie-Raba-Léon, 33035 Bordeaux cedex, France.
| | | | | | | |
Collapse
|
16
|
Ravandi F. New treatments and strategies in acute myeloid leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2011; 11 Suppl 1:S60-4. [PMID: 22035750 PMCID: PMC4096137 DOI: 10.1016/j.clml.2011.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 02/09/2011] [Accepted: 02/22/2011] [Indexed: 10/18/2022]
Abstract
Despite considerable progress in the treatment of acute myeloid leukemia in the past several decades, the prognosis of the majority of patients with this disease remains guarded. Advances in supportive care and better characterization of disease subsets through cytogenetics and molecular analysis have led to significant success in treating specific subsets of patients, such as those with acute promyelocytic leukemia and core binding factor leukemias, particularly among the younger patients who are able to better tolerate the effects of cytotoxic chemotherapy. However, overall, only about 40% of younger patients and <10% of older patients with this disease are alive at 5 years. Current research is focusing on the identification of new cellular targets amenable to specific inhibitors, designing the best strategies for combining these novel agents with traditional chemotherapy regimens, and determining prognostic indicators that may allow us to better stratify therapy.
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Cytokines and microRNA in pediatric B-acute lymphoblastic leukemia. Cytokine Growth Factor Rev 2011; 22:149-56. [DOI: 10.1016/j.cytogfr.2011.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 05/13/2011] [Accepted: 05/24/2011] [Indexed: 01/05/2023]
|
18
|
Inaba H, Cao X, Pounds S, Pui CH, Rubnitz JE, Ribeiro RC, Razzouk BI. Randomized trial of 2 dosages of prophylactic granulocyte-colony-stimulating factor after induction chemotherapy in pediatric acute myeloid leukemia. Cancer 2010; 117:1313-20. [PMID: 21381017 DOI: 10.1002/cncr.25536] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/24/2010] [Accepted: 05/26/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Granulocyte-colony-stimulating factor (G-CSF) is effective in accelerating neutrophil recovery after intensive chemotherapy for acute myeloid leukemia (AML). However, the optimal G-CSF dosage for patients with AML has not been determined. To the authors' knowledge, G-CSF dosages have not been compared in a randomized AML study. METHODS Patients who were enrolled on the St. Jude AML97 protocol and remained on study after window therapy were eligible to participate. The effect of the dosage of G-CSF given after induction chemotherapy Courses 1 and 2 was analyzed in 46 patients who were assigned randomly in a double-blinded manner to receive either 5 μg/kg daily or 10 μg/kg daily of G-CSF. The number of days of G-CSF treatment, neutropenia (an absolute neutrophil count <0.5 × 10(9) /L), and hospitalization; the number of episodes of febrile neutropenia, grade 2 through 4 infection, and antimicrobial therapy; transfusion requirements; the cost of supportive care; and survival were compared between the 2 study arms. RESULTS No statistically significant differences were observed between the 2 arms in any of the endpoints measured. CONCLUSIONS The higher G-CSF dosage (10 μg/kg daily) offered no greater benefit than the lower dosage (5 μg/kg daily) in patients who were receiving intensive chemotherapy for AML.
Collapse
Affiliation(s)
- Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Tavor S, Petit I. Can inhibition of the SDF-1/CXCR4 axis eradicate acute leukemia? Semin Cancer Biol 2010; 20:178-85. [PMID: 20637871 DOI: 10.1016/j.semcancer.2010.07.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 07/08/2010] [Indexed: 02/07/2023]
Abstract
Poor prognosis of acute leukemia with current treatments is mainly due to the relapse of the disease following chemotherapy. In the last decade, an emerging concept has proposed that the leukemia stem cells (LSCs) and their interactions with the BM microenvironment are the major cause of the acute leukemia relapse. Adhesion to the stromal niche is crucial for LSCs as it directly supports self-renewal, proliferation, arrest of differentiation and protects from damaging chemo-agents. One of the key players in this crosstalk between leukemic cells and the BM stroma niche is the chemokine SDF-1. SDF-1 regulates the process of homing and engraftment of LSCs into the BM and inhibition of its receptor CXCR4 induces leukemic cell mobilization into the circulation. However, besides its chemotactic and adhesive functions, SDF-1 is also a pleiotropic cytokine that regulates leukemic cell proliferation as well as their program of differentiation. CXCR4 antagonists are used in combination with chemotherapy in preclinical and clinical studies, which demonstrate that blocking CXCR4 is a novel promising approach of therapy. In this review, we focus on the multifaceted SDF-1/CXCR4 axis in acute leukemia and discuss how targeting this pathway could provide potential interest to eradicate the LSCs.
