The mechanism of cytokine-induced shock remains poorly understood. The combination of IL-2 and IL-12 has synergistic antitumor activity in vivo, yet has been associated with significant toxicity. We examined the effects of IL-2 plus IL-12 in a murine model and found that the daily, simultaneous administration of IL-2 and IL-12 resulted in shock and 100% mortality within 4 to 12 days depending on the strain employed. Mice treated with IL-2 plus IL-12 exhibited NK cell apoptosis, pulmonary edema, degenerative lesions of the gastrointestinal tract, and elevated serum levels of proinflammatory cytokines and acute phase reactants. The actions of TNF-α, IFN-γ, macrophage-inflammatory protein-1α, IL-1, IL-1-converting enzyme, Fas, perforin, inducible nitric oxide synthase, and STAT1 did not contribute to the observed toxicity, nor did B or T cells. However, toxicity and death from treatment with IL-2 plus IL-12 could be completely abrogated by elimination of NK cells. These results suggest that the fatal systemic inflammatory response induced by this cytokine treatment is critically dependent upon NK cells, but does not appear to be mediated by the known effector molecules of this cellular compartment. These data may provide insight into the pathogenesis of cytokine-induced shock in humans.

Activation of monocytes/macrophages by bacteria, fungi, viruses, or their products results in the rapid production of monokines such as TNF-α, IL-1, IL-12, IL-15, and IL-18, which in turn induce NK cell production of IFN-γ and TNF-α (1, 2, 3, 4). IL-12 appears to be pivotal to the NK cell response, as there is only modest NK cell cytokine production in the absence of this factor (5, 6). We have demonstrated previously that the combination of IL-15 and IL-12 exerts a profound synergy upon resting NK cell production of IFN-γ, TNF-α, and MIP-1α3 (6, 7, 8). These proinflammatory cytokines and chemokines play a critical role in the clearance of obligate intracellular pathogens and, in some cases, the promotion of sepsis, shock, and death (9). An identical profile of NK cell cytokine and chemokine production can be induced by the combination of IL-2 and IL-12 (5, 6). This reflects the fact that the heterotrimeric IL-15R and IL-2R share the IL-2Rβ and γ signaling subunits and differ only in the specificity of their high affinity α-chains (10). The ability to obtain synergistic immunomodulatory effects via activation of the IL-2/15R and the IL-12R expressed on NK cells has led to investigations of this cytokine combination for the immunotherapy of neoplastic disease (11, 12). In the current study, we examined the effects of administering IL-2 or IL-15 in combination with IL-12 in a murine toxicity model. While the dose of the individual cytokines was well tolerated, the administration of IL-2 or IL-15 in combination with IL-12 induced a lethal systemic inflammatory response that did not require any of the major proinflammatory factors or signaling pathways felt to be active in the induction of septic shock. However, the lethal toxicity of this cytokine combination therapy was critically dependent upon NK cells, but not B or T cells.

Purified, yeast-derived rhuIL-2 (Chiron, Emeryville, CA) or rhuIL-15 (Immunex, Seattle, WA) was administered at a dose of 3 × 105 U/day via the i.p. route. rIL-12 of murine (mu) origin (Genetics Institute, Cambridge, MA) was administered i.p. at a dose of 1 μg/day. rhuIL-2 and rmuIL-12 were administered daily until the death of the animal. rmuIL-10 was supplied by Schering-Plough (Kenilworth, NJ). Platelet-derived huTGF-β1 (which has activity in murine systems) was purchased from R & D Systems (Minneapolis, MN) and reconstituted according to the manufacturer’s recommendations in 4 mM HCl supplemented with 0.1% BSA (13). Rat IgG1 anti-muIFN-γ mAb (Endogen, Cambridge, MA), and hamster IgG anti-muIFN-γ mAb (Genzyme Diagnostics, Cambridge, MA) were administered at a dose of 100 μg per mouse via the i.p. route 12 h before cytokine therapy and then daily thereafter. Control Abs were purchased from Sigma (St. Louis, MO). A dimeric rhuTNF receptor p80/IgG1 Fc fusion protein (TNFR-Fc; Immunex) was used in TNF-α neutralization experiments (100 μg/mouse/day i.p. beginning 24 h prior cytokine treatment) (14). Human IgG was used as a control for the fusion protein (Baxter, Glendale, CA). Dexamethasone was purchased from Moore Medical (New Britain, CT). Depletion of NK cells was accomplished via i.p. administration of an anti-asialo GM1 Ab (Wako BioProducts, Richmond, VA) every 3 days beginning 2 wk before the injection of cytokines (0.2 mg/mouse) (15). NK cell numbers postdepletion were evaluated via flow-cytometric analysis of murine splenocytes using a phycoerythrin (PE)-labeled pan-NK mAb (clone DX5; PharMingen, San Diego, CA). Mice were depleted of monocytes/macrophages via i.v. and i.p. injection of the F4/80 mAb (an IgG2b mAb) 48 and 24 h before cytokine therapy (16). Macrophage numbers postdepletion were evaluated via the enumeration of plastic-adherent cells following a 2-h culture of PBMCs, splenocytes, bone marrow cells, or peritoneal cells in 24-well plastic culture dishes (1 × 106 cells/well in RPMI 1640 supplemented with 10% FBS) (7). All cytokine reagents contained less than 0.015 EU/ml endotoxin, as measured by the E-Toxate system (Sigma).

Female mice age 4–6 wk were utilized in all experiments. C.B-17 scid/scid (SCID) mice (BALB/c background), splenectomized C.B-17 SCID mice, sham-operated C.B-17 SCID mice, and inbred BALB/c mice were purchased from Taconic Farms (Germantown, NY). Perforin-deficient (−/−) mice, CD3ε transgenic mice, IFN-γ−/− mice, and Faslpr/lpr mice were purchased from The Jackson Laboratory (Bar Harbor, ME) (17, 18, 19, 20). TNFR p55−/− mice, TNFR p75−/− mice, and TNFR p55−/−/TNFR p75−/− mice were provided by Immunex (21). Type I IL-1R−/− mice, type I IL-1R−/−/TNFR p55−/− mice, and type I IL-1R−/−/TNFR p75−/− mice were also provided by Immunex (22). TNFR p55−/− and Faslpr/lpr/TNFR p55−/− mice were produced by Dr. Klaus Pfeffer (Munich, Germany) (20, 23). MIP-1α−/− mice were the gift of Dr. O. Smithies (Chapel Hill, NC) (24). Mice deficient in the enzyme-inducible nitric oxide synthase (iNOS−/−) were the gift of Dr. Ricardo Gazzinelli (Bethesda, MD) (25). Mice deficient in the IL-1β-converting enzyme (ICE−/−) and STAT1−/− mice were produced as described (26, 27). All mice were housed in a specific pathogen-free environment and given food and water ad libitum.

Serum levels of IFN-γ and TNF-α were measured using ELISAs obtained from Endogen (Woburn, MA). IL-1β and IL-6 levels were measured using ELISAs obtained from Biosource International (Camarillo, CA). KC and MIP-2 levels were measured using ELISAs from R & D Systems. Serum chemistries were performed on mouse serum using a Vitros 500 analyzer (Johnson and Johnson, Raritan, NJ). Histopathologic examination of cytokine-treated SCID mice and determination of mean wet-to-dry lung ratios were performed as described (28). NK cells were isolated from the spleens of cytokine-treated C.B-17 SCID mice and analyzed for endonucleolytic cleavage of cellular DNA via a flow-cytometric assay using propidium iodide and DNA gel electrophoresis, as described (12, 29).

Serially diluted serum was analyzed for haptoglobin and α1-acid glycoprotein by immunoelectrophoresis (30). The area under the precipitation peak was quantitated in arbitrary units using the National Institute of Health Image program 1.61. The data for each peak were then converted into mg/ml values by comparison with the values obtained with calibrated mouse acute phase plasma.

An E1A-deleted recombinant adenovirus (rAd) expressing a dominant-negative form of the I-κBα protein was constructed (31). A rAd engineered to express the lacZ gene served as a control. A total of 109 PFUs of rAd/I-κB or rAd/lacZ was administered to mice via tail vein injection 48 h before treatment with IL-2 plus IL-12 (32). In vivo protein expression derived from these adenoviral vectors was confirmed via immunohistochemistry, as described (7).

