Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jun 17.
Published in final edited form as: Ann N Y Acad Sci. 2009 Dec;1182:69–79. doi: 10.1111/j.1749-6632.2009.05069.x

Clinical Use of Interferon-γ

Catriona HT Miller a, Stephen G Maher b, Howard A Young a
PMCID: PMC6574079  NIHMSID: NIHMS1035329  PMID: 20074276

Abstract

Interferon gamma (IFN-γ), a pleotropic cytokine, has been shown to be important to the function of virtually all immune cells and both innate and adaptive immune responses. In 1986, early clinical trials of this cytokine began to evaluate its therapeutic potential. The initial studies focused on the tolerability and pharmacology of IFN-γ and systematically determined its antitumor and anti-infection activities. In the 20-plus years since those first trials, IFN-γ has been used in a wide variety of clinical indications, which are reviewed in this article.

Keywords: Interferon-γ, Actimmune, HuZaf

Introduction

Interferon gamma (IFN-γ), a cytokine with diverse roles in the innate and adaptive immune responses, was discovered in 1965. IFN-γ function has been strongly conserved throughout evolution and across multiple species. T cells, NK cells, and NKT cells are the primary producers of IFN-γ, and it has a myriad of effects in both host defense and immune regulation, including antiviral activity, antimicrobial activity, and antitumor activity. Experimental models in which IFN-γ production has been disrupted have resulted in an increase in autoimmune diseases (reviewed in Ref. 1).

IFN-γ has been shown to be important to the function and maturation of multiple immune cells. IFN-γ is essential for Th1 immune responses and regulates T cell differentiation, activation, expansion, homeostasis, and survival. Killing of intracellular pathogens requires IFN-γ production by T cells. T regulatory cell (Treg) generation and activation requires IFN-γ. IFN-γ stimulates dendritic cells and macrophages to upregulate major histocompatibility complex (MHC) molecules, enhances antigen presentation, and increases expression of costimulatory molecules. IFN-γ stimulated macrophages also produce reactive nitrogen intermediates. NK cells secrete IFN-γ early in host infection, facilitating immune cell recruitment and activation. IFN-γ also activates NK cells, enhancing their cytotoxicity and cell mediatedimmune responses. During a viral infection, IFN-γ mediates IgG2a isotype class switching in B cells. IFN-γ recruits neutrophils, stimulates them to upregulate chemokines and adhesion molecules, and triggers rapid superoxide production and respiratory burst (reviewed in Ref. 1). Due to the pleiotrophic effects of IFN-γ on the immune system, it was thought to have great promise as an immunomodulatory drug.

It was not until 1986 that early clinical trials of this cytokine were first conducted.2 The initial studies were focused on side effects and dose escalation, and then later trials systematically determined its therapeutic potential against cancers and infections. In subsequent years, IFN-γ has been used in a wide variety of clinical indications. The most common adverse therapeutic events occurring with IFN-γ1b therapy are “flu-like,” such as fever, headache, chills, myalgia, or fatigue. Other common side effects include rash, injection site erythema or tenderness, diarrhea and nausea, and leukopenia.38

IFN-γ clinical trials have been conducted using recombinant derived protein (IFN-γ1b, Actimmune), adenovirus vectors which express IFN-γ cDNA (TG-1041, TG-1042), and neutralizing antibodies against IFN-γ (HuZaf and AMG811). Actimmune has been used to treat a wide variety of diseases, including cancer, tuberculosis, hepatitis, chronic granulamotous disease, osteopetrosis, scleroderma, among others. Adeno-IFN-γ has been used to treat cutaenous lymphoma and malignant melanoma. HuZaf has been used against autoimmune diseases, including rheumatoid arthritis and multiple sclerosis. Promising results have been seen with all agents and are reviewed here.

Actimmune

The majority of the clinical trials involving IFN-γ have been performed using Actimmune (InterMune) or IFN-γ1b, a genetically engineered form of human IFN-γ. The naturally occurring IFN-γ produced by human peripheral blood leukocytes (PBLs) has the same primary structure as IFN-γ1b.9,10 However, these two cytokines differ in that: (1) IFN-γ is glycosylated and IFN-γ1b is not, (2) IFN-γ1b is 140 amino acids long while IFN-γ has 143 amino acids, and (3) IFN-γ has blocked pyroglutamate residue N-termini, whereas recombinant IFN-γ1b has methionine at the N-terminus.911

Cancer

The role of IFN-γ in the host response to cancer has been the subject of numerous research studies. Studies have shown that IFN-γ is vital to tumor surveillance by the immune system and a high correlation between IFN-γ production and tumor regression has been seen in immunotherapy. IFN-γ also has direct antitumor effects, as it is anti-angiogenic, inhibits proliferation, sensitizes tumors cells to apoptosis, upregulates MHC class I and II expression, and stimulates antitumor immune activity. There have been mixed results as to the efficacy of IFN-γ in the clinical treatment of various cancers.