Collapse
Affiliation(s)
- Sigal Tavor
- Institute of Hematology and Bone Marrow Transplantation, Sourasky Medical Center, Tel Aviv, Israel.
| | | |
Collapse
|
20
|
Brumatti G, Salmanidis M, Ekert PG. Crossing paths: interactions between the cell death machinery and growth factor survival signals. Cell Mol Life Sci 2010; 67:1619-30. [PMID: 20157838 PMCID: PMC11115775 DOI: 10.1007/s00018-010-0288-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 01/08/2023]
Abstract
Cytokines and growth factors play a crucial role in the maintenance of haematopoietic homeostasis. They transduce signals that regulate the competing commitments of haematopoietic stem cells, quiescence or proliferation, retention of stem cell pluripotency or differentiation, and survival or demise. When the balance between these commitments and the requirements of the organisms is disturbed, particularly when it favours survival and proliferation, cancer may result. Cell death provoked by loss of growth factor signalling is regulated by the Bcl-2 family of apoptosis regulators, and thus survival messages transduced by growth factors must regulate the activity of these proteins. Many aspects of direct interactions between cytokine signalling and regulation of apoptosis remain elusive. In this review, we explore the mechanisms by which cytokines, in particular Interleukin-3 and granulocyte-macrophage colony-stimulating factor, promote cell survival and suppress apoptosis as models of how cytokine signalling and apoptotic pathways intersect.
Collapse
Affiliation(s)
- Gabriela Brumatti
- Children's Cancer Centre, Murdoch Children's Research Institute, Royal Children's Hospital, Department of Paediatrics, University of Melbourne, Flemington Rd Parkville, Melbourne, 3052, Australia.
| | | | | |
Collapse
|
21
|
Abstract
Granulocyte colony-stimulating factor (G-CSF) has been used in the clinic for more than 2 decades to treat congenital and acquired neutropenias and to reduce febrile neutropenia before or during courses of intensive cytoreductive therapy. In addition, healthy stem cell donors receive short-term treatment with G-CSF for mobilization of hematopoietic stem cells. G-CSF has also been applied in priming strategies designed to enhance the sensitivity of leukemia stem cells to cytotoxic agents, in protocols aimed to induce their differentiation and accompanying growth arrest and cell death, and in severe aplastic anemia and myelodysplastic syndrome (MDS) to alleviate anemia. The potential adverse effects of G-CSF administration, particularly the risk of malignant transformation, have fueled ongoing debates, some of which can only be settled in follow-up studies extending over several decades. This specifically applies to children with severe congenital neutropenia who receive lifelong treatment with G-CSF and in which the high susceptibility to develop MDS and acute myeloid leukemia (AML) has now become a major clinical concern. Here, we will highlight some of the controversies and challenges regarding the clinical application of G-CSF and discuss a possible role of G-CSF in malignant transformation, particularly in patients with neutropenia harboring mutations in the gene encoding the G-CSF receptor.
Collapse
|
22
|
Schuster SJ, Venugopal P, Kern JC, McLaughlin P. GM-CSF plus rituximab immunotherapy: Translation of biologic mechanisms into therapy for indolent B-cell lymphomas. Leuk Lymphoma 2009; 49:1681-92. [DOI: 10.1080/10428190802216731] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Grim SA, Clark NM. The role of adjuvant agents in treating fungal diseases. CURRENT FUNGAL INFECTION REPORTS 2009. [DOI: 10.1007/s12281-009-0016-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
24
|
Wheatley K, Goldstone AH, Littlewood T, Hunter A, Burnett AK. Randomized placebo-controlled trial of granulocyte colony stimulating factor (G-CSF) as supportive care after induction chemotherapy in adult patients with acute myeloid leukaemia: a study of the United Kingdom Medical Research Council Adult Leukaemia Working Party. Br J Haematol 2009; 146:54-63. [PMID: 19438472 DOI: 10.1111/j.1365-2141.2009.07710.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The role of granulocyte colony stimulating factor (G-CSF) as supportive therapy following intensive induction chemotherapy for acute myeloid leukaemia (AML) in adults was investigated in a randomized trial. G-CSF (Lenograstim, 263 microg/d) or placebo was administered from day 8 after the end of chemotherapy until neutrophil recovery to 0.5 x 10(9)/l (or for up to 10 d). Eight hundred and three patients were entered. Neutrophil recovery was quicker with G-CSF (P < 0.0001), but this did not lead to differences in the number, severity or duration of infections. There were no substantial supportive care savings, although G-CSF patients spent 2 d less in hospital (P = 0.01). Complete remission (CR) rates were similar between arms (73% G-CSF, 75% placebo, P = 0.5), as were reasons for failure (induction death: P = 0.7; resistant disease: P = 0.5) and, for remitters, 5-year disease-free survival (34% vs. 38%, P = 0.3). Overall survival at 5 years was 29% with G-CSF vs. 36% with placebo (P = 0.10). Both CR rate (P = 0.006) and overall survival (P = 0.006) were worse with G-CSF in patients aged <40 years, but this may be a chance effect. There is some evidence from this trial of an adverse effect of G-CSF but these data need to be viewed in the context of the evidence from the other trials.
