Interferon in Oncological Practice_Review of Interferon

download Interferon in Oncological Practice_Review of Interferon

of 22

Transcript of Interferon in Oncological Practice_Review of Interferon

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    1/22

    Interferon in Oncological Practice: Review of InterferonBiology, Clinical Applications, and Toxicities

    ERIC JONASCH, FRANK G. HALUSKA

    Massachusetts General Hospital, Boston, Massachusetts, USA

    Key Words.Interferon Cancer Clinical applications Toxicity Management

    ABSTRACT

    For the past 40 years, various forms of interferon

    (IFN) have been evaluated as therapy in a number of

    malignant and non-malignant diseases. With the advent

    of gene cloning, large quantities of pure IFN became

    available for clinical study. This paper reviews the biol-

    ogy, pharmacology, and clinical applications of IFN for-

    mulations most commonly used in oncology. It thenreviews the most common side effects seen in patients

    treated with IFN, and makes recommendations for the

    management of IFN-induced toxicity.

    The major oncological indications for IFN include

    melanoma, renal cell carcinoma, AIDS-related Kaposis

    sarcoma, follicular lymphoma, hairy cell leukemia,

    and chronic myelogenous leukemia. Unfortunately,

    IFN therapy is associated with significant toxicity,

    which can be divided into constitutional, neuropsychi-

    atric, hematologic, and hepatic effects. These toxicities

    have a major impact on the patients quality of life, and

    on the physicians ability to optimally treat the patient.

    Careful attention to all aspects of patient care canresult in improved tolerability of this difficult but

    promising therapy. Conclusion: a better understanding

    of IFN biology, indications, side effect profiles, and tox-

    icity management will aid in optimizing its use in the

    treatment of patients with cancer. The Oncologist

    2001;6:34-55

    The Oncologist 2001;6:34-55 www.TheOncologist.com

    Correspondence: Eric Jonasch, M.D., Cox 640, Division of Hematology-Oncology, 55 Fruit Street, Boston, Massachusetts02114, USA. Telephone: 617-724-6582; Fax: 617-726-6974; e-mail: [email protected] Received March 31, 2000;accepted for publication October 3, 2000. AlphaMed Press 1083-7159/2001/$5.00/0

    INTRODUCTION

    In 1957Isaacs andLindeman described a factor that con-

    ferred the property of viral interference [1]. The term inter-

    feron (IFN) was coined to describe this new substance. Over

    the next 40 years, the IFNs have been evaluated as therapeu-

    tic modalities in a large number of malignant and nonmalig-

    nant diseases. Research has revealed diverse mechanisms of

    action, including antiviral, immune-enhancing and cytostatic

    activities.

    In oncology, the IFNs are an important treatment for a

    number of solid tumors and hematological malignancies.

    These include melanoma, renal cell carcinoma, AIDS-

    related Kaposis sarcoma (KS), follicular lymphoma, hairy

    cell leukemia, and chronic myelogenous leukemia (CML).

    IFN therapy is associated with significant side effects

    which have an impact on the patients quality of life and the

    physicians ability to optimally treat the patient. An under-

    standing of IFN biology, interactions, indications for use,

    side-effect profiles, and the management of IFN toxicities

    will aid in the optimal application of these agents in the

    management of patients with cancer.

    In this report, we review the classification of the IFNs and

    summarize their major mechanisms of action. We then

    describe the indications for IFN therapy in oncological prac-

    tice and review the etiology and management of IFN-related

    side effects.

    IFN CLASSIFICATION

    After the discovery of IFN, initial attempts at purifica-

    tion were met with little success, defying efforts at classifi-

    cation. Early clinical trials used crude preparations that were

    less than 1% IFN by weight [2]. It was only in 1978 and

    thereafter that IFN was purified to homogeneity in amounts

    that allowed chemical and physical characterization. The

    initial nomenclature of, , and IFN was used to catego-

    rize the major HPLC peaks, but was quickly adopted to des-

    ignate leukocyte, fibroblast, and immune IFNs, respectively[2]. Table 1 summarizes some of the major properties of the

    various IFN subtypes.

    In 1980, at a meeting jointly sponsored by the National

    Institute of Allergy and Infectious Diseases and the World

    Health Organization, nomenclature was formally adopted

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    2/22

    Jonasch, Haluska 35

    classifying IFNs into three cate-

    gories based on antigenic speci-

    ficity. IFNs were categorized as

    alpha (), beta (), and gamma

    (), corresponding to the previ-ous designations of leukocyte,

    fibroblast and immune IFN [3].

    More recently, IFNs were

    divided into two major sub-

    groups by virtue of their ability

    to bind to common receptor

    types [4-6]. Type I IFNs all bind

    to a type I IFN receptor, and

    include IFN-, IFN-, IFN-,

    and IFN-. IFN-is the sole type

    II IFN, and binds to a distincttype II receptor [7].

    Almost all cell types produce

    type I IFNs. The prototypical

    production sites for IFN- and

    IFN- are leukocytes and fibro-

    blasts, respectively. Their induc-

    tion usually follows exposure to

    viruses, double-stranded RNA,

    polypeptides, and cytokines [8].

    The type II IFN-is produced in

    T cells and natural killer (NK)

    cells following a number of

    immunological stimuli, includ-

    ing T cell-specific antigens,

    staphylococcal enterotoxin A,

    and the combination of phytohemagglutinin and phorbol

    ester [2, 8]. Unlike IFN- and -, it is not directly induced

    in cells following viral infection.

    BIOLOGICAL PROPERTIES OF IFNS

    The IFNs possess a broad spectrum of activity and are

    involved in complex interactions. They display antiviral

    activity, impact cellular metabolism and differentiation, and

    possess antitumor activity. The antitumor effects appear to be

    due to a combination of direct antiproliferative, as well as

    indirect immune-mediated effects. Figure 1 summarizes the

    intracellular signaling, and Figures 2 and 3 summarize the

    major effects of IFN- and -on NK cells, antigen-presenting

    cells, macrophages, T cells, and tumor cells.

    Table 1. IFN classification and properties

    IFN IFN Receptor Prototypic Direct Anti- Stimulates MHC Stimulates MHC Stimulates NKType categories type cell of origin proliferative effects class I expression class II expression cell activation

    Type I Alpha () I Leukocyte Yes Yes No Yes

    Beta () I Fibroblast Yes Yes Slightly Yes

    Omega () I Leukocyte Yes Yes No Yes

    Tao () I OvineTrophoblast

    Type II Gamma () II T-cells Yes Yes Yes Less than type INK cells IFNs, delayed

    P

    PP

    P P

    P

    P

    P

    P

    P

    P P

    P

    P

    P

    P

    P

    P

    P

    P

    P

    IFN-AR1

    IFN-AR

    2

    TYK2

    STAT1

    STAT2

    JAK1

    IFN-

    IFN-

    STAT1

    STAT1

    JAK1

    JAK1

    JAK2

    IFN-G

    R2

    IFN-G

    R2

    IFN

    -GR1

    IFN

    -GR1

    STAT1

    STAT1

    STAT1

    STAT1

    GAS ISGISGISRE

    STAT1

    STAT2

    ISGF3

    P

    Cytoplasm

    Nucleus

    IFN-

    STAT1

    STAT2

    Figure 1. For the type I IFNs, there are two receptor subunits known, IFNAR-1 and IFNAR-2, which bind the

    Janus-activated kinase (Jak) molecules Tyk2 and Jak1, respectively. For IFN-, there are two receptor subunits

    known: IFNGR-1 and IFNGR-2, which associate with Jak1 and Jak2, respectively. Upon binding of IFN to its recep-

    tor, the receptor undergoes oligomerization, with transphosphorylation of Jaks followed by phosphorylation of the

    cytoplasmic tails of the receptor molecules. This provides a docking site for the signal transducers and activators of

    transcription (Stats) which are then phosphorylated by the Jaks. The phosphorylated Stat dimers are released from

    the receptor molecules, and translocate to the nucleus, where they activate transcription of IFN-stimulated genes(ISGs). In the case of type I IFNs, ISGs can be identified by the presence of an IFN-stimulated response element

    (ISRE) in their promoter regions. Enhancers of IFN--inducible genes contain a unique element called the IFN-

    activation site (GAS).

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    3/22

    36 Interferon in Oncological Practice

    CURRENTLY AVAILABLE PREPARATIONS

    The cloning of the IFN genes resulted in the availabil-

    ity of large quantities of pure IFN- and -, allowing sub-

    stantive research to be done on the clinical efficacy of the

    IFNs in malignant and nonmalignant diseases. The clini-

    cal indications for the IFN subtypes are summarized in

    Table 2.

    IFN-2a (Roferon A; Roche Laboratories; Nutley, NJ)and IFN-2b (Intron-A; Schering-Plough Corporation;

    Kenilworth, NJ) are now produced by recombinant (r)DNA

    technology. The molecular sequences of the two rIFNs differ

    from one another by a single amino acid at position 23 [9].

    IFN alfacon-1 (Infergen, Amgen; Thousand Oaks, CA) is a

    recombinant non-naturally occurring IFN, whose 166 amino-

    acid sequence was derived by scanning the sequences of sev-

    eral natural IFN- subtypes and assigning the most frequently

    observed amino acid in each corresponding position. Four

    additional changes were made to facilitate molecular con-

    struction. IFN-n3 (Alferon N; Interferon Sciences Inc.;New Brunswick, NJ) is derived from human leukocytes.

    T-cell

    NK cell/macrophage

    Tumorcell

    Antiangiogenic

    Th1 differentiation

    Growth inhibition?

    MHC I expression

    Tumor-specific antigenexpression

    Adhesion moleculeexpression

    Direct cytostaticeffects

    IFN-production

    Cellular cytotoxicity

    IL-1 production

    Proliferation

    IFN-

    Figure 2. IFN-possesses pleiotropic and potentially antagonistic activity

    in immune cells and tumors. Both stimulatory and inhibitory effects are

    seen in the T-cell population. A stimulatory effect is seen in natural killer

    (NK) cells and macrophages, which may be antagonized by the IFN-medi-

    ated upregulation of major histocompatibility-1 (MHC1) in tumor cells.Direct cytostatic effects are seen in tumor cells, as well as upregulation of

    tumor-specific antigens and adhesion molecules. IFN- may also have

    antiangiogenic effects mediated indirectly by IFN-(Fig. 3).

    T-cell

    MacrophageAPC

    Tumorcell

    Antiangiogenic

    Increased activation(in synergy with IL-2)

    MHC class I upregulation

    Decreased proliferation

    Activation

    MHC class IIupregulation

    IFN-

    Figure 3. IFN-is an activator of T-cells and macrophages, working in concert

    with interleukin-2 (IL-2).IFN-is capable of upregulating both MHC classes I

    and II, and has demonstrated direct inhibitory effects on tumor proliferation. IFN-

    is presumed to induce its antiangiogenic effects through the secretion of IFN-

    inducible protein 10 (IP-10) and monokine induced by IFN-(MIG).