In a study of recurring superficial transitional bladder carcinoma, it was shown that intravesicle instillations of IFN-γ were effective against cancer recurrence.12 There were significant increases in T cells, NK cells, ICAM-1+ B cells, and HLA-DR+ cell infiltrating the tumor. Strong IFN-γ-induced expression of HLA-DR has been correlated with improved prognosis in colorectal cancer patients.13

Ovarian cancer is a leading cause of cancer death and is an ideal target for cytokine targeted treatment.1416 The presence of intratumoral IFN-γ producing CD3+ T cells is associated with better prognosis.17 The standard treatment is platinum-based chemotherapy. IFN-γ is synergistic with platinum chemotherapeutics in vitro at inhibiting ovarian cancer cell proliferation and inducing apoptosis.18 Intraperitoneal IFN-γ has been shown to achieve antitumor responses against ovarian cancer.19 In a recent randomized phase III trial, administration of subcutaneous IFN-γ and cisplatin to patients improved complete response rates from 56% to 68%20 and prolonged progression-free survival. Another study showed that IFN-γ with carboplatin and paclitaxel is safe as a first-line treatment of patients with advanced ovarian cancer. Conversely, a phase III clinical trial was ended prematurely in 2006. Ovarian cancer and peritoneal carcinoma patients were treated with carboplatin/paclitaxel chemotherapy alone or with IFN-γ 1b subcutaneously. In the second interim analysis, it was found that patients treated with IFN-γ and carboplatin/paclitaxel had a significantly shorter survival rate and more adverse events as compared to patients receiving chemotherapy alone. 39.7% of the IFN-γlb and chemotherapy group had died, in contrast to 30.4% of patients in the chemotherapy only group.21

IFN-γ is an approved treatment for adult T cell leukemia (ATL) in Japan. There are several reports which claim that intralesional injections of IFN-γ can induce lasting remissions.22 IFN-γ treatment was first developed and approved in Japan for treatment of Mycosis fungoides (MF).23

Several clinical trials are underway using IFN-γ as an adjuvant for vaccine therapies and chemotherapies ( NCT00428272, NCT0049-9772, NCT00824733, NCT00004016).

Tuberculosis

Tuberculosis (TB), a result of infection with Mycobacterium tuberculosis, primarily affects the respiratory system, and the emergence of multidrug resistant strains (MDR-TB) has led to the need for new therapeutic agents. IFN-γ activates alveolar macrophages, which are important in host immunity against M. tuberculosis.24 Condos et al. performed a clinical trial examining the effects of an IFN-γ aerosol on MDR-TB. Stabilization or an increase in bodyweight was observed in all IFN-γ treated patients. Furthermore, sputum smears became negative, and a decrease in the mycobacterial burden was seen. Two months after cessation of treatment, a reduction in cavitary lesion sizes was observed in all patients.25 Another trial found that co-administration of IFN-γ and anti-TB drugs to tuberculosis patients, resulted in increased levels of STAT1, IRF-1, and IRF-9 in BAL cells from lung segments. IFN-γ actively stimulated signal transduction and gene expresion in alveolar macrophages in TB patients, thus providing a basis for potential use as an adjuvant therapy in this disease.

Other approaches have evaluated IFN-γ as an adjuvant therapy in addition to a chemotherapeutic cocktail. Saurez-Mendez et al. found that intramuscular injection of IFN-γ for 6 months as an adjuvant to chemotherapy led to reduced lesion sizes, negative sputum smears and cultures, and increased body mass index.4

Mycobacterium avium Complex (MAC)

Atypical mycobacteria infections have been rising, especially among older women. MAC infection leads to progressive chronic pneumonia and lung disease. Atypical mycobacteria survive and proliferate within host macrophages. Treatment of MAC pulmonary infections is difficult because of high drug resistance. IFN–γ has been shown to be a critical cytokine in the resistance of infected macrophages. Thirty-two patients were treated with either intramuscular IFN-γ and chemotherapy or chemotherapy and placebo. The overall response in the IFN-γ group was significantly better than those treated with chemotherapy alone (72.2% vs. 37.5% complete responders). During the study, 35.7% of the control group died compared to 11.1% of the IFN-γ treated group.26

Idiopathic Pulmonary Fibrosis

Currently, there is no FDA approved drug treatment for idiopathic pulmonary fibrosis (IPF). IPF is the most frequent of the idiopathic interstitial pneumonias and has the worst prognosis.27 IPF is a chronic condition characterized by progressive scarring, loss of lung function, progressive limitation, and eventual death.28 Alveolar epithelial cells release fibrogenic cytokines, such as TGF-β, PDGF, TNF-α, IL-1, insulin-like growth factor-1, and basic fibroblast factor, in response to injury. The release of these cytokines causes fibroblast proliferation, migration to the lung, and fibroblast differentiation.29,30 Traditional therapies have been ineffective, and new agents are required to halt the progression of disease. IFN-γ therapy of IPF has been explored, as IPF is characterized by an IFN-γ deficit. It was hypothesized that treatment with IFN-γ might halt the progression of IPF.

Unfortunately, the results from IPF studies with IFN-γ treatment are mixed. In an initial randomized clinical trial31 of IPF patients, IFN-γ treatment in combination with prednisolone resulted in increased total lung capacity and increased resting and maximal exertion values of partial pressure of arterial oxygen, compared to prednisolone alone. In a retrospective study of qualified IPF patients, IFN-γ had beneficial effects on forced vital capacity and single breath diffusing capacity for CO2.28 Furthermore, these effects were most pronounced in patients with advanced disease.

