Randomized comparison of single dose of recombinant human IL-12 versus placebo for restoration of hematopoiesis and improved survival in rhesus monkeys exposed to lethal radiation

Zoya Gluzman-Poltorak, Sarita R Mendonca, Vladimir Vainstein, Hue Kha, Lena A Basile, Zoya Gluzman-Poltorak, Sarita R Mendonca, Vladimir Vainstein, Hue Kha, Lena A Basile

Abstract

Background: The hematopoietic syndrome of the acute radiation syndrome (HSARS) is a life-threatening condition in humans exposed to total body irradiation (TBI); no drugs are approved for treating this condition. Recombinant human interleukin-12 (rHuIL-12) is being developed for HSARS mitigation under the FDA Animal Rule, where efficacy is proven in an appropriate animal model and safety is demonstrated in humans.

Methods: In this blinded study, rhesus monkeys (9 animals/sex/dose group) were randomized to receive a single subcutaneous injection of placebo (group 1) or rHuIL-12 at doses of 50, 100, 250, or 500 ng/kg (groups 2-5, respectively), without antibiotics, fluids or blood transfusions, 24-25 hours after TBI (700 cGy).

Results: Survival rates at Day 60 were 11%, 33%, 39%, 39%, and 50% for groups 1-5, respectively (log rank p < 0.05 for each dose vs. control). rHuIL-12 also significantly reduced the incidences of severe neutropenia, severe thrombocytopenia, and sepsis (positive hemoculture). Additionally, bone marrow regeneration following TBI was significantly greater in monkeys treated with rHuIL-12 than in controls.

Conclusions: Data from this study demonstrate that a single injection of rHuIL-12 delivered one day after TBI can significantly increase survival and reduce radiation-induced hematopoietic toxicity and infections. These data significantly advance development of rHuIL-12 toward approval under the Animal Rule as an effective stand-alone medical countermeasure against the lethal effects of radiation exposure.

Figures

Figure 1
Figure 1
Survival of rhesus monkeys following exposure to TBI and treatment 24 hours after TBI with either vehicle or rHuIL-12. Kaplan-Meier plots of survival over the study period are shown for each treatment group. Each dose group comprised 18 animals. Log rank p-values were 0.0305 0.0344, 0.0404, and 0.0265, respectively for the 50 ng/kg, 100 ng/kg, 250 ng/kg and 500 ng/kg dose groups vs. the vehicle-treated control group.
Figure 2
Figure 2
Blood counts over time in rhesus monkeys exposed to lethal TBI and treated 24 hours after TBI with either vehicle or rHuIL-12 (Average ± SEM). A) platelets; B) mean platelet volume; C) neutrophils; D) lymphocytes; E) reticulocytes; F) red blood cells. Normal ranges are as follows: platelets, 252 to 612 × 109/L; mean platelet volume, 6.3 to 9.4 × 109/L; neutrophils, 1.21 to 10.29 × 109/L; lymphocytes, 1.85 to 8.71 × 109/L; reticulocytes, 29.9 to 103.9 × 109/L; red blood cells, 4.95 to 6.94 × 1012/L.
Figure 3
Figure 3
Identification of Bone Marrow Regeneration Islands. (A) Histopathological identification of regenerating bone marrow. Clusters of cells appearing in otherwise ablated bone marrow were scored as one regenerating island. Left panel, ablated bone marrow; middle panel, regenerating bone marrow; right panel, non-irradiated bone marrow. (Olympus BX41 compound microscope; Infinity Analyze software v5.0, magnification: 10×). (B) Quantification of number of islands of regeneration for individual treatment groups (left panel, p < 0.01 for 500 ng/kg group vs. control) and the combined rHUIL-12-treated groups vs. vehicle-treated control (right panel, p < 0.05). (C) Quantification of area of regeneration for individual treatment groups (left panel, p < 0.05 for 50 and 500 ng/kg groups vs. control) and the combined rHuIL-12-treated groups vs. vehicle-treated control (right panel, p < 0.05). (D) Quantification of megakaryocytes for individual treatment groups (left panel) and the combined rHuIL-12-treated groups vs. vehicle-treated control (right panel).

References

    1. Waselenko JK, MacVittie TJ, Blakely WF, Pesik N, Wiley AL, Dickerson WE, Tsu H, Confer DL, Coleman CN, Seed T, Lowry P, Armitage JO, Dainiak N. Medical management of the acute radiation syndrome: recommendations of the strategic national stockpile radiation working group. Ann Intern Med. 2004;140:1037–1051. doi: 10.7326/0003-4819-140-12-200406150-00015.
    1. DiCarlo AL, Maher C, Hick JL, Hanfling D, Dainiak N, Chao N, Bader JL, Coleman CN, Weinstock DM. Radiation injury after a nuclear detonation: medical consequences and the need for scarce resources allocation. Disaster Med Public Health Prep. 2011;5(Suppl 1):S32–44.
    1. Dainiak N, Waselenko JK, Armitage JO, MacVittie TJ, Farese AM. The hematologist and radiation casualties. Hematology Am Soc Hematol Educ Program. 2003;2003 no.1:473–496.
    1. Dumont F, Le Roux A, Bischoff P. Radiation countermeasure agents: an update. Expert Opin Ther Pat. 2010;20:73–101. doi: 10.1517/13543770903490429.
    1. Singh VK, Ducey EJ, Brown DS, Whitnall MH. A review of radiation countermeasure work ongoing at the armed forces radiobiology research institute. Int J Radiat Biol. 2012;88:296–310. doi: 10.3109/09553002.2012.652726.
    1. Drouet M, Herodin F. Radiation victim management and the haematologist in the future: time to revisit therapeutic guidelines? Int J Radiat Biol. 2010;86:636–648. doi: 10.3109/09553001003789604.
    1. Farese AM, Brown CR, Smith CP, Gibbs AM, Katz BP, Johnson CS, Prado KL, MacVittie TJ. The ability of filgrastim to mitigate mortality following LD50/60 total-body irradiation is administration time-dependent. Health Phys. 2014;106(1):39–47. doi: 10.1097/HP.0b013e3182a4dd2c.
    1. Farese AM, Cohen MV, Katz BP, Smith CP, Gibbs A, Cohen DM, MacVittie TJ. Filgrastim improves survival in lethally irradiated nonhuman primates. Radiat Res. 2013;179:89–100. doi: 10.1667/RR3049.1.
    1. Lindemann A, Herrmann F, Oster W, Haffner G, Meyenburg W, Souza LM, Mertelsmann R. Hematologic effects of recombinant human granulocyte colony-stimulating factor in patients with malignancy. Blood. 1989;74:2644–51.
    1. Azoulay E, Attalah H, Harf A, Schlemmer B, Delclaux C. Granulocyte colony-stimulating factor or neutrophil-induced pulmonary toxicity: myth or reality? Systematic review of clinical case reports and experimental data. Chest. 2001;120:1695–1701. doi: 10.1378/chest.120.5.1695.
    1. Basile LA, Ellefson D, Gluzman-Poltorak Z, Junes-Gill K, Mar V, Mendonca S, Miller JD, Tom J, Trinh A, Gallaher TK. HemaMax, a recombinant human interleukin-12, is a potent mitigator of acute radiation injury in mice and non-human primates. PLoS One. 2012;7:e30434. doi: 10.1371/journal.pone.0030434.
    1. Bellone G, Trinchieri G. Dual stimulatory and inhibitory effect of NK cell stimulatory factor/IL-12 on human hematopoiesis. J Immunol. 1994;153:930–937.
    1. Dybedal I, Larsen S, Jacobsen SE. IL-12 directly enhances in vitro murine erythropoiesis in combination with IL-4 and stem cell factor. J Immunol. 1995;154:4950–4955.
    1. Jacobsen SE, Veiby OP, Smeland EB. Cytotoxic lymphocyte maturation factor (interleukin 12) is a synergistic growth factor for hematopoietic stem cells. J Exp Med. 1993;178:413–418. doi: 10.1084/jem.178.2.413.
    1. Chen T, Burke KA, Zhan Y, Wang X, Shibata D, Zhao Y. IL-12 facilitates both the recovery of endogenous hematopoiesis and the engraftment of stem cells after ionizing radiation. Exp Hematol. 2007;35:203–213. doi: 10.1016/j.exphem.2006.10.002.
    1. Basile LA, Gallaher TK, Shibata D, Miller JD, Douer D. Multilineage hematopoietic recovery with concomitant antitumor effects using low dose Interleukin-12 in myelosuppressed tumor-bearing mice. J Transl Med. 2008;6:26. doi: 10.1186/1479-5876-6-26.
    1. Fliedner TM, Friesecke I, Beyrer K, Fliedner TM, Friesecke I, Beyrer K, editor. Medical measurements of radiation accidents. London, UK: The British Institute of Radiology; 2001.
    1. Trinchieri G. Interleukin-12: a cytokine at the interface of inflammation and immunity. Adv Immunol. 1998;70:83–243.
    1. Park HR, Jo SK, Paik SG. Factors effecting the Th2-like immune response after gamma-irradiation: low production of IL-12 heterodimer in antigen-presenting cells and small expression of the IL-12 receptor in T cells. Int J Radiat Biol. 2005;81:221–231. doi: 10.1080/09553000500077088.
    1. Park HR, Jo SK, Paik SG. The NK1.1 + T cells alive in irradiated mice play an important role in a Th1/Th2 balance. Int J Radiat Biol. 2006;82:161–170. doi: 10.1080/09553000600632873.
    1. Colombo MP, Trinchieri G. Interleukin-12 in anti-tumor immunity and immunotherapy. Cytokine Growth Factor Rev. 2002;13:155–168. doi: 10.1016/S1359-6101(01)00032-6.
    1. Williams JP, Brown SL, Georges GE, Hauer-Jensen M, Hill RP, Huser AK, Kirsch DG, Macvittie TJ, Mason KA, Medhora MM, Moulder JE, Okunieff P, Otterson MF, Robbins ME, Smathers JB, McBride WH. Animal models for medical countermeasures to radiation exposure. Radiat Res. 2010;173:557–578. doi: 10.1667/RR1880.1.
    1. Gerosa F, Baldani-Guerra B, Nisii C, Marchesini V, Carra G, Trinchieri G. Reciprocal activating interaction between natural killer cells and dendritic cells. J Exp Med. 2002;195:327–333. doi: 10.1084/jem.20010938.
    1. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 2003;3:133–146. doi: 10.1038/nri1001.
    1. Szabo SJ, Dighe AS, Gubler U, Murphy KM. Regulation of the interleukin (IL)-12R beta 2 subunit expression in developing T helper 1 (Th1) and Th2 cells. J Exp Med. 1997;185(5):817–24. doi: 10.1084/jem.185.5.817.

Source: PubMed

3
Tilaa