    Table 2. IFN clinical indications

    IFN type Trade name Manufacturing technique FDA-approved indications

    IFN-2a Roferon A rDNA NHLHairy cell leukemiaCMLAIDS-related KSChronic hepatitis C

    IFN-2b Intron A rDNA Follicular lymphomaHairy cell leukemia

    AIDS-related KSMalignant melanomaCondyloma accuminataChronic hepatitis BChronic hepatitis C

    IFN alfacon-1 Infergen rDNA Chronic hepatitis C

    IFN-n3 Alferon N Purified from human leukocytes Condyloma accuminata

    IFN-1a Avonex rDNA Relapsing-remitting MS(Chinese hamster ovary cells)

    IFN-1b Betaseron rDNA Relapsing-remitting MS(E. coli)

    IFN- Actimmune rDNA Chronic granulomatous disease

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    4/22

    Jonasch, Haluska 37

    IFN-1b is manufactured by bacterial fermentation of a

    strain ofEscherichia coli that bears a genetically engineered

    plasmid containing the gene for human IFN betaser17. The

    native gene was obtained from human fibroblasts and altered

    in a way that substitutes serine for the cysteine residue found

    at position 17 [10]. IFN-1b has 165 amino acids and an

    approximate molecular weight of 18,500 Da and does not

    include the carbohydrate side chains found in the natural

    product. It is given s.c. every other day [11].

    IFN-1a (Avonex; Biogen Inc; Cambridge, MA) and IFN-

    1b (Betaseron; Berlex; Richmond, CA) are recombinant

    proteins. IFN-1a is a 166-amino acid glycoprotein with a

    predicted molecular weight of approximately 22,500 Da. It

    is produced by mammalian cells (Chinese hamster ovary

    cells) into which the human IFN- gene has been intro-

    duced. The amino acid sequence of IFN-1a is identical to

    that of natural human IFN-. IFN-1a is also U.S. Food and

    Drug Administration (FDA)-approved for treatment of

    relapsing-remitting multiple sclerosis (MS) to reduce fre-

    quency of relapses [12]. It may be preferable to IFN-1b

    because of the convenience of weekly administration, the

    relative lack of injection site reactions, and lower incidence

    of neutralizing antibodies.

    IFN-(Actimmune; Genentech Inc; San Francisco, CA),

    is produced using rDNA technology. It is FDA-approved for

    use in chronic granulomatous disease.

    PHARMACOKINETICS

    As with most peptides, oral delivery of the IFNs is notpractical due to proteolytic degradation. Subcutaneous and

    i.m. absorption of IFN- and IFN- is >80%, and 30%-

    70%, respectively [13]. These routes of administration

    result in a protracted distribution phase, with maximal

    serum or plasma concentrations occurring after 1 to 8 h, fol-

    lowed by measurable concentrations for 4 to 24 h after

    injection for both IFN- and -. After i.m. administration,

    IFN- levels peak between 3 and 15 h, and then decline at

    a rate consistent with a 10-h elimination half-life. At thera-

    peutic dosages of 0.25 mg s.c., serum concentrations of

    IFN- are low or undetectable [14]. Intravenous adminis-

    tration of IFN- or - results in a biexponential decrease in

    serum concentration, and IFN- levels decline monoexpo-

    nentially. Terminal elimination half-lives range from 4-16 h

    for IFN-, 1-2 h for IFN- and 25 to 35 min for IFN-[15-

    17]. Serum concentrations are generally measurable for

    between 8 and 24 h after i.v. injection of IFN- and up to

    4 h after i.v. injection of IFN- and -.

    The relationship between dose and biological response

    varies among disease types. A number of surrogate markers

    have been investigated to better define the dose-response

    relationship of IFN. 25 oligoadenylate synthetase (2-5A),

    an enzyme induced by both IFN- and -, is involved in

    IFN-mediated viral RNA degradation [18]. Introduction of

    2-5A into cells [19] or expression of 2-5A cDNA inhibited

    cell growth rates [20]. Increased 2-5A levels were also

    shown to correlate with decreased cell cycling in mela-

    noma cell cultures treated with IFN [21], suggesting a

    causal role in its antiproliferative action. Using 2-5A activ-

    ity as a marker for response, a two- to sixfold increase in

    2-5A activity was seen over a 300-fold dose range i.m.

    using human lymphoblastoid IFN- [22]. Other biological

    markers used to evaluate IFN response include neopterin

    and 2 microglobulin. Neopterin is a pteridine derivative

    originally found in cultures of stimulated T lymphocytes

    [23] whose serum and urinary levels correlated with thera-

    peutic IFN dosages in the treatment of hairy cell leukemia

    [24, 25]. Small studies in melanoma [26-28] provide con-

    flicting data on the utility of neopterin levels in predicting

    response to immunotherapy. The human MxA protein is a

    GTPase with antiviral activity against orthomyxoviruses

    and certain other negative-strand DNA viruses [29].

    Unlike 2-5A, 2 microglobulin and neopterin, MxA gene

    expression is induced solely by type I IFNs [30, 31], and

    holds promise for future cancer immunology studies. In

    aggregate, these markers provide laboratory confirmation

    of immunological stimulation by IFN, but do not provide

    consistent predictive information on the use of IFN therapy

    in cancer.

    APPLICATION OF IFN THERAPY IN ONCOLOGICALPRACTICE

    The IFNs have been used to treat a number of malig-

    nancies, with varying degrees of success. The following

    section summarizes the most common applications of IFN

    in current oncological practice.

    Hairy Cell Leukemia

    The first report of IFN response in patients with hairy

    cell leukemia was recorded in 1984 [32]. In this study,

    seven patients were treated with 3 106 U of partially puri-

    fied leukocyte IFN- i.m. daily (qd). Three patients had a

    complete remission (CR), four had a partial remission (PR),

    and remissions were maintained for 6 to 10 months. Subse-

    quently, 30 patients with hairy cell leukemia were treated

    with IFN-2a, including seven who were previously

    untreated. Nine (30%) CR and 17 (56%) PR were confirmed

    by bone marrow core biopsies. All patients peripheral blood

    hematologic indices either improved or normalized [33]. A

    multicenter phase II study of 64 patients published in 1986

    confirmed the drugs efficacy in hairy cell leukemia [34].

    IFN-2a and 2b were FDA-approved for use in hairy cell

    leukemia in 1986. IFN- is usually given over a two-year

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    5/22

    38 Interferon in Oncological Practice

    period at 3 106 U/day. Unfortunately, upon cessation of

    therapy, almost all patients will relapse. Although approved

    for use, IFN- has been superceded by more effective thera-

    pies in hairy cell leukemia, including 2-deoxycoformycin

    and cladribine.

    CML

    Early Studies

    Preclinical studies performed in the late 1970s sug-

    gested IFN therapy had antiproliferative effects on granulo-

    cyte precursors [35]. In 1983, Talpaz et al. reported on seven

    patients with CML treated with IFN- 9-15 106 U i.m. qd

    [36], five of whom developed a hematologic remission. In

    1986, the same group reported on 17 patients with early-

    phase CML who were treated with rIFN-2a, 5 106 U/m2

    i.m. qd [37]. Fourteen patients responded to the treatment,

    13 of whom had a complete hematologic remission (CHR)and one had a partial hematologic remission (PHR).

    Cytogenetic analysis revealed that 20%-25% of patients

    experienced long-term suppression of the Philadelphia (Ph)

    chromosome. At least 30 uncontrolled observational studies

    have been published looking at the effect of IFN- therapy

    on patients in chronic-phase CML [38]. These studies vary

    greatly with respect to their reporting of outcome measures,

    and overall survival is often not recorded. Most studies

    record an association between cytogenetic remission and

    improved survival by landmark analysis*, with a minority

    failing to show such an association [39]. (*Landmark analy-

    sis is an assessment of outcome following the stratification

    of patients according to a particular endpoint, in this case the

    achievement of cytogenetic remission.)

    IFN Monotherapy

    In 1994, the Italian Cooperative Group published a

    study of 322 patients with untreated or minimally treated

    CML randomized between IFN-2a and conventional

    chemotherapy [40]. The rate of karyotypic response was

    30% in the IFN group versus 5% in the chemotherapy

    group. Median time to progression and overall survival

    were significantly longer in the IFN groups. Table 3 sum-

    marizes the five randomized studies comparing chemother-apy (busulfan and/or hydroxyurea) to IFN- therapy

    [40-44]. Four of these studies conclude that IFN therapy is

    better than chemotherapy in patients with early chronic-

    phase CML [40-43]. In all but one of these studies, IFN was

    given as monotherapy. TheBenelux study [45] randomized

    patients to IFN plus hydroxyurea as needed to keep the

    Table 3. CML: randomized studies

    Author Year n Study Hematologic response (%) Median

    patients design CHR PHR survival (mos)Broustet[44] 1991 30 IFN 53 N/A N/A

    28 Hydroxurea 82 N/A N/A

    Hehlmann [41] 1994 133 IFN 31 52 66*

    186 Busulfan 23 69 45*

    194 Hydroxyurea 39 51 56

    Italian 1994 218 IFN CHR + PHR = 45% 72Cooperative 104 Hydroxyurea/ CHR + PHR = 46% 52Group [40] Busulfan (p = 0.002)

    Allan [42] 1995 293 IFN 69 18 61

    294 Hydroxyurea/ N/A N/A 41Busulfan (p = 0.0009)

    Ohnishi [43] 1995 80 IFN 39 39 Not reached, but improved79 Busulfan 54 43 survival in IFN arm (p = 0.029)

    Benelux [45] 1998 100 IFN 62 N/A 64Hydroxyurea

    95 Hydroxyurea 38 N/A 68 NS

    Guilhot[50] 1997 361 IFN 55 N/A Not reached, but improvedHydroxyurea survival in IFN-cytarabine

    360 IFN 66 N/A arm (p = 0.02)

    HydroxyureaCytarabine (p = 0.003)

    NS = not significant; *statistically significant

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    6/22

    Jonasch, Haluska 39

    WBC count below 10 109 versus hydroxyurea alone, with

    no survival benefit noted in the IFN arm. Differences in

    trial design, inclusion criteria, treatment and documentation

    of response make direct comparisons between these trials

    difficult, but the evidence supports a benefit to IFN- therapy

    in patients with early chronic-phase CML.

