1
|
Li L, Hou M, Fang S. Application of colony-stimulating factor 3 in determining the prognosis of high-grade gliomas based on magnetic resonance imaging radiomics. Heliyon 2023; 9:e15325. [PMID: 37095939 PMCID: PMC10122032 DOI: 10.1016/j.heliyon.2023.e15325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Rationale and objectives Radiomics is a promising, non-invasive method for determining the prognosis of high-grade glioma (HGG). The connection between radiomics and the HGG prognostic biomarker is still insufficient. Materials and methods In this study, we collected the pathological, clinical, RNA-sequencing, and enhanced MRI data of HGG from TCIA and TCGA databases. We characterized the prognostic value of CSF3. Kaplan-Meier (KM) analysis, univariate and multivariate Cox regression, subgroup analysis, Spearman analysis, and gene set variation analysis enrichment were used to elucidate the prognostic value of the CSF3 gene and the correlation between CSF3 and tumor features. We used CIBERSORT to analyze the correlation between CSF3 and cancer immune infiltrates. Logistic regression (LR) and support vector machine methods (SVM) were used to build the radiomics models for the prognosis prediction of HGG based on the expression of CSF3. Results Based on the radiomics score calculated from LR model, 182 patients with HGG from TCGA database were divided into radiomics score (RS) high and low groups. CSF3 expression varied between tumor and normal group tissues. CSF3 expression was found to be a significant risk factor for survival outcomes. A positive association was found between CSF3 expression and immune infiltration. The radiomics model based on both LR and SVM methods showed high clinical practicability. Conclusion The results showed that CSF3 has a prognostic value in HGG. The developed radiomics models can predict the expression of CSF3, and further validate the predictions of the radiomics models for HGG.
Collapse
Affiliation(s)
- Leina Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Laboratory Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
- Corresponding author. Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Meidan Hou
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University Dalian, Liaoning, China
| | - Shaobo Fang
- Department of Medical Imaging, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Fujita K, Okubo A, Nakamura T, Takeuchi N. Disseminated carcinomatosis of the bone marrow caused by granulocyte colony-stimulating factor: A case report and review of literature. World J Gastrointest Oncol 2022; 14:2077-2084. [PMID: 36310701 PMCID: PMC9611438 DOI: 10.4251/wjgo.v14.i10.2077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/08/2022] [Accepted: 08/21/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Disseminated carcinomatosis of the bone marrow (DCBM) is a widespread metastasis with a hematologic disorder that is mainly caused by gastric cancer. Although it commonly occurs as a manifestation of recurrence long after curative treatment, the precise mechanism of relapse from dormant status remains unclear. Granulocyte colony-stimulating factor (G-CSF) can promote cancer progression and invasion in various cancers. However, the potential of G-CSF to trigger recurrence from a cured malignancy has not been reported.
CASE SUMMARY A 55-year-old Japanese woman was diagnosed with Ewing sarcoma localized on the fifth lumbar vertebrae 6 years after curative gastrectomy for T1 gastric cancer. After palliative surgery to release nerve compression, pathological diagnosis of the resected specimen was followed by curative radiation and chemotherapy. During treatment, G-CSF was administered 32 times for severe neutropenia prophylaxis. Eight months after completing definitive treatment, she complained of severe back pain and was diagnosed as multiple bone metastases with DCBM from gastric cancer. Despite palliative chemotherapy, she died of disseminated intravascular coagulation 13 d after the diagnosis. Immunohistochemical examination of the autopsied bone marrow confirmed a diffuse positive staining for the G-CSF receptor (G-CSFR) in the relapsed gastric cancer cell cytoplasm, whereas the primary lesion cancer cells showed negative staining for G-CSFR. In this case, G-CSF administration may have been the key trigger for the disseminated relapse of a dormant gastric cancer.
CONCLUSION When administering G-CSF to cancer survivors, recurrence of a preceding cancer should be monitored even after curative treatment.