Conversely, in a large (330 patients) one year placebo-controlled clinical trial, subcutaneous IFN-γ administration did not affect progression-free survival or pulmonary function.32 The time to death or disease progression was not significantly altered. However, subgroup analyses, which are at best hypothesis generating, showed a possible survival benefit for patients with mild-to-moderate impairment.33

The results of a double-blind clinical trial of the molecular effects of subcutaneous IFN-γ1b in IPF patients were published in 2004.34 After IFN-γ1b treatment, expression of the immunomodulatory chemoattractant CXCL11 was increased in bronchoalveolar lavage fluid (BALF) and plasma. Levels of neutrophil activating CXCL5, PDGFA (platelet derived growth factor A), and type 1 procollagen were lower in BALF. Gene expression studies showed increases in CXCL11, type III procollagen, and PDGFB in transbronchial biopsy samples. A decrease in elastin was also seen. These changes suggested that IFN-γ 1b would be an effective treatment for IPF via multiple pathways.34

In March of 2007, the INSPIRE trial of Actimmune for treating IPF ( NCT00075998) was forced to end prematurely. INSPIRE was a randomized, double-blind, placebo-controlled Phase 3 study designed to evaluate the safety and efficacy of Actimmune in IPF patients with mild to moderate impairment in lung function. The primary endpoint was survival time. An interim analysis showed that patients who received Actimmune did not benefit. Approximately 14.5% of patients treated with Actimmune died compared to 12.7% of placebo treated patients ( NCT00075998).

Cystic Fibrosis

Cystic fibrosis (CF) is an inherited disorder caused by a mutation in a chloride channel, the cystic fibrosis transmembrane conductance regulator gene. CF is characterized by chronic endobronchial infection and inflammation, destruction of lung tissue, and eventual respiratory failure in 90% of patients. CF patients have inefficient pulmonary clearance of thick secretions and defects in production of nitric oxide resulting in chronic bacterial infections with a thick bacterial biofilm. Bacterial biofilms resist opsonins, phagocytes, antibiotics, and cause chronic neutrophil inflammation. Production of elastase by neutrophils contributes to lung damage. Due to the ability of IFN-γ to activate macrophages, correct deficiencies in NO production in vitro, and inhibit the proliferation of Th2 clones, and based on clinical data showing IFN-γ deficiencies in PBMCs from cystic fibrosis patients,35,36 it was thought to have great potential as a treatment for cystic fibrosis. Sixty-six cystic fibrosis patients received 50 μg–1000 μg aerosolized IFN-γ1b or placebo three times a week for 12 weeks.37 No statistically significant differences were seen in the primary endpoints of the trial, FEV1 (forced expiratory volume in 1 s) and sputum bacterial density over the entire study. At 4 weeks, a slight, but significant reduction was seen in the FEV1 of the 1000 μg IFN-γ treated group versus placebo, and a significant reduction was also seen in bacterial density between the 1000 μg IFN-γ treated group versus placebo. No significant statistical differences were seen in levels of neutrophils, IL-8, elastase, myeloperoxidase, or DNA in the sputum of either IFN-γ or placebo treated patients.

Hepatitis

Liver cirrhosis and hepatocellular carcinoma arise primarily as a result of chronic hepatitis infection.5,38 The current regimen for the treatment of chronic hepatitis B (HBV) and C (HCV) is pegylated IFN-α (peginterferon-α) and ribavirin with a response rate of only 50–60%.6 In light of the large number of non-responders, IFN-γ has been evaluated as a potential alternative treatment. In chronic HBV trials, IFN-γ1b alone was not found to have any significant impact on viral infection but did modulate the immune system.7 Treatment of HCV with IFN-γ1b has also proven generally unsuccessful8,39 but pretreatment with IFN-γ prior to IFN-α treatment resulted in enhanced immunologic activity in HCV patients. The enhanced immunological activity is speculated to enhance IFN-α-mediated viral clearance.40

Fibrosis accounts for the majority of the complications associated with chronic hepatitis. IFN-γ has been shown to have antifibrotic effects and is efficacious against fibrosis in HBV patients.41 In a recent study, the antifibrotic activity of IFN-γ1b in HCV patients was examined.8 Although no overall reduction in fibrosis was seen, select patients had significant fibrosis reductions.

Chronic Granulomatous Disease

In 1991, IFN-γ1b was FDA approved for the treatment of chronic granulomatous disease (CGD). CGD is an inherited disorder of leukocyte function, caused by a defect or the absence of reduced nicotinamide adenine dinucleotide phosphate oxidase. This enzyme is essential for microbiocidal activity and superoxide generation in phagocytes. Consequently, CGD patients suffer recurrent life-threatening bacterial and fungal infections. As IFN-γ is known to enhance the respiratory burst of human phagocytic cells, it was hoped that IFN-γ would reverse the immunological defects observed in CGD patients. In clinical trials, IFN-γ1b reduced the frequency and severity of serious infections in CGD patients.11

Marciano et al. examined the long-term effects of IFN-γ1b administration to CGD patients.42 76 patients were enrolled in an uncontrolled study to assess long-term safety and efficacy of IFN-γ1b therapy. Patients were followed for up to 9 years and received IFN-γ1b subcutaneously thrice weekly. This study concluded that IFN-γ prophylaxis for CGD appears to be effective and well tolerated over a prolonged period of time. Actimmune product literature claims that treatment with their product leads to a 67% reduction in the relative risk of serious infections, 53% fewer primary infections, 64% fewer infections overall, and 67% fewer inpatient hospital days.

