Radiolabelled Peptides and Monoclonal Antibodies

1 downloads 0 Views 380KB Size Report
GPC Biotech ..... 2 diabetes, severe sepsis, multiple sclerosis, Guillain-Barre syn- ...... crucially involved in Guillain-Barré syndrome and experimental al-.
Current Pharmaceutical Design, 2008, 14, 2401-2414

2401

Radiolabelled Peptides and Monoclonal Antibodies for Therapy Decision Making in Inflammatory Diseases G. Malviya1, A. Signore1,2,*, B. Laganà3 and R.A. Dierckx1 1

Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, University of Groningen, The Netherlands; 2Nuclear Medicine Unit, II Faculty of Medicine and Surgery, “Sapienza” University of Rome, Italy and 3Unit of Rheumatology Allergy and Immunology, II Faculty of Medicine and Surgery, “Sapienza” University of Rome, Italy Abstract: Radiolabelled peptides and monoclonal antibodies are an emerging class of radiopharmaceuticals for imaging inflammation with clinical implications for several chronic inflammatory disorders for diagnosis, therapy decision making and follow up. In the last decades, a number of novel monoclonal antibodies and peptides have been introduced for the treatment of different inflammatory disorders and also labelled with a variety of radionuclides depending upon the specific applications, diagnostic or therapeutic, by using direct or indirect methods. These radiopharmaceuticals bind to their targets with high affinity and specificity and therefore have an excellent diagnostic potential for the imaging of patients with chronic inflammatory diseases. In this review article we describe the characteristics of peptides, cytokines and monoclonal antibodies with a particular emphasis on their role in therapy decision making and follow up in different inflammatory diseases.

Key Words: Cytokines, monoclonal antibodies, radiopharmaceuticals, molecular imaging, inflammation, therapy decision making, diagnosis, nuclear medicine. INTRODUCTION Nuclear medicine contributes to the diagnosis of inflammatory diseases and helps in therapy decision making and follow-up of patients. Radiolabelled peptides and monoclonal antibodies provide an ideal scintigraphic tool for the diagnosis of several inflammatory disorders. Therapy decision is generally based on the pathophysiological and patho-biochemical examinations and on clinical history of patients. Nevertheless, therapy, and particularly biological therapy, can often fail due to the absence or low expression of therapeutic target molecules in the inflammatory lesions. Therefore, before the selection of therapy, the clinician must know not only whether the inflammation exists and where it is located, but also which type of cells are involved and, most importantly, what type of receptors or markers are present in the inflammatory lesion. Radiolabelled peptides and monoclonal antibodies are highly specific radiopharmaceuticals for their targets and a positive scintigraphic image shows the presence of their target molecules in the inflammatory lesion. This approach may also provide an explanation for the failure of any particular targeted therapy or the rationale to select specific therapy for the patient with an inflammatory disease. Peptides and cytokines that have been radiolabelled and used as radioactive probes for in vivo histological characterization of tissues include, IL-1, IL-1ra, IL-2, IL-12 p40, IL-8, IFN- and MCP-1 labelled with 123I, 125I, 99mTc or 35S mainly for Graves’ ophtalmopathy, Type 1 Diabetes Mellitus, Celiac disease, Crohn’s disease, thyroid autoimmune diseases, kidney graft rejection, atherosclerosis, lung inflammatory diseases and rheumatoid arthritis. Radiolabelled monoclonal antibodies include anti-TNF-, anti-CD20, antiCD3, anti-CD4, anti-MIF and anti-E-selectin monoclonal antibodies labelled with 99mTc or 111In mainly for rheumatoid arthritis, Crohn’s disease, focal inflammatory lesions, vascular endothelial activation and sentinel lymph node detection (SLN). All these peptides and antibodies will be reviewed in this manuscript with particular emphasis, to their clinical application and use for therapy decision making. *Address correspondence to this author at the Nuclear Medicine Department, “Sapienza” University of Rome, St. Andrea Hospital, Via di Grottarossa 1035, 00189 Rome, Italy; Tel: +39-06-33775471; E-mail: [email protected]

1381-6128/08 $55.00+.00

PATHOPHYSIOLOGY OF INFLAMMATORY PROCESSES Inflammation is a physiological response to invasion by an infection agent, antigen challenge or even just a tissue injury, which could be acute or chronic. An acute inflammation has a rapid onset and it lasts hours or few days, whereas chronic inflammation develops when antigen stimulation persists and it can last months to several years and may lead to late complications. Chronic and acute inflammation is mainly identified by its cause and the time duration of inflammation. Chronic inflammation mainly occurs when any foreign micro-organism or antigen is able to evade clearance by immune system (e.g. tuberculosis) or if the antigen itself continuously causes the activation of T cells (e.g. autoimmune diseases). Chronic inflammation is histologically characterised by little or no oedema and accumulation of activated macrophages, T and B cells at the site of inflammation with induction of cytokine production. Cytokines stimulate the collagen production and fibroblast proliferation, which may result in chronic fibrosis [1]. CYTOKINES IN INFLAMMATORY DISEASES Cytokines are molecules of 5-70 kilo Daltons (kDa) and belong to the class of hormones or growth factor like proteins and glycoproteins. Cytokines play a prominent role in the homeostatic control of the immune system as well as of other organs, both in physiology and pathology. Cytokines are produced by different types of cells (mainly T cells and macrophages). They communicate via specific cell receptors expressed on a known cell population to induce specific cell activities mainly for the regulation of cell function, homing, motility and metastasis. Cytokine receptors, usually of high affinity, are normally present at low levels on non-activated cells, but expression is up regulated during the cell activation. Therefore, these receptors on the affected tissue are suitable targets for the detection of inflammation (Table 1). Interleukin 1 (IL-1) and Interleukin 1 Receptor Antagonist (IL1ra) Interleukin 1 (IL-1), a 17-kD protein, has two forms: IL-1 and IL-1, that bind to two different receptors on a wide variety of cells. Type I IL-1 receptors (80-kDa glycoprotein), are expressed on T cells, hepatocytes, fibroblasts and endothelial cells, whereas type II IL-1receptors (68-kDa glycoprotein) are express on B cells, neutro-

© 2008 Bentham Science Publishers Ltd.

2402 Current Pharmaceutical Design, 2008, Vol. 14, No. 24 Table 1.

Malviya et al.

Radiolabelled Cytokines for Inflammation Imaging

Cytokine

Source

Targets

Activity

Clinical Use

IL-1 /

Mo, M, Fi, T, B, NK, Ep, En, SMS, Ke

IL1RI*=B, Mo, N, IL1RII=En, Fi, He, Ke, T

Acute phase response; activation of T, B, M, NK

Inflammatory processes in animal model

123

I, 125I

[4, 5]

IL-1ra

Mo

IL-1R

Endogenous IL-1 inhibitor without initiating signal transduction. High binding affinity to IL-1R

Inflammatory processes in animal model, Rheumatoid arthritis

123

I, 125I

[7, 9]

IL-2

T

T, B, NK

Growth/differentiation of T, B; activation of NK

Graves’ ophthalmopathy, Type 1 Diabetes, Celiac disease, Crohn’s disease, Thyroid autoimmune disease, Kidney graft rejection, Cutaneous melanoma, Atherosclerosis, Kidney allograft.

I, 125I, Tc, 35 S

[15-39]

Chemoattractant and activator of N, Ba, Lym. Angiogenic activity

Detection of sterile inflammation and osteomyelitis in animal model and in humans

Tc, 123I, I, 131I

[40-45] [49, 50]

Activation of cytotoxic activity of CTL, LAK, M, NK

Lymphocytic infiltrates in animal models, Imaging T lymphocytes

IL-8

Mo, Gr, Lym, Fi, En

N, Ba, T

IL-12

Mo, M, B, NK, Ke

T, NK

MCP-1

En, SMS

Mo, M, Gr

IFN-

T , N K , Ep , SMS,NC

Many Different cells

Chemoattractant chemokine, induces Mo Subacute inflammation in animal models migration and activation, upregulation of the MCP-1 receptor for the CCR-2 peptide. Inhibition of cell growth; activation of Mo, M, NK, Fi, SMS

Lung inflammatory diseases

Isotope

123

Ref.

99m

99m

125

125

I

[56]

Tc

[63, 64]

99m

123

I

[66]

Abbreviations: B = B lymphocytes; Ba = Basophils; Ep = Epithelial cells; En = Endothelial cells; Fi = Fibroblast; Gr = Granulocyte; Ke = Keratinocyte; Lym = Lymphocytes; Mo = Monocytes; M = Macrophage; T = T lymphocytes; N = Neutrophil; NC = Neoplastic Cells; NK = Natural Killer cells; SMS = Smooth cell muscle; *IL1RI, IL1RII, TNF-R1, TNFR2: receptor types I and II.

phils, monocytes and macrophages. Both IL-1 and IL-1 bind to both receptors with affinity in the pico-molar range [2, 3]. Several studies in animal models and in humans demonstrated its specific accumulation at site of inflammation. In inflammatory lesions the accumulation of 125I labelled IL-1 correlates with mRNA expression for the type I and type II IL1-Rs. In neutropenic mice, a significantly lower 125I-IL-1 uptake and type II IL-1R mRNA expression was confirmed [4]. Another study was performed to check if 125I labelled IL-1 specifically localises in the inflammatory foci by binding to its receptors on infiltrated leukocytes. In mice with S. aureus induced inflammation, after i.v. injection of 120 ng (0.4 MBq) of 125I-IL-1, a rapid blood clearance and accumulation of 125I-IL-1 in the inflammatory foci was observed and the uptake reached its maximum value within 2 h after injection. The accumulation of 125I-IL-1 in inflammation was consistently higher than that of 125I-IL-1 . Microscopic autoradiography showed the presence of radiolabelled IL1 in the areas of leukocytic infiltration of inflamed tissue, which confirmed the specificity of its uptake to cell receptors [5]. In addition, Granowitz et al. demonstrated that the accumulation of IL-1 could be inhibited by the use of anti-IL-1 receptor antibodies, as a further proof of specificity of binding [6]. Unfortunately, when IL-1 administered in humans it showed some side effects (such as headache and hypertension) even at very low dose of 10 ng/kg, this limited the clinical application of radiolabelled IL-1. Therefore, as an alternative, the equally sized (17 kDa) naturally occurring IL-1 receptor antagonist (IL-1ra) was radiolabelled for imaging inflammation, which binds to the IL-1 receptor with similar high affinity and showed no biological effects up to a dose of 10 mg/kg [6]. Two studies with radiolabelled IL-1ra demonstrated that it targets to inflamed joints in patients with active rheumatoid arthritis [7, 8] within 1 hour after injection. Disappointingly, authors found a similar uptake in affected joints when using 123I labelled albumin as

control protein [8]. An important autoradiography study concluded that neither in vivo images nor in vitro autoradiography clearly indicate that radiolabelled IL-1ra binds to IL-1 receptors in inflammatory foci, and therefore joint uptake in patients may be due to non-specific mechanisms as well as specific receptor binding [9]. The use of this radiolabelled cytokines need further investigation before including them in the field of molecular imaging of inflammation for diagnostic purposes in humans. Interleukin 2 (IL-2) IL-2 is a single chain glycoprotein of 133 amino acids (mol. weight 15.5 kDa) that is mainly produced by activated T cells [10]. IL-2 interacts with a specific receptor expressed by activated T lymphocytes and plays a prominent role in regulating the immune mediated response through long term T cell proliferation and stimulates the growth and differentiation of B cells, NK cells, monocytes, macrophages, and oligodendrocytes [11-14]. In inflammatory conditions, infiltrating cells express high affinity IL-2 receptors that are targeted by radiolabelled IL-2. Initially, interleukin 2 was labelled with 35S, 14C 125I and 131I for in-vitro studies and in-vivo biodistribution studies in animal models [15-18]. Later, IL-2 was radiolabelled with 123I using Bolton and Hunter method or lactoperoxidase/glucose oxidase technique and its in-vitro receptor binding affinity was evaluated on activated lymphocytes [18]. In animals, 123I-IL-2 was tested in diabetic animal models of rats and mice, which showed higher radioactivity uptake in pancreas and lymph nodes as compared to normal animals [1921]. In these studies authors also confirmed the specificity of receptor binding by autoradiography. In a study in patients with active Crohn’s disease, 123I-IL-2 was able to detect the presence of activated T lymphocytes in the inflamed bowel. Ex-vivo autoradiographic examinations demonstrated specific binding of labelled-IL-2 to IL-2R+ve mononuclear cells infiltrating the gut wall [22]. Another study in patients with celiac disease showed a positive correlation between the uptake of 123I-IL-2 measured by gamma