Statistical significance was analyzed by the Student’s t test.

Intraperitoneal administration of rhuIL-2 (3 × 105 U/day) plus rmuIL-12 (1 μg/day) was lethal to C57BL/6 mice within 4 to 6 days (Fig. 1). The combination of IL-15 plus IL-12 elicited identical results, which was expected given that IL-15 signals through components of the IL-2R (6, 33). No deaths were observed in control mice receiving daily injections of IL-2, IL-15, or IL-12 alone. Similar results were obtained with IL-2 or IL-15 plus IL-12 in several different species of mice, including BALB/c, 129, B6 × 129, CD-1, and C.B-17 mice bearing the scid/scid (SCID) mutation (data not shown). Indeed, SCID mice, which lack B and T cells (34), exhibited 100% mortality within 3 to 5 days of the initiation of treatment and were utilized extensively in the majority of experiments.

FIGURE 1.

Administration of IL-2 or IL-15 in combination with IL-12 is lethal in inbred mice. Four- to six-week-old female C57BL/6 mice were injected daily i.p. with rhuIL-2 (3 × 105 IU/day) plus rmuIL-12 (1 μg/day), or rhuIL-15 (3 × 105 U/day) plus IL-12 and monitored for toxicity. No deaths were observed in mice receiving IL-2, IL-15, or IL-12 alone (not shown). These results are representative of four separate experiments.

FIGURE 1.

Administration of IL-2 or IL-15 in combination with IL-12 is lethal in inbred mice. Four- to six-week-old female C57BL/6 mice were injected daily i.p. with rhuIL-2 (3 × 105 IU/day) plus rmuIL-12 (1 μg/day), or rhuIL-15 (3 × 105 U/day) plus IL-12 and monitored for toxicity. No deaths were observed in mice receiving IL-2, IL-15, or IL-12 alone (not shown). These results are representative of four separate experiments.

Close modal

SCID mice receiving IL-2, IL-12, or IL-2 plus IL-12 were subjected to histopathologic evaluations. IL-2-associated findings included mononuclear cell infiltrates in the portal areas of the liver, splenic extramedullary hemopoiesis, and pulmonary interstitial mononuclear cell infiltrates. IL-12-associated changes included mild hyperplastic and degenerative changes of the gastrointestinal mucosa, and scattered foci of apoptotic lymphoid cells within the lymphoid organs. These IL-2- and IL-12-induced lesions were mild, of late onset (72–96 h), and consistent with those previously described for these cytokines (35, 36). Changes in SCID mice treated with IL-2 plus IL-12 included both significant exacerbation of IL-2- and IL-12-associated lesions as well as novel changes such as fibrinoid necrosis of lymphoid tissue in the spleen and lymph nodes, confluent foci of necrosis in pancreatic exocrine tissue, macrophage activation and proliferation, and fibrinoid necrosis of pulmonary arterioles. Apoptosis of SCID lymphocytes (i.e., NK cells) within the lymphoid tissues (spleen and lymph nodes) was markedly enhanced (Fig. 2,A). Analysis of nonadherent splenic NK cells from cytokine-treated SCID mice by DNA gel electrophoresis and propidium iodide staining (12) confirmed this observation (Fig. 2,B and data not shown). Lesions of the gastrointestinal tract associated with IL-12 administration were also markedly exacerbated by the combination of IL-2 and IL-12 (Fig. 2,C). Pulmonary pathology was prominent and included perivascular and septal mononuclear cell infiltrates associated with multifocal hemorrhage and alveolar edema. The formation of pulmonary edema at the 72-h time point was significantly greater in mice receiving the combination of IL-2 plus IL-12 (p < 0.05, Fig. 2,D) (28). Analysis of serum chemistries revealed significant increases in the liver enzymes ALT, AST, and LDH (5.3-, 9.9-, and 2.4-fold increases over baseline, respectively) as well as acute phase proteins beginning 24–48 h after treatment with IL-2 plus IL-12, but not following the administration of IL-2 or IL-12 alone (Fig. 2 E) (37).

FIGURE 2.

Histopathology and serum chemistries. A, Hemotoxylin/eosin-stained sections of spleens obtained from SCID mice receiving PBS (left panel) or IL-2 plus IL-12 (right panel) for 72 h. Apoptotic splenocytes were noted throughout the spleens of mice receiving IL-2 plus IL-12 (white arrow). Magnification, ×100. B, Nonadherent splenocytes (85–90% NK cells by FACS) from cytokine-treated SCID mice were harvested at 72 h and analyzed for the endonucleolytic cleavage of DNA via gel electrophoresis. Lane 1, ladder; lane 2, PBS-treated SCID mice; lane 3, IL-2- plus IL-12-treated mice; lane 4, IL-2-treated mice; lane 5, IL-12-treated mice. This experiment was repeated twice with similar results. C, Photomicrographs of colon obtained from SCID mice treated with PBS (A), IL-12 alone (B), or IL-2 plus IL-12 (C) for 72 h. Animals treated with IL-12 alone exhibited thinning of the mucosa, depletion of goblet cells, and multifocal single cell necrosis of colonic epithelial cells (arrow). Animals treated with IL-2 plus IL-12 exhibited atrophy of the mucosa, marked depletion of goblet cells, flattening of the surface epithelium, and cystic dilation of glands. D, SCID mice (n = 5 per group) received daily injections of PBS, IL-2 alone, IL-12 alone, or IL-2 plus IL-12 for 72 h. Mouse lungs were harvested, weighed, thoroughly dried, and then reweighed. Mean wet-to-dry lung ratios (±SEM) were calculated for each condition. ∗, Denotes a significant increase in the wet-to-dry lung ratio for mice receiving IL-2 plus IL-12 as compared with mice receiving PBS, IL-2 alone, or IL-12 alone (p < 0.05). E, SCID mice received daily injections of IL-2 alone, IL-12 alone, or IL-2 plus IL-12 for a period of 72 h. Serum was harvested at the indicated time points and analyzed for the presence of haptoglobin and α1-acid glycoprotein via immunoelectrophoresis. Induction of serum haptoglobin by IL-2 plus IL-12 was significantly greater than PBS, IL-2, or IL-12 alone at the 48-h time point (p < 0.002). Induction of serum of α1-acid glycoprotein by IL-2 plus IL-12 was significantly greater than PBS, IL-2, or IL-12 alone at the 48- and 72-h time points (p < 0.02).

FIGURE 2.

Histopathology and serum chemistries. A, Hemotoxylin/eosin-stained sections of spleens obtained from SCID mice receiving PBS (left panel) or IL-2 plus IL-12 (right panel) for 72 h. Apoptotic splenocytes were noted throughout the spleens of mice receiving IL-2 plus IL-12 (white arrow). Magnification, ×100. B, Nonadherent splenocytes (85–90% NK cells by FACS) from cytokine-treated SCID mice were harvested at 72 h and analyzed for the endonucleolytic cleavage of DNA via gel electrophoresis. Lane 1, ladder; lane 2, PBS-treated SCID mice; lane 3, IL-2- plus IL-12-treated mice; lane 4, IL-2-treated mice; lane 5, IL-12-treated mice. This experiment was repeated twice with similar results. C, Photomicrographs of colon obtained from SCID mice treated with PBS (A), IL-12 alone (B), or IL-2 plus IL-12 (C) for 72 h. Animals treated with IL-12 alone exhibited thinning of the mucosa, depletion of goblet cells, and multifocal single cell necrosis of colonic epithelial cells (arrow). Animals treated with IL-2 plus IL-12 exhibited atrophy of the mucosa, marked depletion of goblet cells, flattening of the surface epithelium, and cystic dilation of glands. D, SCID mice (n = 5 per group) received daily injections of PBS, IL-2 alone, IL-12 alone, or IL-2 plus IL-12 for 72 h. Mouse lungs were harvested, weighed, thoroughly dried, and then reweighed. Mean wet-to-dry lung ratios (±SEM) were calculated for each condition. ∗, Denotes a significant increase in the wet-to-dry lung ratio for mice receiving IL-2 plus IL-12 as compared with mice receiving PBS, IL-2 alone, or IL-12 alone (p < 0.05). E, SCID mice received daily injections of IL-2 alone, IL-12 alone, or IL-2 plus IL-12 for a period of 72 h. Serum was harvested at the indicated time points and analyzed for the presence of haptoglobin and α1-acid glycoprotein via immunoelectrophoresis. Induction of serum haptoglobin by IL-2 plus IL-12 was significantly greater than PBS, IL-2, or IL-12 alone at the 48-h time point (p < 0.002). Induction of serum of α1-acid glycoprotein by IL-2 plus IL-12 was significantly greater than PBS, IL-2, or IL-12 alone at the 48- and 72-h time points (p < 0.02).

Close modal

Serum levels of IFN-γ and TNF-α rose rapidly in SCID mice treated with IL-2 plus IL-12, peaked at approximately 24 h, and remained elevated until death (Fig. 3, A and B). Anti-asialo GM1 Ab-treated SCID mice did not exhibit elevated serum levels of IFN-γ or TNF-α after receiving IL-2 plus IL-12, suggesting that this cytokine combination acted directly on NK cells to induce production of IFN-γ and TNF-α (not shown). Serum levels of IL-1β, IL-6, KC, and MIP-2 were also found to be elevated during combined administration of IL-2 with IL-12 (summarized in Fig. 3,C). The elevated levels of proinflammatory cytokines seen with the combination of IL-2 plus IL-12 were not the result of a simple additive effect, since administration of IL-2 or IL-12 alone did not elicit significant cytokine production in SCID mice (with the exception of IFN-γ production in mice receiving IL-12). Of note, elevations in TNF-α and IL-1β occurred early in the course of treatment (i.e., within 1–3 h), whereas peak levels of IL-6, KC, and MIP-2 appeared later in the disease course, as is observed in animal models of septic shock (Fig. 3, B and C, and data not shown) (9, 38).

FIGURE 3.

Serum cytokine levels in SCID mice receiving daily injections of IL-2 plus IL-12. Four- to six-week-old female C.B-17 SCID mice received daily i.p. injections of PBS, IL-2, IL-12, or IL-2 plus IL-12. Serum was obtained from cytokine-treated mice at the indicated times and analyzed for the presence of muIFN-γ (A) and muTNF-α (B) by ELISA. These results are representative of three separate experiments and represent the mean ± SEM of duplicate wells. C, C.B-17 SCID mice received daily i.p. injections of PBS, IL-2, IL-12, or IL-2 plus IL-12. Serum was obtained from cytokine-treated mice at the indicated times and analyzed for the presence of IL-1β, IL-6, KC, and MIP-2 via ELISA. Values are presented as a percentage of maximal cytokine production (the following peak values were obtained: IL-1β = 101 pg/ml, IL-6 = 1037 pg/ml, KC = 5007 pg/ml, and MIP-2 = 59 pg/ml). Peak cytokine levels were less than 5% of maximum in mice receiving IL-2 or IL-12 alone (not shown). These results are representative of three separate experiments and represent the mean ± SEM of duplicate wells.

FIGURE 3.

Serum cytokine levels in SCID mice receiving daily injections of IL-2 plus IL-12. Four- to six-week-old female C.B-17 SCID mice received daily i.p. injections of PBS, IL-2, IL-12, or IL-2 plus IL-12. Serum was obtained from cytokine-treated mice at the indicated times and analyzed for the presence of muIFN-γ (A) and muTNF-α (B) by ELISA. These results are representative of three separate experiments and represent the mean ± SEM of duplicate wells. C, C.B-17 SCID mice received daily i.p. injections of PBS, IL-2, IL-12, or IL-2 plus IL-12. Serum was obtained from cytokine-treated mice at the indicated times and analyzed for the presence of IL-1β, IL-6, KC, and MIP-2 via ELISA. Values are presented as a percentage of maximal cytokine production (the following peak values were obtained: IL-1β = 101 pg/ml, IL-6 = 1037 pg/ml, KC = 5007 pg/ml, and MIP-2 = 59 pg/ml). Peak cytokine levels were less than 5% of maximum in mice receiving IL-2 or IL-12 alone (not shown). These results are representative of three separate experiments and represent the mean ± SEM of duplicate wells.

Close modal

We investigated the mechanism of this fatal cytokine-induced inflammatory response treatment using cytokine neutralization strategies and genetically altered mouse strains. Results are summarized in Table I. Neutralization of IFN-γ or TNF-α did not afford protection to SCID mice treated with IL-2 plus IL-12, nor did genetic deficiencies in IFN-γ or the TNFR complex (p55 and/or p75). We noted somewhat prolonged survival within both the control and experimental groups in our initial experiments with TNFR p55−/− mice. However, in subsequent experiments with the identical strain of TNFR p55−/− mouse and an independently generated strain, we found that all background mice and TNFR p55−/− mice died within 5 to 7 days (Table I and data not shown). These data in conjunction with the results of our TNF-α neutralization experiments led us to conclude that TNF-α was not solely responsible for the toxicity seen in this model. STAT1−/− mice were also susceptible to the toxicity of IL-2 plus IL-12, which is significant because STAT1 is critical for IFN-γ-induced gene regulation and also for the induction of apoptosis by TNF-α (39, 40). IFN-γ and TNF-α were simultaneously neutralized using both an anti-IFN-γ mAb and a TNFR-Fc soluble receptor construct (14, 41). Mortality rates of the experimental and control groups were identical. TNF-α was neutralized in IFN-γ−/− mice, and IFN-γ was neutralized in TNFR p55−/−/TNFR p75−/− mice, without effect. Type I IL-1R−/− mice, IL-1R−/−/TNFR p55−/− mice, and IL-1R−/−/TNFR p75−/− mice all succumbed to the lethal effects of IL-2 plus IL-12, whereas mice receiving IL-2 alone or IL-12 alone exhibited minimal toxicity (not shown). In addition, IL-1R−/−/TNFR p55−/− mice were treated with a neutralizing Ab to muIFN-γ during treatment with IL-2 plus IL-12, yet this intervention did not ameliorate toxicity or prolong survival (not shown). Several alternative effector molecules were considered as possible mediators of toxicity; however, mice with targeted genetic deficiencies in MIP-1α, ICE, perforin, Fas, and iNOS remained completely susceptible to the toxic effects of IL-2 plus IL-12.

Table I.

Role of IFN-γ, TNF-α, and IL-1β in the toxicity of IL-2 and IL-12a

Expt.StrainAb/ConstructMortalityTime to Death (days)
C.B-17 SCID Anti-IFN-γ Abb 12/12 2–5 
 C.B-17 SCID Control Ab 5/5 3–4 
IFN-γ−/−  10/10 3–4 
 C57BL/6c  10/10 4–5 
C.B-17 SCID Sol. TNFR-Fc 6/6 2–5 
 C.B-17 SCID HuIgG 6/6 2–5 
TNFR p55−/−  5/5 10–11 
 TNFR p75−/−  16/16 8–10 
 TNFR p55p75−/−  16/16 5–9 
 129c  8/8 6–12 
 C57BL/6c  8/8 5–11 
TNFR p55−/−d  5/5 3–5 
 C57BL/6c  5/5 5–7 
C.B-17 SCID Anti-IFN-γ Ab & Sol. TNFR-Fc 5/5 2–3 
 C.B-17 SCID Control Ab & HuIgG 5/5 3–4 
INF-γ−/− Sol. TNFR-Fc 6/6 4–5 
 IFN-γ−/− HuIgG 6/6 4–6 
TNFR p55p75−/− Anti-IFN-γ Ab 8/8 4–7 
 TNFR p55p75−/− Control Ab 8/8 5–9 
Type I IL-1R−/−  5/5 
 IL-1R/TNFR p55−/−  5/5 3–7 
 IL-1R/TNFR p75−/−  4/4 
 C57L/6c  5/5 
10 MIP-1α−/−  3/3 4–9 
 C57BL/6c  3/3 9–12 
11 ICE−/−  8/8 3–6 
 B6 × 129c  8/8 6–8 
12 Perforin−/−  8/8 5–9 
 C57BL/6c  8/8 6–12 
13 Fas/TNFR p55−/−  5/5 3–5 
 Fas−/−  5/5 3–7 
 TNFR p55−/−  5/5 3–5 
 C57BL/6c  5/5 5–7 
14 iNOS−/−  6/6 7–10 
 B6 × 129c  6/6 7–8 
15 STAT1−/−  6/6 5–6 
 C57BL/6c  6/6 5–6 
Expt.StrainAb/ConstructMortalityTime to Death (days)
C.B-17 SCID Anti-IFN-γ Abb 12/12 2–5 
 C.B-17 SCID Control Ab 5/5 3–4 
IFN-γ−/−  10/10 3–4 
 C57BL/6c  10/10 4–5 
C.B-17 SCID Sol. TNFR-Fc 6/6 2–5 
 C.B-17 SCID HuIgG 6/6 2–5 
TNFR p55−/−  5/5 10–11 
 TNFR p75−/−  16/16 8–10 
 TNFR p55p75−/−  16/16 5–9 
 129c  8/8 6–12 
 C57BL/6c  8/8 5–11 
TNFR p55−/−d  5/5 3–5 
 C57BL/6c  5/5 5–7 
C.B-17 SCID Anti-IFN-γ Ab & Sol. TNFR-Fc 5/5 2–3 
 C.B-17 SCID Control Ab & HuIgG 5/5 3–4 
INF-γ−/− Sol. TNFR-Fc 6/6 4–5 
 IFN-γ−/− HuIgG 6/6 4–6 
TNFR p55p75−/− Anti-IFN-γ Ab 8/8 4–7 
 TNFR p55p75−/− Control Ab 8/8 5–9 
Type I IL-1R−/−  5/5 
 IL-1R/TNFR p55−/−  5/5 3–7 
 IL-1R/TNFR p75−/−  4/4 
 C57L/6c  5/5 
10 MIP-1α−/−  3/3 4–9 
 C57BL/6c  3/3 9–12 
11 ICE−/−  8/8 3–6 
 B6 × 129c  8/8 6–8 
12 Perforin−/−  8/8 5–9 
 C57BL/6c  8/8 6–12 
13 Fas/TNFR p55−/−  5/5 3–5 
 Fas−/−  5/5 3–7 
 TNFR p55−/−  5/5 3–5 
 C57BL/6c  5/5 5–7 
14 iNOS−/−  6/6 7–10 
 B6 × 129c  6/6 7–8 
15 STAT1−/−  6/6 5–6 
 C57BL/6c  6/6 5–6 
a

Mice received daily injections of rhuIL-2 (3 × 105 IU/day) and rmuIL-12 (1 μg/day) via the i.p. route.

b

Similar results were obtained with a neutralizing Ab from a different commercial source.

c

Background strain.

d

Independently generated TNFR p55−/− strain (23 ).

Dexamethasone, TGF-β1, and ibuprofen have been used effectively to prevent morbidity and death in animal models of septic shock and other inflammatory processes (42, 43, 44). These agents were administered in pharmacologically relevant doses before the start of cytokine treatment and then daily thereafter; however, none was capable of ameliorating the toxicity of IL-2 plus IL-12 (Table II). Inhibition of NF-κB signaling in vivo via overexpression of I-κB in the liver was also ineffective in preventing mortality in this model, in contrast to its ability to protect mice from the lethal effects of endotoxin (45). Indeed, mice expressing the I-κB protein were actually more susceptible to the toxic effects of IL-2 plus IL-12 than were mice treated with the control vector (p < 0.05, Table II). This experiment was repeated in TNFR p55−/− mice because of the role I-κB has in potentiating TNF-mediated apoptosis (31, 46) (M. Karin, unpublished observation). The toxicity of IL-2 plus IL-12 in TNFR p55−/− mice overexpressing I-κB was essentially identical in the experimental and control groups (Table II).

Table II.

Effects of various treatments on the toxicity of IL-2 and IL-12 in SCID mice

Expt.StrainPretreatmentMortalityTime to Death (days)
C.B-17 SCID Dexa (2 mg/kg) 6/6 3–5 
 C.B-17 SCID Dex (6 mg/kg) 5/5 3–5 
 C.B-17 SCID PBS 5/5 3–5 
C.B-17 SCID TGF-βb (50 ng) 5/5 3–5 
 C.B-17 SCID TGF-β (100 ng) 5/5 3–5 
 C.B-17 SCID PBS 5/5 3–5 
C.B-17 SCID Ibuprofenc 6/6 3–5 
 C.B-17 SCID PBS 6/6 3–5 
C.B-17 SCID rAd/IκBd 8/8 
 C.B-17 SCID rAd/lacZ 8/8 4–5 
TNFR p55−/− rAd/IκB 8/8 4–8 
 TNFR p55−/− rAd/lacZ 8/8 5–9 
C.B-17 SCID muIL-10e (5 μg) 2/6 6–7 (n = 2) 
 C.B-17 SCID muIL-10 (20 μg) 2/6 6–7 (n = 2) 
 C.B-17 SCID PBS 6/6 3–4 
Expt.StrainPretreatmentMortalityTime to Death (days)
C.B-17 SCID Dexa (2 mg/kg) 6/6 3–5 
 C.B-17 SCID Dex (6 mg/kg) 5/5 3–5 
 C.B-17 SCID PBS 5/5 3–5 
C.B-17 SCID TGF-βb (50 ng) 5/5 3–5 
 C.B-17 SCID TGF-β (100 ng) 5/5 3–5 
 C.B-17 SCID PBS 5/5 3–5 
C.B-17 SCID Ibuprofenc 6/6 3–5 
 C.B-17 SCID PBS 6/6 3–5 
C.B-17 SCID rAd/IκBd 8/8 
 C.B-17 SCID rAd/lacZ 8/8 4–5 
TNFR p55−/− rAd/IκB 8/8 4–8 
 TNFR p55−/− rAd/lacZ 8/8 5–9 
C.B-17 SCID muIL-10e (5 μg) 2/6 6–7 (n = 2) 
 C.B-17 SCID muIL-10 (20 μg) 2/6 6–7 (n = 2) 
 C.B-17 SCID PBS 6/6 3–4 
a

Mice received daily i.p. injections of dexamethsone beginning 2 days prior to treatment with rhuIL-2 plus rmuIL-12.

b

Mice received daily i.p. injections of TGF-β1 beginning 12 h prior to treatment with IL-2 plus IL-12.

c

Ibuprofen was administered via dietary supplementation (∼2 mg/day) beginning 3 days prior to treatment with IL-2 plus IL-12.

d

rAd was administered via tail vein injeciton 48 h prior to treatment with IL-2 plus IL-12.

e

Mice received daily i.p. injections of muIL-10 beginning 2 days prior to treatment with IL-2 plus IL-12.

SCID mice that lack T and B lymphocytes undergo massive NK cell apoptosis following administration of IL-2 plus IL-12 (Fig. 2, A and B). To determine whether the toxicity of this model was mediated by NK cells, we administered IL-2 plus IL-12 to SCID mice depleted of NK cells by pretreatment with an anti-asialo GM1 Ab (Fig. 4,A). IL-2 (or IL-15) plus IL-12 elicited minimal toxicity when administered to SCID mice depleted of NK cells, and 100% of mice in this group survived, whereas control mice receiving IL-2 plus IL-12 all died within 5 days of the initiation of treatment (Fig. 5,A). To confirm this observation, we administered IL-2 and IL-12 to SCID mice that had been depleted of NK cells by splenectomy and observed 100% survival (47). Sham-operated SCID mice treated with IL-2 plus IL-12 exhibited 100% mortality at 5 days (Fig. 5,B). Furthermore, CD3ε transgenic mice that completely lack mature NK cells (and T cells) due to a developmental block (18) showed absolutely no toxicity when treated with IL-2 plus IL-12. Control mice of the appropriate background all died between 4 and 8 days (Fig. 5 C).