    Combination Therapy

    In the late 1980s, cytarabine (ara-C) was shown to sup-

    press CML clones selectively in vitro [46] and demon-

    strated in vivo antitumor efficacy when given as a low-dose

    continuous infusion [47]. In 1992, Kantarjian et al. pub-

    lished a report on 60 patients with advanced phases of CML

    who received combination therapy with IFN- 5 106

    U/m2 daily, and low-dose ara-C 15 mg/m2 daily for two

    weeks every four weeks until remission, then for one week

    every month as maintenance, compared to historical con-

    trols receiving IFN- therapy alone. Patients receiving

    IFN- plus ara-C had a better CHR rate compared with

    those treated with IFN-, a trend for better Ph suppression

    and longer survival [48]. More recently, Kantarjian et al.

    compared 140 patients with chronic-phase CML receiving

    IFN- at 5 106 and ara-C 10 mg daily to historical con-

    trols receiving IFN- alone or IFN- with intermittent ara-

    C [49]. The study group demonstrated a CHR in 92% of

    patients, with cytogenetic response in 74% of patients, 31%

    of which were complete, results that were significantly bet-

    ter than the control groups. The time to blastic transforma-

    tion and overall survival was the same as in the controlgroups. Several other observational studies report a poten-

    tial benefit to the combination of IFN and ara-C.

    Subsequently, a randomized study by Guilhot et al. indi-

    cated the 360 patients receiving IFN-2b, hydroxyurea, and

    ara-C demonstrated a significant improvement in overall

    survival and cytogenetic response when compared with 361

    patients receiving IFN-2b and hydroxyurea alone [50]

    (Table 3).

    IFN-

    IFN-has been used as therapy in chronic-phase CML.

    In 1987, Kurzrock et al. reported that 6 of 30 patients with

    chronic-phase CML treated with rIFN-demonstrated CHR,

    and varying degrees of cytogenetic improvement [51].

    Subsequent studies using combinations of IFN- and IFN-

    failed to show an improvement over treatment with IFN-

    alone [52, 53], and IFN-is not recommended for treatment

    of CML.

    Bone Marrow Transplantation and IFN

    Concern has been raised about prior IFN- therapy

    having a negative influence on allogeneic bone marrow

    transplant outcomes. Several recent studies have suggested

    this is not the case in patients who received a matched-

    related donor [54-56], but one report suggests adverse out-

    comes for patients who received a greater than six-month

    course of IFN- prior to undergoing a matched-unrelated

    donor transplant [57].

    Summary

    The major decision in otherwise healthy patients with

    CML is whether to treat with IFN- initially, or to proceed

    with early allogeneic bone marrow transplantation. Outcomes

    after transplantation reveal lower overall survival initially

    when compared to IFN therapy, with a crossing-over of sur-

    vival curves at approximately seven years after diagnosis [38].

    There appears to be a survival plateau in the transplant patient

    population, but not in the IFN--treated patient population.

    Unfortunately, most of the transplant data are nonrandomized,

    and there have been no direct valid comparisons between IFN

    therapy and transplantation in CML. The recommendations

    made in an American Society of Hematology-sponsored

    review [38] do not strongly favor IFN- therapy or allogeneic

    bone marrow transplantation therapy, but indicate that both

    should be considered. The patients age, stage of disease,

    comorbid conditions, and the patients and physicians thresh-

    old for risk and treatment-related morbidity need to be taken

    into consideration when making such a decision.

    FOLLICULAR LYMPHOMA

    IFN- was first used in the treatment of patients with fol-licular lymphoma in the early 1980s [58]. Early-phase studies

    showed a 50% objective response rate. Subsequently, two

    phase III trials randomized patients between single alkylat-

    ing agents with or without IFN-, demonstrating improved

    remission duration, but no overall survival improvement [58-

    61]. In addition, two randomized phase III trials evaluated the

    addition of IFN to combination cytoreductive chemotherapy

    [62, 63]. In only one of these studies was there an improved

    median progression-free survival and overall survival in the

    group of patients receiving IFN [63].

    The role of IFN- as maintenance therapy for patients

    with low-grade non-Hodgkins lymphoma (NHL) who

    achieved tumor bulk reduction after cytoreductive chemo-

    therapy has also been evaluated [58, 64-69]. Most studies

    demonstrated an increased failure-free survival in patients

    on maintenance IFN, but no clear-cut increase in overall

    survival. One randomized study of IFN-2b maintenance

    therapy did reveal an overall survival advantage [69].

    Based on the above studies, some authors recommend

    that IFN- therapy be used in combination with chemother-

    apy in clinically aggressive follicular NHL, and as mainte-

    nance therapy in patients with high tumor burden after

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    7/22

    40 Interferon in Oncological Practice

    cytoreductive therapy [58]. These recommendations have

    not been universally accepted. We recommend that IFN-

    be used in an investigational setting to better define its role

    in the treatment of follicular lymphoma.

    RENAL CELL CARCINOMA (RCC)

    IFN has been extensively used in the treatment of RRC.

    Although there is no clear role for its use in the adjuvant set-

    ting, extensive work has been done to define its role in the

    management of metastatic disease. The following sections

    summarize these findings.

    IFN Monotherapy

    Single-agent IFN has been used in over 50 clinical trials

    in the treatment of metastatic RCC. Response rates in these

    trials range between 0% and 50%, with a mean of approxi-

    mately 15% for IFN- and 10% for IFN-. There does not

    appear to be a clear dose-response curve in the treatment ofmetastatic RCC with rIFN-2a or IFN-2b [70]. Stratifi-

    cation of trials among those with average daily doses of 10 million units per day does not reveal any sta-

    tistically significant differences in outcome. A retrospective

    analysis of prognostic markers of response suggested that

    pretreatment nephrectomy and lung metastases as sole site of

    metastatic disease were markers for better outcome. Overall

    performance status is another important prognostic factor

    predicting response to therapy [71].

    Several randomized studies have been published to

    quantify the benefit of IFN in metastatic RCC (Table 4A).The Medical Research Council Renal Cancer Collaborators

    trial randomized 350 patients with metastatic RCC between

    IFN-2b and medroxyprogesterone acetate (MPA) [71]. An

    interim analysis was performed when time-to-progression

    data were available for 335 patients, showing a statistically

    significant improvement in median survival in favor of the IFN

    arm. Pyrhonen et al. prospectively randomized 160 patients

    with locally advanced or metastatic RCC between vinblas-

    tine (VBL) alone and IFN-2a plus VBL for 12 months or

    until progression of disease [72]. Both the response rates and

    median survival were superior in the IFN arm.

    Role of Nephrectomy

    The benefit of preimmunotherapy nephrectomy was

    addressed by the Southwest Oncology Group (SWOG) in a

    randomized trial designed to determine whether nephrec-

    tomy prior to systemic therapy with IFN prolonged survival

    (Table 4A). Patients with operable metastatic renal cancer

    were randomized to two arms: immediate IFN therapy or

    radical nephrectomy followed by IFN therapy. Between

    June 1991 and October 1998, 246 patients were random-

    ized. Median survival was significantly improved in thenephrectomy arm, but the response rate to IFN was a low

    4% in both arms, suggesting the survival advantage was

    mainly due to surgical debulking, and not because surgery

    improved the efficacy of IFN therapy.

    Combination with Interleukin 2 (IL-2)

    Subsequent efforts have been made to evaluate IFN

    together with other biological response modifiers including

    IL-2, or in combination with chemotherapeutic agents, in par-

    ticular 5-fluorouracil (5-FU). The combination of IFN- and

    IL-2 has been extensively investigated in patients withmetastatic RCC. Phase I and II trials have demonstrated

    response rates of approximately 20%, with 5% CR [73].

    Treatment schedules, cytokine doses, patient selection criteria

    and response criteria differ from study to study. Table 4B

    Table 4A. RCC: randomized studies

    Author Year n Study Response Medianpatients design rates (%) survival (mos)

    IFN and Chemo- or Hormonal Therapy

    MRC [71] 1999 174 IFN 10 MU s.c. tiw 12 wks 14 6.0

    176 MPA 300 mg PO qd 12 wks 7 8.5*

    Pyrhonen [72] 1999 81 VBL 0.1 mg/kg q 3 wks 2.5 8.7

    79 VBL 0.1 mg/kg q 3 wks 16.5 15.6

    IFN 3 MU s.c. tiw wk 1 then (p = 0.0025) (p = 0.0049)

    IFn18 MU s.c. tiw

    * statistically significant

    IFN Nephrectomy

    SWOG [156] 2000 123 Nephrectomy 4 8.1

    123 Nephrectomy plus 4 12.5

    IFN 5 MU/m2 s.c. tiw until progression (p = 0.033)

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    8/22

    Jonasch, Haluska 41

    summarizes seven randomized trials assessing the efficacy of

    combination cytokine therapy in metastatic RCC. In a phase

    II trial, Atkins et al. randomly assigned patients to receive

    treatment with either IL-2 alone or IL-2 and IFN-. After 28

    patients were entered onto each arm, the IL-2/IFN arm was

    closed because of a failure to meet predetermined efficacy

    criteria. An additional 43 patients (total 71) were assigned to

    receive IL-2 alone. Responses were seen in 3 of 28 patients

    (11%) on IL-2/IFN- and 12 of 71 patients (17%) on IL-2

    alone. The Groupe Franais dImmunotherapie randomized

    425 patients with metastatic RCC to a continuous i.v. infusion

    of IL-2, s.c. injections of IFN-2a, or both [74]. Response

    Table 4B. RCC: randomized studies

    Author Year n Study Response Medianpatients design rates (%) survival (mos)

    IFN and IL-2

    Atkins [157] 1993 71 IL-2 0.6 MIU/kg i.v. q 8 h 14 11 15.5days 1-5 and 15-19

    28 IL-2 0.36 MIU/kg i.v. q 8 h 14 17 16IFN 3 MU/m2 IV q 8 h 14 NS NSdays 1-5 and 15-19

    Lissoni [78] 1993 15 IL-2 3 MIU s.c. bid 5 days/wk 6 wks 33 Survival at one

    15 IL-2 3 MIU s.c. bid 5 days/wk 6 wks 26 year: NS

    IFN 5 MU/m2 s.c. tiw NS

    Lummen [79] 1996 30 IFN-200 g s.c. q wk 0 13.0

    30 IL-2 4.8 MIU/m2 s.c. qid days 1, 22 23 13.0IL-2 4.8 MIU/m2 s.c. bid days 2, 23IL-2 2.4 MIU/m2 s.c. bid days 3-5, 8-12, (p = 0.01) (p = 0.49)IL-2 15-19, 24-26, 29-33, 36-40

    IFN 3 MU/m2

    days 3, 5, 24, 26IFN 6 MU/m2 days 8, 10, 12, 15, 17,IFN 19, 29, 31, 33, 36, 38, 40

    Negrier[74] 1998 138 IL-2 18 MIU/m2 qd CI 6.5 12days 1-5, 15-19 2 ; then days 1-5 q 3 wks

    147 IFN 18 MU s.c. tiw 10 wks 7.5 13

    140 IL-2 18 MIU/m2 CI qd days 1-5, 15-19 2; 18.6 17then days 1-5 q 3 wk (p = 0.01) (p = 0.55)IFN 6 MU s.c. tiw in the wks IL-2 is given.