Radiolabelled Peptides and Monoclonal Antibodies

camera and the number of histologically determined IL-2 receptorpositive cells in the inflamed jejunal mucosa [23]. However, 123I is a cyclotron radionuclide and therefore is expensive and not readily available, whereas 99mTc readily available at low cost led to the development of 99mTc-IL-2. Chianelli et al. [24] described a two-step synthesis method for 99mTc-IL-2 using the bifunctional chelating agent S-tetrahydrofurfurylacetyl (thio2,3,5,6-tetrafluorophenyl)-adipylglycylglycine. In-vitro binding experiment demonstrated that the binding capability of 99mTc-IL-2 was not altered by the labelling procedure. Also the animal studies in mice model showed no significant uptake of 99mTc-IL-2 in major organs except in the kidneys and, to a lesser extent, in the liver and spleen in normal mice whereas, iodine-labelled IL-2 showed a higher degree of kidney uptake and a lower degree of liver uptake. In particular, Annovazzi et al. [25] performed a comparative scintigraphic study between 99mTc-IL-2 and 99mTc-labeled granulocytes, in detecting the presence and extent of bowel inflammation, in 29 patients with inactive Crohn’s disease (>12 months). This study demonstrated that 18 patients (62%) had a positive 99mTc-IL2 scan and 18 (62%) a positive 99mTc-WBC scan, but only 12 patients (41.4%) were positive on both scans although IL-2 and granulocyte bowel uptake sites were usually located in different parts. Both 99mTc-IL-2 and 99mTc-WBC scintigraphy showed a high negative predictive value for disease recurrence (1.00 and 0.91, respectively) but a weak positive predictive value (0.44 and 0.39, respectively). Signore et al. [26] performed a validation study in 30 patients with melanoma lesions in which they were able to demonstrate that peri-tumoural infiltrating T-cells bind IL-2 to different degree thus providing important in vivo information for therapy decision of the metastatic melanoma with this cytokine. This has also been demonstrated in patients with Head and Neck cancer by Van de Wiele and colleagues [27, 28] but not in patients with hypernephroma [29]. Recently, Annovazzi et al. [30] studied 99mTc-IL-2 scintigraphy in patients with carotid atherosclerosis before and after treatment with a statin or a hypocholesterolaemic diet and concluded that this radiopharmaceutical can identify plaques vulnerable to rupture in which statin treatment may down-regulate the inflammation. This is a beautiful example, how a receptor specific radiolabelled-cytokine can provide in vivo information of the activity state of disease and help for therapy decision making.

Current Pharmaceutical Design, 2008, Vol. 14, No. 24

2403

Similarly, radiolabelled IL-2 showed specific accumulation in inflamed tissue in several other disease including Graves’ disease, melanoma, carotid atherosclerosis, type 1 diabetes mellitus, prediabetes, Hashimoto’s thyroiditis, graves disease, Takayasu’s vasculitis [31-36]. A pilot study conducted in patients with Graves’ ophtalmopathy (GO) showed the very specific accumulation of 99mTc labelled IL-2 in the inflamed region of the eye (as also published by others [3739]) (Fig. 1). In particular we evaluated the presence of CD25 (IL-2 receptor) antigen in the inflamed eyes by scintigraphic examination with radiolabelled IL-2 in GO patients, before starting anti CD25 monoclonal antibody (Simulect®) therapy and three months after the therapy. In this study, we found that the patients who showed high uptake of 99mTc-IL-2 before unlabelled anti-CD25 monoclonal antibody (mAb) therapy responded much better than the patients who showed very less uptake of 99mTc-IL-2 before starting antiCD25 mAb therapy (Fig. 2). This is an excellent demonstration of how a receptor specific radiolabelled-cytokine can provide in vivo information of the disease activity state and also help for therapy decision before starting targeted monoclonal antibody therapy. These and other studies (both in animals and in humans) make the labelled-IL-2 the most deeply and carefully investigated radiopharmaceutical for imaging chronic T-cell mediated inflammatory diseases. At present, although not yet commercially available (a company is evaluating its commercialization), radiolabelled IL-2 is used by many European centres for diagnostic purposes and for therapy decision making in order to determine the appropriateness of antiIL-2R antibody therapy in several inflammatory diseases and graft rejection, as well as for therapy follow-up. Interleukin 8 (IL-8) Interleukin 8 (IL-8) is a chemotactic cytokine or “chemokine”, member of CXC subfamily, in which first two cysteine residues are separated by one amino acid residue. IL-8 has a molecular weight of only 8.5 kDa, binds to CXCR1 and CXCR2 receptors on neutrophils with high affinity (0.3-4x10-9 mol/L) and promotes chemotaxis of these cells. In 1994, Hay et al. reported for the first time the use of radiolabelled IL-8 for imaging inflammation in carrageenaninduced sterile inflammation in rats [40]. They showed that the radiopharmaceutical highly accumulates in lesions at 1-3 h post

Fig. (1). Transaxial image of the head of a patient with severe Graves’ exophthalmos injected with 99mTc labelled IL-2 (left) and its corresponding MRI image (right) and fused images (centre). The scintigraphic image shows the presence of CD-25 receptor in the muscles of the left eye (much less in the right eye) thus providing in vivo histological imaging of activated T-cells infiltrating the retro-bulbar tissues an indication to be treated with unlabelled anti-CD25 mAb (Simulect®).

2404 Current Pharmaceutical Design, 2008, Vol. 14, No. 24

Malviya et al.

Fig. (2). Images of inflamed eye region of 2 patients with severe Graves’ exophthalmos after 99mTc labelled IL-2 administration. Left images (A and C) were acquired before therapy and right images (B and D) were acquired three months after therapy with anti-CD25 mAb (Simulect®). Patient 1 (image A and B) was clinically responding to anti-CD25 mAb therapy and indeed showed high IL-2 accumulation in the right eye (a sign of active infiltration) whereas patient 2 (image C and D) did not respond to therapy and indeed showed no uptake ofradiolableed IL-2 before therapy.

injection. The target-to-background (T/B) ratio could not exceed 2.5 whereas the level of accumulation of another chemokine NAP-2 (neutrophil activating peptide 2) showed a higher T/B ratio of 5.3 at 72 h p.i. [40, 41]. A study with 123I labelled human recombinant IL8 in eight patients with osteomyelitis, demonstrated that 123I-IL-8 accumulates in the inflammatory foci [42]. Although, systemic administration of IL-8 may induce pronounced side-effects on the numbers and subtypes of circulating leukocytes [43, 44], studies with radiolabelled IL-8 demonstrated very specific accumulation within few hours of injection both in animal models and in humans [45, 46] with minor side-effects. It has been reported that the labelling method largely affects the biodistribution of radiolabelled IL-8. IL-8 radioiodinated via the Bolton-Hunter method cleared much earlier from the background as compared to IL-8 iodinated with Iodogen or Chloramine T method [47]. IL-8 has also been radiolabelled with 99mTc using HYNIC as a chelating agent, with a high specific activity (SA) and it was further optimised by using alternative co-ligands such as nicotinic acid and tricine, to stabilize the 99mTc-IL8 complex [48]. The high SA of 99m Tc-IL-8 reduces substantially the dose of protein to be injected and therefore possible side effects. Studies to evaluate in-vivo imaging capability of 99mTc-labelled IL-8 in rabbit models of osteomyelitis and colitis demonstrated the highly specific accumulation of 99m Tc-IL-8 in the inflammatory lesions [49]. In another study, with 99m Tc-HYNIC-IL8 in the rabbit model of chemically induced colitis, this radiopharmaceutical allowed a meticulous evaluation of disease severity within 4h after administration [50].

Nevertheless, due to its binding to neutrophils, radiolabelled IL8 finds its main clinical application for diagnosis and follow-up on bacterial infections rather than sterile chronic inflammatory diseases. A radiolabelled analogue of IL-8 is being investigated by a pharmaceutical company in order to commercialise it. Interleukin-12 (IL-12) Interleukin-12 is a heterodimeric cytokine of 75 kDa, formed by two covalently linked glycosylated chains of approximately 40 kDa and 35 kDa (IL-12 p40 and IL-12 p35, respectively). It induces IFN- production by T cells and NK cells, enhances NK and ADCC activity and co-stimulates peripheral blood lymphocyte proliferation [51-53]. IL-12 plays a central role in the induction of T helper1 (Th1) cell development, acting in antagonism with other cytokines (such as IL-4, IL-10) that favour differentiation of Th2 cells [54,55]. IL-12 p40 was radiolabelled with 125I by enzymatic method with a labelling efficiency of 58-79%. In-vitro binding assay with KIT225 cell and Scatchard plot analysis demonstrated that it retains its specific binding capacity after labelling. In-vivo biodistribution and targeting experiments showed its specific uptake to the Th1+ve cells [56]. These promising results reveal that the radiolabelled IL12 could be used as a prominent radiopharmaceutical for the diagnosis of Th1-mediated inflammatory disorders. Recently Annovazzi et al., radiolabelled IL-12 with 99m-technetium, by using succinimidyl-6-hydrazinopyridine-3-carboxylate (HYNIC-NHS) as a co-ligand [56]. The labelling efficiency of 99m Tc-IL-12 ranged between 75% to 85% and saturation binding analysis showed a Kd of 2.09 nM. 99mTc-IL-12 demonstrated sig-

Radiolabelled Peptides and Monoclonal Antibodies

nificant uptake in the inflamed colon of mice with TNBS-induced colitis, which also correlates with the degree of bowel lymphocytic infiltration at histology. Since this cytokine is involved in the first steps of inflammation induction and in the cascade of events that lead to chronic inflammation, several modern therapeutic approaches aim to block the effects of this cytokine. In this context, the availability of a radiolabelled IL-12 could be of great value for therapy follow-up but it remains a research product. Monocyte Chemotactic Protein-1 (MCP-1) Although MCP-1 is a chemotactic peptides not expressed in normal vessels, several stimulations such as TNF- and other cytokines, induce the expression and secretion of MCP-1 by endothelial cells, vascular smooth muscle cells (SMCs), or cardiomyocytes, which, by triggering and sustaining leukocyte accumulation, may in turn promote chronic inflammation [57-60]. Elevated levels of circulating MCP-1 have been found in patients with congestive heart failure and coronary artery disease and acute coronary syndromes [61, 62]. Hartung et al. performed a study with 99mTc-labelled MCP-1 to detect the concentration of MCP-1 receptors on inflammatory cells in vulnerable atherosclerotic plaques [63]. They induced atherosclerotic lesions by balloon de-endothelialisation of the abdominal aorta, followed by a high cholesterol diet in the experimental model of rabbits. Imaging 3 hours after the i.v. injection of 99m Tc-labelled MCP-1, showed uptake of radioactivity in the atherosclerotic lesions. The mean quantitative MCP-1 accumulation in atherosclerotic lesions was 4-fold higher than in control healthy rabbit aorta. These data were also confirmed by histological examination. Another study was performed to check the imaging capability of MCP-1 to image subacute and chronic inflammation [64] induced in rats by intramuscular injection of turpentine in the thigh. Imaging performed 1 h after i.v. injection of 37-148 MBq (1-4 mCi) of 99mTc-labelled MCP-1, demonstrated an elevated binding of MCP-1 in inflamed thigh than in control on days 1-5 after turpentine injection. Moreover, an autoradiographic examination in animals co-injected with 125I-BSA (bovine serum albumin) showed specific location of MCP-1 to infiltrating monocytes/macrophages but not of BSA. In the same study, 99mTc-labelled annexin-V was also used for imaging apoptotic cells and the authors concluded that 99m Tc-labeled MCP-1 and 99mTc-labelled annexin-V both accumulate in the inflammatory lesions in relation to the density of monocytes/macrophages and the presence of apoptotic cells, respectively. Both the above studies demonstrate the possible use of 99mTclabelled MCP-1 for the detection of inflammatory lesions, with particular regard to macrophage rich lesions such as atherosclerosis. Also in this case, radiolabelled MCP-1 remains a research product. IFN- Interferons (IFN) are a group of cytokines with antiviral activity. These molecules are produced at site of inflammation or injury and mainly act locally. IFN- is known for its capability to induce cells to block or inhibit the replication of different variety of viruses. Mainly T cells and natural killer (NK) cells produce IFN-, in response to different kind of stimulations. It plays an important role in the regulation of mononuclear phagocytes, B-cell switching to certain IgG classes and inhibition or support of the development of T helper (Th) cells [65]. IFN- was radiolabelled with 123I with a specific activity of 48 mCi/mg and a study was performed in 10 normal subjects to determine the dose response and radiation absorption of 123I labelled IFN- and also its biodistribution in human and safety [66]. An inhalation scintigraphic study with 123I-IFN-, using a Pari-Master nebuliser, demonstrated that the maximum lung deposition was achieved within 10 min after administration and the uptake of 123IIFN- was inversely correlated to lung function and probably as a