Collapse
Affiliation(s)
- Keith Wheatley
- Birmingham Clinical Trials Unit, School of Cancer Sciences, Robert Aitken Institute, University of Birmingham, Edgbaston, Birmingham, UK.
| | | | | | | | | |
Collapse
|
25
|
Nowak D, Stewart D, Koeffler HP. Differentiation therapy of leukemia: 3 decades of development. Blood 2009; 113:3655-65. [PMID: 19221035 PMCID: PMC2943835 DOI: 10.1182/blood-2009-01-198911] [Citation(s) in RCA: 264] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 02/09/2009] [Indexed: 12/27/2022] Open
Abstract
A characteristic feature of leukemia cells is a blockade of differentiation at a distinct stage in cellular maturation. In the 1970s and 1980s, studies demonstrating the capabilities of certain chemicals to induce differentiation of hematopoietic cell lines fostered the concept of treating leukemia by forcing malignant cells to undergo terminal differentiation instead of killing them through cytotoxicity. The first promising reports on this notion prompted a review article on this subject by us 25 years ago. In this review, we revisit this interesting field of study and report the progress achieved in the course of nearly 3 decades. The best proof of principle for differentiation therapy has been the treatment of acute promyelocytic leukemia with all-trans retinoic acid. Attempts to emulate this success with other nuclear hormone ligands such as vitamin D compounds and PPARgamma agonists or different classes of substances such as hematopoietic cytokines or compounds affecting the epigenetic landscape have not been successful on a broad scale. However, a multitude of studies demonstrating partial progress and improvements and, finally, the new powerful possibilities of forward and reverse engineering of differentiation pathways by manipulation of transcription factors support the continued enthusiasm for differentiation therapy of leukemia in the future.
Collapse
Affiliation(s)
- Daniel Nowak
- Division of Hematology and Oncology, Cedars Sinai Medical Center, University of California Los Angeles (UCLA) School of Medicine, CA 90048, USA.
| | | | | |
Collapse
|
26
|
Márkász L, Hajas G, Kiss A, Lontay B, Rajnavölgyi É, Erdődi F, Oláh É. Granulocyte Colony Stimulating Factor Increases Drug Resistance of Leukaemic Blast Cells to Daunorubicin. Pathol Oncol Res 2008; 14:285-92. [DOI: 10.1007/s12253-008-9057-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Accepted: 04/17/2008] [Indexed: 11/28/2022]
|
27
|
Levenga TH, Timmer-Bonte JNH. Review of the value of colony stimulating factors for prophylaxis of febrile neutropenic episodes in adult patients treated for haematological malignancies. Br J Haematol 2007; 138:146-52. [PMID: 17593021 DOI: 10.1111/j.1365-2141.2007.06653.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chemotherapy-induced neutropenia is a major dose-limiting toxicity of systemic cancer chemotherapy that can lead to fever and infection, requiring prompt analysis and in-patient treatment with broad-spectrum antibiotics. Complicated neutropenia may lead to reduction and/or delay of systemic anti-cancer treatment, which may compromise outcome. Haematopoietic growth factors have the ability to augment haematopoietic cell cycling and are used to facilitate more dose-intense treatments and to decrease treatment-related complications. This review focuses on randomised trials that investigated the use of colony-stimulating factors (CSF) to prevent treatment-related febrile complications in haematological malignancies in (younger) adult patients. In general, these studies demonstrated that CSF reduced the duration of severe neutropenia but not always its febrile complications; therefore inconsistent results regarding clinically relevant reduction of hospitalisation, duration of therapeutic antibiotics, infection-related or disease-related mortality and economic effects were reported. Current developments in treatment of haematological malignancies will pose new challenges as a shift in infectious pathogens can be expected.
Collapse
|
28
|
Ravandi F, Faderl S, Kebriaei P, Kantarjian H. Modern treatment programs for adults with acute lymphoblastic leukemia. Curr Hematol Malig Rep 2007; 2:169-75. [DOI: 10.1007/s11899-007-0023-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
29
|
John Wiley & Sons, Ltd.. Current Awareness in Hematological Oncology. Hematol Oncol 2006. [DOI: 10.1002/hon.755] [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]
|