FIGURE 4.

Analysis of NK cell and macrophage depletion following the administration of Ab. A, Splenocytes were isolated from the spleens of SCID mice that had received injections of PBS (left-hand panel) or an anti-asialo GM1 Ab (right-hand panel). NK cell numbers were evaluated via flow-cytometric analysis using a PE-labeled pan-NK mAb. The fluorescence of splenocytes stained with a PE-labeled isotype control Ab fell within the first log (not shown). These results are representative of three separate determinations. B, PBMCs, splenocytes, bone marrow cells, and peritoneal cells were isolated from SCID mice that had received injections of the F4/80 mAb or a control Ab. These cell preparations were resuspended in RPMI 1640 media supplemented with 10% FBS and plated at a density of 1 × 106 cells/well in 24-well plastic culture dishes. Following a 2-h culture, nonadherent cells were removed and the number of adherent cells was determined (7).

FIGURE 4.

Analysis of NK cell and macrophage depletion following the administration of Ab. A, Splenocytes were isolated from the spleens of SCID mice that had received injections of PBS (left-hand panel) or an anti-asialo GM1 Ab (right-hand panel). NK cell numbers were evaluated via flow-cytometric analysis using a PE-labeled pan-NK mAb. The fluorescence of splenocytes stained with a PE-labeled isotype control Ab fell within the first log (not shown). These results are representative of three separate determinations. B, PBMCs, splenocytes, bone marrow cells, and peritoneal cells were isolated from SCID mice that had received injections of the F4/80 mAb or a control Ab. These cell preparations were resuspended in RPMI 1640 media supplemented with 10% FBS and plated at a density of 1 × 106 cells/well in 24-well plastic culture dishes. Following a 2-h culture, nonadherent cells were removed and the number of adherent cells was determined (7).

Close modal
FIGURE 5.

Death induced by administration of IL-2 plus IL-12 is critically dependent upon the NK cell compartment. A, C.B-17 SCID mice were depleted of NK cells by pretreatment with an anti-asialo GM1 Ab (see Materials and Methods). Control mice were pretreated with injections of PBS. Mice in both groups subsequently received daily i.p. injections of rhuIL-2 (3 × 105 IU/day) plus rmuIL-12 (1 μg/day) and were monitored for survival. This experiment was repeated three times with similar results. B, Splenectomized SCID mice or sham-operated controls received daily i.p. injections of IL-2 and IL-12. This experiment was repeated twice with similar results. C, Transgenic mice expressing the CD3ε subunit of the human TCR received daily injections of IL-2 and IL-12 via the i.p. route. Normal mice of the identical background served as controls. This experiment was repeated twice with similar results. D, SCID mice were partially depleted of macrophages by pretreatment with the F4/80 mAb. Control mice were pretreated with injections of a control mAb. Mice in both groups subsequently received daily i.p. injections of IL-2 plus IL-12 and were monitored for survival. This experiment was repeated twice with similar results. All experiments depicted in this utilized at least six mice per group.

FIGURE 5.

Death induced by administration of IL-2 plus IL-12 is critically dependent upon the NK cell compartment. A, C.B-17 SCID mice were depleted of NK cells by pretreatment with an anti-asialo GM1 Ab (see Materials and Methods). Control mice were pretreated with injections of PBS. Mice in both groups subsequently received daily i.p. injections of rhuIL-2 (3 × 105 IU/day) plus rmuIL-12 (1 μg/day) and were monitored for survival. This experiment was repeated three times with similar results. B, Splenectomized SCID mice or sham-operated controls received daily i.p. injections of IL-2 and IL-12. This experiment was repeated twice with similar results. C, Transgenic mice expressing the CD3ε subunit of the human TCR received daily injections of IL-2 and IL-12 via the i.p. route. Normal mice of the identical background served as controls. This experiment was repeated twice with similar results. D, SCID mice were partially depleted of macrophages by pretreatment with the F4/80 mAb. Control mice were pretreated with injections of a control mAb. Mice in both groups subsequently received daily i.p. injections of IL-2 plus IL-12 and were monitored for survival. This experiment was repeated twice with similar results. All experiments depicted in this utilized at least six mice per group.

Close modal

Given the ability of NK cell-derived cytokines to potentiate macrophage effector functions (1, 2, 3, 48), the role of macrophages in the toxicity of this model was investigated. SCID mice were depleted of monocytes and macrophages by approximately 50% in the peripheral blood, spleen, and bone marrow, and by >95% in the peritoneal cavity by injecting the F4/80 mAb (16) via the i.v. and i.p. routes 48 and 24 h before the administration of IL-2 plus IL-12 (Fig. 4,B). Mice receiving the F4/80 mAb tolerated the administration of IL-2 plus IL-12 significantly better than mice receiving the control Ab and exhibited a 50% survival rate (p < 0.05). Control mice all died within 3 to 4 days (Fig. 5,D). IL-10 is a potent macrophage deactivator (49). Pretreatment of SCID mice with rmuIL-10 afforded significant protection from the toxicity of IL-2 plus IL-12 (66% survival overall, p < 0.02, Table II). Taken together, these data suggest that monocytes/macrophages, in addition to NK cells, have a role in mediating the lethal toxicity of IL-2 plus IL-12.

Administration of IL-12 in combination with IL-2 or IL-15 induced a systemic inflammatory response that rapidly progressed to a fatal shocklike state. The NK cell compartment was the only lymphocyte population responsible for mediating this toxicity because mice that underwent depletion of NK cells or had congenital absence of NK cells were completely protected from the lethal effects of this cytokine combination. Treatment of SCID mice with IL-2 plus IL-12 resulted in high serum levels of IFN-γ and TNF-α, and production of these cytokines was dependent upon the presence of NK cells. However, neither IFN-γ nor TNF-α was required for the fatal inflammatory reaction induced by IL-2 plus IL-12. Other effector molecules of the NK cell and macrophage compartments, namely MIP-1α, IL-1, ICE, Fas, perforin, iNOS, and the STAT1 pathway of signal transduction, were eliminated as critical mediators of toxicity in this model.

Our histopathologic findings and analysis of serum cytokine levels indicated that the administration of IL-2 plus IL-12 had induced a severe systemic inflammatory response. High circulating levels of IL-1β, IL-6, and the IL-8 homologues (KC and MIP-2) appeared only in the serum of mice receiving the combination of IL-2 plus IL-12, which is significant in that the production of these cytokines is limited to periods of inflammation, tissue injury, and immunologic challenge (38). Indeed, the sequence of cytokine induction was highly reminiscent of that observed in experimental models of endotoxemia and in humans diagnosed with septic shock (9, 38). The presence of high serum levels of IL-6 at the time of death is particularly significant in that they represent the net effect of biologically active IL-1β and TNF-α, and have been found to correlate inversely with survival in patients with septic shock (50). Thus, treatment of mice with IL-2 plus IL-12 induces many of the same proinflammatory mediators that are active in sepsis. The presence of increased pulmonary edema, multiple organ system toxicities, and an acute phase response in mice receiving the combination of IL-2 and IL-12 lends additional support to this observation (9, 51). Induction of proinflammatory mediators has also been implicated in the pathogenesis of the shocklike states associated with high dose cytokine therapy in humans, and we initially hypothesized that the lethal reaction to IL-2 plus IL-12 might be the result of additive toxicities induced by the overlapping actions of known proinflammatory factors (52, 53). However, no protection was afforded by simultaneous neutralization of TNF-α and IFN-γ or utilization of IL-1R−/−/TNFR p55−/− and IL-1R−/−/TNFR p75−/− mice. The data also suggest that the fatal toxicity of IL-2 plus IL-12 was not mediated via NF-κB signaling within the liver; however, it is possible that NF-κB was only partially inhibited and therefore still able to activate the transcription of NF-κB-responsive genes during this intense inflammatory response (45). We cannot exclude the possibility that other cytokines, cytokine receptors, or effector molecules might combine with IL-1, TNF-α, or IFN-γ following administration of IL-2 plus IL-12 to induce a lethal inflammatory response. However, the data suggest that stimulation of the NK cell compartment with IL-2 plus IL-12 might result in the production of novel factors or factors that have yet to be characterized as proinflammatory. Such a hypothesis is supported by our characterization of several unique pathologic lesions that have not been observed in other models of shock (9, 51).