    Jayson [77] 1998 29 IL-2 18 MIU s.c. days 1-5 3-4 wks 7 14.6

    29 IL-2 18 MIU s.c. days 1-5 3-4 wks 0 12.5IFN 9 MU tiw 3-4 wks (p = 0.98)

    Boccardo [75] 1998 22 IL-2 18 MIU/m2 CI qd days 1-4 wks 1,3, 22.7 307, 9, 13, 17, 21, 25

    22 IFN 6 MU/m2 IM days 1, 3, 5 52 wks 9.0 18

    22 IL-2 18 MIU/m2 CI qd days 1-4 wks 1,5, 9, 9.0 1313, 17, 21 NS NS

    IFN 6 MU/m2 i.m. days 1, 3, 5 wks 2-4, 6-8,10-12, 14-16, 18-20, 22-24

    Henriksson [76] 1998 63 Tamoxifen 40 mg PO qd 3.2* 13.3

    65 Tamoxifen 40 mg PO qd 7.7* 11.8

    IL-2 4.8 MIU/m2 q 8 h days 1, 22 NSIL-2 4.8 MIU/m2 q 12 h days 2, 23IL-2 2.4 MIU/m2 q 12 h day 3-5, 8-12,IL-2 15-19, 24-26, 29-33, 36-40

    IFN 3 MU/m2 days 3, 5, 24, 26,IFN 6 MU/m2 days 8, 10, 12, 15, 17,IFN 19, 29, 31, 33, 36, 38, 40

    NS = not significant; *complete response only

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    9/22

    42 Interferon in Oncological Practice

    rates and event-free survival rates were significantly better

    in the combination therapy group, but overall survival rates

    were not significantly different. The other randomized stud-

    ies assessing the role of combination IFN and IL-2 therapy

    failed to demonstrate a survival advantage to this combina-

    tion when compared with cytokine monotherapy [75-79]

    (Table 4B). In summary, as numerous studies have failed to

    show a survival advantage for patients with metastatic RCC

    receiving combination cytokine therapy, it cannot be rec-

    ommended as standard treatment.

    Combination with IL-2 and 5-FU

    Combinations of IFN-, IL-2, and 5-FU have been

    evaluated in metastatic RCC. In vitro data suggest

    improved efficacy of 5-FU when given in combination with

    IFN- [80, 81]. An overall response rate of approximately

    30% has been seen in most studies [73, 82-90]. Two recent

    randomized studies presented in abstract form address theefficacy of chemoimmunotherapy in metastatic RCC [91,

    92] (Table 4C). Negrier et al. randomized 131 patients

    between IL-2, IFN and IL-2, IFN and 5-FU, and reported no

    significant difference in response between the two treatment

    arms. Atzpodien et al. randomized patients between

    Tamoxifen and IFN, IL-2 and i.v. 5-FU, and found a highly

    significant improvement in response rate and survival in the

    combination therapy arm. The results from the Atzpodien

    study are encouraging, but in the absence of a confirmatory

    study, are insufficient to make IL-2, IFN, and 5-FU standard

    care in metastatic RCC.

    Summary

    In conclusion, IFN monotherapy in patients with metasta-

    tic RCC provides a modest but significant prolongation of

    survival with manageable side effects. A subset of patients,

    in particular those with good performance status, lung-only

    disease and resected primaries, may benefit from combina-

    tion chemoimmunotherapy, but more data are needed to

    determine whether combination therapy is superior to IFN

    alone. At the current time, the best approach is to enter

    patients into well-designed phase III trials that will help

    answer these questions.

    MELANOMA

    In 1980, Bart et al. reported on the inhibition of B16

    melanoma in vitro and in vivo by murine IFN [93]. Subsequent

    phase I studies using partially purified leukocyte IFN at doses

    below 10 106 U/day resulted in few responses [94]. Other

    reports using rIFN-2a 12 106 U/m2 three times a week (tiw)

    [95], and 50 106 tiw i.m., respectively [96], for three months

    revealed response rates at both dose levels of around 20%. IFN

    monotherapy in patients with metastatic melanoma inducesresponses in approximately 15% and CR rates on the order of

    5% [97]. Predictors of favorable response were uninterrupted

    schedules of therapy regardless of route, with no clear advan-

    tage of lower or higher dosages in the range between 10 106

    U/m2 and 50 106 U/m2 qd or tiw. Intermittent cyclic therapy

    was associated with a poorer outcome [97]. Suggestions of

    response durability set IFN apart from DTIC as a therapy for

    metastatic melanoma [98].

    Combination Therapy

    More recently, IFN- has been used in the context of com-bination therapy for the treatment of metastatic melanoma.

    Numerous trials have been performed combining IFN with

    IL-2, with single-agent chemotherapy, and with Tamoxifen

    (Table 5A). The addition of IFN to IL-2 did not result in a

    Table 4C. RCC: randomized studies

    Author Year n Study Response Medianpatients design rates (%) survival (mos)

    Chemoimmunotherapy

    Negrier[91] 1997 70 IL-2 9 MIU s.c. days 1-6 wks 1, 3, 5, 7 1.4 N/AIFN 6 MU s.c. days 1, 3, 5 wks 1,3, 5, 7

    61 IL-2 9 MIU s.c. days 1-6 wks 1, 3, 5, 7 8.2IFN 6 MU s.c. days 1, 3, 5 wks 1,3, 5, 75-FU 600 mg/m2 CI days 1-5 wks 1, 5 (p = 0.10)

    Atzpodien [92] 1997 41 IL-2 10 MIU/m2 s.c. bid days 3-5 wks 39 >42 mos1, 4; 5 MIU/m2 s.c. days 1, 3, 5 wks 2,3IFN 6 MU/m2 s.c. day 1 wks 1, 4 anddays 1, 3, 5 wks 2, 3IFN 9 MU/m2 days 1, 3, 5 wks 5-85-FU 1,000 mg/m2 q wk wks 5-8

    37 Tamoxifen 45 mg/m2 PO bid wks 1-8 0 14(p < 0.04)

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    10/22

    Jonasch, Haluska 43

    significantly greater response rate or survival, with signifi-

    cantly more side effects [99]. An early study looking at the

    combination of IFN and DTIC chemotherapy was promising

    [100], but subsequent investigations failed to demonstrate

    an advantage to this combination [101-103].

    Chemoimmunotherapy has been assessed in patients

    with metastatic melanoma [104-109]. Table 5B summarizessome of the major studies looking at this treatment modality.

    Although increased response rates have been shown in most

    of these trials, this has not translated into an increase in over-

    all survival. A recent abstract from the M.D. Anderson

    Cancer Center randomized patients between cisplatin, VBL

    and DTIC (CVD) and CVD plus sequential infusion IL-2 and

    IFN therapy. This study demonstrated a survival improve-

    ment in the chemoimmunotherapy arm that approached sta-

    tistical significance [110]. The major question in treating

    patients with metastatic melanoma who are eligible for these

    trials is whether the severe toxicities encountered with

    chemoimmunotherapy are worth the as-yet unproven survival

    benefits. The Eastern Cooperative Oncology Group (ECOG)

    3695/Cancer and Leukemia Group B (CALGB) 509802, a

    study currently accruing patients and randomizing them to

    CVD or concomitant CVD plus IL-2 and IFN, will hopefully

    provide information to help answer this question.

    Adjuvant Therapy

    Several trials looked at the role of IFN- in the adjuvant

    setting for melanoma patients at high risk for relapse, includ-

    ing patients with deep primary lesions and those with lymph

    node involvement [111-116]. Studies using relatively low

    doses of IFN (3 106 U tiw) failed to show an overall survival

    benefit in this patient group. In 1996, Kirkwood et al. pub-

    lished the results of ECOG 1684, which demonstrated both a

    disease-free and overall survival benefit for patients with

    stage III (lymph node-positive) melanoma treated with maxi-

    mally tolerated doses of IFN- [113]. The regimen used in

    Table 5A. Melanoma: randomized studies

    Author Year n Study Response Medianpatients design rates (%) survival (mos)

    IL-2 and IFN

    Sparano [99] 1993 44 IL-2 6 MU/m2 q 8 h i.v. 14 doses days 1-5, 15-19 5 10.2

    41 IL-2 4.5 MU/m2 i.v. q 8 h 14 doses days 1-5, 15-19 10 9.7IFN- 3 MU/m2 i.v. q 8 h 14 doses days 1-5, 15-19 NS

    IFN and chemotherapy

    Falkson [100] 1991 31 DTIC 200 mg/m2 i.v. days 1-5 q 28 days 20 9.6

    30 IFN- 15 MU/m2 i.v. M-F 3 wk, then 53 17.610 MU/m2 s.c tiw (p < 0.05) (p < 0.01)DTIC 200 mg/m2 i.v. days 1-5 q 28 d starting wk 4

    Thomson [103] 1993 83 DTIC 800 mg/m2 i.v. q 21 days 17 7.8

    87 DTIC (200-800 mg2) i.v. q 21 days 21 9.0IFN- 9 MU s.c. qd NS NS

    Bajetta [102] 1994 82 DTIC 800 mg/m2 i.v. q 21 days 20 11

    76 DTIC 800 mg/m2

    i.v. q 21 days 28 13IFN- 3 MU s,c, tiw

    84 DTIC 800 mg/m2 i.v. q 21 days 23 11IFN- 3 MU IM days 1-3, 6 MU days 4-6 NS NSthen 9 MU IM qd

    Falkson [101] 1998 69 DTIC 200 mg/m2 i.v. days 1-5 q 28 days 15 9.99

    68 IFN 15 MU/m2 i.v. days 1-5 wks 1-3, then 21 9.3510 MU/m2 s.c. tiwDTIC 200 mg/m2 i.v. days 1-5 q 28 days,starting day 22

    66 DTIC 200 mg/m2 i.v. days 1-5 q 28 days 19 9.54Tamoxifen 20 mg PO qdIFN 15 MU/m2 i.v. days 1-5 wks 1-3, then10 MU/m2 s.c. tiw

    68 DTIC 200 mg/m2 i.v. days 1-5 q 28 days 18 7.97Tamoxifen 20 mg PO qd NS (p = 0.85)

    NS = not significant

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    11/22

    44 Interferon in Oncological Practice

    ECOG 1684 involved administration of IFN- at 20 106 U/m2

    i.v. qd for five days/week for four weeks, followed by 10 106

    U/m2 s.c. tiw for 11 months. Based on the findings from this

    study, the FDA approved the uses of IFN- in stage III and

    stage IIB melanoma. The follow-up study ECOG 1690 showed

    a disease-free survival advantage in similar patient groups, but

    an overall survival advantage was not seen [114]. Of note, over-

    all survival of both the IFN and control groups in ECOG 1690

    is higher than the IFN arm in ECOG 1684. This may be due toa number of reasons, including patient crossover, or a favorable

    patient cohort in the control arm of ECOG 1690. Results from

    ECOG 1694, which randomized patients between standard

    high-dose IFN and GM-K, a ganglioside vaccine developed at

    the Memorial Sloan-Kettering Cancer Center, will be available

    soon. This study was closed prematurely, because a signifi-

    cantly greater number of relapses occurred in the vaccine arm.