Current Pharmaceutical Design, 2008, Vol. 14, No. 24

2405

consequence of binding to alveolar macrophages. Highest deposition was found in the central middle parts of lungs, although, a homogeneous lung deposition was also present in peripheral areas. Nevertheless, 50% of the total dose was present in the lungs within 30 min from inhalation, and was reduced to 6% after 24 hours. No serious side-effects were observed during this study but despite this, this radiolabelled cytokines did not stimulate the interest of many centres neither pharmaceutical companies for possible commercialisation. However, studies with 123I labelled IFN- could be very helpful in patients with lung diseases, particularly who are selected for IFN- therapy. RADIOLABELLED MONOCLONAL ANTIBODIES IN INFLAMMATORY DISEASES In the last decade, radiolabelled monoclonal antibodies (mAbs) and their Fab’ fragments were developed as a new class of radiopharmaceuticals for radioimmuno-scintigraphy. These radiolabelled-mAbs allow an excellent molecular imaging of target molecules involved in several immune-mediated disorders. In particular, biological therapies are now widely used to suppress TNF, CD20, CD3 or CD4 antigens. The availability of these radiolabelled mAb is therefore extremely useful for therapy decision making. Generally, these monoclonal antibodies are named with a suffix ‘-mab’ which indicates their animal origin. Different classes of antibodies can be identified by considering their infix, like murine mAbs have ‘-o-’, chimeric mAbs use ‘-xi-’ and humanised mAbs use ‘-zu-’, in their names. However, there is no special meaning for the prefix in the name of mAbs, but it should be different from other drugs (Table 2). Anti-TNF Monoclonal Antibodies Infliximab (Remicade®) is a chimeric IgG1 mAb, with murine variable (Fv) domain of mouse anti-human TNF- antibody and constant (Fc) sequences of human IgG1, produced by recombinant cell culture technique. It specifically recognises and binds to both soluble and membrane-bound TNF- with high avidity and high affinity (Ka = 1010 M-1) forming stable non-dissociating immune complexes [67]. Infliximab has a median half-life of 9.5 days. This binding neutralises the biological activity of TNF- by inhibiting the binding of TNF- to its receptor [68, 69]. Studies demonstrated that infliximab can induce cell lysis in transmembrane TNF- expressing cells in-vitro [68] and in-vivo [70], mediated by ADCC or CDC [71, 72]. Infliximab is licensed for the treatment of moderate to severely active rheumatoid arthritis, Crohn’s disease, ankylosing spondylitis, psoriatic arthritis and ulcerative colitis. Infliximab has been labelled with 99mTc, using direct radiolabelling method with a high labelling efficiency (LE) of more than 97%. High performance liquid chromatography (HPLC) analysis of 99m Tc-labelled infliximab showed only one peak in the radiogram profile corresponding to the radiolabelled antibody [73]. A scintigraphic imaging study was reported by Conti et al., in a patient with arthritis to assess the degree of TNF- mediated inflammation in the affected knee [74]. The patient underwent scintigraphic examination with 99mTc-labelled infliximab before and 4 months after intra-articular infliximab therapy. After the injection of 99mTc-infliximab (15 mCi), planar image of the inflamed joint were acquired at 6 h and 24 h. Scintigraphy showed an intense accumulation of 99mTc-infliximab in the affected knee at diagnosis, which represents the high levels of intra-lesional TNF-, but no uptake was detectable in the joint after therapy. Another pilot study was performed on seven RA patients [75]. In this study, nine inflamed joints were examined with 99mTcinfliximab scintigraphy. The RA patients underwent radiolabelled infliximab scintigraphic examination before and three months after the intra-articular infliximab therapy. Images were acquired at 3 h, 6 h and 24 h after the injection of 99mTc-infliximab (15 mCi). In this study, post-treatment scintigraphy demonstrated that different

2406 Current Pharmaceutical Design, 2008, Vol. 14, No. 24 Table 2.

Malviya et al.

Radiolabelled Monoclonal Antibodies for Imaging Inflammation

mAb Name (Commercial Name)

Company

Type

Target

Infliximab (Remicade®)

Centocor/ Schering-Plough

Chimeric IgG1

TNF-

Isotope

Clinical Use

Ref.

99m

Crohn’s Disease, Rheumatoid Arthritis

[73-75, 77]

Tc

Adalimumab (Humira®)

Abbott

Fully human IgG1

TNF-

99m

Rheumatoid Arthritis (RA)

[81, 83]

1D09C3

GPC Biotech

Humanised IgG4

HLA-DR

99m

Tc

Experimental stage

[132]

CD25

211

At

Therapy and imaging T-cell leukemia

[147]

Therapy and imaging of T-cell leukemia

[145, 146, 148, 149]

Basiliximab (Simulect®)

Novartis

Chimeric IgG1

Tc

18

F, 99mTc, 111In, 125 212 I, Bi, 67Ga

Daclizumab (Zenapax®)

Roche

Humanised IgG1

CD25

Muromonab

Ortho Pharma

Murine IgG2a

CD3

99m

RA in animal model and Humans

[118, 119]

Visilizumab (Nuvion®)

Protein Design Labs

Humanized IgG2

CD3

99m

T cell traffic in animal model

[132]

MAX.16H5

--

Murine IgG1

CD4

99m

RA in animal model and Humans

[114, 115]

Tc Tc Tc

125

Anti-MIF

--

--

MIF

I

Inflammation imaging in animal model

[151]

Anti E-Selectin

--

Murine IgG1

E-Selectin

111

In

RA in animal model and Humans

[84, 90-94]

Rituximab (Mabthera®)

Roche

Chimeric IgG1

CD20

99m

Tc

RA, sentinel lymph node (SLN)

[107-109]

amounts of radiopharmaceutical can be detected in the treated joints after therapy: 3 joints showed a significant decrease in uptake whereas in other 4 joints the uptake was only slightly reduced and was unchanged in 2 joints. Thus, post-therapy scintigraphy can be useful in evaluating residual presence of TNF- in joints and provide a rationale for a new cycle of therapy. In a comparative study, in experimental colitis model of rats, scintigraphic images and histological examination was performed after 99mTc-infliximab and 99mTc-tin-colloid-labelled-leukocytes. In the inflamed colon, 99mTc-infliximab accumulation was much less than 99mTc-leucocyte accumulation (T/B of 2.6+0.3 versus 41.2+ 16.1 at 4 h post injection respectively) suggesting a limited role of labelled-infliximab in colitis [76]. In order to evaluate this aspect in humans, we have recently published a study with 99mTc labelled infliximab in patients with active Crohn’s Disease (CD), to investigate in-vivo bio-kinetics of the anti-TNF- antibody, to predict the clinical response of anti-TNF therapy and to compare these scans with 99mTc-labelled-leukocytes [77]. We found little TNF- in the affected bowel of patients with active CD and almost never in the same regions were leukocytes accumulate. Therefore, we concluded that, the clinical benefit that patients have from anti-TNF- therapy is unlikely to be the consequence of a local reduction of TNF-. The mechanism of action, in therapeutic doses, therefore deserves further investigation. Nevertheless infliximab, being a chimeric mAb may trigger the human anti-chimeric antibody (HACA) response, with possible reduction of the therapeutic benefit and efficacy of therapy and possible induction of false negatives at scintigraphy. The first “fully human” monoclonal antibody against TNF-, adalimumab was engineered through the phase display technology. Adalimumab is a recombinant human IgG1 antibody, composed of two kappa light chains (24 kDa each) and two IgG1 heavy chains (49 kDa each), expressed in CHO cells [78]. Adalimumab is indicated for the treatment of rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis and moderate to severely active Crohn’s disease. It recognizes both soluble and membrane-bound TNF- with high specificity and high affinity (KD=6x10-10M) and inhibits its biological activity by blocking interaction with p55 and p75 cell surface TNF receptors [79, 80]. This fully human antibody minimised potential side effects and antigenicity of previous chimeric and humanised mAbs.

Also adalimumab has been labelled with 99mTc [81], using an indirect radiolabelling method as described by Abrams et al. [82]. Labelling efficiency achieved by using this method was greater than 95% and HPLC analysis demonstrated only a minimal release of 95% has been determined by using both methods. Rituximab and other monoclonal antibodies against CD20 have been also labelled with 90Y (ibritumomab, Zevalin®) and 131I (tositumomab, Bexxar®), and these radiopharmaceuticals are successfully used for radioimmunotherapy of B-cell lymphoma [110, 111].

Fig. (4). Multiple joint involvement in a patient with Rheumatoid Arthritis as demonstrated by scintigraphy with 99m Tc-Adalimumab.

Anti-E-Selectin Monoclonal Antibody E-selectin is an endothelial-specific, cytokine-inducible adhesion molecule [84], which is exclusively expressed on the luminal surface of vascular endothelium during the inflammatory response. Its expression has been demonstrated by immunohistochemistry in a variety of acute and chronic inflammatory diseases [85]. E-selec-

Recently, 99mTc-labelled rituximab was used in mice as well as in breast cancer patients for sentinel node scintigraphy. Results demonstrated that the radiopharmaceutical is very reliable in showing the status of sentinel lymph nodes (SLN) both in mice and in breast cancer patients with a specificity for metatstatic nodes versus non affected nodes of 95% [109]. More recently, we labelled rituximab with high labelling efficiency (>98%) by using a direct radiolabelling method and also performed scintigraphic studies in patients with active RA. In this

2408 Current Pharmaceutical Design, 2008, Vol. 14, No. 24

study, we found that the 99mTc-rituximab allows clear visualisation of the diseased joints within 2 hours after administration. Most interestingly, when compared to 99mTc-adalimumab scintigraphy, in the same patient, a different degree of uptake can be seen in different joints with both tracers (Fig. 5), indicating the different pathogenetic mechanism underlying different joint inflammation and the need for a targeted individualized therapy based on scintigraphic evaluation of joint activity. This molecular approach to diagnosis and therapy decision making will be certainly more developed in the near future as a base for biological treatments and follow-up. Conjugated anti-CD20 mAb is commercially available for labelling with 111In (Zevalin®), not yet for 99mTc labelling.

Malviya et al.