Osteopetrosis

Congenital osteopetrosis (OP) is a rare osteosclerotic bone disease caused by a defect in osteoclast function and bone resorption.43 Severe, malignant OP is characterized by an overgrowth of body structures which results in infection, anemia, thrombocytopenia, blindness, deafness, and ultimately early death.4446 The granular leukocytes of OP patients are defective in superoxide production,47,48 resulting in frequent, severe infections. As IFN-γ1b reduces infection in CGD patients by increasing neutrophil superoxide production, Key et al. hypothesized that IFN-γ1b might stimulate osteoclasts in a similar manner.49 IFN-γ1b administration to OP patients significantly increased osteoclastic bone resorption, increased superoxide production in PBLs, and reduced infection.50 In vitro studies with OP patient blood cultures, have shown that IFN-γ1b enhances osteoclast generation and normalizes superoxide production.45 Presently, the only effective treatment for osteopetrosis is hematopoietic stem cell transplant (HSCT), which has high treatment related morbidity and mortality.51

In 2000, the FDA approved Actimmune for delaying the time to disease progression in patients with severe malignant OP. In a 1999 phase III clinical trial, 15 osteopetrosis patients received either Actimmune or control vitamin D. The length of time to disease progression was significantly delayed in patients treated with Actimmune (165 days) compared to patients treated with the control (65 days). Evidence of increased bone resorption, enhanced bone marrow activity, and a reduction in serious infections was observed.50

Scleroderma

Scleroderma is a connective tissue disease that affects multiple organ systems, including skin, heart, lungs, and kidneys.52 The mechanisms of fibrosis in scleroderma are not fully understood. It is known that soluble mediators, such as TGF-β, PDGF, IL-4, IL-6, and TNF-α, can affect the behavior of fibroblast growth, proliferation, collagen synthesis, and chemotaxis.5254 IFN-γ has been used in the treatment of scleroderma because of its antifibrotic activity, its ability to reduce collagen production in vitro, and to inhibit fibroblast cell proliferation. In most clinical trials, either subcutaneous or intramuscular administration of IFN-γ to scleroderma patients has resulted in modest improvements.5558

Invasive Fungal Infections/Immunosuppressed Patients

Invasive fungal infections are an increasing problem, especially in immunocompromised patients, such as leukemia patients, HIV patients, and transplant patients. The most common infectious agents are candida and aspergillus. There are several new drugs available to treat fungal infection, including triazoles and immunotherapeutics, such as colony stimulating factors, granulocyte transfusions and IFN-γ. GM-CSF and IFN-γ are given in combination to treat patients with serious refractory fungal infections and non-neutropenic infections. It has been demonstrated that IFN-γ increases the anti-fungal activity of macrophages and neutrophils.26 (ISRCTN70900209)

Cryptococcus neoformans is responsible for the most common central nervous system infection in HIV patients, acute cryptococcal meningitis, and is the most common cause of fungal meningitis worldwide. In a double-blind clinical trial, patients received either 100 or 200 μg of IFN-γ1b or placebo in addition to standard anti-fungal therapy. ( NCT00012467) Among 75 patients, 13% of placebo patients, 36% of 100 μg IFN-γ1b, and 32% of 200 μg IFN-γ1b had fungus clean cerebrospinal fluid cultures after 2 weeks.59

The efficacy of IFN-γ1b to reduce opportunistic infections in advanced HIV was tested in a 12-month double-blind phase III trial of HIV patients on antiretroviral drugs. Eighty-four patients were treated with either IFN-γ or placebo subcutaneously for 48 weeks. Patients on placebo had an average of 3.45 opportunistic infections in the first 48 weeks, while patients with IFN-γ1b therapy had an average of 1.71. Three-year survival in the IFN-γ arm was 28% compared to 18% in the placebo group, although this difference was not statistically significant. IFN-γ1b treatment was especially effective against candida, herpes, and cytomegalovirus infections.60

Adeno-IFN-γ

Cutaneous B and T Cell Lymphomas

Primary cutaneous lymphomas (CL) are characterized by an accumulation of clonal T or B lymphocytes in the skin. Typically, CLs are chronic indolent diseases. TG-1042, or adeno-IFN-γ, was investigated in a phase I trial, where patients with advanced primary cutaneous B or T CL were repeatedly injected intratumorally with TG-1042.61,62 TG-1042 is a nonreplicating adenovirus vector containing a human IFN-γ cDNA insert. Five of nine treated patients had local clinical responses. Three patients had complete responses with the clearance of non-injected skin lesions. Two patients had partial responses. IFN-γ from TG-1042 message was detected in injected lesions in seven patients after the first treatment cycle and remained detectable for several cycles. Humoral antitumor immune responses were also detected. Adeno-IFN-γ is now being tested in a phase II trial ( NCT00394693) against chronic BCL.61

Malignant Melanoma

A phase I clinical trial was conducted in 2003 using TG-1041, a recombinant adeno-IFN-γ vector to treat malignant melanoma. TG-1041 is a replication deficient adenovirus with the cDNA for IFN-γ inserted in the E1 region of the viral genome. Patients were given three intratumoral injections of adeno-IFN-γ. Out of 11 treated patients, no complete or partial responses were seen. However five patients had minor decreases in injected tumor nodules, eight patients had local inflammation, one patient had significant necrosis of the injected nodule, one patient had inflammation of distant nodules, and one patient had disease stabilization.63

Anti-Interferon-γ Antibodies (HuZaf and AMG811)

Fontolizumab (HuZAF) is a humanized monoclonal antibody that binds IFN-γ and inhibits expression of IFN-γ regulated genes. Fontolizumab is being explored for the treatment of autoimmune diseases, such as Crohn’s disease, lupus, rheumatoid a arthritis, and multiple sclerosis. Adverse side effects are generally mild and rare, and include abdominal pain, vomiting, headache, nausea, arthalgia, asthenia, and cough.64,65 AMG811, a fully human monoclonal antibody that binds and neutralizes IFN-γ, is being evaluated by Amgen.

