Partial marrow shielding and total-body irradiation

1963 ◽  
Vol 18 (3) ◽  
pp. 629-632 ◽  
Author(s):  
W. D. Noyes ◽  
C. A. Finch ◽  
H. Wasserman ◽  
K. Glickman

The effects of shielding one femur on bone marrow morphology and function were studied in rats receiving total-body irradiation. Animals were exposed to 550 r from a Co60 source. Shielding consisted of 100 mm of lead, reducing exposure to one femur to 16 r. Marrow cell counts, radioiron localization in femurs, plasma iron turnover, red cell radioiron utilization, and peripheral cell counts were observed. Phlebotomy in some animals served as a functional stress. There was no observed morphological change in shielded marrow despite marked depopulation of nucleated cells in heavily irradiated marrow. In phlebotomized shielded animals, the rate of hemoglobin synthesis was nearly half that of unirradiated controls although retarded by several days whereas in unshielded animals, hemoglobin synthesis was further delayed and less than a third of control values. Initially the erythropoietic function resides primarily in shielded marrow as evidenced by radioiron localization and marrow cell counts with no evidence of indirect depression. Cellular repopulation appears more rapidly in partially shielded animals and compares favorably with the effectiveness of marrow transfusion. Submitted on December 5, 1962

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4203-4203 ◽  
Author(s):  
Fatima S.F. Kaya-Aerts ◽  
Trudi P. Visser ◽  
Chris L. Reading ◽  
James M. Frincke ◽  
Monique M.A. Verstegen ◽  
...  

Abstract 5-androstene-3β,17β-diol, a naturally occurring adrenal steroid, has been shown to protect mice exposed to γ ray total body irradiation (TBI), as apparent by enhanced survival, stimulated myelopoiesis and elevated nadirs of neutrophils and platelets. In the present study, rhesus monkeys (Macaca mulatta; n=8) were subjected to the midlethal dose of 6 Gy 6 MV TBI at a dose rate of 35 cGy/minute, after which 4 randomly selected monkeys (2 males and 2 females) received 15 mg/kg IM of the testing compound HE2100 for 5 consecutive days, starting at 2 hours after irradiation. Placebo monkeys (also 2 males, 2 females) received the carrier IM for 5 days. TBI resulted in profound pancytopenia in all monkeys, which lasted 3 to 4 weeks in the placebo control monkeys. However, treatment with HE2100 reduced the period of severe neutropenia, with an 8 days earlier neutrophil recovery to levels >0.5x109/L than the placebo group, i.e. 12.8 days (range 11–15) vs 20.8 days (range 17–22), respectively. The neutrophil recovery was also reflected by accelerated recovery of CD11b+ cells. CD16/56+NK cells trailed the neutrophils. Recovery of reticulocytes was markedly enhanced in the HE2100 group to reach levels of >1% in peripheral blood (PB) by day 18.8 (range 18–20), whereas placebo control monkeys did not reach this level until day 26.3 (range 23–32). Reticulocyte reconstitution was accompanied by a transient erythroblastosis in HE2100 monkeys, emphasizing the pronounced stimulation of red cell reconstitution. A most prominent effect of HE2100 was noted for platelet recovery, since HE2100 effectively counteracted the need for transfusions, with only 1 (range 0–2) transfusion needed to maintain platelets at the used high threshold level of >40x109/L as apposed to 4 (range 2–8) in the placebo controls. The reconstitution of PB cells was preceded by a markedly accelerated recovery of immature bone marrow (BM) cells in HE2100 treated monkeys. At 2 weeks post-TBI, numbers of BM CD34+ cells approached pre-irradiation levels, whereas placebo monkeys did not reach such levels before the 4th week after TBI. Consequently, CD34+ cells in BM of HE2100 monkeys differed as much as 2 logs from those in placebo controls at 2 weeks after TBI, which was also reflected in the clonogenic progenitor cells GM-CFU and BFU-E, in accelerated normalization of BM cellularity and in PB CD34+ levels. Direct local or systemic toxic effects were not observed during administration of the steroid, but HE2100 monkeys displayed an increase of, on average, 10% in body weight due to fluid retention starting in the 2nd week after TBI, resulting in transient edema, which resolved without sequelae. This preclinical study characterizes HE2100 as a most potent novel agent to promote stem cell reconstitution and multilineage myelopoiesis after cytoreductive radiation exposure, resulting in enhanced reticulocyte, neutrophil and platelet recovery and profoundly decreased needs for transfusions and antibiotic treatment. The mechanisms involved are subject of further analyses.


Blood ◽  
2012 ◽  
Vol 120 (13) ◽  
pp. 2600-2609 ◽  
Author(s):  
Maegan L. Capitano ◽  
Michael J. Nemeth ◽  
Thomas A. Mace ◽  
Christi Salisbury-Ruf ◽  
Brahm H. Segal ◽  
...  

Abstract Neutropenia is a common side effect of cytotoxic chemotherapy and radiation, increasing the risk of infection in these patients. Here we examined the impact of body temperature on neutrophil recovery in the blood and bone marrow after total body irradiation (TBI). Mice were exposed to either 3 or 6 Gy TBI followed by a mild heat treatment that temporarily raised core body temperature to approximately 39.5°C. Neutrophil recovery was then compared with control mice that received either TBI alone heat treatment alone. Mice that received both TBI and heat treatment exhibited a significant increase in the rate of neutrophil recovery in the blood and an increase in the number of marrow hematopoietic stem cells and neutrophil progenitors compared with that seen in mice that received either TBI or heat alone. The combination treatment also increased G-CSF concentrations in the serum, bone marrow, and intestinal tissue and IL-17, IL-1β, and IL-1α concentrations in the intestinal tissue after TBI. Neutralizing G-CSF or inhibiting IL-17 or IL-1 signaling significantly blocked the thermally mediated increase in neutrophil numbers. These findings suggest that a physiologically relevant increase in body temperature can accelerate recovery from neutropenia after TBI through a G-CSF–, IL-17–, and IL-1–dependent mechanism.


2009 ◽  
Vol 85 (12) ◽  
pp. 1089-1100 ◽  
Author(s):  
John E. Baker ◽  
Brian L. Fish ◽  
Jidong Su ◽  
Steven T. Haworth ◽  
Jennifer L. Strande ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document