    Summary

    Although concerns have been raised regarding the toxicity

    of IFN therapy, the authors recommend its use in the adjuvant

    setting. For stage IV disease, the role of IFN- is less clear.

    The promising response rates of chemoimmunotherapy have

    not translated into a hoped-for prolongation in survival.

    Nevertheless, in patients with good performance status, low

    disease burden and favorable disease sites (lung, skin),

    chemoimmunotherapy should be considered, preferably in

    the setting of a clinical trial. Results from several studies will

    be available in the near future to help clarify the role of IFN

    therapy in both the adjuvant and metastatic settings.

    KS

    IFN has been used in the treatment of HIV-associated

    KS since 1981 [117]. Initial treatment regimens employed

    doses in the 20 106 U/d range, which were associated with

    significant response rates, but also high levels of toxicity

    [118]. In subsequent studies [119], the probability of

    responding to therapy was correlated with CD4 count.

    Patients with CD4 counts greater than 400/mm3 responded,

    whereas none of those with CD4 counts of less than 150/mm3

    had a response.

    Table 5B. Melanoma: randomized studies

    Author Year n Study Response Medianpatients design rates (%) survival (mos)

    Chemoimmunotherapy

    Keilholz [105] 1997 66 Starting d 3: IL-2 18 MIU/m2 i.v. CI over 6 h, 18 9IL-2then 18 MIU/m2 i.v. CI over 12 h, thenIL-218 MIU/m2 i.v. CI over 24 h, thenIL-24.5 MIU/m2 i.v. CI qd 3 daysIFN- 10 MU/m2 s.c. days 1-5

    60 Starting d 3: IL-2 18 MIU/m2 i.v. CI over 6 h, 33 9IL-2then 18 MIU/m2 i.v. CI over 12 h, thenIL-218 MIU/m2 i.v. CI over 24 h, then (p = 0.04) NSIL-24.5 MIU/m2 i.v. CI qd 3 daysIFN- 10 MU/m2 s.c. days 1-5Cisplatin 100 mg/m2 day 1

    Rosenberg [109] 1999 52 Cisplatin 25 mg/m2 i.v. days 2-4 and 23-25 27 15.8DTIC 220 mg/m2 i.v. days 2-4 and 23-25Tamoxifen 20 mg PO qd starting day 1

    50 Cisplatin 25 mg/m2

    i.v. days 2-4 and 23-25 44 10.7DTIC 220 mg/m2 i.v. days 2-4 and 23-25Tamoxifen 20 mg PO qd starting day 1IFN- 6 MU/m2 s.c. days 5-8 and 26-29 (p = 0.071) (p = 0.052)IL-2 0.72 MIU/kg i.v. q 8 h days 5-8 and 26-29to patient tolerance

    Eton [110] 2000 92 Cisplatin 20 mg/m2 i.v. qd days 1-4, 22-25 25 9.5DTIC 800 mg/m2 i.v. day 1, 22VBL 2 mg/m2 i.v. days 1-4, 22-25

    91 Cisplatin 20 mg/m2 i.v. qd days 1-4, 22-25 48 11.8DTIC 800 mg/m2 i.v. day 1, 22VBL 1.5 mg/m2 i.v. days 1-4, 22-25 (p = 0.001) (p = 0.055)IL-2 9 MIU/m2 CI days 5-8, 17-20, 26-29IFN 5 MU/m2 s.c. days 5-9, 17-21, 26-30

    NS = not significant; CI = continuous infusion

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    12/22

    Jonasch, Haluska 45

    The combination of lower dosages of IFN- and zidovu-

    dine (AZT) was found to be effective in treating HIV-associ-

    ated KS, including those with lower CD4 counts [120], but

    with dose-limiting hematological side effects. In 1991,

    Scadden et al. reported that the use of GM-CSF as an adjunct

    to combination IFN-/AZT therapy resulted in an improved

    end-of-study absolute neutrophil count [121, 122]. Another

    report indicated that although GM-CSF decreased the inci-

    dence of neutropenia in patients on IFN- and AZT, other

    IFN-related side effects precluded any dose escalation [123].

    More recently, Shepherd et al. reported on treatment of HIV-

    related KS with AZT and IFN- at two dose levels. All

    patients received AZT 500 mg daily and were randomized to

    receive IFN- 1 106 U or 8 106 U s.c. qd. Response was

    reported in 31% of high-dose therapy and 8% of low-dose

    therapy patients (p = .011). Response at both dose levels was

    higher for patients with CD4 (+) counts greater than 150/mm3.

    The median time to progression was longer for patients in the

    8-million U arm (18 versus 13 weeks;p = .002). Both hema-

    tologic and nonhematologic toxicities were higher in the high-

    dose arm; 50 of 54 patients who received 8 106 U required

    dose alterations in the first four months compared with only

    19 of 53 patients who received 1 106 U (p = .0002) [124].

    A phase I/II study assessed effectiveness of IFN-therapy

    in the treatment of HIV-associated KS, and showed clinical

    response in 3 of 17 patients [125]. There does not appear to be

    any role for IFN-in the treatment of KS.

    For widespread, symptomatic KS, combination cyto-

    toxic chemotherapy is usually the treatment of choice[126]. Paclitaxel has recently been evaluated in the treat-

    ment of AIDS-related KS demonstrating promising results

    [127, 128]. The exciting initial findings with paclitaxel will

    prompt direct comparisons between paclitaxel and IFN-.

    Further study will allow us to determine the definitive role

    for IFN- therapy in KS.

    In summary IFN-, either as monotherapy or in combi-

    nation with AZT, shows activity in HIV-positive patients

    provided they possess relatively elevated CD4 (+) lympho-

    cyte counts (i.e., greater than 150/mm3). For more advanced

    disease, combination chemotherapy is currently the accepted

    means of treatment for KS.

    TOXICITIES

    Treatment with IFN is always considered in the context of

    its significant side-effect profile. Four major side effect groups

    occur: constitutional, neuropsychiatric, hematologic, and

    hepatic. These vary in degree, persistence and our ability to

    manage them. Side effects can also be divided into those that

    occur acutely but decrease over time, and those that are chronic.

    The severity of many side effects appears to be directly related

    to the dose and duration of IFN therapy [129]. This section

    summarizes the major types of toxicity and their etiology, and

    concludes with proposed toxicity management guidelines.

    Acute Toxicity

    Toxicity can be divided into acute and chronic manifesta-

    tions. In the acute setting, the patients experience fevers, chills

    and rigors, usually between 3 and 6 h after receiving IFN.

    Patients may also experience headaches, myalgias, and

    malaise. With prolonged and uninterrupted administration of

    IFN, tolerance can develop to these symptoms. However,

    treatment breaks as short as a few days can result in the rede-

    velopment of rigors and chills. Transaminitis and neutropenia

    may occur within the first few days of treatment, and can be

    controlled by adjusting the IFN dose. Both resolve rapidly

    upon cessation of treatment. If the transaminitis is not followed

    closely, it can result in fatal hepatotoxicity [113].

    Chronic Toxicity

    The chronic symptoms experienced by patients on IFN

    include fatigue (70%-100% of patients), anorexia (40%-70%),

    and neuropsychiatric symptoms (up to 30%). These symp-

    toms appear to be dose-related, and cumulative, worsening

    over time [9].

    Mechanisms of Toxicity

    The mechanisms of IFN-induced toxicity are unclear, but

    are most likely multifactorial. These will be summarized in

    the following section.

    Fatigue

    Fatigue is the most common symptom associated with

    chronic use of IFN-, occurring in more than 70% of patients.

    It is frequently the dose-limiting toxicity [130]. The exact eti-

    ology of the fatigue is unclear, but there appears to be both a

    psychological and neuromuscular component [130, 131].

    Chronic fatigue generally worsens with continued therapy,

    does not exhibit tolerance, is dose-related, and does not

    respond to therapy with steroids or antiinflammatory drugs.

    Little research has been done to analyze the neuromuscular

    axis in patients on IFN to determine whether there are clear

    structural or biochemical changes that can be quantified.

    Assessment with muscle biopsy and other diagnostic modali-

    ties has not been performed. Clearly, further study into the eti-

    ology of IFN-induced fatigue is needed to better manage this

    prevalent and debilitating side effect. The first step will be to

    build into clinical trials an accurate, objective and reproducible

    evaluation of patient fatigue.

    Neurological Toxicities

    Central nervous system toxicities include somnolence,

    confusion, lethargy, dizziness, and impaired mental status

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    13/22

    46 Interferon in Oncological Practice

    [9]. Peripheral nervous system toxicities include numbness

    and tingling.

    Duration of therapy appears to be the most important

    factor in determining the degree of cognitive impairment

    [132, 133]. Absolute dose appears to be less important.

    Caraceni et al. evaluated neurotoxicities of 113 patients

    receiving IFN- 3 106 U s.c. tiw for 36 months as part of a

    randomized trial. Particular emphasis was placed on the devel-

    opment of extrapyramidal signs and symptoms, psychiatric

    symptoms, attention, memory, reasoning capability, psycho-

    logical adaptation, and quality of life [134]. Significant differ-

    ences between study and control groups included a higher

    incidence of action tremor in the treatment group. Cognitive

    performance did not differ between the two groups, but a

    higher level of anxiety was recorded in the IFN group. In the

    quality-of-life assessment, there was a significantly greater

    number of patients who experienced fatigue.

    Mood Disorders

    Patients with cancer in general have a higher risk of

    developing clinical depression [133]. A significant minority

    of patients on IFN therapy becomes depressed, in some

    cases leading to suicide. There are also several reports of

    mania in patients on IFN [135-138]. Mood disorders can

    occur in patients without predisposing factors or past history

    of psychiatric problems.

    In non-cancer patients, depression is associated with alter-

    ations in a number of bodily systems, including the endocrine

    system [133]. The mechanism by which IFN causes psychi-atric disturbances is poorly understood, and research into the

    mechanisms underlying endogenous depression has been

    performed in patients receiving IFN to better understand the

    etiology of IFN-induced mood disturbances. Neuroendocrine

    disturbances, including perturbation of the hypothalamic-

    thyroid-adrenal axis and alteration in dopamine and sero-

    tonin production have all been implicated [139], as have

    alterations in the secretion of secondary cytokines, espe-

    cially IL-1 [133].

    Endocrine Dysfunction

    In 1983,Ernstoff et al. reported that human leukocyte

    IFN administration resulted in an increase in cortisol lev-

    els via adrenocorticotropic hormone (ACTH) stimulation

    [140]. More recent studies revealed that IFN- and IFN-

    both induced ACTH, prolactin, growth hormone and corti-

    sol levels in patients [141], although other investigators did

    not find any effect of IFN- on anterior pituitary function

    [142]. An assessment of IFN--induced endocrine stimu-

    lation in patients with myeloproliferative disorders

    revealed that on day 1 of therapy, a significant stimulation

    of the hypothalamic-pituitary axis was apparent, an effect

    that had disappeared by the third week of therapy [143].