Several studies have been performed in patients with different autoimmune diseases using radiolabelled anti-CD4 monoclonal antibodies that demonstrated high specificity for inflammation. One of the first scintigraphic studies was published by Becker et al. in 1990, in six patients with active, severe RA [114]. The CD4 monoclonal antibody (MAX. 16H5) was labelled with 99mTc by direct method using 2-mercaptoethanol. Each patient received a sub-therapeutic dose of 200-300 micrograms of 99mTc-labelled CD4 specific antibody (555 MBq) and they were examined at 1.5 h, 4 h and 24 h after the injection of radiopharmaceutical. The localisation of diseased joints was also correlated with clinical signs and 3phase MDP (methylene di-phosphonate) bone scan. This study demonstrated that 99mTc-labelled anti-CD4 antibodies can specifically detect CD4+ cells in diseased joints of patients with active RA and the obtained information is clinically more helpful than with 99m Tc-MDP, because of the higher specificity and sensitivity in early joint disease. In another study, focused to investigate the specificity of binding of anti-CD4 mAb to CD4+ cells, performed by Kinne et al., a direct comparison was made between radiolabelled anti-CD4 mAb and radiolabelled non-specific human immunoglobulins (HIG) for imaging inflamed joints in RA patients [115]. Eight patients with active or severe RA were intravenously injected with a subtherapeutic dose of 200-300 micrograms (370-550 MBq) of 99mTcmurine anti-human CD4 mAb (MAX.16H5) or 1 mg (370 MBq) of polyclonal 99mTc-HIG. Whole body and joint images were acquired at 1 h, 4 h and 24 h post-injection. Authors concluded that the antiCD4 mAb allows highly specific detection of inflammatory infiltrates which are rich in CD4-positive cells. These papers offered for the first time the possibility to perform “evidence-based biological therapy” of autoimmune diseases, and indeed patients with positive 99mTc-anti-CD4 scan were successfully treated with anti-CD4 therapy. In view of these exiting data, a pharmaceutical company has produced a kit for 99mTc-labelling of CD4 mAb fragments and phase 2 studies are now being performed to evaluate its possible commercialisation.

Fig. (5). Images of shoulders of a Rheumatoid Arthritis patient injected with 99m Tc-Rituximab (above) and, one week later with 99mTc-Adalimumab (below). This study clearly indicates the different pathogenesis mechanism underlying different joint inflammation and the need for a targeted individualized therapy based on scintigraphic evaluation of joint activity in this patient. More receptor-bound TNF than CD20 in indeed present in the joints and this type of therapy should be preferred.

Anti-CD4 Monoclonal Antibody CD4 is a 55 kDa monomeric membrane glycoprotein expressed on T lineage cells, including the majority of thymocytes and a subset of peripheral T cells and monocytes. The extra-cellular domains of CD4 bind to the conserved regions of MHC II molecules on antigen-presenting cells (APCs). CD4+T cells constitute the helper subset which regulates T and B cell function during T cell dependent responses. CD4+T cells and their cytokine products play an important role also in rheumatoid arthritis (RA) [112] and several, if not all, autoimmune diseases. Indeed, a number of anti-CD4 monoclonal antibodies are available for the management of different autoimmune diseases including murine and primatised CD4 mAbs [113]. These monoclonal antibodies bind with very high affinity to human CD4, induce CD4 receptor down regulation, and, at pharmacological doses, are potent inhibitor of T cell response.

Anti-CD3 Monoclonal Antibody Muromonab-CD3 (Orthoclone OKT®3) is a murine monoclonal antibody to the CD3 antigen of human T cells. The antibody is a biochemically purified IgG2a immunoglobulin. Muromonab-CD3 reacts with and blocks the function of CD3 receptor on the cell membrane of human T lymphocytes. This receptor in antigen recognition by T cells and is essential for signal transduction. Binding of muromonab-CD3 to T lymphocytes result in early activation of T cells, which leads to cytokine release, followed by blocking of T cell functions. In vivo, muromonab-CD3 reacts with most peripheral blood T cells and tissue infiltrating T cells, but has not been found to react with other haematopoietic elements or other tissues of the body. When binding to activated T-cells may induce their dead by inducing apoptosis. In a study performed by Martines et al., OKT3 (a murine antihuman CD3 mAb) was labelled with technetium-99m according to the technique developed by Martins & Gutfilen [116, 117]. The use of 99mTc-OKT3 for scintigraphic diagnosis of acute rejection in renal transplants was subsequently evaluated. Among 22 patients who underwent renal transplantation, they reported an increased 99m Tc-OKT3 kidney uptake with time in 3 patients with rejecting allograft, differentiating them from those without rejection, and these findings agreed with those of biopsies. In this study, the authors concluded that the 99mTc-OKT3 scans may be used as a diagnostic method to identify kidney allograft rejection, possibly allowing a balance between adequate immunosuppression to prevent rejection of an allograft and excessive immunosuppression that may have severe side effects.

Radiolabelled Peptides and Monoclonal Antibodies

In another study, OKT-3 was labelled with 99m-technetium by reducing the disulphide bonds; a high labelling efficiency (LE) of more than 95% was achieved and HPLC analysis showed less than 5% colloid formation [118]. In this study, 99mTc-OKT-3 (185 MBq) was intravenously injected to 7 patients with RA and in 2 patients with psoriatic arthritis. Results showed that all 34 joints with moderate to severe pain had moderate to marked uptake of radioactivity and the authors concluded that 99mTc-OKT-3 imaging could be useful as a measurement of therapeutic effectiveness in RA. Recently, 99mTc-OKT3 mAb was used in 38 patients with active RA, in order to monitor disease activity [119]. Two sets of planar anterior images of the patients’ wrists, metacarpophalangeal and interphalangeal joints, elbows, shoulders and knees joints were obtained 1 h and 3 h after the injection of 99mTc-OKT3. The scintigraphic findings showed significant correlation between the accumulation of 99mTc-OKT3 and swollen joints, tender joints and the visual analogue scale. Interestingly, there was no correlation between the radiopharmaceutical accumulation and the patients’ age, gender, duration of disease or erythrocyte sedimentation rate. In this study, authors concluded that, 99mTc-OKT3 scintigraphy is a reliable and objective method for detecting synovial activity, and can be used to define disease prognosis. Another a new generation of genetically engineered anti-CD3 mAbs, visilizumab (Nuvion®), has been developed in recent years by grafting murine complementarity-determining regions (CDRs) derived from M291 hybridoma into human non-CDR region of IgG2 and introducing non-FcR-binding mutations at amino acid residues 234 and 237 (ValAla) into the IgG2 Fc portion [120124]. This non-FcR binding humanised mAb directed towards the CD3 antigen on T lymphocytes binds with human CD3- chain with high specificity and high avidity (Ka= 0.5x109 M-1) [125]. Furthermore, non-FcR-binding mAbs do not activate resting T cells [126, 127] and have limited potential for inducing cytokine release and acute toxicity in vivo [128-130]. Visilizumab has therefore been proposed for treatment of several autoimmune diseases and clinical trials have been performed for the treatment of steroid-resistant Graft-versus-Host Disease (GvHD) and ulcerative colitis [131]. We have recently radiolabelled visilizumab with 99m-technetium [132], using an indirect radiolabelling method as described by Abrams et al. [82]. Briefly, the mAb was conjugated with a bifunctional chelator, succinimyl-hydrazinicotinamide (S-HYNIC), then labelled using tricine as co-ligand and stannous chloride as reducing agent. High labelling efficiency was achieved by using this method, which was always greater than 95%. We performed several animal experiments in Balb/c and SCID irradiated mice reconstituted with human lymphocytes. We conclude that 99mTc-labelled visilizumab can allow specific in vivo imaging of CD3 cells in tissues thus providing a rationale for therapy with unlabelled visilizumab and may also allow early follow-up of the efficacy of therapy. Nuvion® (visilizumab) has been withdrawn from production, as a therapeutic agent, after phase 3 trials, but it could still be produced by the same company as a diagnostic tool. Anti-DR Monoclonal Antibody The HLA-DR antigens play important roles in the cellular interaction involved in immune response. The HLA-DR is normally expressed on B lymphocytes, activated T lymphocytes, macrophages, monocytes, dendritic cells, activated NK cells and progenitor haemopoietic cells. During the resting state of T lymphocytes DR is not expressed and is therefore very specific for T cell activation. It is well known that this activation antigen is normally expressed on tissue infiltrating lymphocytes in inflamed tissues, in a high percentage of cells and for a longer time span compared to other activation markers, such as CD25 (IL2 receptor). It is therefore, a stable and reliable marker for detection of T cell mediated inflammation. Isobe et al., described that other tissues, such as vessel endothelium, may express DR following the release of local

Current Pharmaceutical Design, 2008, Vol. 14, No. 24

2409

inflammatory molecules [133]. Using 111In-labelled anti-DR monoclonal antibody, Isobe et al. found the expression of MHC class II antigens in a rat model of heart rejection and in mouse kidney allograft rejection. Indeed the scintigraphy revealed the presence of DR molecules on both the graft endothelium and the infiltrating mononuclear cells [134]. We have recently, radiolabelled labelled anti-DR monoclonal antibody (1D09C3) with technetium-99m. Using the 2-ME method, a high labelling efficiency of 98% and specific activity of 150 mCi/mg was obtained (unpublished data). 1D09C3 is a fully humanised IgG4 monoclonal antibody and does not induce Fc-portionmediated side effects, like chimeric antibodies do [135, 136]. Studies demonstrated that 1D09C3 has in-vivo as well as in-vitro tumouricidal activity and it can act selectively on tumour-transformed and activated cells via a non-apoptotic mechanism [137]. The 99m Tc- labelled Anti-DR monoclonal antibody can provide a useful tool for imaging of inflammation and several cancer types, mainly leukaemia and lymphoma cells. Furthermore, Rimsza et al. demonstrated that the HLA-DR can be used as a prognostic marker in the patients with diffuse large Bcell lymphoma (DLBCL) [138]. They demonstrated that the HLADR protein status predicts the survival in patients with DLBCL treated with the MACOP-B chemotherapy regimen. These findings have suggested the use of anti-DR monoclonal antibodies for diagnostic and therapeutic purposes. 1D09C3 is currently produced by a pharmaceutical company as potential therapeutic agent. Anti-CD25 Monoclonal Antibody Daclizumab (Zenapax®) is a humanised IgG1 monoclonal antibody that specifically binds to the Tac subunit (CD25) part of the high-affinity IL-2 receptor complex and inhibits the binding of IL-2 and the cascade of pro-inflammatory events involved in organ transplant rejection and several autoimmune diseases [139-141]. Similarly, basiliximab (Simulect®) is a chimeric IgG1 monoclonal antibody, produced by recombinant DNA technology, against the IL-2 receptor (CD25) expressed on T lymphocytes. Basiliximab has a molecular weight of 144 kDa which bind with high affinity (Ka= 1x1010 M-1) to the alpha chain of the high affinity IL-2 receptor complex. Interestingly, CD25 antigen is expressed on activated but not on resting T lymphocytes [142-144]. Anti-CD25 monoclonal antibodies and their fragments have been radiolabelled with 67Ga, 211At, 212Bi, 125I, 99mTc, 111In, 88Y and 18 F for different diagnostic and therapeutic purposes. Several studies in animals as well as in humans demonstrated the complete blockade of IL-2 receptors by using anti-CD25 monoclonal antibodies [145-149]. Hartmann et al., performed a study with 212Bi-anti-Tac-antibody in nude mice model bearing human IL-2 receptor -expressing lymphoma. This study demonstrated that radiobismuth-labelled humanised anti-Tac is a less immunogenic agent, which kills Tacexpressing malignancies in vivo and could be very useful in the treatment of HTLV-I-associated adult T-cell leukaemias and other Tac expressing leukaemias where large numbers of IL-2R receptors are expressed on circulating malignant cells [148]. Another study was performed with 18F and 125I labelled anti-Tac disulfide-stabilized Fv fragments (dsFv) in tumour bearing nude mice to compare both radiopharmaceuticals for their biodistribution and pharmacokinetics. dsFv were genetically engineered from a murine monoclonal antibody targeted against the IL-2 receptor. Labelling was performed with 18F using N-succinimidyl 4-([18F] fluoromethyl)benzoate or with 125I using the lodo-gen method. In this study authors found, that the blood clearance for both preparations was rapid, less than 10% retained in the blood by 15 minutes. Highest accumulation in tumours occurred between 45 to 90 mins and peaked at a mean of 4.2% injected dose/g (18F) and 5.6% of