Collapse
Affiliation(s)
- Kengo Fujita
- Department of Medical Oncology, Ina Central Hospital, Nagano 396-8555, Japan
| | - Ayaka Okubo
- Department of Medical Oncology, Ina Central Hospital, Nagano 396-8555, Japan
| | - Toshitsugu Nakamura
- Department of Diagnostic Pathology, Ina Central Hospital, Nagano 396-8555, Japan
| | - Nobumichi Takeuchi
- Department of Medical Oncology, Ina Central Hospital, Nagano 396-8555, Japan
| |
Collapse
|
3
|
High Neutrophil-to-Lymphocyte Ratio Facilitates Cancer Growth-Currently Marketed Drugs Tadalafil, Isotretinoin, Colchicine, and Omega-3 to Reduce It: The TICO Regimen. Cancers (Basel) 2022; 14:cancers14194965. [PMID: 36230888 PMCID: PMC9564173 DOI: 10.3390/cancers14194965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Several elements that are composed of, or related to, neutrophils, have been shown to inhibit strong immune responses to cancer and promote cancers’ growth. This paper presents the collected data showing these elements and how their coordinated actions as an ensemble facilitate growth in the common cancers. The paper goes on to present a drug regimen, TICO, designed to reduce the cancer growth enhancing effects of the neutrophil related elements. TICO uses four already marketed, readily available generic drugs, repurposed to inhibit neutrophil centered growth facilitation of cancer. Abstract This paper presents remarkably uniform data showing that higher NLR is a robust prognostic indicator of shorter overall survival across the common metastatic cancers. Myeloid derived suppressor cells, the NLRP3 inflammasome, neutrophil extracellular traps, and absolute neutrophil count tend to all be directly related to the NLR. They, individually and as an ensemble, contribute to cancer growth and metastasis. The multidrug regimen presented in this paper, TICO, was designed to decrease the NLR with potential to also reduce the other neutrophil related elements favoring malignant growth. TICO is comprised of already marketed generic drugs: the phosphodiesterase 5 inhibitor tadalafil, used to treat inadequate erections; isotretinoin, the retinoid used for acne treatment; colchicine, a standard gout (podagra) treatment; and the common fish oil supplement omega-3 polyunsaturated fatty acids. These individually impose low side effect burdens. The drugs of TICO are old, cheap, well known, and available worldwide. They all have evidence of lowering the NLR or the growth contributing elements related to the NLR when clinically used in general medicine as reviewed in this paper.
Collapse
|
4
|
Battram AM, Oliver-Caldés A, Suárez-Lledó M, Lozano M, Bosch I Crespo M, Martínez-Cibrián N, Cid J, Moreno DF, Rodríguez-Lobato LG, Urbano-Ispizua A, Fernández de Larrea C. T cells isolated from G-CSF-treated multiple myeloma patients are suitable for the generation of BCMA-directed CAR-T cells. Mol Ther Methods Clin Dev 2022; 26:207-223. [PMID: 35859694 PMCID: PMC9271987 DOI: 10.1016/j.omtm.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/16/2022] [Indexed: 10/29/2022]
Abstract
Autologous cell immunotherapy using B cell maturation antigen (BCMA)-targeted chimeric antigen receptor (CAR)-T cells is an effective novel treatment for multiple myeloma (MM). This therapy has only been used for relapsed and refractory patients, at which stage the endogenous T cells used to produce the CAR-T cells are affected by the immunosuppressive nature of advanced MM and/or side effects of previous therapies. An alternative pool of "fitter" T cells is found in leukocytoapheresis products that are routinely collected to obtain hematopoietic progenitor cells for autologous stem cell transplantation (ASCT) early in the treatment of MM. However, to mobilize the progenitor cells, patients are dosed with granulocyte colony-stimulating factor (G-CSF), which is reported to adversely affect T cell proliferation, function, and differentiation. Here, we aimed to first establish whether G-CSF treatment negatively influences T cell phenotype and to ascertain whether previous exposure of T cells to G-CSF is deleterious for anti-BCMA CAR-T cells. We observed that G-CSF had a minimal impact on T cell phenotype when added in vitro or administered to patients. Moreover, we found that CAR-T cell fitness and anti-tumor activity were unaffected when generated from G-CSF-exposed T cells. Overall, we showed that ASCT apheresis products are a suitable source of T cells for anti-BCMA CAR-T cell manufacture.