    The acute stimulatory effect of IFN- on cortisol release

    appears to be mediated by the release of hypothalamic

    corticotropin releasing hormone [143].

    There have been reports of alterations in the levels of sex

    hormones during IFN therapy, and male sexual dysfunction

    does occur [139]. The hypothalamic-pituitary-gonadal axis

    can be suppressed during acute and chronic illness, and this

    effect appears to be cytokine-mediated [139]. Thus it is pos-

    sible that IFN-induced alteration in sex hormone levels is

    mediated via direct or indirect pathways.

    Autoimmune-Mediated Thyroid Dysfunction

    Thyroid dysfunction occurs in 8%-20% of patients

    receiving IFN- therapy [139]. Numerous studies report the

    development of overt or subclinical hyper- or hypothyroidism

    in patients on IFN- for various malignancies [144-149]. The

    pattern demonstrated by most patients is one of an autoim-mune thyroiditis, with a period of hyper- followed by hypo-

    thyroidism [139]. A few patients with hepatitis C have

    developed symptoms of Graves disease while on IFN-

    [139]. Investigation into etiologies of IFN--induced thy-

    roiditis suggests an indirect effect via activation of other

    cytokines, including IFN-. Preexisting autoimmune dis-

    ease or baseline serological abnormalities appear to predis-

    pose patients to developing overt autoimmune disease

    while on IFN- [147, 148]. Antithyroid antibodies occur in

    up to 16% of women and 1.5% to 3% of men in the general

    population, and individuals expressing baseline antithyroidantibodies have a 60% risk of developing clinical thyroid

    disease during the course of therapy [139]. Treatment of MS

    patients with IFN- was shown to induce antithyroid

    autoantibodies and symptoms of overt hypothyroidism in a

    small series of patients [150]. Treatment with IFN-para-

    doxically did not appear to induce the same number of

    autoimmune side effects as IFN- [151]. A recent study

    correlated the degree of autoimmune disease with improved

    prognosis in renal cancer patients treated with IFN- and

    IL-2, suggesting that autoimmune disease while on cytokine

    therapy is a marker for breaking immunologic tolerance in

    patients with cancer [152].

    Although it is unclear how much of the IFN-mediated

    toxicity is due to endocrine dysfunction, a serum thyroid

    stimulating hormone (TSH) determination is indicated in a

    patient on IFN who complains of severe and persistent

    fatigue, cold sensitivity, or other symptoms suggestive of

    hypothyroidism.

    STUDIES AND SIDE-EFFECT PROFILES

    Due to the variation in dosing and schedule, dose adjust-

    ment, duration of follow-up, data presentation, and disease

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    14/22

    Jonasch, Haluska 47

    type in which IFN has been administered, it is difficult

    to make direct comparisons of side-effect profiles seen in

    various studies.

    Some general trends can be observed. In Table 6, side

    effects resulting from treatment of melanoma, NHL and KS

    with high-dose levels of IFN (10 106 to 50 106 U/m2) are

    compared. Fatigue symptoms at this dose level occur at levels

    approaching 90% in the studies that categorized this symptom.

    A very high incidence of fevers is also recorded. The one study

    that categorized depression recorded a rate of 47%. Other sideeffects are difficult to compare due to the incompleteness of the

    data, but in general it appears that the incidence of severe toxi-

    cities (grade III or greater) is much greater at these higher doses.

    Table 7 summarizes the side-effect profiles of patients

    who received IFN dosages in the range of 3 106 U to

    5 106 U/m2 for a variety of cancers. Although direct com-

    parisons may not be quantitative, and several studies only

    recorded the incidence of grade 3 and higher toxicities, in

    general the degree of constitutional toxicities appears to be

    considerably lower than is seen in the studies with the higher

    doses seen in Table 6, and the incidence of dose-limiting

    hepatic, neuropsychiatric and hematologic toxicities appears

    to be negligible.

    QUALITY OF LIFE

    Due to the substantial toxicities encountered during IFN

    therapy, a number of groups have looked at the quality of life

    of patients undergoing IFN therapy. Cole et al. evaluated the

    quality of life of patients receiving high-dose IFN therapy in

    the ECOG 1684 trial for stage III melanoma using the

    Quality-Adjusted Time Without Symptoms and Toxicity(Q-TWiST) methodology [153], which estimates the mean

    time spent in a series of clinical health states that differ in

    terms of quality of life. Q-TWiST adjusts each time period

    depending on how much value the patient places on the

    quality of life of each health state [153]. Even if patients

    were to place a very low value on the time spent on IFN,

    there would still be a net prolongation in overall survival on

    the ECOG 1684 trial.

    The same Q-TWiST methodology was used to evaluate

    the quality-of-life-adjusted survival in the previously men-

    tioned trial randomizing patients to doxorubicin-based

    Table 6. Summary of toxicities in patients receiving high-dose IFN for malignancies

    Study Real[117] Creagan [96] Kirkwood[113] Creagan [116] Sertoli [159] Creagan [95] OConnell[160]

    Disease/stage Kaposis/HIV Melanoma IV Melanoma III Melanoma III Melanoma IV Melanoma IV NHL, CLL

    Type/dose IFN-2a 36 IFN-2a 50 IFN-2b 20 IFN-2a 20 IFN-2b IFN-2a 12 IFN- 12

    MU IM qd MU/m2

    i.m. MU/m2

    IV MU/m2

    i.m. tiw 10 MU/m2

    i.m. MU/m2

    i.m. MU/m2

    i.m. tiw 4 wks, then tiw 12 wks 5 days/wk 12 wks tiw tiw 12 wks 8 wks, thentiw 4 wks, then until disease weekly if response,

    10 MU/m2 s.c. progression and 25 MU/m2 tiwtiw 11 mo 4 wks if stable

    disease

    n patients on IFN 34 31 143 131 21 30 20

    Degree of All Mod- All Grade All Grade All Grade Mod- All Severeseverity Grades Severe Grades III Grades III Grades III Severe Grades

    % % % % % % % % % % %

    Constitutional 97 48Fatigue 88 87 89 20 100 43 50 90 10Anorexia 74 58 55 3 13 47 55 10Weight loss 13Fevers 94 83 24 100 10 80 85 5Myalgias 74 63 7 27Headache 62 44 5 45 0

    Hepatic 64 16 N/A N/A 19 0 0 10 5

    Neuropsychiatric 83 28 11 2 5 3Depression 21Dizziness/vertigo 47Confusion 23 15 5

    5 5

    Hematologic 64 24 N/A N/A 29 0Thrombocytopenia 35 0Anemialeukopenia 45 0

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    15/22

    48 Interferon in Oncological Practice

    combination chemotherapy with or without IFN-2b for

    the treatment of advanced follicular lymphoma [63, 154].

    An advantage in terms of quality-adjusted survival was seen

    in the patients receiving IFN regardless of the value placed

    on the time patients were undergoing IFN therapy [154].

    Kilbridge et al. interviewed 107 patients with superficial

    melanomas who did not require IFN therapy, asking them

    what value they place on a prolongation of life secondary to

    IFN therapy, were they to need it. The authors assessed patient

    preference for a particular health state as a function of toxicity

    and outcome. A great majority of respondents indicated

    they preferred experiencing even severe treatment-related

    side effects for a gain in disease-free survival [155].

    MANAGING IFN TOXICITY

    Communication among all members of the treating team

    is imperative in managing patients on IFN therapy. Each

    team member is in a position to obtain complementary infor-

    mation that, when put together, gives a complete picture of

    the patients side-effect profile. The following section will

    summarize management of the major IFN side effects.

    Constitutional Side Effects

    Constitutional side effects are managed with copious

    hydration and acetaminophen. Acetominophen is useful in

    controlling the shakes, fevers and myalgias experienced by the

    patient at the onset of therapy. Nonsteroidal anti-inflammatory

    medications may be used if acetaminophen is not adequate

    in controlling these symptoms, but should be used only if

    adequate control is not achieved with acetaminophen alone.

    There is no recognized pharmacological antidote for

    IFN-induced fatigue. It is equally unclear what can be done

    to alleviate this symptom short of dose decrement, or cessa-

    tion of treatment. Suggestions include mild exercise, good

    nutritional intake, copious fluid ingestion, and stress man-

    agement techniques. The treating team needs to remember

    that a treatment-induced decrement in functional status can be

    a serious blow to a patients self-esteem and sense of indepen-

    dence [130], and that patients need encouragement and positive

    feedback from their treating team. On the other hand, patients

    also need to be told that it is acceptable to draw on social

    resources, which include family, friends, or hospital-based

    social services if the patients feel they are unable to cope.

    Table 7. Summary of toxicities in patients receiving low-dose IFN for malignancies

    Study Real[117] Grob [158] Doveil[115] Ozer [39] Kantarjian [49]

    Disease/stage Kaposis/HIV Melanoma II Melanoma III CML chronic-phase CML chronic-phase

    Type/dose IFN IFN-2a 3 IFN-2a 3 Mu IFN-2b 3 IFN-2b 5 IFN- 5MU i.m. qd s.c. tiw 18 mos MU i.m. MU/m2 s.c. qd MU/m2 s.c.

    4 wks, then tiw 12 qd + Ara-Ctiw until disease mos 10 mg s.c. qdprogression until progression

    n patients on IFN 36 248 50 107 140

    Degree of severity All Grades All Grades Grade III Grade III Grade III Grade III% % % % % %

    ConstitutionalFatigue 83 48

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    16/22

    Jonasch, Haluska 49

    Mood Disorders

    Forty percent of patients on IFN will experience some

    degree of mood impairment, and up to 10% may become

    overtly depressed [113, 130]. It is important that the treatment

    team be aware of the potential for depression, and to incorpo-

    rate questions about the patients mood and emotional state

    into the patients routine evaluation. Early psychiatric referral

    is important, and IFN-related mood disorders can often be

    managed with measures similar to those used to treat endoge-

    nous depression. The occurrence of mania in patients on IFN

    also needs to be kept in mind, especially after patients

    undergo significant dose-reduction or treatment breaks [138].

    The clinicians natural instinct is to interdict the use of IFN

    in patients with any past history of depression, or a strong fam-

    ily history of mood disorders. We recommend that patients

    with any of the above risk factors be evaluated prior to treat-

    ment by a psychiatrist versed in the assessment and manage-

    ment of cancer patients, and ideally with prior experience in

    treating IFN-related mood disorders [138]. These patients

    should not be denied treatment, but should be followed closely

    for signs of depression, with mood-related questions as part of

    the review of systems at each visit.

    Hepatotoxicity

    In patients receiving high-dose i.v. IFN for stage III

    melanoma, liver function tests (LFTs) need to be drawn at

    least weekly, assessing for grade III transaminitis.