2410 Current Pharmaceutical Design, 2008, Vol. 14, No. 24

injected dose/g (125I). The kidneys were found to be the major route of elimination. Authors suggested that the fast pharmacokinetics and rapid targeting ability of radiolabelled anti-Tac dsFv could allow diagnostic imaging either using 18F, 99mTc or short-lived isotopes of iodine such as 123I. Moreover, the targeting may be more adequate for imaging purposes rather than therapy due to too low absolute concentration of these radiopharmaceuticals in tumours [149]. As mentioned in a previous chapter of this review article, CD25 is a very interesting molecule to be targeted for immuno-scintigraphy and approaches have been made using bothe the radiolabelled natural ligand of CD25 (IL-2) and mAbs against the CD25 (anti-TAC). Both approaches seem to be adequate for diagnostic purposes, in order to evaluate the presence and extent of CD25+ cells in target tissues or CD25+ leukaemia, and for therapy decision making to plan and follow-up therapy with unlabelled anti-CD25. Several anti-CD25 mAbs are commercially available as therapeutic agents and can be easily radiolabelled but need to be used in humans under regulations and authorisation of local Ethic Committes. Anti-MIF Monoclonal Antibody Macrophage migration inhibitor factor (MIF) is a lymphokine produced by activated lymphocytes and involved in delayed type hypersensitivity and various macrophage functions [150-153]. MIF acts by aggregating and concentrating macrophages in the inflammatory lesions in delayed-type hypersensitivity reaction, moreover, a correlation between macrophage activating factor has been observed [154, 155]. However, at the site of inflammation in endothelial cells lining, MIF can also promote macrophage rolling and transmigration by up regulating P-selection expression [156, 157]. Several studies demonstrated higher level of MIF in plasma or serum in patients with inflammatory diseases, including rheumatoid arthritis, Crohn’s disease, ulcerative colitis, acute pancreatitis, type 2 diabetes, severe sepsis, multiple sclerosis, Guillain-Barre syndrome [158-164]. Anti-MIF monoclonal antibodies have been radiolabelled with radioiodine Na125I, by the Iodogen technique, with >95% labelling efficiency and high specific activity (29.56 GBq/μmol) [165]. In this study, biodistribution of Na125I labelled anti-MIF monoclonal antibody and its in-vivo binding capacity was determined by organ counting assay and by whole-body autoradiography examinations in mice. Animals were divided into three groups, in first and second group inflammation was induced by intramuscular injection of 2x107-108 CFU (colony forming units) of S. aureus and E. coli, whereas in the third group a sterile inflammation was induced by intramuscular injection of turpentine oil, into left thigh muscle. Mice were injected i.p. with 3.7 MBq125I-anti-MIF mAb and three mice of each group were sacrificed at 30 minutes, 4 hours, 24 hours, 48 hours, and 72 hours after injection, and samples of two thigh muscles (left as target, right as control), lungs, heart, liver, spleen, kidney, bone and blood were collected. The highest uptake was found in S. aureus group and the lowest was in E. coli group. The uptake in turpentine oil group was in between the two. Wholebody autoradiography showed that all inflammation foci could be visualised clearly from 24 hours after injection, but 48 hours images were much clearer in accordance with the high T/NT (target/nontarget) ratio. This study demonstrated the in-vivo targeting capability of this radiopharmaceutical, which could also be used for imaging of several inflammatory disorders, although it use has been proposed only for research purposes. CONCLUDING REMARKS More and more antibodies are nowadays available for biological therapy of immune mediated inflammatory diseases. Therefore, new molecules/receptors are regularly identified as possible therapeutic targets. The possibility to image in vivo the location and

Malviya et al.

quantify the extent of these target molecules in inflamed tissues is therefore extremely important to plan an appropriate therapy and to follow-up the efficacy of therapy. Moreover, considering that biological therapies with mAbs could be extremely expensive especially in chronic inflammatory diseases, such as RA, a pre-therapy scintigraphic approach with radiolabelled peptides or monoclonal antibodies for therapy decision making may provide a cost-effective solution. The majority of target molecules identified so far are functional receptors, cytokines or cytokine receptors. To specifically image these molecules we can use two different approaches: to radiolabel the natural ligand of the receptor molecule or a monoclonal antibody directed against the target molecule. Radiolabelled receptor ligands (such as cytokines) can provide an excellent solution for molecular imaging due to their short plasma half-life, low background uptake, low accumulation in excretory organs and fast blood kinetics. Radiolabelled cytokines however can have side effects due to their biological action. On the other hand, radiolabelled antibodies are well known for their high specificity against target molecules and offers exciting possibilities for the selection of candidate patients for therapy. Most of these cytokines and mAbs are now commercially available for therapeutic purposes and in some cases also as kit for easy radiolabelling with 99mTc or 111In. In this review we have summarised several different radiopharmaceuticals and their use for imaging inflammation in different diseases. In the present scenario, we can conclude that there is no single ideal radiopharmaceutical for imaging inflammatory diseases but clinicians must have the possibility to choose between different options according to the purpose and clinical requirements. These radiopharmaceuticals not only provide us better diagnosis than other diagnostic tools but also provide essential clinical information for therapy decision making and follow-up in different inflammatory diseases. ACKNOWLEDGEMENT The authors wish to thank International Society of Radiolabelled Blood Elements (ISORBE) for providing the scientific materials and support to prepare this manuscript. REFERENCES References 166-168 are related articles recently published. [1] [2] [3] [4]

[5]

[6]

[7]

[8]

Kindt TJ, Goldsby RA, Osborne BA. Kuby Immunology, 6th ed., WH Freeman and Company, New York 2007; 340-346. Dinarello CA. Interleukin-1 and interleukin-1 antagonism. Blood 1991; 77: 1627-52. Lowenthal JW, Mac Donald HR. Binding and internalization of interleukin 1 by T cells. J Exp Med 1986; 164:1060-74. Van der Laken CJ, Boerman OC, Oyen WJG, Laverman P, Van de Ven MTP, Corstens FHM, et al. In-vivo expression of IL-1 receptors during various experimentally induced inflammatory conditions. J Infect Dis 1998; 177: 1398-401. Van der Laken CJ, Boerman OC, Oyen WJG, Van de Ven MTP, Chizzonite R, Corstens FHM, et al. Preferential localization of systemically administered radiolabelled interleukin-1alpha in experimental inflammation in mice by binding to the type II receptor. J Clin Invest 1997; 100 (12): 2970-76. Granowitz EV, Porat R, Mier JW, Pribble JP, Stiles DM, Bloedow DC, et al. Pharmacokinetics, safety and immunomodulatory effects of human recombinant interleukin-1 receptor antagonist in healthy humans. Cytokine 1992; 4: 353-60. Van der Laken CJ, Barrera P, Boerman OC, Oyen WJG, Van der Ven MTP, Van der Meer JWM, et al. Radiolabelled interleukin-1 receptor antagonist targets to inflamed joints in patients with active rheumatoid arthritis. A report of preliminary results [abstract]. Eur J Nucl Med 1998; 25: 897. Wilbur DS, Hadley SW, Hylarides MD, Abrams PG, Beaumier PA, Morgan AC, et al. Development of a stable radioiodinating reagent

Radiolabelled Peptides and Monoclonal Antibodies

[9]

[10]

[11]

[12]

[13] [14] [15]

[16]

[17]

[18]

[19]

[20]

[21]

[22]

[23]

[24]

[25]

[26]

[27]

[28]

to label monoclonal antibodies for radiotherapy of cancer. J Nucl Med 1989; 30: 216-26. Barrera P, van der Laken CJ, Boerman OC, Oyen WJ, van de Ven MT, van Lent PL, et al. Radiolabelled interleukin-1 receptor antagonist for detection of synovitis in patients with rheumatoid arthritis. Rheumatology (Oxf) 2000; 39: 870-4. Morgan DA, Ruscetti FW, Gallo RC. Selective in vitro growth factor of T lymphocytes from normal human bone marrow. Science 1976; 193: 1007-9. Miyajima A, Miyatake S, Schreurs J, De Vries J, Arai N, Yokota T, et al. Coordinate regulation of immune and inflammatory responses by T cell-derived lymphokines. FASEB J 1988; 2(9): 2462-73. Muraguchi A, Kehrl JH, Longo DL, Volkman DJ, Smith KA, Fauci AS. Interleukin 2 receptors on human B cells. Implications for the role of interleukin 2 in human B cell function. J Exp Med 1985; 161(1):181-97. Smith KA. Interleukin-2. Curr Opin Immunol 1992; 4(3): 271-6. Smith KA. Interleukin-2: inception, impact and implications. Science 1988; 240: 1169-76. Koths K, Halenbech R. In: Sorg C, Schimpl A, Landy M, Eds, Pharmacokinetic studies on 35S-labelled recombinant interleukin-2 in mice. Cellular and molecular biology of lymphokines: International Lymphokine Workshop 1984, at Schloss Elmau, West Germany. Orlando (FL): Academic Press, 1985: 779-83. Robb RJ, Mayer PC, Garlick R. Retention of biological activity following radioiodination of human interleukin-2: comparison with biosynthetically labelled growth factor in receptor binding assay. J Immunol Meth 1985; 81: 15-30. Gennuso R, Spigelman MK, Vallabhajosula S, Moore F, Zappulla R, Nieves J, et al. Systemic biodistribution of radioiodinated interleukin-2 in the rat. J Biol Resp Mod 1989; 8: 375-84. Sabo J, Ni G, Nadeau R, Liberato DJ, Loh A. Comparative tissue distribution of 125I and U-14C labelled recombinant human interleukin-2 in the rat. Lymphokine Cytokine Res 1992; 11:229. Signore A, Chianelli M, Toscano A, Monetini L, Ronga G, Nimmon CC, et al. A radiopharmaceutical for imaging areas of lymphocytic infiltration: 123I-interleukin-2. Labelling procedure and animal studies. Nucl Med Commun 1992; 13(10): 713-22. Signore A, Beverley PC, Parman A, Negri M, Pozzilli P. Labelling of interleukin-2 (IL-2) with 123-iodine with retention of its capacity to bind to activated lymphocytes. Exp Clin Endocrinol 1987; 89(3): 301-6. Chianelli M, Signore A, Toscano A, Tonnarini GF, Pozzilli P, Negri M, et al. In: Schmidt HAE, Schattauer HR, Eds, HPLC purification of 123I-labelled interleukin-2 with high specific activity for human studies. Nuclear Medicine in Research and practice. Vienna 1991; 143-146. Signore A, Chianelli M, Annovazzi A, Bonanno E, Spagnoli LG, Pozzilli P, et al. 123Iinterleukin-2 scintigraphy for in vivo assessment of intestinal mononuclear cell infiltration in Crohn's disease. J Nucl Med 2000; 41(2): 242-49. Signore A, Chianelli M, Annovazzi A, Rossi M, Maiuri L, Greco M, et al. Imaging of active lymphocytic infiltration in Celiac disease with 123I-interleukin 2 and its response to diet. Eur J Nucl Med 2000; 27:18-24. Chianelli M, Signore A, Fritzberg AR, Mather SJ. The development of technetium-99m-labelled interleukin-2: a new radiopharmaceutical for the in vivo detection of mononuclear cell infiltrates in immune-mediated diseases. Nucl Med Biol 1997; 24(6):579-86. Annovazzi A, Biancone L, Caviglia R, Chianelli M, Capriotti G, Mather SJ, et al. 99mTc-interleukin-2 and (99m)Tc-HMPAO granulocyte scintigraphy in patients with inactive Crohn's disease. Eur J Nucl Med Mol Imaging 2003; 30(3): 374-82. Signore A, Annovazzi A, Barone R, Bonanno E, D'Alessandria C, Chianelli M, et al. 99mTc-interleukin-2 scintigraphy as a potential tool for evaluating tumor-infiltrating lymphocytes in melanoma lesions: a validation study. J Nucl Med 2004; 45(10): 1647-52. Loose D, Signore A, Bonanno E, Vermeersch H, Dierckx R, Deron P, et al. Prognostic Value of CD25 Expression on Lymphocytes and Tumor Cells in Squamous-Cell Carcinoma of the Head and Neck. Cancer Biother Radiopharm. 2008; 23(1): 25-33. Loose D, Signore A, Staelens L, Bulcke KV, Vermeersch H, Dierckx RA, et al. (123)I-Interleukin-2 uptake in squamous cell carci-