The appearance of IL-1β, IL-6, and other macrophage-derived cytokines in the circulation, and the presence of activated and proliferating macrophages in the splenic bed following the administration of IL-2 plus IL-12 implied that this treatment had directly or indirectly activated the macrophage compartment (54). The importance of macrophages in the toxicity of this model was confirmed by studies in which partial depletion of the macrophage compartment resulted in a 50% survival rate for mice receiving IL-2 plus IL-12. The protection afforded by IL-10 pretreatments also suggested a role for the macrophage compartment in the toxicity of this model; however, we cannot rule out the possibility that IL-10 may exert its protective effects via some other pathway (49). The prevention of death by the congenital absence or depletion of NK cells and the improved survival by the partial depletion of macrophages suggested that the toxicity of this model is the result of interactions between these two cell populations. NK cells constitutively express the IL-2/15R, and activation of this receptor complex results in further up-regulation of NK cell IL-12R expression (55). Thus, we suspect that the toxicity of this model first involves stimulation of NK cells by IL-2/15 plus IL-12, followed by an NK cell-dependent activation of macrophages. The inability of IL-2 plus IL-12 administration to induce TNF-α in NK-depleted SCID mice also supports our proposed sequence of events, as NK cells and macrophages are both sources of this cytokine. Thus, mobilization of macrophage effector function may occur as a result of continuous and uncontrolled production of NK cell factors in response to IL-2 plus IL-12 (2), although the precise cellular source of these proinflammatory cytokines cannot be determined based upon measurements of serum cytokines. Our postulate that chronic stimulation of the NK cell compartment via daily injection of IL-2 plus IL-12 results in macrophage activation is not an unprecedented one (1, 2, 56, 57). However, the inability of IFN-γ and TNF-α neutralization strategies to prevent death was unexpected and suggests that NK cells may be able to activate macrophage effector functions via alternate pathways.

To the best of our knowledge, the present model represents the first example of a cytokine-induced shock syndrome that is mediated by components of the innate immune system. Our current understanding of the role of the NK cell in other models of shock is quite limited, but there is evidence that NK cells can be activated during septic events and may contribute to the pathogenesis of this condition via the secretion of IFN-γ, which acts primarily to augment macrophage function. One well-studied example of this phenomena is the Shwartzman reaction in which an intradermal priming dose of LPS is followed 24 h later by an i.v. LPS challenge (58). The first LPS dose induces the production of IL-12, which stimulates the release of IFN-γ (presumably by NK cells) and permits the priming of macrophages. Upon subsequent LPS challenge, sensitized macrophages release massive amounts of TNF-α and IL-1β, which mediate the lethal effects of this treatment (58). Heremans et al. have demonstrated that depletion of NK cells before the induction of the generalized Shwartzman reaction leads to a 70% reduction in mortality and significantly lower levels of IFN-γ and TNF-α following the systemic injection of LPS (59). In contrast, mice depleted of CD4+ or CD8+ T cells were still highly susceptible to the lethal effects of the Shwartzman reaction. Ozmen et al. analyzed the Shwartzman reaction in detail and found that simple coinjection of TNF-α plus IL-1β or TNF-α plus IFN-γ was sufficient to induce lethality following priming of mice with IL-12 or IFN-γ alone (60). However, regardless of the priming event, no mortality was observed if IL-1β and IFN-γ were administered in the absence of TNF-α. Thus, in distinct contrast to our model, TNF-α is crucial for the lethality of the Shwartzman reaction. Furthermore, our experiments with IFN-γ−/− and STAT1−/− mice would suggest that IFN-γ priming (following IL-12 administration) is not a critical event in our model.

The induction of NK cell apoptosis in the present model was dependent upon the coadministration of IL-2 with IL-12. Nonadherent splenocytes from cytokine-treated SCID mice did not exhibit endonucleosomal DNA degradation or the morphologic features of apoptosis unless mice had received both cytokines, results that were confirmed in vitro (not shown). Apoptosis of NK cells has also been observed following coactivation with IL-2 and engagement of the low affinity FcRγ (or cross-linking of CD94), and after cell-mediated lysis of leukemic targets cells in the presence of IL-2 (61, 62, 63, 64). We have demonstrated previously that in vitro stimulation of resting human NK cells with IL-2/15 and IL-12 leads to the production of large amounts of IFN-γ and TNF-α, followed by the induction of programmed cell death at approximately 48–72 h (12). Activation-induced apoptosis appears to be a common strategy for the removal of activated effector cells and attenuation of the immune response to specific pathogens (65). The ability of IL-2 plus IL-12 to induce NK cell apoptosis in vivo is further evidence for the central role of the NK cell compartment in the toxicity of this treatment. The existence of this suicide pathway following NK cell activation further suggests that unbridled amplification of NK cell effector functions may be deleterious to the organism. This apoptotic regulatory mechanism was apparently insufficient to limit toxicity in the present model, possibly due to the intensity and rapidity of the NK cell response to administration of IL-2/15 plus IL-12. Activation-induced apoptosis associated with the induction of a shocklike state has also been described following experimental activation of T cells with staphylococcal enterotoxins (superantigens) or anti-CD3 Ab (66, 67). However, in contrast to our model, the toxicity of these T cell-mediated shock syndromes is mediated primarily by TNF-α.

In summary, we have demonstrated that the administration of IL-2 or IL-15 in combination with IL-12 results in a fatal systemic inflammatory response that is critically dependent upon the NK cell compartment. This lethal shocklike reaction does not appear to be mediated by any of the known cytokine products of the NK cell compartment, nor any of the effector molecules associated with NK cell cytotoxic activity. This is the first time that the NK cell (or any cellular compartment) has been identified as an important component of cytokine-induced systemic inflammation. Elucidation of the factors involved in this novel inflammatory pathway may therefore have relevance for understanding the complications of high dose cytokine therapy as well as other forms of shock.

We thank Dr. Ricardo Gazzinelli for his assistance with experiments involving the inducible nitric oxide synthase-deficient murine strain, and Dr. Peter Kantor for analysis of serum chemistries.

1

This work was supported by National Institute of Health Grants CA68326 and CA68458, and in part by Grant P30 CA16058.

3

Abbreviations used in this paper: MIP, macrophage-inflammatory protein; hu, human; I-κB, inhibitor of NF-κB; ICE, IL-1β-converting enzyme; iNOS, inducible nitric oxide synthase; KC, ??; mu, murine; PE, phycoerythrin; rAd, recombinant adenovirus.