Collapse
Affiliation(s)
- Anthony M Battram
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Aina Oliver-Caldés
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Maria Suárez-Lledó
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Miquel Lozano
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy & Hemostasis, ICMHO (Institut Clínic de Malalties Hematològiques i Oncològiques), Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
| | - Miquel Bosch I Crespo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Núria Martínez-Cibrián
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Joan Cid
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Apheresis & Cellular Therapy Unit, Department of Hemotherapy & Hemostasis, ICMHO (Institut Clínic de Malalties Hematològiques i Oncològiques), Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
| | - David F Moreno
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Alvaro Urbano-Ispizua
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, 08036 Barcelona, Spain.,Department of Haematology, University of Barcelona, 08036 Barcelona, Spain
| | - Carlos Fernández de Larrea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain.,Department of Haematology, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
5
|
Tian W, Wang Y, Zhou Y, Yao Y, Deng Y. Effects of Prophylactic Administration of Granulocyte Colony-Stimulating Factor on Peripheral Leukocyte and Neutrophil Counts Levels After Chemotherapy in Patients With Early-Stage Breast Cancer: A Retrospective Cohort Study. Front Oncol 2022; 12:777602. [PMID: 35547875 PMCID: PMC9084938 DOI: 10.3389/fonc.2022.777602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background Both chemotherapy-induced neutropenia (CIN) and febrile neutropenia (FN) frequently occur and can lead to dose-limiting toxicity and even fatal chemotherapy side effects. The prophylactic use of recombinant human granulocyte colony-stimulating factor (rhG-CSF), including pegylated rhG-CSF (PEG-rhG-CSF), significantly reduces the risks of CIN and FN during chemotherapy in early-stage breast cancer (ESBC) patients. However, whether the prophylactic use of granulocyte colony-stimulating factor (G-CSF), especially PEG-rhG-CSF, can influence white blood cell (WBC) counts and absolute neutrophil counts (ANCs) after finishing the chemotherapy remains unknown. Therefore, exploring the development and recovery tendency of WBC counts and ANCs during and after chemotherapy is crucial. Objective We aimed to investigate the variation tendency and recovery of WBC counts and ANCs during and after chemotherapy and evaluate the independent factors influencing leukopenia and neutropenia lasting longer after chemotherapy. We also aimed to provide individualized prophylactically leukocyte elevation therapy for breast cancer patients. Methods This single-center retrospective cohort study evaluated 515 ESBC patients who received rhG-CSF or PEG-G-CSF for prophylaxis after adjuvant or neoadjuvant chemotherapy. Blood test reports were analyzed during chemotherapy, and on a 12-month follow-up period after finishing the chemotherapy. The WBC counts and ANCs were measured to assess their variation tendency characteristics and to identify independent factors that influenced the occurrence of leukopenia and neutropenia lasting longer than 12 months after chemotherapy. Results Prophylaxis with rhG-CSF or PEG-rhG-CSF kept the mean values of WBC counts and ANCs within the normal range during chemotherapy, but a significant difference in WBC levels was detected before the end of the last chemotherapy compared to the prechemotherapy period (baseline) (p < 0.001). During the 12-month follow-up after the end of the last chemotherapy, WBC counts and ANCs gradually recovered, but the group that used only PEG-rhG-CSF (long-acting group, p WBC = 0.012) or rhG-CSF (short-acting group, p WBC = 0.0005) had better leukocyte elevation effects than the mixed treatment group (PEG-rhG-CSF mixed rhG-CSF). Besides, the short-acting group had a better neutrophil elevation effect than the longer-acting (p ANC = 0.019) and mixed (p ANC = 0.002) groups. Leukopenia was still present in 92 (17.9%) patients and neutropenia in 63 (12.2%) 12 months after the end of the last chemotherapy. The duration of leukopenia over 12 months was closely associated with the baseline WBC level (p < 0.001), G-CSF types (p = 0.027), and surgical method (p = 0.041). Moreover, the duration of neutropenia over 12 months was closely related to the baseline ANC (p < 0.001), G-CSF types (p = 0.043), and molecular typing (p = 0.025). Conclusion The prophylactic application of G-CSF effectively stabilized the WBC counts and ANCs during chemotherapy in ESBC patients. Nevertheless, the recovery of WBC counts and ANCs after chemotherapy varied between different G-CSF treatment groups. The risk of leukopenia and neutropenia persisting for more than 12 months after chemotherapy was associated with G-CSF types, the baseline level of WBC count/ANCs, surgical method, and molecular typing.
Collapse
Affiliation(s)
- Wei Tian
- Department of Breast Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Breast Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunxiang Zhou
- Department of Breast Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yihan Yao
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Breast Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Mouchemore KA, Anderson RL. Immunomodulatory effects of G-CSF in cancer: Therapeutic implications. Semin Immunol 2021; 54:101512. [PMID: 34763974 DOI: 10.1016/j.smim.2021.101512] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/23/2021] [Indexed: 01/04/2023]
Abstract
Numerous preclinical studies have reported a pro-tumour role for granulocyte colony-stimulating factor (G-CSF) that is predominantly mediated by neutrophils and MDSCs, the major G-CSF receptor expressing populations. In the presence of G-CSF (either tumour-derived or exogenous) these myeloid populations commonly exhibit a T cell suppressive phenotype. However, the direct effects of this cytokine on other immune lineages, such as T and NK cells, are not as well established. Herein we discuss the most recent data relating to the effect of G-CSF on the major immune populations, exclusively in the context of cancer. Recent publications have drawn attention to the other tumour-promoting effects of G-CSF on myeloid cells, including NETosis, promotion of cancer stemness and skewed differentiation of bone marrow progenitors towards myelopoiesis. Although G-CSF is safely and commonly used as a supportive therapy to prevent or treat chemotherapy-associated neutropenia in cancer patients, we also discuss the potential impacts of G-CSF on other anti-cancer treatments. Importantly, considerations for immune checkpoint blockade are highlighted, as many publications report a T cell suppressive effect of G-CSF that may diminish the effectiveness of this immunotherapy.