Multiple Sclerosis

Multiple sclerosis (MS) is an autoimmune disorder where the immune system attacks the myelin sheath of the central nervous system. Disturbances in cytokine synthesis, particularly IFN-γ, play critical roles in the initiation and prolongation of MS.66 IFN-β is currently used in the treatment of MS and may work via inhibition of IFN-γ-mediated immune activation. In a randomized study of patients with progressive MS, those patients who received a short course of neutralizing IFN-γ antibody had a significant delay in disability progression.65 MRIs showed decreased numbers of active lesions. The cytokine profile produced by activated blood cells from treated patients changed, with decreased IL-1β, TNF-α, and IFN-γ and increased TGF-β. These data indicate that neutralizing IFN-γ may be a new treatment option for the management of progressive MS.

Crohn’s Disease

Recent studies have examined the safety and efficacy of HuZAF in the treatment of moderate to severe Crohn’s disease (CD).67 Crohn’s disease is a chronic inflammatory disease of the gastrointestinal tract. IFN-γ has been implicated in the inflammation observed in CD and increased levels are found in the mucosa.64 In several models of experimental colitis, increased mucosal IFN-γ levels were detected. In a randomized double-blind phase II clinical trial, 42 patients received one dose, and 91 patients received two doses of HuZAF.67 There was no difference in response between HuZAF and placebo groups after a single dose. In contrast, those patients receiving two doses of fontolizumab had a doubled response rate at day 56 compared to placebo controls. Given the long half-life (18 days) and low immunogenicity, this study concluded that treatment of active CD with anti-IFN-γ antibody warrants further investigation.67 Based on the results with Crohn’s disease, HuZAF is being considered as a treatment for pediatric inflammatory bowel disease.

Rheumatoid Arthritis

Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by a predominant Th1-associated autoimmunity. Early clinical studies tested IFN-γ as a therapeutic for rheumatoid arthrititis. In these studies, 54 patients were treated with IFN-γ, while 51 received placebo in a double-blind study.68 While greater improvement was seen in the IFN-γ treated group, it was not statistiscally significant. Recently, antibodies to IFN-γ have been found to be beneficial in the treatment of RA, in a randomized double-blind trial.69 Thirty patients with active RA received intramuscular injections of either anti-IFN-γ, anti-TNF-α, or placebo. Based on a physical examination, nine patients receiving anti-TNF-α, seven receiving anti-IFN-γ and two receiving placebo, appeared to have an improvement of their condition.

Lupus

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by self-reactive antibodies, often against nucleic acids, which form immune complexes and collect in body organs and joints. Because SLE patients have been shown to have increased levels of serum IFN-γ,70 IFN-γ has been shown to exacerbate SLE,71 and in mouse models, IFN-γ receptor is required for development of SLE.72 Amgen has recently begun a phase Ib clinical trial, using AMG-811, a fully human monoclonal antibody that binds IFN-γ to treat SLE ( NCT00818948). This trial will be focused on safety and pharmokinetics.

Summary

IFN-γ has proven to be a key immunoregulatory molecule whose effects on immune system development, maturation and function is widespread, affecting a myriad of cell types. While IFN-γ affects numerous disease processes, the clinical applications of this important molecule are currently limited. Given the critical importance of IFN-γ in immunity, clinical use of IFN-γ will depend upon a more precise understanding of its basic biology and localized effects in order to better define how to use this molecule in the context of the disease setting.

Acknowledgment

This project has been funded in whole or in part with federal funds from the National Cancer Institute, National Institutes of Health, under Contract No. HHSN261200800001E. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.