Current Pharmaceutical Design, 2008, Vol. 14, No. 24

[29]

[30]

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

2411

noma of the head and neck carcinoma. Eur J Nucl Med Mol Imaging. 2008; 35(2): 281-6. Renard V, Staelens L, Signore A, Van Belle S, Dierckx RA, Van De Wiele C. Iodine-123-interleukin-2 scintigraphy in metastatic hypernephroma: a pilot study. Q J Nucl Med Mol Imaging. 2007; 51(4): 352-6. Annovazzi A, Bonanno E, Arca M, D'Alessandria C, Marcoccia A, Spagnoli LG, et al. 99mTc-interleukin-2 scintigraphy for the in vivo imaging of vulnerable atherosclerotic plaques. Eur J Nucl Med Mol Imaging 2006; 33(2): 117-26. Signore A, Picarelli A, Chianelli M, Biancone L, Annovazzi A, Tiberti C, et al. 123I-Interleukin-2 scintigraphy: a new approach to assess disease activity in autoimmunity. J Pediatric Endocrinol Metab 1996; 9 (Suppl 1): 139-44. Signore A, Picarelli A, Annovazzi A, Britton KE, Grossman AB, Bonanno E, et al. 123I-interleukin-2: biochemical characterization and in vivo use for imaging autoimmune diseases. Nucl Med Commun 2003; 24: 305-16. Barone R, Chianelli M, Procaccini E, Chianelli M, Bottoni U, Panetta C, et al. 99mTc-IL-2 scintigraphy in patients with cutaneous melanoma: detection of lymphocytic infiltration. (abstract) Eur J Nucl Med (1998); 25: 896. Lucia P, Parisella MG, Danese C, Bruno F, Manetti LL, Capriotti G, et al. Diagnosis and followup of Takayasu's arteritis by scintigraphy with radiolabelled interleukin 2. J Rheumatol 2004; 31(6): 1225-7. Procaccini E, Chianelli M, Parisell MG, Barone R, Di Leve G, Grossmann A, et al. 99mTc-IL2 scintigraphy in autoimmune thyroid diseases. (abstract) Eur J Nucl Med 1998; 25: 977. Nadeau RW, Satoh H, Scheide S, Crowl R, Conroy R, Garland WA, et al. A comparison of mass balance, pharmacokinetics and deposition of [14C(U)] and [125 I] recombinant human interleukin-2 in cynomolgus monkeys. Drug Metabol Dispos 1995; 23(9): 904909. Rendl JE, Guthoff R, Schirbel A, Brechtelsbauer D, Schiller D, Seybold S, et al. Iodine-123-Interleukin-2 (I-123-IL-2) Scintigraphy in Graves´ Ophthalmopathy (GO): A new Approach to assess disease activity. Endocr J 2000; 47: 0-4. Schirbel A, Schiller D, Rendl J, Reiners C. Radiosynthese von [123I]Interleukin-2 für die Szintigraphie von Patienten mit endokriner Orbitopathie (EO). Nuklearmedizin 2001; 40: V73. Rendl J, Guthoff R, Schirbel A, Brechtelsbauer D, Schiller D, Seybold S, et al. Iod-123-Interleukin-2(I-123-IL-2)-Szintigraphie bei der Endokrinen Orbitopathie (EO): Ein neues verfahren zur beurteilung der entzündlichen Aktivität. Nuklearmedizin 2001; 40: V140. Hay RV, Skinner RS, Newman OC, Kunkel SL, Lyle LR, Shapiro B, et al. Scintigraphy of acute inflammatory lesions in rats with radiolabelled cytokines (abstract). J Nucl Med 1994; 35:256P. Hay RV, Skinner RS, Newman OC, Kunkel SL, Lyle LR, Shapiro B, et al. Scintigraphy of acute inflammatory lesions in rats with radiolabelled recombinant human neutrophil-activating peptide-2. Nucl Med Commun 2002; 23(4): 367-72. Gross MD, Shapiro B, Skinner RS, Shreve P, Fig LM, Hay RV. Scintigraphy of osteomyelitis in man with human recombinant interleukin-8. J Nucl Med 1996; 37: 25P. Laterveer L, Lindley IJD, Heemskerk DPPM, Camps JA, Pauwels EK, Willemze R, et al. Rapid mobilization of hematopoietic progenitor cells in Rhesus monkeys by a single intravenous injection of interleukin-8. Blood 1996; 87: 781-88. Van Zee KJ, Fischer E, Hawes AS, Hebert CA, Terrell TG, Baker JB, et al. Effects of intravenous IL-8 administration in nonhuman primates. J Immunol 1992; 148: 1746-52. Hay RV, Skinner RS, Newman OC, Kunkel SL, Lyle LR, Shapiro B, et al. Scintigraphy of acute inflammatory lesions in rats with radiolabelled recombinant human interleukin-8. Nucl Med Commun 1997; 18: 367-78. Shapiro B, Gross MD, Shreve PD, Skinner RS, Hay R, Fig LM, et al. Imaging of inflammation and infection with radiolabelled cytokines (abstract). Nucl Med Commun 1997; 18: 472. Van der Laken CJ, Boerman OC, Oyen WJG, Van de Ven MTP, Van der Meer JWM, Corstens FHM. The kinetics of radiolabeled interleukin-8 in infection and sterile inflammation. Nucl Med Commun 1998; 19: 271-81.

2412 Current Pharmaceutical Design, 2008, Vol. 14, No. 24 [48]

[49]

[50]

[51] [52] [53] [54]

[55]

[56]

[57] [58]

[59]

[60]

[61]

[62]

[63]

[64]

[65] [66]

[67]

[68]

[69] [70]

Rennen HJ, van Eerd JE, Oyen WJ, Corstens FH, Edwards DS, Boerman OC. Effects of coligands variation on the in vivo characteristics of Tc-99m-labeled interleukin-8 in detection of infection. Bioconjug Chem 2002; 13: 370-77. Gratz S, Rennen HJ, Boerman OC, Oyen WJ, Burma P, Corstens FH. (99m)Tc-interleukin-8 for imaging acute osteomyelitis. J Nucl Med 2001; 42: 1257-64. Gratz S, Rennen HJ, Boerman OC, Oyen WJ, Corstens FH. Rapid imaging of experimental colitis with (99m)Tc-interleukin-8 in rabbits. J Nucl Med 2001; 42: 917-23. Scott P. IL-12: initiation cytokine for cell-mediated immunity. Science 1993; 260: 496-7. Stern AS, Magram J, Presky DH. Interleukin-12 an integral cytokine in the immune response. Life Sci 1996; 58: 639-54. Adorini L. Interleukin-12, a key cytokine in Th1-mediated autoimmune diseases. Cell Mol Life Sci 1999; 55: 1610-25. Hsieh CS, Macatonia SE, Tripp CS, Wolf SF, O’Garra A, Murphy KM. Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages. Science 1993; 260: 547- 49. Manetti R, Parronchi P, Giudizi MG, Piccinni MP, Maggi E, Trinchieri G, et al. Natural Killer cell stimulatory factor (interleukin 12 [IL12]) induces T helper type 1 (Th1)-specific immune responses and inhibits the development of IL-4-producing Th cells. J Exp Med 1993; 177: 1199-204. Annovazzi A, D’Alessandria, Bonanno E, Mather SJ, Cornelissen B, Van de Wiele C, et al. Synthesis of 99mTc-HYNIC-interleukin12, a new specific rediopharmaceutical for imaging T lymphocytes. Eur J Nucl Med Mol Imaging 2006; 33(4): 474-82. Charo IF, Taubman MB. Chemokines in the pathogenesis of vascular disease. Circ Res. 2004; 95: 858-66. Weber C, Schober A, Zernecke A. Chemokines: key regulators of mononuclear cell recruitment in atherosclerotic vascular disease. Arterioscler Thromb Vasc Biol 2004; 24: 1997-08. Nelken NA, Coughlin SR, Gordon D, Wilcox JN. Monocyte chemoattractant protein-1 in human atheromatous plaques. J Clin Invest. 1991; 88: 1121-27. Yla-Herttuala S, Lipton BA, Rosenfeld ME, Sarkioja T, Yoshimura T, Leonard EJ, et al. Expression of monocyte chemoattractant protein 1 in macrophage-rich areas of human and rabbit atherosclerotic lesions. Proc Natl Acad Sci USA 1991; 88: 5252-6. Aukrust P, Ueland T, Müller F, Andreassen AK, Nordøoy I, Aas H, et al. Elevated circulating levels of C-C chemokines in patients with congestive heart failure. Circulation. 1998; 97: 1136-43. Behr TM, Wang X, Aiyar N, Coatney RW, Li X, Koster P, et al. Monocyte chemoattractant protein-1 is upregulated in rats with volume-overload congestive heart failure. Circulation 2000; 102: 1315-22. Hartung D, Petrov A, Haider N, Fujimoto S, Blankenberg F, Fujimoto A, et al. Radiolabeled Monocyte Chemotactic Protein 1 for the detection of inflammation in experimental atherosclerosis. J Nucl Med 2007; 48(11): 1816-21. Blankenberg FG, Tait JF, Blankenberg TA, Post AM, Strauss HW. Imaging macrophages and the apoptosis of granulocytes in a rodent model of subacute and chronic abscesses with radiolabeled monocyte chemotactic peptide-1 and annexin V. Eur J Nucl Med 2001; 28(9): 1384-93. Vilcek J, Feldmann M. Historical review: Cytokine as therapeutics and targets of therapeutics. Trends Pharma Sci 2004; 201-9. Virgolini I, Kurtaran A, Leimer M, Smith-Jones P, Agis H, Angelberger P, et al. Inhalation scintigraphy with Iodine-123-labeled interferon g-1b: pulmonary deposition and dose escalation study in healthy volunteers. J Nucl Med 1997; 38:1475-81. Knight DM, Trinh H, Le J, Siegel S, Shealy D, McDonough M, et al. Construction and initial characterization of a mouse-human chimeric anti-TNF antibody. Mol Immunol 1993; 30:1443-53. Scallon BJ, Moore MA, Trinh H, Knight DM, Ghrayeb J. Chimeric anti-TNF monoclonal antibody cA2 binds recombinant transmembrane TNF and activates immune effector functions. Cytokine 1995; 7: 251-9. Remicade® (infliximab) package insert. Malvern, Pennsylvania: Centocor, Inc.; revised April, 2007. ten Hove T, van Montfrans C, Peppelenbosch MP, van Deventer SJ. Infliximab treatment induces apoptosis of lamina propria T lymphocytes in Crohn’s disease. Gut 2002; 50: 206-11.

Malviya et al. [71]

[72]

[73]

[74]

[75]

[76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

[84] [85]

[86]

[87]

[88]

[89]

[90] [91]

Siegel SA, Shealy DJ, Nakada MT, Le J, Woulfe DS, Probert L, et al. The mouse/human chimeric monoclonal antibody cA2 neutralizes TNF in vitro and protects transgenic mice from cachexia and TNF lethality in vivo. Cytokine 1995; 7: 15-25. Barone D, Krantz C, Lambert D, Maggiora K, Mohler K. Comparative analysis of the ability of Etanercept and infliximab to lyse TNF-expressing cells in a complement dependent fashion. Arthritis Rheum 1999; 42(Suppl): S90. Annovazzi A, D’Alessandria C, Caprilli R, Viscido A, Corsetti F, Parisella MG, et al. Radiolabelling of a monoclonal anti-TNF- antibody with 99mTc: in vitro studies. Q J Nucl Med 2002; 46(Suppl 1): 27. Conti F, Priori R, Chimenti MS, Coari G, Annovazzi A, Valesini G, et al. Successful treatment with intraarticular infliximab for resistant knee monarthritis in a patient with spondylarthropathy a role for scintigraphy with 99m Tc-infliximab. Arthritis Rheum 2005; 52 (4):1224-26. Chianelli M, D’Alessandria C, Conti F, Priori R, Valesini G, Annovazzi A, et al. New radiopharmaceuticals for imaging rheumatoid arthritis. Q J Nucl Med Mol Imaging 2006; 50: 217-25. Tsopelas C, Penglis S, Ruskiewicz A, Bartholomeusz DL. Scintigraphic imaging of experimental colitis with technetium-99minfliximab in the rat. Hell J Nucl Med 2006; 9(2): 85-9. D’Alessandria C, Malviya G, Viscido A, Aratari A, Maccioni F, Amato A, et al. Use of a 99m-Technetium labelled anti-TNF monoclonal antibody in Crohn’s Disease: in vitro and in vivo studies. Q J Nucl Med Mol Imaging 2007; 51: 1-9. Paul J, Anderson PJ. Tumor necrosis factor inhibitors: clinical implications of their different immunogenicity profiles. Semin Arthritis Rheum 2005; 34(Suppl): 19-22. Rau R. Adalimumab (a fully human anti-tumour necrosis factor a monoclonal antibody) in the treatment of active rheumatoid arthritis: the initial results of five trials. Ann Rheum Dis 2002; 61(Suppl II): 70-73. Adalimumab (Humira). European Public Assessment Report - Scientific Discussion. http://www.emea.europa.eu/humandocs/PDFs/ EPAR/humira/Humira-H-481-II-43-AR.pdf (assessed on 01.07.08). Barrera P, Oyen WJG, Boerman OC, van Riel PLCM. Scientigraphic detection of tumour necrosis factor in patients with rheumatoid arthritis. Ann Rheum Dis 2003; 62: 825-8. Abrams MJ, Juweid M, ten Kate CI, Schwartz DA, Hauser MM, Gaul FE, et al. Technetium-99m-human polyclonal IgG radiolabelled via the hydrazino nicotinamide derivative for imaging focal sites of infection in rats. J Nucl Med 1990; 31: 2022-8. Annovazzi A, D’Alessandria C, Lenza A, Lanzolla T, Conti F, Priori R, et al. Radiolabelled anti-TNF- antibodies for therapy decision making and follow-up in rheumatoid arthritis. Eur J Nucl Med 2006; 33(Suppl 2): S146. Bevilacqua MP. Endothelial-leukocytes adhesion molecules. Ann Rev Immunol 1993; 11: 767-804. Mason JC, Haskard DO. The clinical importance of leucocytes and endothelial cell adhesion molecules in inflammation. Vasc Med Rev 1994; 5: 249-75. Bevilacqua MP, Stengelin S, Gimbrone MA Jr, Seed B. Endothelial leucocyte adhesion molecule-1: an inducible receptor for neutrophils related to complement proteins and lectins. Science 1989; 243: 1160-65. Bhushan M, Bleiker TO, Ballsdon AE, Allen MH, M Sopwith, Robinson MK, et al. Anti-E-selectin is ineffective in the treatment of psoriasis: a randomized trial. Br J Dermatol 2002; 146: 824-31. Pober JS, Bevilacqua MP, Mendrik DL, Lapierre LA, Fiers W, Gimborne MA Jr. Two distinct monokines, interleukin 1 and tumor necrosis factor, each in dependently induce biosynthesis and transient expression of the antigen on the surface of cultured human vascular endothelial cells. J Immunol 1986; 136:1680-87. Wellicome SM, Thornhill MH, Pitzalis C, Thomas DS, Lanchbury JS, Panayi GS, et al. A monoclonal antibody that detects a novel antigen on endothelial cells that is induced by tumor necrosis factor, IL-1 or lipopolysaccharide. J Immunol 1990; 144: 2558-65. Bevilacqua MP, Nelson RM. Selectins. J Clin Invest 1993; 91: 37987. Jamar F, Chapman PT, Harrison AA, Binns RM, Haskard DO, Peters AM. Inflammatory arthritis: imaging of endothelial cell acti-

Radiolabelled Peptides and Monoclonal Antibodies

[92]

[93]

[94]

[95]

[96]

[97]

[98]

[99] [100]

[101]

[102]

[103]

[104]

[105] [106] [107]

[108]

[109]

[110]

[111]

[112]

vation with an indium-111-labelled F(ab’)2 fragment of anti-Eselectin monoclonal antibody. Radiology 1995; 194: 843-50. Chapman PT, Jamar F, Keelan ET, Peters AM, Haskard DO. Use of monoclonal antibody against E-selectin for imaging endothelial activation in rheumatoid arthritis. Arthritis Rheum 1996; 39: 137175. Jamar F, Chapman PT, Manicourt D-H, Glass DM, Haskard DO, Peters AM. A comparison between 111In-anti-E-selectin mAb and 99 Tcm-labelled human non-specific immunoglobulin in radionuclide imaging of rheumatoid arthritis. Brit J Radiol 1997; 70: 473-81. Jamar F, Houssiau FA, Devogelaer JP, Chapman PT, Haskard DO, Beaujean V, et al. Scintigraphy using a technetium 99m-labelled anti-E-selectin Fab fragment in rheumatoid arthritis. Rheumatology 2002; 41: 53-61. Keelan ETM, Harrison AA, Chapman PT, Binns RM, Peters AM, Haskard DO. Imaging vascular endothelial activation: an approach using radiolabelled monoclonal antibodies against the endothelial cell adhesion molecule E-selectin. J Nucl Med 1994; 35: 276-81. van der Lubbe PAHM, Arndt JW, Calame W, Ferreira TC, Pauwels EK, Breedveld FC. Measurement of synovial inflammation in rheumatoid arthritis with technetium-99m labelled human polyclonal immunoglobulin G. Eur J Nucl Med 1991; 18: 119-23. de Bois MH, Arndt JW, van der Velde EA, van der Lubbe PA, Westedt ML, Pauwels EK, et al.. 99mTc human immunoglobulin scintigraphy- a reliable method to detect joint activity in rheumatoid arthritis. J Rheumatol 1992; 19: 1371-6. Liberatore M, Clemente M, Iurilli AP, Zorzin L, Marini M, Di Rocco E, et al. Scintigraphic evaluation of disease activity in rheumatoid arthritis: a comparison of technetium-99m human nonspecific immunoglobulins, leucocytes and albumin nanocolloids. Eur J Nucl Med 1992; 19: 853-57. Rituxan® (Rituximab), package insert. Genentech Inc., San Francisco, CA; revised February 21, 2007. Reff ME, Carner K, Chambers KS, Chinn PC, Leonard JE, Raab R, et al. Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood 1994; 83: 435-45. Stashenko P, Nadler LM, Hardy R, Schlossman SF. Characterization of a human B lymphocyte-specific antigen. J Immunol 1980; 125: 1678-85. Tedder TF, Boyd AW, Freedman AS, Nadler LM, Schlossman SF. The B cell surface molecule B1 is functionally linked with B-cell activation and differentiation. J Immunol 1985; 135(2): 973-9. Dorner, T, Rumester, G. The role of B-cells in rheumatoid arthritis: mechanisms and therapeutic targets. Curr Op Rheum 2003; 15: 246-52. Stern M, Herrmann R. Overview of monoclonal antibodies in cancer therapy: present and promise. Crit Rev Oncol/Hematol 2005; 54:11-29. Johnson PW, Glennie MJ. Rituximab: mechanisms and applications. Br J Cancer 2001; 85: 1619-23. Olszewski AJ, Grossbard ML. Empowering targeted therapy: lessons from Rituximab. Sci STKE 2004; 241: 30. Stopar TG, Mlinaric-Rascan I, Fettich J, Hojker S, Mather SJ. 99mTc-rituximab radiolabelled by photo-activation: a new nonHodgkin’s lymphoma imaging agent. Eur J Nucl Med 2006; 33: 53-59. Stalteri MA, Mather SJ. Technetium-99m labelling of the antitumour antibody PR1A3 by photo activation. Eur J Nucl Med 1996; 23: 178-87. Wang XJ, Lin BH, Yang Z, Ouyang T, Li JF, Xu B, et al. Preliminary study on a new sentinel lymphoscintigraphy agent 99mTcRituximab for breast patient. Zhonghua Zhong Liu Za Zhi (Chinese J Oncol) 2006; 28(3): 200-3. Lindén O, Kurkus J, Garkavij M, Cavallin-Ståhl E, Ljungberg M, Nilsson R, et al. A Novel platform for radioimmunotherapy: extracorporeal depletion of biotinylated and 90Y-labelled rituximab in patients with refractory B-cell lymphoma. Can Biother Radiopharma 2005; 20(4): 457-66. Jacene HA, Filice R, Kasecamp W, Wahl RL. Comparison of 90Yibritumomab tiuxetan and 131I-tositumomab in clinical practice. J Nucl Med. 2007; 48(11): 1767-76. Pohlers D, Schmidt-Weber CB, Franch A, Kuhlmann J, Bräuer R, Emmrich F, et al. Differential clinical efficacy of anti-CD4 monoclonal antibodies in rat adjuvant arthritis is paralleled by differen-

Current Pharmaceutical Design, 2008, Vol. 14, No. 24

[113]

[114]

[115]

[116]

[117]

[118]

[119]

[120]

[121]

[122]

[123]

[124] [125] [126]

[127]

[128]

[129]

[130]

[131]

[132]

[133]

2413

tial influence on NF-B binding activity and TNF- secretion of T cells. Arthritis Res 2002; 4:184-89. Reddy MP, Kinney CAS, Chaikin MA, Payne A, Fishman-Lobell J, Tsui P, et al. Elimination of Fc receptor-dependent effector functions of a modified IgG4 monoclonal antibody to human CD4. J Immunol 2000; 164: 1925-33. Becker W, Emmrich F, Horneff G, Burmester G, Seiler F, Schwarz A, et al. Imaging rheumatoid arthritis specifically with technetium 99m CD4-specific (T-helper lymphocytes) antibodies. Eur J Nucl Med 1990; 17(3-4): 156-9. Kinne RW, Becker W, Schwab J, Horneff G, Schwarz A, Kalden JR, et al. Comparison of 99Tcm-labelled specific murine anti-CD4 monoclonal antibodies and nonspecific human immunoglobulin for imaging inflamed joints in rheumatoid arthritis. Nucl Med Commun 1993; 14(8): 667-75. Martins FPP, Gutfilen B. Estudo da marcação do anticorpo monoclonal OKT3 com tecnécio-99m: aplicações clinicas. Radiologia Brasileira 2002; 35: 286. Martins FPP, Souza SAL, Gonçalves RT, Fonseca LMB, Gutfilen B. Preliminary results of [99mTc]OKT3 scintigraphy to evaluate acute rejection in renal transplants. Transplant Proc 2004; 36: 2664- 67. Marcus C, Thakur ML, Huynh TV, Louie JS, Leibling M, Minami C, et al. Imaging rheumatic joint diseases with anti-T lymphocyte antibody OKT-3. Nucl Med Commun 1994; 15(10): 824-30. Martins FPP, Gutfilen B, DE Souza SAL, DE Azevedo MNL, Cardoso LR, Fraga R, et al. Monitoring rheumatoid arthritis synovitis with 99mTc-anti-CD3. Br J Radiol 2008; 81: 25-29. Woodle ES, Xu D, Zivin RA, Auger J, Charette J, O’Laughlin R, et al. Phase I trial of a humanized, Fc receptor nonbinding OKT3 antibody, huOKT31(Ala-Ala) in the treatment of acute renal allograft rejection. Transplantation 1999; 68: 608-16. Cole MS, Stellrecht KW, Shi JD, Homola M, Hsu DH, Anasetti C, et al. HuM291, a humanized anti-CD3 antibody, is immunosuppressive to T cells while exhibiting reduced mitogenicity in vitro. Transplantation 1999; 68(4): 563-71. Routledge EG, Falconer ME, Pope H, Lloyd IS, Waldmann H. The effect of aglycosylation on the immunogenicity of a humanized therapeutic CD3 monoclonal antibody. Transplantation 1995; 60(8): 847-53. Cole M, Anasetti C, Tso JY. Human IgG2 variants of chimeric anti-CD3 are non-mitogenic to T cells. J Immunol 1997; 159: 3613-21. Trajkovic V. Nuvion protein design labs. Curr Opin Investig Drugs 2002; 3(3): 411-4. Dingermann T, Zündorf I. Macromolecular immunosuppressants. Biotechnol J 2006; 1: 47-57. Alegre ML, Tso JY, Sattar HA, Smith J, Desalle F, Cole M, et al. An anti-murine CD3 monoclonal antibody with a low affinity for Fc g receptors suppresses transplantation responses while minimizing acute toxicity and immunogenicity. J Immunol 1995; 155(3): 1544-55. Smith JA, Tang Q, Bluestone JA. Partial TCR signals delivered by FcR-nonbinding anti-CD3 monoclonal antibodies differentially regulate individual Th subsets. J Immunol 1998; 160: 4841-9. Chatenoud L, Ferran C, Reuter A, Legendre C, Gevaert Y, Kreis H, et al. Systemic reaction to the anti-T cell mAb OKT3 in relation to serum levels of tumor necrosis factor and interferon-gamma. N Engl J Med 1989; 320(21): 1420-1. Kan EAR, Wright SD, Welte K, Wang CY. Fc receptors on monocytes cause OKT3-treated lymphocytes to internalize T3 and to secrete IL-2. Cell Immunol 1986; 98(1): 181-7. Clement LT, Tilden AB, Dunlap NE. Analysis of the monocyte Fc receptors and Ab-mediated cellular interactions required for the induction of T cell proliferation by anti-T3 Abs. J Immunol 1985; 135(1): 165-71. Carpenter PA, Sanders JE. Steroid-refractory graft-vs.-host disease: past, present and future. Pediatr Transplant 2003; 7(Suppl 3): 1931. Malviya G, De Vries EFJ, Dierckx RA, Signore A. Radiopharmaceuticals for imaging chronic lymphocytic inflammation. Bra Arch Bio Tech 2007; 50: 1-13. Isobe M, Narula J, Southern JF, Strauss HW, Khaw BA, Haber E. Imaging the rejecting heart. In vivo detection of major histocom-

2414 Current Pharmaceutical Design, 2008, Vol. 14, No. 24

[134]

[135]

[136]

[137]

[138]

[139] [140] [141]

[142] [143]

[144]

[145]

[146]

[147]

[148]

[149]

[150]

patibility complex class II antigen induction. Circulation 1992; 85(2): 738-46. Isobe M. Scintigraphic imaging of MHC class II antigen induction in mouse kidney allografts: a new approach to non-invasive detection of early rejection. Transpl Int 1993; 6(5): 263-9. Billing R, Chatterjee S. Prolongation of skin allograft survival in monkeys treated with anti-Ia and anti-blast/monocyte monoclonal antibodies. Transplant Proc 1983; 15: 649-50. Jonker M, Nooij FJM, den Butter G, van Lambalgen R, Fuccello AJ. Side effects and immunogenicity of murine lymphocytespecific monoclonal antibodies in subhuman primates. Transplantation 1988; 45: 677-82. Nagy ZA, Hubner B, Löhning C, Rauchenberger R, Reifert S, Thomassen Wolf E, et al. Fully human, HLA-DR-specific monoclonal antibodies efficiently induce programmed death of malignant lymphoid cells. Nat Med 2002; 8: 801-7. Rimsza LM, Farinha P, Fuchs DA, Masoudi H, Connors JM, Gascoyne RD. HLA-DR protein status predicts survival in patients with diffuse large B-cell lymphoma treated on the MACOP-B chemotherapy regimen. Leuk Lymphoma 2007; 48(3): 542-46. Prescribing information. Zenapax (daclizumab). Roche. New Jersey, USA; Revised: September 2005. Mottershead M, Neuberger. Daclizumab. Expert Opin Biol Ther 2007; 7(10):1583-96. Mertelsmann R, Trepel M. Identification of their epitope reveals the structural basis for the mechanism of action of the immunosuppressive antibodies basiliximab and daclizumab. Cancer Res 2007; 15:67(8):3518-23. Prescribing information. Simulect (basiliximab). Novartis. New Jersey 07936; Revised: November 2003. Binder M, Vögtle FN, Michelfelder S, Müller F, Illerhaus G, Sundararajan S, et al. A monoclonal antibody (anti-Tac) reactive with activated and functionally mature human T cells. II. Expression of Tac antigen on activated cytotoxic killer T cells, suppressor cells, and on one of two types of helper T cells. J Immunol 1981; 126:1398-403. Queen C, Schneider WP, Selick HE, Payne PW, Landolfi NF, Duncan JF, et al. A humanized antibody that binds to the interleukin 2 receptor. Proc Natl Acad Sci USA 1989; 86, 10029-33. Wu C, Jagoda E, Brechbiel M, Webber KO, Pastan I, Gansow O, et al. Biodistribution and catabolism of Ga-67-labeled anti-tac dsFv fragment. Bioconjug Chem 1997; 8: 365-9. Kobayashi H, Yoo TM, Drumm D, Kim MK, Sun BF, Le N, et al. Improved biodistribution of 1251-labeled anti-tac disulfidestabilized Fv fragment by blocking its binding to the  subunit of the interleukin 2 receptor in the circulation with preinjected humanized anti-tac IgG. Cancer Res 1997; 57: 1955-1961. Zhang M, Yao Z, Zhang Z, Garmestani K, Talanov VS, Plascjak PS, et al. The Anti-CD25 monoclonal antibody 7G7/B6, armed with the -emitter 211 At, provides effective radioimmunotherapy for a murine model of leukemia. Cancer Res 2006; 66(16): 822732. Hartmann F, Horak EM, Garmestani K, Wu C, Brechbiel MW, Kozak RW, et al. Radioimmunotherapy of nude mice bearing a human interleukin 2 receptor alpha expressing lymphoma utilizing the alpha-emitting radionuclide-conjugated monoclonal antibody 212Bi-anti-Tac. Cancer Res 1994; 54(16): 4362-70. Choi CW, Lang L, Lee JT, Webber KO, Yoo TM, Chang HK, et al. Biodistribution of 18F- and 125I-labeled anti-Tac disulfidestabilized Fv fragments in nude mice with interleukin 2 alpha receptor-positive tumor xenografts. Cancer Res 1995; 55(22): 5323-9. Bloom BR, Bennett B. Mechanism of a reaction in vitro associated with delayed-type hypersensitivity. Science 1966; 153(3731): 8082.

Malviya et al. [151]

[152]

[153]

[154]

[155]

[156]

[157]

[158]

[159]

[160]

[161]

[162]

[163]

[164]

[165]

[166]

[167]

[168]

Kawaguchi T, Mednis A, Golde DW, David JR, Remold HR. A monoclonal antibody against migration inhibitory factor (MIF) obtained by immunization with MIF from the human lymphoblast cell line Mo. J Leuko Biol 1986; 39: 223-32. David JR. Delayed hypersensitivity in vitro: its mediation by cellfree substances formed by lymphoid cell-antigen interaction. Proc Nat Acad Sci USA 1966; 56(1): 72-77. Bernhagen J, Calandra T, Mitchell RA, Martin SB, Tracey KJ, Voelter W, et al. MIF is a pituitary-derived cytokine that potentiates lethal endotoxaemia. Nature 1993; 365(6448): 756-59. David JR, David RA. Cellular hypersensitivity and immunity: Inhibition of macrophase migration and lymphocyte mediators. Prog. Allergy 1972; 16: 300. Churchill WH, Mednis AD, Remold H. In: Oppenheim JJ, Cohen S, Eds, Guinea pig macrophage activating factor can be distinguished from migration inhibitory factor by its sensitivity to trypsin. Proceedings of the Third International Lymphokines Workshop. New York: Academic Press 1983; 269. Calandra T, Roger T. Macrophage migration inhibitory factor: a regulator of innate immunity. Nat Rev Immun 2003; 3(10): 791800. Denkinger CM, Metz C, Fingerle-Rowson G, Denkinger MD, Forsthuber TG. Macrophage migration inhibitory factor and its role in autoimmune diseases. Arch Immunol Therap Exp 2004; 52(6): 389-400. Leech M, Metz C, Hall P, Hutchinson P, Gianis K, Smith M, et al. Macrophage migration inhibitory factor in rheumatoid arthritis: evidence of proinflammatory function and regulation by glucocorticoids. Arth Rheu 1999; 42(8): 1601-08. Ohkawara T, Nishihira J, Takeda H, Hige S, Kato M, Sugiyama T, et al. Amelioration of dextran sulfate sodium-induced colitis by anti-macrophage migration inhibitory factor antibody in mice. Gastroenterol 2002; 123(1): 256-70. Sakai Y, Masamune A, Satoh A, Nishihira J, Yamagiwa T, Shimosegawa T. Macrophage migration inhibitory factor is a critical mediator of severe acute pancreatitis. Gastroenterol 2003; 124(3): 725-36. Bozza FA, Gomes RN, Japiass A M, Soares M, Castro-Faria-Neto HC, Bozza PT, et al. Macrophage migration inhibitory factor levels correlate with fatal outcome in sepsis. Shock 2004; 22(4): 309-13. Niino M, Ogata A, Kikuchi S, Tashiro K, Nishihira J. Macrophage migration inhibitory factor in the cerebrospinal fluid of patients with conventional and optic-spinal forms of multiple sclerosis and neuro-Behçet’s disease. J Neurol Sci 2000; 179(1-2): 127-31. Yabunaka N, Nishihira J, Mizue Y, Tsuji M, Kumagai M, Ohtsuka Y, et al. Elevated serum content of macrophage migration inhibitory factor in patients with type 2 diabetes. Diabetes Care 2000; 23(2): 256- 58. Nicoletti F, Créange A, Orlikowski D, Bolgert F, Mangano K, Metz C, et al. Macrophage migration inhibitory factor (MIF) seems crucially involved in Guillain-Barré syndrome and experimental allergic neuritis. J Neuroimmunol 2005; 168(1-2): 168-74. Zhang C, Hou G, Han J, Song J, Liang T. Radioiodine labeled Anti-MIF McAb: a potential agent for inflammation imaging. Mediators Inflamm 2007; 2007: 50180. Chakravarty R, Pandey U, Manolkar RB, Dash A, Venkatesh M, Pillai MR. Development of an electrochemical 90 Sr-90 Y generator for separation of 90 Y suitable for targeted therapy. Nucl Med Biol 2008; 35(2): 245-53. Collier TL, Waterhouse RN, Kassiou M. Imaging sigma receptors: applications in drug development. Curr Pharm Des 2007; 13(1): 5172. Wadas TJ, Wong EH, Weisman GR, Anderson CJ. Copper chelation chemistry and its role in copper radiopharmaceuticals. Curr Pharm Des 2007; 13(1): 3-16.