Collapse
Affiliation(s)
- Kellie A Mouchemore
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
7
|
Hiraga T, Ito S, Mizoguchi T. Opposing Effects of Granulocyte Colony-Stimulating Factor on the Initiation and Progression of Breast Cancer Bone Metastases. Mol Cancer Res 2021; 19:2110-2119. [PMID: 34465584 DOI: 10.1158/1541-7786.mcr-21-0243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/09/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022]
Abstract
Granulocyte colony stimulating factor (G-CSF), an essential cytokine regulating granulopoiesis, is expressed in a substantial proportion of breast cancers, and it has been implicated in cancer progression. Here, we examined effects of G-CSF on the development of bone metastases of breast cancer using immunocompetent mouse models. The expression of CXC chemokine ligand 12 (CXCL12) in bone marrow stromal cells, which plays a critical role in the maintenance of hematopoietic stem cells and also in cancer cell homing to bone, was markedly decreased in mice treated with G-CSF. Flow cytometric analysis revealed that pretreatment of mice with G-CSF reduced the number of bone-homing cancer cells. G-CSF also increased the population of myeloid-derived suppressor cells (MDSCs) in bone marrow. Depletion of MDSCs using anti-Gr-1 antibody treatment significantly decreased the metastatic tumor burden in bone. The overall effects of G-CSF on bone metastases were finally examined using two different treatment protocols. When mice were treated with G-CSF prior to the tumor cell inoculation, G-CSF did not change bone metastatic-tumor burden. In contrast, when G-CSF treatment was started after the tumor cells had homed to bone, G-CSF significantly accelerated bone metastases formation. These results suggest that G-CSF suppressed cancer cell homing to bone by downregulating CXCL12 expression in bone marrow stromal cells, whereas G-CSF stimulated the progression of bone metastases at least in part by MDSC-mediated mechanisms. IMPLICATIONS: G-CSF had opposing effects on the initiation and progression of bone metastases of breast cancer and the balance may regulate the metastatic tumor burden.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Shiojiri-shi, Nagano, Japan.
| | - Susumu Ito
- Division of Instrumental Analysis, Research Center for Human and Environmental Sciences, Shinshu University, Matsumoto-shi, Nagano, Japan
| | - Toshihide Mizoguchi
- Institute for Oral Science, Matsumoto Dental University, Shiojiri-shi, Nagano, Japan.,Oral Health Science Center, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
8
|
Jurgensen KJ, Skinner WKJ, Oronsky B, Abrouk ND, Graff AE, Landes RD, Culp WE, Summers TA, Cary LH. RRx-001 Radioprotection: Enhancement of Survival and Hematopoietic Recovery in Gamma-Irradiated Mice. Front Pharmacol 2021; 12:676396. [PMID: 33967816 PMCID: PMC8100686 DOI: 10.3389/fphar.2021.676396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/06/2021] [Indexed: 01/03/2023] Open
Abstract
The present studies evaluate the in vivo prophylactic radioprotective effects of 1-bromoacetyl-3, 3-dinitroazetidine (RRx-001), a phase III anticancer agent that inhibits c-myc and downregulates CD-47, after total body irradiation (TBI), in lethally and sublethally irradiated CD2F1 male mice. A single dose of RRx-001 was administered by intraperitoneal (IP) injection 24 h prior to a lethal or sublethal radiation dose. When irradiated with 9.35 Gy, the dose lethal to 70% of untreated mice at 30 days (LD70/30), only 33% of mice receiving RRx-001 (10 mg/kg) 24 h prior to total body irradiation (TBI) died by day 30, compared to 67% in vehicle-treated mice. The same pretreatment dose of RRx-001 resulted in a significant dose reduction factor of 1.07. In sublethally TBI mice, bone marrow cellularity was increased at day 14 in the RRx-001-treated mice compared to irradiated vehicle-treated animals. In addition, significantly higher numbers of lymphocytes, platelets, percent hematocrit and percent reticulocytes were observed on days 7 and/or 14 in RRx-001-treated mice. These experiments provide proof of principle that systemic administration of RRx-001 prior to TBI significantly improves overall survival and bone marrow regeneration.
Collapse
Affiliation(s)
- Kimberly J Jurgensen
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States.,Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University, Bethesda, MD, United States
| | - William K J Skinner
- Department of Radiation Oncology, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | | | | | - Andrew E Graff
- Department of Radiation Oncology, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - William E Culp
- Director, Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Thomas A Summers
- Department of Pathology, Uniformed Services University, Bethesda, MD, United States
| | - Lynnette H Cary
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|