1
Bancroft, G. J..
1993
. The role of natural killer cells in innate resistance to infection.
Curr. Opin. Immunol.
5
:
503
2
Orange, J. S., B. Wang, C. Terhorst, C. A. Biron.
1995
. Requirement for natural killer cell-produced interferon γ in defense against murine cytomegalovirus infection and enhancement of this defense pathway by interleukin 12 administration.
J. Exp. Med.
182
:
1045
3
Fearon, D. T., R. M. Locksley.
1996
. The instructive role of innate immunity in the acquired immune response.
Science
272
:
50
4
Ahn, H. J., S. Maruo, M. Tomura, J. Mu, T. Hamaoka, K. Nakanishi, S. Clark, M. Kurimoto, H. Okamura, H. Fujiwara.
1997
. A mechanism underlying synergy between IL-12 and IFN-γ-inducing factor in enhanced production of IFN-γ.
J. Immunol.
159
:
2125
5
Chan, S. H., B. Perusia, J. W. Gupta, M. Kobayashi, M. Popisil, H. A. Young, S. F. Wolf, D. Young, S. C. Clark, G. Trinchieri.
1991
. Induction of IFN-γ production by NK cell stimulatory factor (NKSF): characterization of the responder cells and synergy with other inducers.
J. Exp. Med.
173
:
869
6
Carson, W. E., J. G. Giri, M. J. Lindemann, M. L. Linett, M. Ahdieh, D. Anderson, J. Eisenmann, K. Grabstein, M. A. Caligiuri.
1994
. Interleukin-15 is a novel cytokine which activates human natural killer cells via components of the interleukin-2 receptor.
J. Exp. Med.
180
:
1395
7
Carson, W. E., M. E. Ross, R. A. Baiocchi, M. J. Marien, N. Boiani, K. Grabstein, M. A. Caligiuri.
1995
. Endogenous production of interleukin 15 by activated human monocytes is critical for optimal production of interferon-γ by natural killer cells in vitro.
J. Clin. Invest.
96
:
2578
8
Bluman, E., K. J. Bartynski, B. R. Avalos, M. A. Caligiuri.
1996
. Human natural killer cells produce abundant macrophage inflammatory protein-1α in response to monocyte-derived cytokines.
J. Clin. Invest.
97
:
2722
9
Hack, C. E., L. A. Aarden, L. G. Thijs.
1997
. Role of cytokines in sepsis.
Adv. Immunol.
66
:
101
10
Anderson, D. A., S. Kumaki, M. Ahdieh, J. Bertles, M. Tometsko, A. Loomis, J. Giri, N. G. Copeland, D. J. Gilbert, N. A. Jenkins, V. Valentine, D. N. Shapiro, S. W. Morris, L. S. Park, D. Cosman.
1995
. Functional characterization of the human interleukin-15 receptor chain and close linkage of IL15RA and IL2RA genes.
J. Biol. Chem.
270
:
29862
11
Wigginton, J. M., K. L. Komschlies, T. C. Back, J. L. Franco, M. J. Brunda, R. H. Wiltrout.
1996
. Administration of interleukin 12 with pulse interleukin 2 and the rapid and complete eradication of murine renal carcinoma.
J. Natl. Cancer Inst.
88
:
38
12
Ross, M. E., M. A. Caligiuri.
1997
. Cytokine-induced apoptosis of human natural killer cells identifies a novel mechanism to regulate the innate immune response.
Blood
89
:
1910
13
Su, H. C., R. Ishikawa, C. A. Biron.
1993
. Transforming growth factor-β expression and natural killer cell responses during virus infection of normal, nude, and SCID mice.
J. Immunol.
151
:
4874
14
Mohler, K. M., D. S. Torrance, C. A. Smith, R. G. Goodwin, K. E. Stremler, V. P. Fung, H. Madain, M. B. Widmer.
1993
. Soluble tumor necrosis factor (TNF) receptors are effective therapeutic agents in lethal endotoxemia and function simultaneously as both TNF carriers and TNF antagonists.
J. Immunol.
151
:
1548
15
Baiocchi, R. A., M. A. Caligiuri.
1994
. Low-dose interleukin 2 prevents the development of Epstein-Barr virus (EBV)-associated lymphoproliferative disease in scid/scid mice reconstituted i.p. with EBV-seropositive human peripheral blood lymphocytes.
Proc. Natl. Acad. Sci. USA
91
:
5577
16
Salazar-Mather, T. P., R. Ishikawa, C. A. Biron.
1996
. NK cell trafficking and cytokine expression in splenic compartments after IFN induction and viral infection.
J. Immunol.
157
:
3054
17
Kagi, D., B. Ledermann, K. Burki, P. Seiler, B. Odermatt, K. J. Olsen, E. R. Podack, R. M. Zinkernagel, H. Hengartner.
1994
. Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-deficient mice.
Nature
369
:
31
18
Wang, B., C. Biron, J. She, K. Higgins, M. J. Sunshine, E. Lacy, N. Lonberg, C. A. Terhorst.
1994
. Block in both early T lymphocyte and natural killer cell development in transgenic mice with high-copy numbers of the human CD3E gene.
Proc. Natl. Acad. Sci. USA
91
:
9402
19
Dalton, D. K., S. Pitts-Meek, S. Keshav, I. S. Figari, A. Bradley, T. A. Stewart.
1993
. Multiple defects of immune cell function in mice with disrupted interferon-γ genes.
Science
259
:
1739
20
Watanabe-Fukunaga, R., C. I. Brannan, N. G. Copeland, N. A. Jenkins, S. Nagata.
1992
. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis.
Nature
356
:
314
21
Peschon, J. J., D. S. Torrance, K. L. Stocking, M. B. Glaccum, C. Otten, C. R. Willis, K. Charrier, P. J. Morrisey, C. B. Ware, K. M. Mohler.
1998
. TNF receptor-deficient mice reveal divergent roles for p55 and p75 in several models of inflammation.
J. Immunol.
160
:
943
22
Glaccum, M. B., K. L. Stocking, K. Charrier, J. L. Smith, C. R. Willis, C. Maliszewski, D. J. Livingston, J. J. Peschon, P. J. Morrissey.
1997
. Phenotypic and functional characterization of mice that lack the type I receptor for IL-1.
J. Immunol.
159
:
3364
23
Pfeffer, K., T. Matsuyama, T. M. Kundig, A. Wakeham, K. Kishira, A. Shahinian, K. Wiegmann, P. S. Ohashi, M. Kronke, T. W. Mak.
1993
. Mice deficient for the 55kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L.
monocytogenes infection. Cell
73
:
457
24
Cook, D. N., M. A. Beck, T. M. Coffman, S. L. Kirby, J. F. Sheridan, I. B. Pragnell, O. Smithies.
1995
. Requirement of MIP-1α for an inflammatory response to viral infection.
Science
269
:
1583
25
MacMicking, J. D., C. Nathan, G. Hom, N. Chartrain, D. S. Fletcher, M. Trumbauer, K. Stevens, Q.-W. Xie, K. Sokol, N. Hutchinson, H. Chen, J. S. Mudgett.
1995
. Altered responses to bacterial infection and endotoxic shock in mice lacking inducible nitric oxide synthase.
Cell
81
:
641
26
Kuida, K., J. A. Lippke, G. Ku, M. W. Harding, D. J. Livingston, M. S. Su, R. A. Flavell.
1995
. Altered cytokine export and apoptosis in mice deficient in interleukin-1β converting enzyme.
Science
267
:
2000
27
Durbin, J. E., R. Hackenmiller, M. C. Simon, D. E. Levy.
1996
. Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease.
Cell
84
:
443
28
Goldblum, S. E., M. Jay, K. Yoneda, D. A. Cohen, C. J. McClain, M. N. Gillespie.
1987
. Monokine-induced lung injury in rabbits.
J. Appl. Physiol.
63
:
2093
29
Fehniger, T. A., W. E. Carson, E. Mrozek, M. A. Caligiuri.
1997
. Stem cell factor enhances interleukin-2-mediated expansion of murine natural killer cells in vivo.
Blood
90
:
3647
30
Lai, C. F., J. Ripperger, K. K. Morella, J. Jurlander, T. S. Hawley, W. E. Carson, T. Kordula, M. A. Caligiuri, R. Hawley, G. H. Fey, H. Baumann.
1996
. Receptors for IL-10 and IL-6-type cytokines use similar signaling mechanisms for inducing transcription through IL-6 response elements.
J. Biol. Chem.
271
:
13969
31
Iimuro, Y., T. Nishiura, C. Hellerbrand, K. E. Behrns, R. Schoonhoven, J. W. Grisham, D. A. Brenner.
1998
. NFκB prevents apoptosis and liver dysfunction during liver regeneration.
J. Clin. Invest.
101
:
802
32
Rose, J. A., K. I. Berns, M. D. Hoggan, F. J. Koczot.
1969
. Evidence for a single-stranded adenovirus-associated virus genome: formation of a DNA density hybrid on release of viral DNA.
Proc. Natl. Acad. Sci. USA
64
:
863
33
Giri, J. G., S. Kumaki, M. Ahdieh, D. J. Friend, A. Loomis, K. Shanebeck, R. DuBose, D. Cosman, L. S. Park, D. M. Anderson.
1995
. Identification and cloning of a novel IL-15 binding protein that is structurally related to the α chain of the IL-2 receptor.
EMBO J.
14
:
3654
34
Bosma, G. C., R. P. Custer, M. J. Bosma.
1983
. A severe combined immunodeficiency mutant in the mouse.
Nature
301
:
527
35
Mattory, Y. L., A. E. Chang, E. H. Lipford, R. Braziel, C. L. Hyatt, H. D. McDonald, S. A. Rosenberg.
1985
. Toxicity of recombinant human interleukin-2 in rats following intravenous infusion.
J. Biol. Response Modif.
4
:
377
36
Trinchieri, G..
1994
. Interleukin 12: a cytokine produced by antigen-presenting cells with immunoregulatory functions in the generation of T helper cells type I and cytotoxic lymphocytes.
Blood
84
:
4008
37
Gauldie, J., H. Baumann.
1991
. Cytokines and acute phase protein expression. E. S. Kimball, ed.
Cytokines and Inflammation
275
CRC Press, Boca Raton.
38
Dinarello, C. A..
1996
. Cytokines as mediators in the pathogenesis of septic shock.
Curr. Top. Microbiol. Immunol.
216
:
134
39
Sadowski, H. B., K. Shuai, J. E. Darnell, M. Z. Gilman.
1993
. A common nuclear signal transduction pathway activated by growth factor and cytokine receptors.
Science
261
:
1739
40
Kumar, A., M. Commane, T. W. Flickinger, C. M. Horvath, G. R. Stark.
1997
. Defective TNF-α-induced apoptosis in STAT1-null cells due to low constitutive levels of caspases.
Science
278
:
1630
41
Theodos, C. M., K. L. Sullivan, J. K. Griffiths, S. Tzipori.
1997
. Profiles of healing and nonhealing Cryptosporidium parvum infection in C57BL/6 mice with functional B and T lymphocytes: the extent of γ interferon modulation determines the outcome of infection.
Infect. Immun.
65
:
4761
42
Papa, M. Z., J. T. Vetto, S. E. Ettinghausen, J. J. Mule, S. A. Rosenberg.
1986
. Effects of corticosteroids on antitumor activity of lymphokine-activated killer cells and interleukin-2 in mice.
Cancer Res.
46
:
5618
43
Yamamoto, H., M. Hirayama, C. Genyea, J. Kaplan.
1994
. TGF-β mediates natural suppressor activity of IL-2-activated lymphocytes.
J. Immunol.
152
:
3842
44
Eberlein, T. J., D. D. Schoof, H. R. Michie, A. F. Massaro, U. Burger, D. W. Wilmore, R. E. Wilson.
1989
. Ibuprofen causes reduced toxic effects of interleukin 2 administration in patients with metastatic cancer.
Arch. Surg.
124
:
542
45
Bohrer, H., F. Qiu, T. Zimmerman, Y. Zhang, T. Jllmer, D. Mannel, B. W. Bottiger, D. M. Stern, R. Waldherr, H.-D. Saeger, R. Zeigler, A. Bierhaus, E. Martin, P. P. Nawroth.
1997
. Role of NFκB in the mortality of sepsis.
J. Clin. Oncol.
100
:
972
46
Wang, C. Y., M. W. Mayo, A. S. Baldwin, Jr.
1996
. TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-κB.
Science
274
:
784
47
Frederica, C., M. Forni, C. Riccardi, A. Soleti, F. Di Pierro, G. Forni.
1992
. Growth and spread of human malignant T lymphoblasts in immunosuppressed nude mice: a model for meningeal leukemia.
Blood
80
:
1279
48
Trinchieri, G., M. Kubin, G. Bellone, M. C. Cassatella.
1993
. Cytokine cross-talk between phagocytic cells and lymphocytes: relevance for differentiation/activation of phagocytic cells and regulation of adaptive immunity.
J. Cell. Biochem.
7
:
145
49
Bogdan, C., Y. Vodovotz, C. Nathan.
1991
. Macrophage deactivation by interleukin 10.
J. Exp. Med.
174
:
1549
50
Damas, P., D. LeDoux, M. Nijs, Y. Vrindts, D. De Groote, P. Franchimont, M. Lamy.
1992
. Cytokine serum levels during severe sepsis in humans: IL-6 as a marker of severity.
Ann. Surg.
215
:
356
51
Dietch, E. A..
1992
. Multiple organ failure: pathophysiology and potential future therapy.
Ann. Surg.
216
:
117
52
Mier, J. W..
1993
. Pathogenesis of interleukin-2-induced vascular leak syndrome. M. B. Atkins, Jr, and J. W. Mier, Jr, eds.
Therapeutic Applications of Interleukin-2
363
Marcel Dekker, New York.
53
Leonard, J. P., M. L. Sherman, G. L. Fisher, L. J. Buchanan, G. Karsen, M. B. Atkins, J. A. Sosman, J. P. Dutcher, N. J. Vogelzang, J. L. Ryan.
1997
. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-γ production.
Blood
90
:
2541
54
Nathan, C. F..
1987
. Secretory products of macrophages.
J. Clin. Invest.
79
:
319
55
Wu, C. Y., R. R. Warrier. D. M. Carvajal, A. O. Chu, L. J. Minetti, R. Chizzonite, P. K. Mongini, A. S. Stern, U. Gubler, D. H. Presky, M. K. Gately.
1996
. Biological function and distribution of human interleukin-12 receptor β chain.
Eur. J. Immunol.
26
:
345
56
Unanue, E. R..
1996
. Macrophages, NK cells and neutrophils in the cytokine loop of Listeria resistance.
Res. Immunol.
147
:
499
57
Gazzinelli, R. T., S. Hieny, T. A. Wynn, S. Wolf, A. Sher.
1993
. Interleukin 12 is required for the T-lymphocyte-independent induction of interferon γ by an intracellular parasite and induces resistance in T-cell-deficient hosts.
Proc. Natl. Acad. Sci. USA
90
:
6115
58
Heremans, H., J. Van Damme, C. Dillen, R. Dijkmans, A. Billiau.
1990
. Interferon γ, a mediator of lethal lipopolysaccharide-induced Shwartzman-like shock reactions in mice.
J. Exp. Med.
171
:
1853
59
Heremans, H., C. Dillen, J. van Damme, A. Billiau.
1994
. Essential role for natural killer cells in the lethal lipopolysaccharide-induced Shwartzman-like reaction in mice.
Eur. J. Immunol.
24
:
1155
60
Ozmen, L., M. Pericin, J. Hakimi, R. A. Chizzonite, M. Wysocka, G. Trinchieri, M. Gately, G. Garotta.
1994
. Interleukin 12, interferon γ, and tumor necrosis factor α are the key cytokines of the generalized Shwartzman reaction.
J. Exp. Med.
180
:
907
61
Ortaldo, J. R., A. Mason, J. J. O’Shea.
1995
. Receptor-induced death in human natural killer cells: involvement of CD16.
J. Exp. Med.
181
:
339
62
Eischen, C. M., J. D. Schilling, D. H. Lynch, P. H. Krammer, P. J. Leibson.
1996
. Fc receptor-induced expression of Fas ligand on activated NK cells facilitates cell-mediated cytotoxicity and subsequent autocrine NK cell apoptosis.
J. Immunol.
156
:
2693
63
Ida, H., M. J. Robertson, S. Voss, J. Ritz, P. Anderson.
1997
. CD94 ligation induces apoptosis in a subset of IL-2-stimulated NK cells.
J. Immunol.
159
:
2154
64
Yamauchi, A., K. Taga, H. S. Mostowski, E. T. Bloom.
1996
. Target cell-induced apoptosis of interleukin-2-activated human natural killer cells: roles of cell surface molecules and intracellular events.
Blood
87
:
5127
65
Van Parijs, L., A. K. Abbas.
1998
. Homeostasis and self-tolerance in the immune system: turning lymphocytes off.
Science
280
:
243
66
Miethke, T., C. Wahl, K. Heeg, B. Echtenacher, P. H. Kramer, H. Wagner.
1992
. T cell-mediated lethal shock triggered in mice by the superantigen staphylococcal enterotoxin B: critical role of tumor necrosis factor.
J. Exp. Med.
175
:
91
67
Chatenoud, L..
1993
. OKT3-induced cytokine-release syndrome: prevention effect of anti-tumor necrosis factor monoclonal antibody.
Transplant. Proc.
25
:
47