Footnotes

Given the large number of publications on IFN-γ, it is inevitable that some authors may believe that their work was not properly cited or included in this review. The authors of this review apologize to those investigators whose work was not cited as any exclusion of relevant work was not intentional.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  • 1.Young HA, Romero-Weaver AL, Savan R, et al. 2007. Interferon-gamma. Interferon-γ in Class II Cytokines Zdanov A, Ed.: 51–106. Research Signpost, Kerala, India. [Google Scholar]
  • 2.Jaffe HS & Sherwin SA. 1986. The early clinical trials of recombinant human interferon-gamma. Interferons as Cell Growth Inhibitors and Antitumor Factors Freedman R, Merigan T & Sreevalson T, Eds.: 37–46. Alan R. Liss; New York. [Google Scholar]
  • 3.Condos R, Hull FP, Schluger NW, et al. 2004. Regional deposition of aerosolized interferon-gamma in pulmonary tuberculosis. Chest 125: 2146–2155. [DOI] [PubMed] [Google Scholar]
  • 4.Suarez-Mendez R, Garcia-Garcia I, Fernandez-Olivera N, et al. 2004. Adjuvant interferon gamma in patients with drug-resistant pulmonary tuberculosis: A pilot study. BMC Infect. Dis 4: 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Soza A, Heller T, Ghany M, et al. 2005. Pilot study of interferon gamma for chronic hepatitis C. J. Hepatol 43: 67–71. [DOI] [PubMed] [Google Scholar]
  • 6.Di Bisceglie AM & Hoofnagle JH. 2002. Optimal therapy of hepatitis C. Hepatology 36: S121–S127. [DOI] [PubMed] [Google Scholar]
  • 7.Lau JY, Lai CL, Wu PC, et al. 1991. A randomised controlled trial of recombinant interferon-gamma in Chinese patients with chronic hepatitis B virus infection. J. Med. Virol 34: 184–187. [DOI] [PubMed] [Google Scholar]
  • 8.Muir AJ, Sylvestre PB & Rockey DC. 2006. Interferon gamma-1b for the treatment of fibrosis in chronic hepatitis C infection. J. Viral Hepat 13: 322–328. [DOI] [PubMed] [Google Scholar]
  • 9.Rinderknecht E, O’Connor BH & Rodriguez H. 1984. Natural human interferon-gamma. Complete amino acid sequence and determination of sites of glycosylation. J. Biol. Chem 259: 6790–6797. [PubMed] [Google Scholar]
  • 10.Rinderknecht E & Burton LE. 1985. Biochemical characterization of natural and recombinant IFN-gamma. The Biology of the Interferon System Kirchner H & Schellekens H, Eds.: 397–402. Elsevier; Amsterdam. [Google Scholar]
  • 11.Czarniecki CW & Sonnenfeld G. 2006. Clinical applications of interferon-gamma. The Interferons: Characterization and Application Meager A, Ed.: 309–336. Wiley-VCH; Weinheim. [Google Scholar]
  • 12.Giannopoulos A, Constantinides C, Fokaeas E, et al. 2003. The immunomodulating effect of interferon-gamma intravesical instillations in preventing bladder cancer recurrence. Clin. Cancer Res 9: 5550–5558. [PubMed] [Google Scholar]
  • 13.Matsushita K, Takenouchi T, Shimada H, et al. 2006. Strong HLA-DR antigen expression on cancer cells relates to better prognosis of colorectal cancer patients: Possible involvement of c-myc suppression by interferon-gamma in situ. Cancer Sci 97: 57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chen CK, Wu MY, Chao KH, et al. 1999. T lymphocytes and cytokine production in ascitic fluid of ovarian malignancies. J. Formos. Med. Assoc 98: 24–30. [PubMed] [Google Scholar]
  • 15.Punnonen R, Teisala K, Kuoppala T, et al. 1998. Cytokine production profiles in the peritoneal fluids of patients with malignant or benign gynecologic tumors. Cancer 83: 788–796. [DOI] [PubMed] [Google Scholar]
  • 16.Wall L, Burke F, Smyth JF & Balkwill F. 2003. The anti-proliferative activity of interferon-gamma on ovarian cancer: in vitro and in vivo. Gynecol. Oncol 88: S149–S151. [DOI] [PubMed] [Google Scholar]
  • 17.Marth C, Fiegl H, Zeimet AG, et al. 2004. Interferon-gamma expression is an independent prognostic factor in ovarian cancer. Am. J. Obstet. Gynecol 191: 1598–1605. [DOI] [PubMed] [Google Scholar]
  • 18.Marth C, Windbichler GH, Hausmaninger H, et al. 2006. Interferon-gamma in combination with carboplatin and paclitaxel as a safe and effective first-line treatment option for advanced ovarian cancer: results of a phase I/II study. Int. J. Gynecol. Cancer 16: 1522–1528. [DOI] [PubMed] [Google Scholar]
  • 19.Pujade-Lauraine E, Guastalla JP, Colombo N, et al. 1996. Intraperitoneal recombinant interferon gamma in ovarian cancer patients with residual disease at second-look laparotomy. J. Clin. Oncol 14: 343–350. [DOI] [PubMed] [Google Scholar]
  • 20.Windbichler GH, Hausmaninger H, Stummvoll W, et al. 2000. Interferon-gamma in the first-line therapy of ovarian cancer: a randomized phase III trial. Br. J. Cancer 82: 1138–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Alberts DS, Marth C, Alvarez RD, et al. 2008. Randomized phase 3 trial of interferon gamma-1b plus standard carboplatin/paclitaxel versus carboplatin/paclitaxel alone for first-line treatment of advanced ovarian and primary peritoneal carcinomas: Results from a prospectively designed analysis of progression-free survival. Gynecol. Oncol 109: 174–181. [DOI] [PubMed] [Google Scholar]
  • 22.Tamura K, Makino S, Araki Y, et al. 2006. Recombinant interferon beta and gamma in the treatment of adult T-cell leukemia. Cancer 59: 1059–1062. [DOI] [PubMed] [Google Scholar]