    Withholding treatment until LFTs decrease to a grade I tox-

    icity and restarting with a 30%-50% dose reduction is astandard approach. Recently, a report was published sug-

    gesting that less aggressive dose reduction can be per-

    formed with no decrease in patient safety or compliance

    [161]. Once a steady-state level of treatment is achieved,

    LFTs are fairly stable and assessment of liver function can

    be decreased to monthly or bimonthy blood draws.

    Hematologic Toxicity

    Weekly blood draws assessing for grade III neutropenia

    need to be performed in patients receiving high-dose i.v.

    IFN. Dose-reduction algorithms are the same as those for

    hepatotoxicity. In patients on s.c. IFN regimens, once a

    steady-state of treatment is established, neutrophil counts

    are fairly stable, although they may drift down further in

    some patients. In general, blood draws can be decreased to

    a monthly or even bimonthly schedule once stable dosing

    occurs.

    Endocrine Toxicity

    Recognition of the occurrence of hypothyroidism is

    important in the management of patients on IFN. Initially,

    monthly TSH measurements should be performed, and if

    three sequential values are stable and within a normal range,

    bimonthly or quarterly evaluations may be sufficient.

    SUMMARY AND RECOMMENDATIONS

    IFN is a promising but incompletely understood anti-

    cancer agent. Clinical trials have established a number of

    indications for the IFNs in both the hematological and solid

    tumor arenas, although the effectiveness of the IFNs is modest

    in most applications of the agent.

    The side-effect profiles of the IFNs are substantial, and at

    high doses, daunting, making the choice of IFN therapy one

    that has to be weighed against the near certainty of a decrement

    in quality of life while on the agent. It is important that the

    treating physician be aware of the four major categories of IFN

    toxicity: constitutional, neuropsychiatric, hepatic, and hemato-

    logic. An ongoing dialogue must occur between the patient

    and the treating team to ensure that all aspects of IFN toxicity

    are addressed. A number of steps can be taken to minimize themorbidity of IFN therapy, resulting in an improvement in both

    quality of life and patient compliance.

    IFN has been FDA-approved for use in a number of clin-

    ical applications. Treatment with IFN outside of these recom-

    mendations should be performed in the context of a clinical

    trial designed to assess immunological endpoints and to care-

    fully measure type and degree of toxicities. Companion stud-

    ies designed to ask specific questions regarding the etiology

    of IFN-induced toxicities should also be encouraged, as this

    knowledge will be extremely useful in the evolution of IFN as

    a useful agent in the oncological armamentarium.

    REFERENCES

    1 Isaacs A, Lindenman J. Virus interference I. The interferon.

    Proc R Soc 1957;147:258-267.

    2 Pestka S. The human interferon-alpha species and hybrid

    proteins. Semin Oncol 1997;24(suppl 9):S9-4-S9-17.

    3 Interferon nomenclature. Nature 1980;286:110.

    4 Haque SJ, Williams BR. Signal transduction in the inter-

    feron system. Semin Oncol 1998;25(suppl 1):14-22.

    5 Aguet M, Grobke M, Dreiding P. Various human interferon

    alpha subclasses cross-react with common receptors: their

    binding affinities correlate with their specific biological

    activities. Virology 1984;132:211-216.

    6 Merlin G, Falcoff E, Aguet M. 125I-labelled human inter-

    ferons alpha, beta and gamma: comparative receptor-binding

    data. J Gen Virol 1985;66:1149-1152.

    7 Pfeffer LM, Dinarello CA, Herberman RB et al. Biologicalproperties of recombinant alpha-interferons: 40th anniversaryof the discovery of interferons. Cancer Res 1998;58:2489-2499.

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    17/22

    50 Interferon in Oncological Practice

    8 Sreevalsan T. Biological therapy with Interferon-alfa and beta:

    preclinical studies. In: DeVita VTJ, Hellman SMD, Rosenberg

    SA, eds. Biologic Therapy of Cancer, Second Ed. Philadelphia:

    J.B. Lippincott Company, 1995;347-364.

    9 Weiss K. Safety profile of interferon-alpha therapy. Semin

    Oncol 1998;25(suppl 1):9-13.

    10 Mark DF, Lu SD, Creasey AA et al. Site-specific mutagene-

    sis of the human fibroblast interferon gene. Proc Natl Acad

    Sci USA 1984;81:5662-5666.

    11 Witt PL, Storer BE, Bryan GT et al. Pharmacodynamics of

    biological response in vivo after single and multiple doses of

    interferon-beta. J Immunother 1993;13:191-200.

    12 Jacobs LD, Cookfair DL, Rudick RA et al. Intramuscular inter-

    feron beta-1a for disease progression in relapsing multiple scle-

    rosis. The Multiple Sclerosis Collaborative Research Group

    (MSCRG) [published erratum appears in Ann Neurol

    1996;40:480]. Ann Neurol 1996;39:285-294.

    13 Wills RJ. Clinical pharmacokinetics of interferons. Clin

    Pharmacokinet 1990;19:390-399.

    14 Goldstein D, Sielaff KM, Storer BE et al. Human biologic

    response modification by interferon in the absence of measur-

    able serum concentrations: a comparative trial of subcutaneous

    and intravenous interferon-beta serine. J Natl Cancer Inst

    1989;81:1061-1068.

    15 Sarna G, Pertcheck M, Figlin R et al. Phase I study of recom-

    binant beta ser 17 interferon in the treatment of cancer. Cancer

    Treat Rep 1986;70:1365-1372.

    16 Rinehart JJ, Malspeis L, Young D et al. Phase I/II trial of

    human recombinant interferon gamma in renal cell carcinoma.

    J Biol Response Mod 1986;5:300-308.

    17 Shah I, Band J, Samson M et al. Pharmacokinetics and tolerance

    of intravenous and intramuscular recombinant alpha 2 interferon

    in patients with malignancies. Am J Hematol 1984;17:363-371.

    18 Zhou A, Hassel BA, Silverman RH. Expression cloning of 2-

    5A-dependent RNAase: a uniquely regulated mediator of

    interferon action. Cell 1993;72:753-765.

    19 Hovanessian AG, Wood JN. Anticellular and antiviral

    effects of pppA(2p5A)n. Virology 1980;101:81-90.

    20 Rysiecki G, Gewert DR, Williams BR. Constitutive expression

    of a 2,5-oligoadenylate synthetase cDNA results in increased

    antiviral activity and growth suppression. J Interferon Res

    1989;9:649-657.21 Creasey AA, Eppstein DA, Marsh YV et al. Growth regula-

    tion of melanoma cells by interferon and (2 -5)oligoadeny-

    late synthetase. Mol Cell Biol 1983;3:780-786.

    22 Merritt JA, Ball LA, Sielaff KM et al. Modulation of 2,5-

    oligoadenylate synthetase in patients treated with alpha-inter-

    feron: effects of dose, schedule, and route of administration.

    J Interferon Res 1986;6:189-198.

    23 Huber C, Fuchs D, Hausen A et al. Pteridines as a new

    marker to detect human T cells activated by allogeneic or

    modified self major histocompatibility complex (MHC)

    determinants. J Immunol 1983;130:1047-1050.

    24 Gastl G, Aulitzky W, Tilg H et al. A biological approach to

    optimize interferon treatment in hairy cell leukemia.

    Immunobiology 1986;172:262-268.

    25 Huber C, Aulitzky W, Tilg H et al. Studies on the optimal

    dose and the mode of action of alpha-interferon in the treat-

    ment of hairy cell leukemia. Leukemia 1987;1:355-357.

    26 Buzaid AC, Grimm EA, Ali-Osman F et al. Mechanism ofthe anti-tumour effect of biochemotherapy in melanoma:

    preliminary results. Melanoma Res 1994;4:327-330.

    27 Anderson CM, Buzaid AC, Sussman J et al. Nitric oxide and

    neopterin levels and clinical response in stage III melanoma

    patients receiving concurrent biochemotherapy. Melanoma Res

    1998;8:149-155.

    28 Bernengo MG, Quaglino P, Cappello N et al. Macrophage-

    mediated immunostimulation modulates therapeutic efficacy

    of interleukin-2 based chemoimmunotherapy in advanced

    metastatic melanoma patients. Melanoma Res 2000;10:55-65.

    29 Roers A, Hochkeppel HK, Horisberger MA et al. MxA gene

    expression after live virus vaccination: a sensitive marker forendogenous type I interferon. J Infect Dis 1994;169:807-813.

    30 Simon A, Fah J, Haller O et al. Interferon-regulated Mx genes

    are not responsive to interleukin-1, tumor necrosis factor, and

    other cytokines. J Virol 1991;65:968-971.

    31 von Wussow P, Jakschies D, Hochkeppel HK et al. The human

    intracellular Mx-homologous protein is specifically induced by

    type I interferons. Eur J Immunol 1990;20:2015-2019.

    32 Quesada JR, Reuben J, Manning JT et al. Alpha interferon for

    induction of remission in hairy-cell leukemia. N Engl J Med

    1984;310:15-18.

    33 Quesada JR, Hersh EM, Manning J et al. Treatment of hairy

    cell leukemia with recombinant alpha-interferon. Blood1986;68:493-497.

    34 Golomb HM, Jacobs A, Fefer A et al. Alpha-2 interferon ther-

    apy of hairy-cell leukemia: a multicenter study of 64 patients.

    J Clin Oncol 1986;4:900-905.

    35 Verma DS, Spitzer G, Gutterman JU et al. Human leukocyte

    interferon preparation blocks granulopoietic differentiation.

    Blood 1979;54:1423-1427.

    36 Talpaz M, McCredie KB, Mavligit GM et al. Leukocyte inter-

    feron-induced myeloid cytoreduction in chronic myelogenous

    leukemia. Blood 1983;62:689-692.

    37 Talpaz M, Kantarjian HM, McCredie K et al. Hematologic

    remission and cytogenetic improvement induced by recombinant

    human interferon alpha A in chronic myelogenous leukemia.

    N Engl J Med 1986;314:1065-1069.

    38 Silver RT, Woolf SH, Hehlmann R et al. An evidence-based

    analysis of the effect of busulfan, hydroxyurea, interferon, and

    allogeneic bone marrow transplantation in treating the chronic

    phase of chronic myeloid leukemia: developed for the

    American Society of Hematology. Blood 1999;94:1517-1536.

    39 Ozer H, George SL, Schiffer CA et al. Prolonged subcutaneous

    administration of recombinant alpha 2b interferon in patients

    with previously untreated Philadelphia chromosome-positive

    chronic-phase chronic myelogenous leukemia: effect on remis-

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    18/22

    Jonasch, Haluska 51

    sion duration and survival: Cancer and Leukemia Group B

    study 8583. Blood 1993;82:2975-2984.

    40 Interferon alfa-2a as compared with conventional chemother-

    apy for the treatment of chronic myeloid leukemia. The Italian

    Cooperative Study Group on Chronic Myeloid Leukemia. N

    Engl J Med 1994;330:820-825.

    41 Hehlmann R, Heimpel H, Hasford J et al. Randomized com-

    parison of interferon-alpha with busulfan and hydroxyurea

    in chronic myelogenous leukemia. The German CML Study

    Group. Blood 1994;84:4064-4077.

    42 Allan NC, Richards SM, Shepherd PC. UK Medical Research

    Council randomised, multicentre trial of interferon- alpha n1

    for chronic myeloid leukaemia: improved survival irrespective

    of cytogenetic response. The UK Medical Research Councils

    Working Parties for Therapeutic Trials in Adult Leukaemia.

    Lancet 1995;345:1392-1397.

    43 Ohnishi K, Ohno R, Tomonaga M et al. A randomized trial

    comparing interferon-alpha with busulfan for newly diag-

    nosed chronic myelogenous leukemia in chronic phase. Blood1995;86:906-916.

    44 Broustet A, Reiffers J, Marit G et al. Hydroxyurea versus

    interferon alfa-2b in chronic myelogenous leukaemia: prelim-

    inary results of an open French multicentre randomized study.

    Eur J Cancer 1991;27(suppl 4):S18-S21.

    45 Randomized study on hydroxyurea alone versus hydroxyurea

    combined with low-dose interferon-alpha 2b for chronic

    myeloid leukemia. The Benelux CML Study Group. Blood

    1998;91:2713-2721.

    46 Sokal JE, Leong SS, Gomez GA. Preferential inhibition by

    cytarabine of CFU-GM from patients with chronic granulocytic

    leukemia. Cancer 1987;59:197-202.47 Robertson MJ, Tantravahi R, Griffin JD et al. Hematologic

    remission and cytogenetic improvement after treatment of sta-

    ble-phase chronic myelogenous leukemia with continuous infu-

    sion of low-dose cytarabine. Am J Hematol 1993;43:95-102.

    48 Kantarjian HM, Keating MJ, Estey EH et al. Treatment of

    advanced stages of Philadelphia chromosome-positive

    chronic myelogenous leukemia with interferon-alpha and

    low-dose cytarabine. J Clin Oncol 1992;10:772-778.

    49 Kantarjian HM, OBrien S, Smith TL et al. Treatment of

    Philadelphia chromosome-positive early chronic phase chronic

    myelogenous leukemia with daily doses of interferon alpha

    and low-dose cytarabine. J Clin Oncol 1999;17:284-292.50 Guilhot F, Chastang C, Michallet M et al. Interferon alfa-2b

    combined with cytarabine versus interferon alone in chronic

    myelogenous leukemia. French Chronic Myeloid Leukemia

    Study Group. N Engl J Med 1997;337:223-229.

    51 Kurzrock R, Talpaz M, Kantarjian H et al. Therapy of chronic

    myelogenous leukemia with recombinant interferon-gamma.

    Blood 1987;70:943-947.

    52 Kloke O, Wandl U, Opalka B et al. A prospective randomized

    comparison of single-agent interferon (IFN)-alpha with the

    combination of IFN-alpha and low-dose IFN-gamma in chronic

    myelogenous leukaemia. Eur J Haematol 1992;48:93-98.

    53 Wandl UB, Opalka B, Kloke O et al. Treatment of chronic

    myelogenous leukemia with different cytokines. Semin Oncol

    1992;19(suppl 4):88-94.

    54 Rodriguez J, Cortes J, Smith T et al. Determinants of prog-

    nosis in late chronic-phase chronic myelogenous leukemia.

    J Clin Oncol 1998;16:3782-3787.

    55 Tomas JF, Lopez-Lorenzo JL, Requena MJ et al. Absence ofinfluence of prior treatment with interferon on the outcome of

    allogeneic bone marrow transplantation for chronic myeloid

    leukemia. Bone Marrow Transplant 1998;22:47-51.

    56 Zuffa E, Bandini G, Bonini A et al. Prior treatment with

    alpha-interferon does not adversely affect the outcome of

    allogeneic BMT in chronic phase chronic myeloid leukemia.

    Haematologica 1998;83:231-236.

    57 Morton AJ, Gooley T, Hansen JA et al. Association between

    pretransplant interferon-alpha and outcome after unrelated

    donor marrow transplantation for chronic myelogenous

    leukemia in chronic phase. Blood 1998;92:394-401.

    58 Ozer H, Wiernik PH, Giles F et al. Recombinant interferon-alpha therapy in patients with follicular lymphoma [published

    erratum appears in Cancer 1998;83:593]. Cancer 1998;82:1821-

    1830.

    59 Price CG, Rohatiner AZ, Steward W et al. Interferon-alpha 2b

    in the treatment of follicular lymphoma: preliminary results of

    a trial in progress. Ann Oncol 1991;2(suppl 2):141-145.

    60 Rohatiner A, Crowther D, Radford J et al. The role of inter-

    feron in follicular lymphoma. Proc Am Soc Clin Oncol

    1996;15:418.

    61 Peterson BA, Petroni GR, Oken MM et al. Cyclophos-

    phamide versus cyclophosphamide plus interferon alfa-2b in

    follicular low-grade lymphomas: an intergroup phase III trial

    (CALGB 8691 and EST 7486). Proc Am Soc Clin Oncol

    1997;16:14a.

    62 Smalley RV, Andersen JW, Hawkins MJ et al. Interferon alfa

    combined with cytotoxic chemotherapy for patients with non-

    Hodgkins lymphoma. N Engl J Med 1992;327:1336-1341.

    63 Solal-Celigny P, Lepage E, Brousse N et al. Doxorubicin-con-

    taining regimen with or without interferon alfa-2b for

    advanced follicular lymphomas: final analysis of survival and

    toxicity in the Groupe dEtude des Lymphomes Folliculaires

    86 Trial. J Clin Oncol 1998;16:2332-2338.

    64 McLaughlin P, Cabanillas F, Hagemeister FB et al. CHOP-

    Bleo plus interferon for stage IV low-grade lymphoma. Ann

    Oncol 1993;4:205-211.

    65 Hiddemann W, Unterhalt M, Koch P et al. New aspects in the

    treatment of advanced low-grade non-Hodgkins lymphomas:

    prednimustine/mitoxantrone versus cyclophosphamide/vincris-

    tine/prednisone followed by interferon alfa versus observation

    onlya preliminary update of the German Low-Grade Lym-

    phoma Study Group. Semin Hematol 1994;31(suppl 3):32-35.

    66 Unterhalt M, Herrmann R, Tiemann M et al. Prednimustine,

    mitoxantrone (PmM) vs cyclophosphamide, vincristine, pred-

    nisone (COP) for the treatment of advanced low-grade non-

    Hodgkins lymphoma. German Low-Grade Lymphoma Study

    Group. Leukemia 1996;10:836-843.

  • 7/30/2019 Interferon in Oncological Practice_Review of Interferon

    19/22

    52 Interferon in Oncological Practice

    67 Unterhalt M, Herrmann R, Nahler M et al. Significant prolon-

    gation of disease-free survival in advanced low grade non-

    Hodgkins lymphomas (NHL) by interferon alfa maintenance.

    Blood 1995;86:439a.

    68 Hagenbeek A, Carde P, Somers R et al. Interferon-alfa-2a vs

    control as maintenance therapy for low grade non-Hodgkins

    lymphoma: results from a prospective randomized clinicaltrial. Proc Am Soc Clin Oncol 1995;14:386.

    69 Aviles A, Duque G, Talavera A et al. Interferon alpha 2b as

    maintenance therapy in low grade malignant lymphoma

    improves duration of remission and survival. Leuk Lymphoma

    1996;20:495-499.

    70 Savage PD, Muss HB. Renal cell cancer. In: De Vita VT Jr,

    Hellman S, Rosenberg SA, eds. Biologic Therapy of Cancer

    Second Ed. Philadelphia: J.B. Lippincott Company,

    1995;373-387.

    71 Interferon-alpha and survival in metastatic renal carcinoma:

    early results of a randomised controlled trial. Medical Research

    Council Renal Cancer Collaborators. Lancet 1999;353:14-17.

    72 Pyrhonen S, Salminen E, Ruutu M et al. Prospective ran-

    domized trial of interferon alfa-2a plus vinblastine versus

    vinblastine alone in patients with advanced renal cell cancer.

    J Clin Oncol 1999;17:2859-2867.

    73 Bukowski RM. Natural history and therapy of metastatic

    renal cell carcinoma: the role of interleukin-2. Cancer

    1997;80:1198-1220.

    74 Negrier S, Escudier B, Lasset C et al. Recombinant human

    interleukin-2, recombinant human interferon alfa-2a, or both in

    metastatic renal-cell carcinoma. Groupe Francais dImmuno-

    therapie. N Engl J Med 1998;338:1272-1278.

    75 Boccardo F, Rubagotti A, Canobbio L et al. Interleukin-2,

    interferon-alpha and interleukin-2 plus interferon-alpha in

    renal cell carcinoma. A randomized phase II trial. Tumori

    1998;84:534-539.

    76 Henriksson R, Nilsson S, Colleen S et al. Survival in renal

    cell carcinomaa randomized evaluation of tamoxifen vs

    interleukin 2, alpha-interferon (leucocyte) and tamoxifen. Br

    J Cancer 1998;77:1311-1317.

    77 Jayson GC, Middleton M, Lee SM et al. A randomized phase

    II trial of interleukin 2 and interleukin 2- interferon alpha in

    advanced renal cancer. Br J Cancer 1998;78:366-369.

    78 Lissoni P, Barni S, Ardizzoia A et al. A randomized study of

    low-dose interleukin-2 subcutaneous immunotherapy versus

    interleukin-2 plus interferon-alpha as first line therapy formetastatic renal cell carcinoma. Tumori 1993;79:397-400.

    79 Lummen G, Goepel M, Mollhoff S et al. Phase II study of inter-

    feron-gamma versus interleukin-2 and interferon-alpha 2b in

    metastatic renal cell carcinoma. J Urol 1996;155:455-458.

    80 Houghton JA, Morton CL, Adkins DA et al. Locus of the inter-

    action among 5-fluorouracil, leucovorin, and interferon-alpha

    2a in colon carcinoma cells. Cancer Res 1993;53:4243-4250.

    81 Morita T, Tokue A. Biomodulation of 5-fluorouracil by

    interferon-alpha in human renal carcinoma cells: relation-

    ship to the expression of thymidine phosphorylase. Cancer

    Chemother Pharmacol 1999;44:91-96.

    82 Lopez Hanninen E, Kirchner H, Atzpodien J. Interleukin-2

    based home therapy of metastatic renal cell carcinoma: risks

    and benefits in 215 consecutive single institution patients.

    J Urol 1996;155:19-25.

    83 Hofmockel G, Langer W, Theiss M et al. Immunochemo-

    therapy for metastatic renal cell carcinoma using a regimen

    of interleukin-2, interferon-alpha and 5-fluorouracil. J Urol1996;156:18-21.