  • 23.Nagatani T, Okazawa H, Inomata N, et al. 2004. PUVA and interferon-gamma combination therapy for plaque stage mycosis fungoides. Nishi Nihon Hifuka 66: 274–279. [Google Scholar]
  • 24.Condos R, Raju B, Canova A, et al. 2003. Recombinant gamma interferon stimulates signal transduction and gene expression in alveolar macrophages in vitro and in tuberculosis patients. Infect. Immun 71: 2058–2064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Condos R, Rom WN & Schluger NW. 1997. Treatment of multidrug-resistant pulmonary tuberculosis with interferon-gamma via aerosol. Lancet 349: 1513–1515. [DOI] [PubMed] [Google Scholar]
  • 26.Milanes-Virelles M, Garcia-Garcia I, Santos-Herrera Y, et al. 2008. Adjuvant interferon gamma in patients with pulmonary atypical Mycobacteriosis: A randomized, double-blind, placebo-controlled study. BMC Infect. Dis 8: 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Katzenstein AL & Myers JL. 1998. Idiopathic pulmonary fibrosis: clinical relevance of pathologic classification. Am. J. Respir. Crit. Care Med 157: 1301–1315. [DOI] [PubMed] [Google Scholar]
  • 28.Nathan SD, Barnett SD, Moran B, et al. 2004. Interferon gamma-1b as therapy for idiopathic pulmonary fibrosis. An intrapatient analysis. Respiration 71: 77–82. [DOI] [PubMed] [Google Scholar]
  • 29.Kahlil N & O’Connor R. 2004. Idiopathic pulmonary fibrosis: current understanding of the pathogenesis and the status of treatment. CMAJ 171: 153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Selman M, Thannickal VJ, Pardo A, et al. 2004. Idiopathic pulmonary fibrosis: pathogenesis and therapeutic approaches. Drugs 64: 405–430. [DOI] [PubMed] [Google Scholar]
  • 31.Ziesche R, Hofbauer E, Wittmann K, et al. 1999. A preliminary study of long-term treatment with interferon gamma-1b and low-dose prednisolone in patients with idiopathic pulmonary fibrosis. N. Engl. J. Med 341: 1264–1269. [DOI] [PubMed] [Google Scholar]
  • 32.Raghu G, Brown KK, Bradford WZ, et al. 2004. A placebo-controlled trial of interferon gamma-1b in patients with idiopathic pulmonary fibrosis. N. Engl. J. Med 350: 125–133. [DOI] [PubMed] [Google Scholar]
  • 33.Shah NR, Noble P, Jackson RM, et al. 2005. A critical assessment of treatment options for idiopathic pulmonary fibrosis. Sarcoidosis. Vasc. Diffuse. Lung Dis 22: 167–174. [PMC free article] [PubMed] [Google Scholar]
  • 34.Strieter RM, Starko KM, Enelow RI, et al. & the other members of the Idiopathic Pulmonary Fibrosis Biomarkers Study Group. 2004. Effects of interferon-{gamma} 1b on biomarker expression in patients with idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med 170: 133–140. [DOI] [PubMed] [Google Scholar]
  • 35.Knutsen AP, Hutchinson PS, Albers GM, et al. 2003. Increased sensitivity to IL-4 in cystic fibrosis patients with allergic bronchopulmonary aspergillosis allergy. Allergy 59: 81–87. [DOI] [PubMed] [Google Scholar]
  • 36.Moss RB, Hsu YP & Olds L. 2009. Cytokine dysregulation in activated cystic fibrosis (CF) peripheral lymphocytes. Clin. Exp. Immunol 120: 518–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Moss RB, Mayer-Hamblett N, Wagener J, et al. 2004. Randomized, double-blind, placebo-controlled, dose-escalating study of aerosolized interferon gamma-1b in patients with mild to moderate cystic fibrosis lung disease. Pediatr. Pulmonol 39: 209–218. [DOI] [PubMed] [Google Scholar]
  • 38.Brown RS Jr. & Gaglio PJ. 2003. Scope of world-wide hepatitis C problem. Liver Transpl 9: S10–S13. [DOI] [PubMed] [Google Scholar]
  • 39.Saez-Royuela F, Porres JC, Moreno A, et al. 1991. High doses of recombinant alpha-interferon or gamma-interferon for chronic hepatitis C: A randomized, controlled trial. Hepatology 13: 327–331. [DOI] [PubMed] [Google Scholar]
  • 40.Katayama K, Kasahara A, Sasaki Y, et al. 2001. Immunological response to interferon-gamma priming prior to interferon-alpha treatment in refractory chronic hepatitis C in relation to viral clearance. J. Viral Hepat 8: 180–185. [DOI] [PubMed] [Google Scholar]
  • 41.Weng HL, Cai WM & Liu RH. 2001. Animal experiment and clinical study of effect of gamma-interferon on hepatic fibrosis. World J. Gastroenterol 7: 42–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Marciano BE, Wesley R, De Carlo ES, et al. 2004. Long-term interferon-gamma therapy for patients with chronic granulomatous disease. Clin. Infect. Dis 39: 692–699. [DOI] [PubMed] [Google Scholar]
  • 43.Blin-Wakkach C, Wakkach A, Sexton PM, et al. 2004. Hematological defects in the oc/oc mouse, a model of infantile malignant osteopetrosis. Leukemia 18: 1505–1511. [DOI] [PubMed] [Google Scholar]
  • 44.Key LL Jr., Wolf WC, Gundberg CM & Ries WL. 1994. Superoxide and bone resorption. Bone 15: 431–436. [DOI] [PubMed] [Google Scholar]
  • 45.Madyastha PR, Yang S, Ries WL & Key LL Jr. 2000. IFN-gamma enhances osteoclast generation in cultures of peripheral blood from osteopetrotic patients and normalizes superoxide production. J. Interferon Cytokine Res 20: 645–652. [DOI] [PubMed] [Google Scholar]
  • 46.Shapiro F, Glimcher MJ, Holtrop ME, et al. 1980. Human osteopetrosis: A histological, ultrastructural, and biochemical study. J. Bone Joint Surg. Am 62: 384–399. [PubMed] [Google Scholar]
  • 47.Beard CJ, Key L, Newburger PE, et al. 1986. Neutrophil defect associated with malignant infantile osteopetrosis. J. Lab Clin. Med 108: 498–505. [PubMed] [Google Scholar]
  • 48.Reeves JD, August CS, Humbert JR & Weston WL. 1979. Host defense in infantile osteopetrosis. Pediatrics 64: 202–206. [PubMed] [Google Scholar]
  • 49.Key LL Jr., Ries WL, Rodriguiz RM & Hatcher HC. 1992. Recombinant human interferon gamma therapy for osteopetrosis. J. Pediatr 121: 119–124. [DOI] [PubMed] [Google Scholar]
  • 50.Key LL Jr., Rodriguiz RM, Willi SM, et al. 1995. Long-term treatment of osteopetrosis with recombinant human interferon gamma. N. Engl. J. Med 332: 1594–1599. [DOI] [PubMed] [Google Scholar]
  • 51.Stark Z & Savarirayan R. 2009. Osteopetrosis. Orphanet J. Rare Dis 4: 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sapadin AN & Fleischmajer R. 2002. Treatment of scleroderma. Arch. Dermatol 138: 99–105. [DOI] [PubMed] [Google Scholar]
  • 53.Fleischmajer R, Perlish JS, Krieg T & Timpl R. 1981. Variability in collagen and fibronectin synthesis by scleroderma fibroblasts in primary culture. J. Invest. Dermatol 76: 400–403. [DOI] [PubMed] [Google Scholar]
  • 54.Jimenez SA, Hitraya E & Varga J. 1996. Pathogenesis of scleroderma. Collagen. Rheum. Dis. Clin. North Am 22: 647–674. [DOI] [PubMed] [Google Scholar]
  • 55.Grassegger A, Schuler G, Hessenberger G, et al. 1998. Interferon-gamma in the treatment of systemic sclerosis: a randomized controlled multicentre trial. Br. J. Dermatol 139: 639–648. [DOI] [PubMed] [Google Scholar]
  • 56.Hein R, Behr J, Hundgen M, et al. 1992. Treatment of systemic sclerosis with gamma-interferon. Br. J. Dermatol 126: 496–501. [DOI] [PubMed] [Google Scholar]
  • 57.Hunzelmann N, Anders S, Fierlbeck G, et al. 1997. Systemic scleroderma. Multicenter trial of 1 year of treatment with recombinant interferon gamma. Arch. Dermatol 133: 609–613. [DOI] [PubMed] [Google Scholar]
  • 58.Polisson RP, Gilkeson GS, Pyun EH, et al. 1996. A multicenter trial of recombinant human interferon gamma in patients with systemic sclerosis: Effects on cutaneous fibrosis and interleukin 2 receptor levels. J. Rheumatol 23: 654–658. [PubMed] [Google Scholar]
  • 59.Pappas P, Bustamante B, Ticona E, et al. 2004. Recombinant interferon-γ1b as adjunctive therapy for AIDS-related acute cryptococcal meningitis. J. Infect. Dis 189: 2185–2191. [DOI] [PubMed] [Google Scholar]
  • 60.Riddell LA, Pinching AJ, Hill S, et al. 2001. A phase III study of recombinant human interferon gamma to prevent opportunistic infections inadvanced HIV disease. AIDS Res. Hum. Retroviruses 17: 789–797. [DOI] [PubMed] [Google Scholar]
  • 61.Dummer R 2005. Emerging drugs in cutaneous T-cell lymphomas. Expert Opin. Emerg. Drugs 10: 381–392. [DOI] [PubMed] [Google Scholar]
  • 62.Dummer R, Hassel JC, Fellenberg F, et al. 2004. Adenovirus-mediated intralesional interferon-{gamma} gene transfer induces tumor regressions in cutaneous lymphomas. Blood 104: 1631–1638. [DOI] [PubMed] [Google Scholar]
  • 63.Khorana AA, Rosenblatt JD, Sahasrabudhe DM, et al. 2003. A phase I trial of immunotherapy with intratumoral adenovirus-interferon-gamma (TG1041) in patients with malignant melanoma. Cancer Gene Ther 10: 251–259. [DOI] [PubMed] [Google Scholar]
  • 64.Fuss IJ, Neurath M, Boirivant M, et al. 1996. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J. Immunol 157: 1261–1270. [PubMed] [Google Scholar]
  • 65.Skurkovich S, Boiko A, Beliaeva I, et al. 2001. Randomized study of antibodies to IFN-gamma and TNF-alpha in secondary progressive multiple sclerosis. Mult. Scler 7: 277–284. [DOI] [PubMed] [Google Scholar]
  • 66.Skurkovich B & Skurkovich S. 2003. Anti-interferon-gamma antibodies in the treatment of autoimmune diseases. Curr. Opin. Mol. Ther 5: 52–57. [PubMed] [Google Scholar]
  • 67.Hommes DW, Mikhajlova TL, Stoinov S, et al. 2006. Fontolizumab, a humanised anti-interferon gamma antibody, demonstrates safety and clinical activity in patients with moderate to severe Crohn’s disease. Gut 55: 1131–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cannon GW, Pincus SH, Emkey RD, et al. 1989. Double-blind trial of recombinant gamma-interferon versus placebo in the treatment of rheumatoid arthritis. Arthritis Rheum 8: 964–973. [DOI] [PubMed] [Google Scholar]
  • 69.Sigidin YA, Loukina GV, Skurkovich B & Skurkovich S. 2001. Randomized, double-blind trial of anti-interferon-gamma antibodies in rheumatoid arthritis. Scand. J. Rheumatol 30: 203–207. [DOI] [PubMed] [Google Scholar]
  • 70.Robak E, Smolewski P, Wozniacka A, et al. 2004. Relationship between peripheral blood dendritic cells and cytokines involved in the pathogenesis of systemic lupus erythematosus. European Cytokine Network 15: 222–230. [PubMed] [Google Scholar]
  • 71.Machold KP & Smolen JS. 1990. Interferon-gamma induced exacerbation of systemic lupus erythematosus. J. Rheumatol 17: 831–832. [PubMed] [Google Scholar]
  • 72.Theofilopoulos A, Koundouris S, Kono D & Lawson B. 2001. The role of IFN-gamma in systemic lupus erythematosus: a challenge to the Th1/Th2 paradigm in autoimmunity. Arthritis Res 3: 136–141. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES