US20020102208A1 - Radiolabeling kit and binding assay - Google Patents

Radiolabeling kit and binding assay Download PDF

Info

Publication number
US20020102208A1
US20020102208A1 US09/259,337 US25933799A US2002102208A1 US 20020102208 A1 US20020102208 A1 US 20020102208A1 US 25933799 A US25933799 A US 25933799A US 2002102208 A1 US2002102208 A1 US 2002102208A1
Authority
US
United States
Prior art keywords
antibody
cells
dtpa
kit
radiolabeling
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/259,337
Inventor
Paul Chinn
Ronald Morena
Michael Labarre
John E. Leonard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biogen Inc
Original Assignee
Idec Pharmaceuticals Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Idec Pharmaceuticals Corp filed Critical Idec Pharmaceuticals Corp
Priority to US09/259,337 priority Critical patent/US20020102208A1/en
Assigned to IDEC PHARMACEUTICALS CORPORATION reassignment IDEC PHARMACEUTICALS CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LABARRE, MICHAEL, CHINN, PAUL, LEONARD, JOHN E., MORENA, RONALD
Priority to MYPI20000754A priority patent/MY138674A/en
Priority to SK91-2010A priority patent/SK288096B6/en
Priority to SK1216-2001A priority patent/SK287745B6/en
Priority to MEP-2009-21A priority patent/ME00782B/en
Priority to NZ530868A priority patent/NZ530868A/en
Priority to AU35052/00A priority patent/AU776747B2/en
Priority to YU61601A priority patent/RS51778B/en
Priority to AT00913645T priority patent/ATE433108T1/en
Priority to SI200031085T priority patent/SI2112512T1/en
Priority to SI200031036T priority patent/SI1192463T1/en
Priority to MXPA01008837A priority patent/MXPA01008837A/en
Priority to CZ20013126A priority patent/CZ303262B6/en
Priority to CNB008057397A priority patent/CN1235639C/en
Priority to KR1020017011093A priority patent/KR100729247B1/en
Priority to DE60042326T priority patent/DE60042326D1/en
Priority to BRPI0017596A priority patent/BRPI0017596B8/en
Priority to UA2001096636A priority patent/UA78484C2/en
Priority to IL14510900A priority patent/IL145109A0/en
Priority to EEP200100465A priority patent/EE05650B1/en
Priority to PT00913645T priority patent/PT1192463E/en
Priority to NZ513788A priority patent/NZ513788A/en
Priority to PL352531A priority patent/PL205780B1/en
Priority to CZ2010-746A priority patent/CZ306959B6/en
Priority to EP00913645A priority patent/EP1192463B1/en
Priority to PT91585141T priority patent/PT2112512E/en
Priority to DK09158514.1T priority patent/DK2112512T3/en
Priority to JP2000602634A priority patent/JP4704570B2/en
Priority to BRPI0008719-0 priority patent/BRPI0008719B8/en
Priority to CA2742153A priority patent/CA2742153C/en
Priority to DK00913645T priority patent/DK1192463T3/en
Priority to EP09158514.1A priority patent/EP2112512B1/en
Priority to ES00913645T priority patent/ES2328100T3/en
Priority to ES09158514.1T priority patent/ES2526723T3/en
Priority to HU1300275A priority patent/HU230516B1/en
Priority to CA2362119A priority patent/CA2362119C/en
Priority to HU0202522A priority patent/HU229627B1/en
Priority to PCT/US2000/005061 priority patent/WO2000052473A2/en
Priority to RU2001126400/15A priority patent/RU2251110C2/en
Priority to TW089103582A priority patent/TWI294969B/zh
Priority to ZA200106943A priority patent/ZA200106943B/en
Priority to NO20014245A priority patent/NO329142B1/en
Priority to BG105957A priority patent/BG65690B1/en
Priority to HR20010714A priority patent/HRP20010714B1/en
Publication of US20020102208A1 publication Critical patent/US20020102208A1/en
Priority to HK02107258.0A priority patent/HK1045730B/en
Assigned to BIOGEN IDEC INC. reassignment BIOGEN IDEC INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: IDEC PHARMACEUTICALS CORPORATION
Priority to US11/033,439 priority patent/US7608241B2/en
Priority to CY20091100912T priority patent/CY1109335T1/en
Priority to HK09110560.0A priority patent/HK1131208A1/en
Priority to NO20100717A priority patent/NO330705B1/en
Priority to CY20151100160T priority patent/CY1116265T1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/534Production of labelled immunochemicals with radioactive label
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/60Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances involving radioactive labelled substances

Definitions

  • the present invention relates to antibody binding assays and radiolabeling kits, lyophilized cell preparations, reagents and protocols for testing the clinical efficacy of therapeutic antibodies for the treatment/imaging of tumors and tumor cells.
  • the kits of the present invention are used for making and evaluating radiolabeled antibody conjugates that will be used for the treatment and imaging of B-cell lymphoma tumors by targeting the B cell surface antigen BP35 (“CD20”).
  • the immune system of vertebrates (for example, primates, which include humans, apes, monkeys, etc.) consists of a number of organs and cell types which have evolved to: accurately and specifically recognize foreign microorganisms (“antigen”) which invade the vertebrate-host; specifically bind to such foreign microorganisms; and, eliminate/destroy such foreign microorganisms.
  • Lymphocytes as well as other types of cells, are critical to the immune system. Lymphocytes are produced in the thymus, spleen and bone marrow (adult) and represent about 30% of the total white blood cells present in the circulatory system of humans (adult).
  • T cells are responsible for cell mediated immunity, while B cells are responsible for antibody production (humoral immunity).
  • B cells are responsible for antibody production (humoral immunity).
  • T cells and B cells can be considered as interdependent—in a typical immune response, T cells are activated when the T cell receptor binds to fragments of an antigen that are bound to major histocompatability complex (“MHC”) glycoproteins on the surface of an antigen presenting cell; such activation causes release of biological mediators (“interleukins”) which, in essence, stimulate B cells to differentiate and produce antibody (immunoglobulins”) against the antigen.
  • MHC major histocompatability complex
  • Each B cell within the host expresses a different antibody on its surface—thus one B cell will express antibody specific for one antigen, while another B cell will express antibody specific for a different antigen. Accordingly, B cells are quite diverse, and this diversity is critical to the immune system. In humans, each B cell can produce an enormous number of antibody molecules (i.e., about 10 7 to 10 8 ). Such antibody production most typically ceases (or substantially decreases) when the foreign antigen has been neutralized. Occasionally, however, proliferation of a particular B cell will continue unabated; such proliferation can result in a cancer referred to as “B cell lymphoma.”
  • T cells and B cells both comprise cell surface proteins which can be utilized as “markers” for differentiation and identification.
  • One such human B cell marker is the human B lymphocyte-restricted differentiation antigen Bp35, referred to as “CD20.”
  • CD20 is expressed during early pre-B cell development and remains until plasma cell differentiation. Specifically, the CD20 molecule may regulate a step in the activation process which is required for cell cycle initiation and differentiation and is usually expressed at very high levels on neoplastic (“tumor”) B cells.
  • CD20 by definition, is present on both “normal” B cells as well as “malignant” B cells, i.e., those B cells whose unabated proliferation can lead to B cell lymphoma.
  • the CD20 surface antigen has the potential of serving as a candidate for “targeting” of B cell lymphomas.
  • such targeting can be generalized as follows: antibodies specific to the CD20 surface antigen of B cells are, e.g., injected into a patient. These anti-CD20 antibodies specifically bind to the CD20 cell surface antigen of (ostensibly) both normal and malignant B cells; the anti-CD20 antibody bound to the CD20 surface antigen may lead to the destruction and depletion of neoplastic B cells. Additionally, chemical agents or radioactive labels having the potential to destroy the tumor can be conjugated to the anti-CD20 antibody such that the agent is specifically “delivered” to, e.g., the neoplastic B cells. Irrespective of the approach, a primary goal is to destroy the tumor: the specific approach can be determined by the particular anti-CD20 antibody which is utilized and, thus, the available approaches to targeting the CD20 antigen can vary considerably.
  • Murine (mouse) monoclonal antibody 1F5 an anti-CD20 antibody
  • 1F5 an anti-CD20 antibody
  • Extremely high levels (>2 grams) of 1F5 were reportedly required to deplete circulating tumor cells, and the results were described as being “transient.” Press et al., “Monoclonal Antibody 1F5 (Anti-CD20) Serotherapy of Human B-Cell Lymphomas,” Blood 69/2:584-591 (1987).
  • non-human monoclonal antibodies typically lack human effector functionality, i.e., they are unable to, inter alia, mediate complement dependent lysis or lyse human target cells through antibody dependent cellular toxicity or Fc-receptor mediated phagocytosis.
  • non-human monoclonal antibodies can be recognized by the human host as a foreign protein; therefore, repeated injections of such foreign antibodies can lead to the induction of immune responses leading to harmful hypersensitivity reactions.
  • HAMA Human Anti-Mouse Antibody response
  • these “foreign” antibodies can be attacked by the immune system of the host such that they are, in effect, neutralized before they reach their target site.
  • lymphoma cells are inherently sensitive to radiotherapy. Therefore, B cell malignancies are attractive targets for radioimmunotherapy (RIT) for several reasons: the local emission of ionizing radiation of radiolabeled antibodies may kill cells with or without the target antigen (e.g., CD20) in close proximity to antibody bound to the antigen; penetrating radiation, i.e., beta emitters, may obviate the problem of limited access to the antibody in bulky or poly vascularized tumors; and, the total amount of antibody required may be reduced.
  • target antigen e.g., CD20
  • penetrating radiation i.e., beta emitters
  • Radiolabeled antibodies include the use of a radioactive substance which may require the need for precautions for both the patient (i.e., possible bone marrow transplantation) as well as the health care provider (i.e., the need to exercise a high degree of caution when working with radioactivity).
  • “Chimeric” antibodies i.e., antibodies which comprise portions from two or more different species (e.g., mouse and human) have been developed as an alternative to “conjugated” antibodies.
  • Mouse/human chimeric antibodies have been created, and shown to exhibit the binding characteristics of the parental mouse antibody, and effector functions associated with the human constant region. See e.g., Cabilly et al., U.S. Pat. No. 4,816,567; Shoemaker et al., U.S. Pat. No. 4,978,745; Beavers et al., U.S. Pat. No. 4,975,369; and Boss et al., U.S. Pat. No.
  • chimeric antibodies are constructed by preparing a genomic gene library from DNA extracted from pre-existing murine hybridomas. Nishimura et al. (1987) Cancer Research 47: 999. The library is then screened for variable region genes from both heavy and light chains exhibiting the correct antibody fragment rearrangement patterns. The cloned variable region genes are then ligated into an expression vector containing cloned cassettes of the appropriate heavy or light chain human constant region gene. The chimeric genes are then expressed in a cell line of choice, usually a murine myeloma line.
  • Liu, A. Y., et al. “Production of a Mouse-Human Chimeric Monoclonal Antibody to CD20 with Potent Fc-Dependent Biologic Activity”, J. Immun. 139/10:3521-3526 (1987), describes a mouse/human chimeric antibody directed against the CD20 antigen. See also, PCT Publication No. WO 88/04936. However, no information is provided as to the ability, efficacy or practicality of using Liu's chimeric antibodies for the treatment of B cell disorders in the reference.
  • “In2B8” conjugate comprises a murine monoclonal antibody, 2B8, specific to human CD20 antigen, that is attached to Indium[111] ( 111 In) via a bifunctional chelator, i.e., MX-DTPA (diethylenetriaminepentaacetic acid), which comprises a 1: 1 mixture of 1-isothiocyanatobenzyl-3-methyl-DTPA and 1-methyl-3-isothiocyanatobenzyl-DTPA.
  • MX-DTPA diethylenetriaminepentaacetic acid
  • Indium-[111] is selected as a diagnostic radionuclide because it emits gamma radiation and finds prior usage as an imaging agent.
  • Patents relating to chelators and chelator conjugates are known in the art.
  • U.S. Pat. No. 4,831,175 of Gansow is directed to polysubstituted diethylenetriaminepentaacetic acid chelates and protein conjugates containing the same, and methods for their preparation.
  • U.S. Pat. Nos. 5,099,069, 5,246,692, 5,286,850, and 5,124,471 of Gansow also relate to polysubstituted DTPA chelates. These patents are incorporated herein in their entirety.
  • the specific bifunctional chelator used to facilitate chelation in application Ser. Nos. 08/475,813, 08/475,815 and 08/478,967 was selected as it possesses high affinity for trivalent metals, and provides for increased tumor-to-non-tumor ratios, decreased bone uptake, and greater in vivo retention of radionuclide at target sites, i.e., B-cell lymphoma tumor sites.
  • target sites i.e., B-cell lymphoma tumor sites.
  • other bifunctional chelators are known in the art and may also be beneficial in tumor therapy.
  • the Y2B8 conjugate comprises the same anti-human CD20 murine monoclonal antibody, 2B8, attached to yttrium-[90] ( 90 Y) via the same bifunctional chelator.
  • This radionuclide was selected for therapy for several reasons. The 64 hour half-life of 90 Y is long enough to allow antibody accumulation by the tumor and, unlike e.g.
  • 131 I is a pure beta emitter of high energy with no accompanying gamma irradiation in its decay, with a range of 100 to 1000 cell diameters.
  • the minimal amount of penetrating radiation allows for outpatient administration of 90 Y-labeled antibodies.
  • internalization of labeled antibodies is not required for cell killing, and the local emission of ionizing radiation should be lethal for adjacent tumor cells lacking the target antigen.
  • the Y2B8 conjugate possesses the same advantages discussed above, e.g., increased retention of radionuclide at a target site (tumor).
  • a diagnostic “imaging” radionuclide such as 111 In, can be used for determining the location and relative size of a tumor prior to and/or following administration of therapeutic chimeric or 90 Y-labeled antibodies for the purpose of tumor reduction.
  • indium-labeled antibody enables dosimetric assessment to be made.
  • radionuclides which have been used in clinical diagnosis include 131 I, 125 I, 123 I, 99 Tc, 67 Ga, as well as 111 In.
  • Antibodies have also been labeled with a variety of radionuclides for potential use in targeted immunotherapy (Peirersz et al. (1987) The use of monoclonal antibody conjugates for the diagnosis and treatment of cancer. Immunol. Cell Biol. 65: 111-125).
  • radionuclides include 188 Re and 186 Re as well as 90 Y, and to a lesser extent 199 Au and 67 Cu. I-[131] has also been used for therapeutic purposes.
  • U.S. Pat. No. 5,460,785 provides a listing of such radioisotopes and is herein incorparted by reference.
  • murine antibodies may find particular utility as diagnostic reagents.
  • a therapeutically effective dosage of the yttrium-labeled anti-CD20 antibody following administration of Rituxan® is sufficient to (a) clear any remaining peripheral blood B cells not cleared by the chimeric anti-CD20 antibody; (b) begin B cell depletion from lymph nodes; or (c) begin B cell depletion from other tissues.
  • conjugation of radiolabels to cancer therapeutic antibodies provides a valuable clinical tool which may be used to assess the potential therapeutic efficacy of such antibodies, create diagnostic reagents to monitor the progress of treatment, and devise additional therapeutic reagents which may be used to enhance the initial tumor-killing potential of the chimeric antibody.
  • additional therapeutic reagents which may be used to enhance the initial tumor-killing potential of the chimeric antibody.
  • radiolabeled proteins may be inherently unstable, particularly those labeled with radiolytic isotopes such as 90 Y, which have the tendency to cause damage to the antibody the longer they are attached to it in close proximity.
  • radiolysis causes unreliable efficiency of the therapeutic due to loss of radiolabel and/or reduced binding to the target antigen, and may lead to undesired immune responses directed at denatured protein.
  • clinicians have had no choice but to order therapeutic antibodies already labeled, or have them labeled off site at a related facility and transported in following labeling for administration to the patient. All such manipulations add precious time to the period between labeling and administration, thereby contributing to the instability of the therapeutic, while in effect decreasing the utility of radiolabeling kits in the clinical setting.
  • kits of the present invention allow rapid labeling which may be affected in approximately a half an hour or as little as five minutes depending on the label. Moreover, the kit protocols of the present invention have a labeling efficiency of over 95% thereby foregoing the need for further purification. By foregoing the need for further purification, the half-life of the radiolabel and the integrity of the antibody is reserved for the therapeutic purpose for which it is labeled.
  • kits and methods whereby diagnostic and therapeutic antibodies may be radiolabeled and administered to a patient in a reproducible, reliable and convenient manner.
  • the kits of the present invention transform the process of radiolabeling antibodies into a hassle-free, worry-free standardized process, which greatly facilitates patient treatment protocols.
  • the present kits provide advantages over the prior art in that the optimum parameters for labeling and administering therapeutic or diagnostic have been determined, thereby reducing the cost of goods. Since the kits described herein provide the optimum parameters according to the particular label, use of a kit designed for a particular label will also minimize cannibalization, i.e., which occurs when an inappropriate kit is used for a particular label. Avoiding cannibalization in turn also provides for optimum labeling efficiency. Moreover, the protocols and sterile, pyrogen-free ingredients included with each kit make for a more user-friendly process, since sterility, pyrogen testing and post-labeling purification of the reagents are obviated.
  • the present invention includes a kit for radiolabeling a diagnostic or therapeutic antibody before administration to a patient comprising at least (i) a vial containing a chelator-conjugated antibody, (ii) a vial containing formulation buffer for stabilizing and administering the radiolabeled antibody, and (iii) instructions for radiolabeling the antibody, wherein said vial components are supplied in such an amount and at such a concentration that when they are combined with a radiolabel of sufficient purity and activity according to the kit instructions, no further purification of the labeled antibody is required before administration to said patient.
  • such isotope incorporation may reach levels higher than 95%, and even as high as 98% or higher.
  • the antibody included in the kit is most preferably an anti-CD20 antibody.
  • the antibody is supplied in a form whereby it is attached to a bifunctional chelator.
  • the antibody is conjugated to MX-DTPA, but other chelators such as phenyl- or benzyl-conjugated DTPA, cyclohexyl-DTPA, EDTA derivatives and DOTA may be used.
  • a chelator according to the present invention may be any chelator that is at least bifunctional, i.e., which possesses at least two binding sites (at least one site for chelating a metallic ion and at least one site for coupling to a protein ligand).
  • the conjugated antibody is typically supplied at a concentration of 0.5 to 30 mg/ml, more preferably 2 mg/ml.
  • the volume of conjugated antibody will depend on the concentration and the amount required for optimum labeling depending on the radiolabel.
  • the conjugated antibody is to be supplied in such a volume and concentration that the entire volume will be added to the reaction vial using a sterile syringe and aseptic technique. This will allow for increased reproducibility and ease of use. All reagents of the kits disclosed herein are sterile and pyrogen-free, and specifically designed for simplicity and speed in advancing directly from antibody testing to administration. With some labels, the need for testing labeling efficiency may not be required.
  • a particularly advantageous component of the kit is the formulation buffer for stabilizing against the effects of radiolysis and administering the radiolabeled conjugated antibody to a patient.
  • the formulation buffer is a pharmaceutically acceptable carrier which serves as both a diluent for the labeled antibody and an administration buffer.
  • any pharmaceutically acceptable diluent may be used for administering therapeutic or diagnostic antibodies to patient, the formulation buffer of the present invention is particularly suited for administering radiolabeled antibodies.
  • the formulation buffer of the present invention comprises a radioprotectant such as human serum albumin (HSA) or ascorbate, which minimize radiolysis due to yttrium, and to a lesser degree, indium.
  • a radioprotectant such as human serum albumin (HSA) or ascorbate
  • HSA human serum albumin
  • Other radioprotectants are known in the art and could also be used in the formulation buffer of the present invention, i.e., free radical scavengers (phenol, sulfites, glutathione, cysteine, gentisic acid, nicotinic acid, ascorbyl palmitate, HOP(:O)H 2 , glycerol, sodium formaldehyde sulfoxylate, Na 2 S 2 O 5 , Na 2 S 2 O 3 , and SO 2 , etc.).
  • free radical scavengers phenol, sulfites, glutathione, cysteine, gentisic acid, nicotinic acid,
  • radioprotectants are generally employed in the formulation buffer to protect the antibody from radiolysis, it may be possible to affect further protection by including the radioprotectant in the reaction buffer as well. This has generally not been done before, i.e., with HSA, due to the presence of metals which would interfere with the labeling process. However, it may be possible to “clean” the HSA using a chelating resin such that it could be included in the reaction buffer as well. Ascorbate or other radioprotectants may also need to be treated to remove contaminating metals.
  • the formulation buffer of the present invention also comprises excess unconjugated chelator.
  • the purpose for including unconjugated chelator is that this chelator serves to scavenge any non-protein bound radiolabel in the patient, and effects excretion of the radiolabel thereby reducing uptake of “bone-seeking” isotopes, i.e., 90 Y, by the bones of the patient.
  • excess DTPA or any other chelator may be included in the formulation buffer.
  • the formation buffer is also preferably supplied in a volume such that the entire contents are transferred to the reaction vial. As discussed above, this results in increased ease of use and reproducibility because exact volumes do not have to be measured and transferred.
  • a preferred formulation buffer comprises phosphate buffered or physiological saline, human serum albumin and DTPA.
  • the human serum albumin is preferably at a concentration of between about 1 to 25% (w/v), and more preferably at a concentration of about 7.5% (w/v).
  • the concentration of DTPA is preferably about 1 mM.
  • Ascorbate may be used as an alternative to human serum albumin, and is typically used at a concentration of about 1 to 100 mg/ml. Although a wider range of concentrations may be used without compromising patient safety.
  • the antibody of the radiolabeling kit is readily labeled with a radioisotope of choice via a bifunctional chelator according to the methods of the present invention.
  • the kit of the present invention may also include a vial containing a buffer for adjusting the pH of the radioisotope solution, and a sterile glass reaction vial for performing the labeling and subsequently for resuspending the final radiolabeled antibody in formulation buffer.
  • a 10 ml reaction vial is typically sufficient, but vials capable of holding 5 to 20 mls may also be used.
  • the buffer is preferably a low-metal sodium acetate solution at a concentration of 10 to 1000 mM, most preferably 50 mM.
  • a specific kit of the present invention comprises the MX-DTPA conjugated antibody, 2B8-MX-DTPA.
  • 2B8 is an anti-CD20 antibody shown to affect B cell depletion upon administration to lymphoma patients.
  • the radiolabeling kit of the present invention may be optimized for the radiolabeling of other anti-CD20 antibodies, or any other antibody which has been conjugated to DTPA or other polyvalent chelator.
  • the preferred kit of the present invention may comprise at least (i) a vial containing the MX-DTPA conjugated 2B8 antibody, either in solution or lyophilized (requiring reconstitution); and (ii) a vial containing formulation buffer for administering the radiolabeled antibody to a patient.
  • the preferred kit will also contain (iii) a buffer for adjusting isotope pH, and (iv) a reaction vial.
  • the buffer is supplied in the reaction vial, thereby eliminating the steps of measuring and transferring the buffer and increasing the simplicity, consistency and sterility of the kit components.
  • the buffer is added to the isotope vial first, and the buffered isotope is then transferred to the reaction vial.
  • the reaction vial could be supplied with the required antibody volume.
  • the isotope/buffer vial could be made large enough to accommodate addition of the antibody conjugate, i.e., directly to the supplier's vial. This would eliminate the need for the reaction vial.
  • the reaction vial itself contains the required volume of conjugated antibody (i.e., 1 or 1.5 mL for 111 In and 90 Y, respectively).
  • the antibody may be supplied in a buffer that provides the appropriate radiolabeling pH according to the specific desired isotope (i.e., pH 3-6 for 111 In, pH 3-5 for 90Y). Different buffers may be used, depending on the isotope (i.e., sodium acetate for 90 Y, sodium citrate for 111 In).
  • the pH and composition of the buffer may also vary depending on the nature of the binding ligand to be labeled (i.e., labeling peptides may permit ⁇ pH 3 to be used).
  • the isotope would be transferred directly to the reaction vial, as would the formulation buffer. Limiting use of the kit to two transfer steps would further increase reproducibility and simplicity, and further decrease the chance for contamination of sterility during manipulation of the kit components.
  • the radiolabeling kits of the present invention may further comprise a vial of radioisotope, or radioisotope may be ordered separately from an appropriate supplier.
  • Preferred radioisotopes of the present invention are 111 In chloride and 90 Y chloride in HCl although the disclosed methods are not limited to these isotopes.
  • Other radionuclides that have been used for imaging applications are known in the art, i.e., as described in U.S. Pat. Nos. 4,634,586, 5,460,785 and 5,766,571, which are herein incorporated by reference.
  • Indium-[111] is particularly advantageous for imaging B cell tumors and beta emitters such as 90 Y are particularly useful as radiotherapeutic agents.
  • other radioisotopes suitable for these or other purposes i.e., alpha emitters, may be used depending on the chelator used for antibody conjugation.
  • kits embodiment will also include a separate vial of chimeric antibody, i.e., Rituxan®, to be administered before or after the radiolabeled anti-CD20 antibody.
  • chimeric antibody i.e., Rituxan®
  • the HAMA response which might generally occur in response to administration of a murine anti-CD20 antibody may be significantly decreased, thereby increasing the therapeutic utility of radiolabeled murine antibodies.
  • chimeric anti-CD20 is employed to clear circulating B cells, subsequent diagnostic images achieved with 111 In-labeled antibodies may be much clearer.
  • kits of the present invention may include separate, perhaps color-coded, buffer vials specifically formulated according to the optimum pH requirements for radiolabeling antibodies with the specific radioisotopes to be used. Such a system would ensure that the appropriate buffer was used for each label, and would allow the clinician the same ease in radiolabeling the two antibodies as if two kits had been purchased. Such a kit in effect combines the components from two radiolabeling kits into one.
  • the components of the radiolabeling kit of the present invention are supplied at the appropriate concentration and pH so that sterility is readily maintained before antibody administration and there is little need for additional buffers or media.
  • some of the reagents can be prepared, sterilized and tested for sterility on site.
  • variations of the kit of the invention are envisioned depending on the budget and preference of the consumer.
  • the radiolabeling kit of the present invention may be used in a method for radiolabeling a chelator-conjugated antibody for administration to a patient.
  • a method for radiolabeling a chelator-conjugated antibody for administration to a patient comprises, in general, (i) mixing a chelator-conjugated antibody with a solution containing a radioisotope; (ii) incubating the mixture for an appropriate amount of time at appropriate temperature; and (iii) diluting the labeled antibody to an appropriate concentration in formulation buffer, such that the radiolabeled antibody may be administered directly to a patient without further purification.
  • the antibody is an anti-CD20 antibody, and in particular, the anti-CD20 antibody may be 2B8.
  • the antibody may be conjugated to any appropriate chelator, i.e., MX-DTPA, CHX-DTPA, phenyl- or benzyl-DTPA, DOTA, EDTA derivatives, etc. MX-DTPA is preferred. Methods for affecting antibody conjugation are known in the art (Kozak et al. (1989); Mirzadeh et al. (1990), Brachbiel et al. (1986)).
  • the present inventors have found that the method of radiolabeling a chelator-conjugated antibody works best wherein the solution containing the radiolabel is adjusted to a pH of between about 3.0 and 6.0, and more preferably to about 4.2 before it is mixed with the chelator-conjugated antibody.
  • Low-metal sodium acetate is particularly preferred for adjusting the pH, although other buffers may be used.
  • the sodium acetate is at a concentration of between about 10 and 1000 mM, and more preferably 50 mM.
  • the volume quantity of 111 In chloride which should be used to prepare a single administrative dose is typically about 5.5 mCi divided by the radioactivity concentration at the time of labeling.
  • a typical diagnostic dose of 111 In is about 2 to 10 mCi.
  • the quantity of sodium acetate used for adjusting the pH varies depending on the sodium acetate concentration and the isotope carrier solution, and may therefor be quite broad.
  • the concentration of sodium acetate is 50 mM
  • the amount required for adjusting the pH is typically about 1.2 times the volume quantity of 111 In chloride used although larger volumes may be used.
  • the ratio of sodium acetate to HCl is what is important, and the amount of sodium acetate used would change depending on the amount and concentration of HCl in the buffer.
  • About 1 ml of a chelator-conjugated antibody at a concentration of about 2 mg/ml is then mixed with the radiolabel acetate solution, and the mixture is incubated for about 30 minutes, or for a time sufficient to achieve optimal labeling of the antibody. Such time may range from about 30 seconds to about 60 minutes.
  • Formulation buffer is then added in an amount necessary to achieve a total final volume of about 10 ml.
  • the optimum time required for labeling the antibody may vary depending on the antibody, the particular radiolabel and the particular conjugate employed.
  • An underlying factor in the optimization of the time allotted for radiolabeling is the chelator to antibody ratio of the reagent which is to be labeled.
  • the chelator to antibody ratio must be high enough to achieve a therapeutically useful level of incorporation, i.e., 90 to 95% depending on the radioisotope, but must also not be too high such that the structural integrity or immunoreactivity of the antibody is compromised. This requires a certain balancing process that in some cases may lead to a lower level of conjugated chelator and longer labeling time.
  • the volume quantity of 90 Y chloride used for preparing a single administrative dose typically ranges from about 10 to 50 mCi, and is preferably about 45 mCi, divided by the radioactivity concentration at the time of labeling.
  • the quantity of sodium acetate used for adjusting the pH varies depending on the sodium acetate concentration and the concentration of isotope carrier, and may therefor be quite broad.
  • the concentration of sodium acetate is 50 mM and the 90 Y is supplied in 50 mM HCl
  • the amount required for adjusting the pH is typically about 1.2 times the volume quantity of 90Y chloride used.
  • a chelator-conjugated antibody at a concentration of about 2 mg/ml is then mixed with the radiolabel acetate solution, and incubated for about 5 minutes, or for a time sufficient to achieve optimal labeling of the antibody. Such time may range from about 30 seconds to about 60 minutes.
  • Formulation buffer is added in an amount necessary to achieve a total final volume of about 10 ml.
  • the radiolabeling method of the invention is performed using the radiolabeling kit described herein.
  • the preferred components and conditions are merely acceptable guidelines for practicing the method of the invention, and may be altered to some degree with appropriate optimization. Conditions which depart from those preferred but still accomplish the purpose of the method are considered to be within the scope of the invention.
  • the radiolabeling kit of the present invention may also be supplied with reagents suitable for conveniently verifying the binding affinity of the antibody following radiolabeling.
  • the kit of the invention may also be used for determining the percent binding of a radiolabeled antibody to its target cell before administering the antibody to a patient.
  • the present inventors have also found that the particular binding assay kit disclosed may be useful for testing the affinity of any antibody generally for which purified antigen is not available. Accordingly, the binding assay components may also be sold as a separate kit.
  • a binding assay and radiolabeling kit comprises (i) at least one vial of lyophilized cells which express the antigen which is recognized by the antibody in the kit; (ii) a vial containing chelator-conjugated antibody; (iii) a vial containing formulation buffer, and (iv) instructions for radiolabeling the antibody such that the radiolabeled antibody may be administered directly to a patient without the need for subsequent purification.
  • this kit may also comprise a vial containing a buffer for adjusting the pH of the radioisotope, and a sterile glass reaction vial.
  • the buffer is a low-metal sodium acetate solution at a concentration of between about 10 and 1000 mM, and the glass reaction vial holds a volume of at least 5 ml.
  • the antibody is preferably an anti-CD20 antibody, and the chelator is preferably MX-DTPA. Other chelators may be used as described previously.
  • the preferred conjugated antibody is 2B8-MX-DTPA, although any chelator-conjugated antibody may be labeled and its affinity assessed.
  • the formulation buffer is phosphate buffered saline comprising a radioprotectant and unconjugated chelator as described above, and radioisotope may or may not be included and is preferably 111 In chloride or 90 Y chloride. Other radioisotopes may be used depending on the chelator.
  • the difference between the binding assay/radiolabeling kit and the radiolabeling kit described above is the inclusion of antigen-positive cells to serve as a substrate target for testing antibody affinity.
  • antigen is CD20
  • preferred CD20-positive cells are SB cells (ATCC # CCL 120) but any CD20-positive cells may be used.
  • the binding assay and radiolabeling kit may further include antigen-negative cells for use as a negative control.
  • Preferred CD20-negative cells are HSB cells (ATCC # CCL 120.1) but any CD20-negative cells may be used.
  • the combined radiolabeling and binding assay kit may further comprise a vial of chimeric anti-CD20 antibody in addition to the antibody to be labeled for the purposes of affecting a combined therapeutic regimen, or for clearing peripheral B cells prior to diagnostic imagery.
  • a separate antibody is preferably Rituxan®, but may be any antibody shown to effectuate tumor cell killing.
  • two different types of antibodies may be combined in one kit, i.e., antibodies directed to two different B cell antigens, so long as the combined therapeutic regimen serves to target the same type of cell, i.e., the B cell lymphoma.
  • the binding assay and radiolabeling kit of the present invention is particularly suited for the commercial setting in that the target cells are provided in lyophilized form. This allows the verification of antibody efficacy to proceed simply and systematically, and negates the hassle and expense involved in maintaining tissue culture facilities.
  • the lyophilized cells are generally supplied in aliquots of between 0.5 and 500 ⁇ 10 6 cells per vial according to the methods of the invention.
  • the present invention also provides for a binding assay kit for determining the percent binding of a radiolabeled antibody to its target cell.
  • a binding assay kit for determining the percent binding of a radiolabeled antibody to its target cell.
  • Such a kit includes at least one vial of fixed and/or lyophilized antigen-positive cells, and may optionally contain antigen-negative cells as described above for the binding assay and radiolabeling kit.
  • variations of such a kit may include an unlabeled control antibody for verifying the binding specificity of the consumer's antibody via a competitive assay.
  • the CD20-positive cells are preferably SB cells (ATCC # CCL 120) and the CD20-negative cells are preferably HSB cells (ATCC # CCL120. 1), which are supplied in lyophilized form in aliquots of between 0.5 and 50 ⁇ 10 6 cells.
  • the antibody is preferably an MX-DTPA conjugate of 2B8 labeled with 111 In or 90 Y.
  • the present inventors have also found that the method used for preparing fixed and lyophilized cells for the binding assay kits of the present invention is particularly suitable for preparing cells for commercial kits.
  • Cells may be fixed prior to lyophilization to improve structure/stability.
  • the cells of the present invention demonstrate high reproducibility when used for antibody binding assays.
  • the present invention includes a method of preparing lyophilized cells comprising (i) harvesting cells at a cell density of 0.5 to 2 ⁇ 10 6 cells per ml by centrifugation; (ii) washing cells at least one time in a balanced salt solution, i.e., HBSS; (iii) resuspending pelleted cells in a lyophilization buffer comprising a balanced salt solution containing carrier protein and at least one type of sugar; (iv) dispensing an aliquot of resuspended cells into a microfuge tube or a glass vial; and (v) lyophilizing the cells 12-96 h and more preferably 24-72 h at about 30-60 millitor.
  • the method is particularly suitable for preparing lyophilized cells wherein said cells are SB cells (ATCC # CCL 120) or HSB cells (ATCC # CCL 120.1), but is likely applicable to other cell types as well.
  • the buffer generally contains bovine serum albumin as the carrier protein at a concentration of 1% (w/v) and mannitol at a concentration of 10%.
  • carrier proteins i.e., HSA, and other sugars may be used.
  • the cells are harvested by centrifugation at a speed of about 1300 rpm, and the salt solution HBSS (Hank's balanced salt solution) is added.
  • the cells are generally resuspended at a concentration of 50 ⁇ 10 6 cells per ml.
  • the above conditions may be modified slightly without significantly compromising cell viability.
  • the above conditions may be supplemented by additional procedures designed to optimize the process for larger quantities of cells, e.g., tangential flow diafiltration to exchange cells into the lyophilization buffer.
  • the binding assay kits of the present invention may be used in an assay for assessing the binding affinity of a radiolabeled antibody. Such an assay is also a subject of the present invention.
  • a binding assay for determining the percent binding of a radiolabeled antibody to its target cell comprises in general the following steps: (i) mixing at least one aliquot of a radiolabeled antibody with at least one aliquot of antigen positive cells; (ii) mixing at least one aliquot of a radiolabeled antibody identical to the aliquot of step (i) with at least one aliquot of dilution buffer of the same volume as the aliquot of antigen-positive cells in step (i) as a control; (iii) pelleting the cells by centrifugation; (iv) measuring the radioactivity in the supernatant of the pelleted cells and the control; and (v) comparing the quantity of radioactivity in the cell supernatant to the quantity of radioactivity in the control.
  • the binding assay of the present invention is typically performed with antibodies labeled with 111 In or 90 Y.
  • 111 In is the radiolabel
  • radioactivity in the assay tubes is measure using a gamma counter.
  • 90 Y is the label, radioactivity is measured using a scintillation counter, although a gamma counter could be used.
  • the preferred antibody is an anti-CD20 antibody
  • the anti-CD20 antibody is preferably 2B8, wherein the 2B8 antibody is labeled using the radiolabeling kit of the present invention.
  • any radiolabeled antibody may be tested provided cells expressing the particular antigen are available.
  • CD20 is the antigen
  • the preferred cells for performing the assay are SB cells (ATCC # CCL 120), however, the assay may also be optimized and performed with any radiolabeled antibody and appropriate target cell.
  • the dilution buffer used for the assay should maintain binding of the antibody, i.e., physiological buffer, possibly containing a carrier protein, e.g. BSA, to minimize non-specific binding to cells.
  • a carrier protein e.g. BSA
  • a further control may be included in the assay by using antigen-negative cells.
  • the binding assay further comprises the following steps: (i) mixing at least one aliquot of a radiolabeled antibody with at least one aliquot of antigen-negative cells; (ii) pelleting the antigen-negative cells by centrifugation; (iv) measuring the radioactivity in the supernatant of the antigen-negative pelleted cells; and (v) comparing the quantity of radioactivity in the antigen-negative cell supernatant to the quantity of radioactivity in the supernatant of the antigen-positive cell supernatant and the control. Comparing the radioactivity obtained from this tube to the dilution buffer control will serve as a measure of the amount of non-specific binding to antigen-positive cells.
  • CD20 negative cells are preferably HSB cells (ATCC # CCL 120.1).
  • the lyophilized cells of the present invention provide a simple, efficient and reproducible standard for testing the binding efficacy of a radiolabeled antibody. Therefore, the binding assay of the present invention is preferably performed using the lyophilized cells included in the binding assay kits of the present invention.
  • the radiolabeling assays of the present invention may be combined with the binding assays of the present invention, wherein the antibody is first labeled by the method of labeling an chelator-conjugated antibody as described in the present invention. Most preferably, the binding assay of the present invention is performed using one of the binding assay and radiolabeling kits described herein.
  • the present invention also encompasses a competitive binding assay for assessing affinity of a test antibody to a target cell, comprising (i) preparing a ruthenium-labeled control antibody using a known antibody specific for the same antigen; (ii) incubating increasing amounts of test antibody and increasing amounts of unlabeled control antibody with a fixed concentration of target cells and a trace amount of ruthenium-labeled antibody wherein each separate concentration of test antibody and each separate concentration of control antibody are in separate tubes, respectively; (iii) determining the quantity of binding in each reaction tube based on relative electrochemiluminescence (ECL) using ORIGEN instrumentation; and (iv) calculating the average affinity value of the test antibody.
  • ECL electrochemiluminescence
  • the average affinity value may be calculated from the EC50 values and the known concentration of trace antibody using the method of Muller (J. Immunological Methods (1980) 34:345) or any other appropriate method. It should be noted that this assay may also be used to test the affinity of radiolabeled antibodies, or any antibody for which antigen cannot be purified and cells are required as an antigen source.
  • the fixed, lyophilized cells of the present invention may be used as target cells.
  • the control antibody may be 2B8, or any other unconjugated anti-CD20 antibody.
  • the control antibody may be a chelator-conjugated antibody.
  • the test antibody may also be a chelator-conjugate of the control antibody.
  • the test antibody may be another anti-CD20 antibody whose binding affinity to CD20 as compared to 2B8 is of interest.
  • the assay may be adapted for use with antibodies having other specificities so long as an appropriate target cell is available.
  • the preferred target cells are CD20-positive cells, more preferably SB cells (ATCC # CCL 120), and are more preferably resuspended lyophilized SB cells prepared according to the method of the present invention. Cells lyophilized using other methods or fixed cells may also be used.
  • the ruthenium-labeled antibody is typically prepared by a process comprising incubating the control antibody with N-hydroxysuccinimide ester of ruthenium (II) tris-bypyridine chelator (TAG-NHS), although other known method of labeling antibodies are also envisioned.
  • TAG-NHS N-hydroxysuccinimide ester of ruthenium (II) tris-bypyridine chelator
  • the control antibody and TAG-NHS are preferably incubated at about a 1:15 molar ratio.
  • FIG. 1 Immunoreactivity of native 2B8 was compared to commercially available anti-CD20 antibodies Bi (Coulter) and Leu 16 (Becton Dickinson) by direct competition in a radioimmunoassay using 125 I-labeled B1.
  • Antigen-positive SB cells 100,000 were added to each well of V&P filter plates; 10 ng of radiolabeled B1 was mixed with various concentrations of unlabeled competition and the mixture added to the cells.
  • the antibodies were incubated with the cells for one hour at ambient temperature; determinations were performed in triplicate. Subsequently, the wells were washed, dried and the filter-associated radioactivity determined. The data shown were corrected for background radioactivity and are the means of triplicate determinations.
  • FIG. 2 Increasing amounts of unconjugated 2B8 were analyzed for binding to human B-cells (SB) using FACS analysis. Comparisons were made with a commercially available anti-CD20 monoclonal antibody (B1) and with two irrelevant isotype antibodies. Goat anti-mouse IgG-FITC F(ab)′ 2 was used as the secondary reagent. The results show that 2B8 is specific for the CD20 antigen and that it exhibits greater binding than B1.
  • FIG. 3 Human B-cells (SB) were incubated with increasing amounts of 125 I-labeled 2B8. Triplicate samples were incubated for one hour and cell-bound radioactivity was determined after filtration to collect cells. Scatchard analysis allowed calculation of an apparent affinity constant of 4.3 ⁇ 10 ⁇ 9 M.
  • FIG. 4 Immunoreactivity of native 2B8, 2B8-MX-DTPA, and B1.
  • the B1 antibody was radiolabeled as described in the Methods section. Ten nanograms of radiolabeled B1 were mixed with increasing concentrations of the competitor and the mixture added to wells of V&P filter plates containing 100,000 antigen-positive SB cells each; all determinations were performed in triplicate. Following a one hour incubation at ambient temperature, the wells were washed extensively. Subsequently, the filters were dried and the associated radioactivity determined by gamma counting; all values were corrected for background. Values shown are the means of triplicate determinations.
  • FIG. 5 Antibody 2B8 was formulated at a final concentration of 10 mg/mL in normal saline or normal saline containing 10 mM glycine-HCl, pH 6.8. Duplicate sets of samples were then placed in screw-capped vials, the vials purged with nitrogen, and then capped. The samples were then incubated at 4° C. or 30° C. for 12 weeks; the immunoreactivity of the samples was evaluated weekly. No loss of immunoreactivity was observed with any of the 2B8 samples throughout the 12-week study. Immunoreactivities at week 1 (FIG. 5A), week 6 (FIG. 5B) and week 12 (FIG. 5C) are depicted.
  • FIG. 6 Binding assay for determination of immunoreactivity of 111 In-labeled 2B8-MX-DTPA incubated in PBS, pH 7.4 containing 50 mg/mL human serum albumin (48 h incubation).
  • FIG. 6A A constant amount of radiolabeled antibody (5 ng/mL) was incubated with increasing volumes of SB cells (20 ⁇ 106 cells/mL). The amount of radioactivity (cpm) bound to cells was plotted against the volume of cell suspension added.
  • FIG. 6B Total applied radioactivity over bound radioactivity (AT/B) was plotted. Linear extrapolation allowed calculation of the y-intercept (0.997). The reciprocal of the y-intercept ⁇ 100 yielded an immunoreactivity value of 100% at infinite antigen excess.
  • FIG. 7 Autoradiograms obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and lmM DTPA. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions, denaturing conditions (SDS). The samples were loaded at 5 ⁇ L (lanes 1,2), 10 ⁇ L (lanes 5,6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • SDS denaturing conditions
  • FIG. 8 Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Trib-glycine gel using non-reducing conditions. Samples were loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 96.2%.
  • FIG. 9 Densitometric scan of 48 h autoradiogram obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. Samples were loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 95.5%.
  • FIG. 10 Autoradiograms obtained from SDS-PAGE analysis of 111 In-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 50 mg/mL human serum albumin. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions. The samples were loaded at 5 ⁇ L (lanes 1, 2), 10 ⁇ L (lanes 3, 4), and 20 ⁇ L (lanes 5, 6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed. (Note: The 48 h autoradiogram was obtained using intensifying screens resulting in a more intense signal compared to the time zero autoradiogram).
  • FIG. 11 Densitometry scan of time zero autoradiogram obtained from SDS-PAGE analysis of 111 In-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 50 mg/mL human serum albumin. The sample was electrophoresed on a 4-20% Tris-glycine gel under non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#3) was 95.9%.
  • FIG. 12 Densitometry scan of 48 h autoradiogram obtained from SDS-PAGE analysis of 111 In-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 50 mg/mL human serum albumin. The sample was electrophoresed on a 4-20% Tris-glycine gel under non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes scanned using a densitometer. The relative area of the radiolabeled conjugate was 97.0% (combined areas of peaks #2, 3, and 4).
  • FIG. 13 Autoradiograms obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non- reducing conditions. The samples were loaded at 5 ⁇ L (lanes 1, 2), 10 ⁇ L (lanes 3, 4), and 20 ⁇ L (lanes 5, 6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • FIG. 14 Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ,L, and 20 ⁇ L in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 97.9%.
  • FIG. 15 Densitometric scan of 98 h autoradiogram obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 94.7%.
  • FIG. 16 Autoradiograms obtained from SDS-PAGE analysis of 111 In-labeled 2B8-MX-DTPA incubate at 37° C. in human serum. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions. The samples were loaded at 5 ⁇ L (lanes 1, 2), 10 ⁇ L (lanes 3, 4), and 20 ⁇ L (lanes 5, 6). The gels were exposed to x-ray film for approximately 16-20 h at ambient temperature and photographed.
  • FIG. 17 Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of 111 In-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 16-20 h at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#3) was 95.3%.
  • FIG. 18 Densitometric scan of the 96 h autoradiogram obtained from SDS-PAGE analysis of 111 In-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 16-20 h at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#3) was 94.0%.
  • FIG. 19 Cynomolgus monkeys were injected intravenously every 48 hours for a total of seven injections; the amounts administered are shown. Circulating T- and B-cell levels were determined by FACS analysis using anti-CD2 (T-cell), anti-Mo-IgG (2B8), anti-CD20 (Leu 16), and anti-human-IgG (B-cell). No effect was observed on circulating T-cell levels. (Group V animals were given a single dose).
  • FIG. 20 The recovery of circulating B-cell levels in animals receiving 2B8 was followed by FACS analysis using the fluorescently-labeled antibodies described in the brief description of FIG. 19. The animals in Groups III and IV were not monitored as they were sacrificed on day 13.
  • FIG. 21 Cynomolgus monkeys were injected intravenously with 89y 2B8-MX-DTPA which had been prepared using clinical-grade 2B8-MX-DTPA. The animals were dosed every 48 hours with the amounts shown above for a total of seven doses. On days 0, 2, 7, 10 and 14 the monkeys were bled and evaluated for serum chemistries hematology and circulating B-cell levels (day 10 sera were not analyzed for B-cell content). Other than decreased total lymphocyte count in all animals, except one individual in groups II, no significant abnormalities were noted during the course of the study.
  • FIG. 22 The clearance of murine anti-CD20 antibody 2B8 from cynomolgus monkeys was determined by ELISA following a single injection of 10 mg/kg on day zero. As shown in panel A, the antibody exhibited at ⁇ t 1 ⁇ 2 value of approximately 4.5 days. The clearance of the 2B8 antibody and its MX-DTPA conjugate from the circulation of BALB/c mice are shown in panel B. Mice were injected intravenously with 25 ⁇ g of native or conjugated 2B8 and blood samples taken at various times up to 264 hours following injection; sera was subsequently analyzed by enzyme immunoassay using SB cells as the capture agent. Both the native and conjugated antibodies exhibited clearance values of 8.75 days.
  • FIG. 23 Twenty BALB/c mice were each injected with 1.1 ⁇ Ci of radiolabeled conjugate (100 ⁇ L) formulated in PBS, pH 7.4, containing 50 mg/mL HSA. Groups of five mice each were sacrificed at 1, 24, 48, and 72 hours and then blood and various tissues prepared and analyzed for associated radioactivity.
  • FIG. 24 Twenty BALB/c mice were each injected intravenously with approximately 1.0 ⁇ Ci (in 100 ⁇ l) of radiolabeled conjugate formulated in 1 ⁇ PBS, pH 7.4, containing 75 mg/mL human serum albumin and 1 m MDPA. Groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their blood and various tissues prepared and analyzed for associated radioactivity.
  • FIG. 25 Athymic mice bearing Ramos B-cell tumors were injected intravenously with 24 ⁇ Ci of 111 -In-2B8-MX-DTPA and groups of three mice each were sacrificed at 0, 24, 48 and 72 hours. Following tissue preparation and determination of associated radioactivity, the percent injected dose per gram tissue values were determined and plotted as shown.
  • FIG. 26 Binding assay for determination of immunoreactivity of “mix-&-shoot” 90 Y-labeled 2B8-MX-DTPA incubated in PBS, pH 7.4 containing 50-75 mg/mL human serum albumin (48 h incubation).
  • Panel A A constant amount of 90 Y-labeled antibody (approximately 1 ng/ml) was incubated with increasing amounts of SB cells. The amount of radioactivity (cpm) bound to cells was plotted against the cell concentration.
  • Panel B Total applied 90 Y radioactivity over bound radioactivity (AT/B) was plotted. Linear extrapolation allowed calculation of the y-intercept (1.139). The reciprocal of the y-intercept ⁇ 100 yielded an immunoreactivity value of 87.9% at infinite antigen excess. No binding was observed with CD20-negative cells (HSB).
  • FIG. 27 Autoradiograms obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions, denaturing conditions (SDS). The samples were loaded at 5 ⁇ L (lanes 1,2), 10 ⁇ L (lanes 5,6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • FIG. 28 Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Trib-glycine gel using non-reducing conditions. Samples were loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 96.1%.
  • FIG. 29 Densitometric scan of 48 h autoradiogram obtained from SDS-PAGE analysis of 90 Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. Samples were loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 94.1%.
  • FIG. 30 Autoradiograms obtained from SDS-PAGE analysis of “mix-&-shoot” 90 Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions. The samples were loaded at 5 ⁇ L (lanes 1, 2), 10 ⁇ L (lanes 3, 4), and 20 ⁇ L (lanes 5, 6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • FIG. 31 Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of “mix-&-shoot” 90 Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 89.1%.
  • FIG. 32 Densitometric scan of 72 h autoradiogram obtained from SDS-PAGE analysis of “mix-&-shoot” 90 Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 ⁇ L, 10 ⁇ L, and 20 ⁇ L in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 88.8%.
  • FIG. 33 Twenty BALB/c mice were each injected intravenously with 5 ⁇ Ci 90 Y-labeled 2B8-MX-DTPA formulated in 1 ⁇ PBS, pH 7.4, containing 75 mg/mL human serum albumin and 1 mM DTPA. Groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their blood and various tissues prepared and analyzed for associated radioactivity.
  • FIG. 34 Increasing amounts of CHO-derived 2B8 antibody labeled were incubated with a fixed concentration of freshly harvested CD20-positive B-cells (SB) or CD20-negative T-cells (HSB). Antibody binding to cells was quantified using FACS analysis using goat anti-mouse IgG-FITC F(ab)′ 2 as described herein. Comparison was made to an irrelevant isotype antibody (S004). Only the CHO-derived 2B8 antibody showed any appreciable binding to CD20-positive SB cells.
  • FIG. 35 The immunoreactivity of CHO-derived 2B8 was compared to the 2B8-49 parent antibody produced in a hybridoma cell line by direct competition in an ORIGEN assay. Increasing amounts of antibody was incubated with a fixed concentration of CD20-positive B-cells (SB) and a trace amount of ruthenium-labeled CHO 2B8. After incubation for three hours at ambient temperature, binding, expressed as relative electrochemiluminescence (ECL), was determined using the ORIGEN instrument as described in the Materials and Methods. Values represent the means of duplicate determinations. Average affinity constants for CHO 2B8 and 2B8-49 were calculated to be 1.3 ⁇ 10 ⁇ 10 M and 2.5 ⁇ 10 ⁇ 10 M, respectively. An irrelevant isotype antibody (S004), was included for comparison.
  • S004 CD20-positive B-cells
  • FIG. 36 The binding of 2B8-MX-DTPA conjugates prepared from CHO-derived 2B8 was compared to the unconjugated antibody by direct competition in an ORIGEN assay.
  • Conjugates were prepared by incubation of 2B8 with MX-DTPA for 8, 17, and 24 h before removal of unreacted chelate.
  • antibodies were incubated with a fixed concentration of CD20-positive B-cells (SB) and a trace amount of ruthenium-labeled CHO 2B8. After incubation for three hours at ambient temperature, binding, expressed as relative electrochemiluminescence (ECL), was determined using the ORIGEN instrument as described in the Materials and Methods. Values represent the means of duplicate determinations.
  • Conjugate preparations exhibited similar binding compared to unconjugated 2B8 antibody.
  • FIG. 37 A) SB cells were washed and resuspended to 90 ⁇ 10 6 cells/mL with dilution buffer (1 ⁇ PBS, pH 7.4 containing 1% (w/v) bovine serum albumin. Increasing concentrations of cells were incubated for 3 h with 7.5 ng/mL In2B8 prepared using 2B8-MX-DTPA lot 0165A. B) Double-inverse plot of cell concentration vs. bound radioactivity/total radioactivity (B/AT). Immunoreactivity was calculated as 1/y-intercept ⁇ 100. Immunoreactivity and correlation coefficient (R) values were 80.6% and 0.981, respectively.
  • FIG. 38 A) SB cells were washed and resuspended to 90 ⁇ 10 6 cells/mL with dilution buffer (1 ⁇ PBS, pH 7.4 containing 1% (w/v) bovine serum albumin. Increasing concentrations of cells were incubated for 3 h with 2 ng/mL Y2B8 prepared using 2B8-MX-DTPA lot # 0165A. B) Double-inverse plot of cell concentration vs. bound radioactivity/total radioactivity (B/AT). Immunoreactivity was calculated as 1/y-intercept x 100. Immunoreactivity and correlation coefficient (R) values were 72.2% and 0.999, respectively.
  • low metal refers to reagents treated to reduce metal contamination to a level which does not impact radioincorporation
  • antigen positive means expresses antigen that is recognized by particular antibody of the invention in such a way that the antibody is capable of binding.
  • % radioincorporation refers to the amount of radiolabel from a radiolabeling reaction that is conjugated to the antibody relative to the total amount of radiolabel initially added to the reaction.
  • % binding refers to the amount of antibody from a sample which binds to the target antigen, with or without specificity.
  • % immunoreactivity or binding specificity refers to that amount of an antibody sample which binds to the target antigen with specificity.
  • diagnostic antibody refers to an antibody conjugated to a radiolabel such as 111 In which can effect diagnostic imaging of tumors and antigen positive cells.
  • therapeutic antibody refers to an antibody conjugated to a alpha or beta emitting radiolabel (such as 90 Y) which can effect cell killing when bound to the targeted antigen.
  • a alpha or beta emitting radiolabel such as 90 Y
  • the human cell lines SB and HSB were obtained from the American Type Culture Collection and cultured in RPMI-1640 containing 10% fetal bovine serum.
  • the CD20-positive SB cell line is a B lymphoblastoid cell line derived from the peripheral blood buffy coat of a patient with acute lymphoblastic leukemia (1).
  • the antigen-negative cell line HSB is a T lymphoblastoid cell line developed from tumors induced in newborn Syrian hamsters (2).
  • the murine myeloma cell line SP2/0 was similarly maintained in RPMI-1640 containing 10% fetal bovine serum.
  • the anti-CD20 antibodies Bi and Leu 16 were purchased from Coulter Immunology and Becton/Dickinson, respectively.
  • the 125 I-labeled goat anti-mouse IgG and goat anti-human IgG antibodies were obtained from ICN.
  • Goat F(ab′)2 anti-mouse IgG was obtained from Cappel.
  • Freund's complete and incomplete adjuvants were purchased from Sigma Chemical Company. Polyethylene glycol, HAT concentrate, and HT concentrate were all obtained from Boehringer Mannheim. Fluorescein isothiocyanate (FITC) was purchased from Sigma Chemical Company. Indium-[ 111 ] chloride and 90 Y chloride were obtained from Amersham or NEN Dupont. Yttrium-[89] chloride was purchased from Aldrich Chemical Company. All other reagents were obtained from standard sources.
  • FITC Fluorescein isothiocyanate
  • Reagents used for conjugation and radiolabeling protocols were processed to remove contaminating heavy metal ions which could compete with the radioisotopes during the radiolabeling step.
  • Reagents were typically processed by passing the solutions through a column of Chelex 100 ion exchange resin (BioRad Industries) or batch processing by addition of Chelex 100 to a prepared solution.
  • Low metal-containing water, either Milli-Q-purified or Water for Irrigation (WFIr) was used for all preparations and dilutions.
  • the metal-free solutions were sterile-filtered and collected in sterile plastic containers.
  • mice Ten BALB/c mice were immunized with 20 million SB cells suspended in PBS containing Freunds complete adjuvant. The cells were injected both s.c and i.p at multiple sites on the animal. After a 2 week rest period the mice were injected a second time with SB cells emulsified in Freund′ s incomplete adjuvant. Subsequent immunization boosters were performed on a weekly schedule with SB cells suspended in PBS. Mice were immunized for a period of 6 weeks to 4 months.
  • Radiolabeled antibody was placed in each well of a previously blocked filter assay plate (blocking buffer: dilution buffer containing 10% FBS) along with 50 ⁇ L of hybridoma supernatant from test wells and 100,000 SB cells suspended in 50 ⁇ L dilution buffer. The suspension was incubated for one hour at ambient temperature. The plates were washed thoroughly with wash buffer (PBS, pH 7.4, containing 0.2% gelatin and 0.02% sodium azide) on a V&P Scientific vacuum manifold and filter bottoms containing trapped SB cells were transferred to a gamma counter. Wells containing only HAT media and labeled B1 antibody were used as background controls and identical wells containing SB cells were used as positive controls. Inhibition controls contained radiolabeled B1 and various amounts of unlabeled B1 antibody ranging from 2 ⁇ g to 2 ng.
  • blocking buffer dilution buffer containing 10% FBS
  • fluorescein was coupled to purified 2B8 antibody through the reaction of amino groups with fluorescein isothiocyanate (FITC). Briefly, 2B8 antibody (1.2 mg/mL) was incubated in pH 9.5, 0. 1M sodium carbonate buffer with 150-200 ⁇ g FITC per mg protein. The solution was incubated at room temperature for 2 hours and the resulting 2B8-FITC conjugate was purified on a Sephadex G-25 column. Other reagents used in these studies such as B1 and Leu 16 were purchased as fluorescein conjugates directly from Coulter or Becton Dickinson.
  • FITC fluorescein isothiocyanate
  • Cells to be analyzed were harvested and washed three times with PBS containing 0.2% BSA and 0.1% sodium azide. Viability was determined by trypan blue exclusion with a viability requirement of >90%. Cell concentrations were adjusted to 3 million per ml with 50 ⁇ L added per well into 96 well U-bottom plates. Primary antibody (50 ⁇ L) was added to appropriate wells and the mixture incubated for 30 min. to 1 h. at ambient temperature; subsequently the cells were washed 5 times with 200 ⁇ L/well of PBS containing 0.2% BSA and 0.02% sodium azide. Cells were centrifuged in the plates at 1300 RPM for 1 min.
  • the cells were washed twice by centrifugation and resuspension in HBSS. Fetal bovine serum (2 mL) was added and the cells resuspended. One hundred microliters of the resuspended cells were then distributed to each of 6, 15 ml conical centrifuge tubes.
  • Tube #1 Murine anti-CD2-FITC (AMAC), 2.5 ⁇ g/mL, 5 ⁇ g; Tube #2: Goat anti-Human IGM-FITC (Fisher) 2.5 ⁇ g/mL, 5 ⁇ g; Tube#3: Goat anti-mouse IgG-RPE (Fisher) 2.5 ⁇ g/mL, 5 ⁇ g; Tube #4: Goat anti-Human IgM-FITC + Goat anti-mouse IgG-RPE (absorbed), 2.5 ⁇ g/mL, 5 ⁇ g; Tube #5: anti-human CD20-FITC (anti-Leu 16, Becton Dickinson), 5 ⁇ g; Tube #6: Cells only (auto-fluorescence).
  • AMAC Murine anti-CD2-FITC
  • Tube #2 Goat anti-Human IGM-FITC (Fisher) 2.5 ⁇ g/mL, 5 ⁇ g
  • Tube#3 Goat anti-mouse IgG-RPE (Fisher) 2.5 ⁇
  • pre-study lymphocyte levels were determined for each cynomolgus monkey and used as baseline values. The percentage of T- and B-cells and T:B ratios were calculated and used as depletion references. The pre-study B cell population was enumerated with Leu 16 and anti-human IgM antibodies.
  • the extract was centrifuged in a microfuge at high speed for 30 min and the supernatants were transferred to new tubes.
  • Protein A-Sepharose 300 ⁇ L was added to each tube and the resin pelleted by centrifugation. The protein A-Sepharose was then washed 20 times to remove non specifically bound iodinated protein. When the bead-to-supernatant radioactivity ratio reached a value of 100, the pellet was extracted with SDS PAGE sample buffer and heated to boiling. After cooling, approximately 15,000 cpm of each of the extracts were added to wells of a 10% polyacrylamide gel. A low molecular weight pre-stained standard (BioRad Inc.) was added to a separate well and used for molecular weight estimation. The proteins were resolved by electrophoresis and the gel was dried and exposed to a sheet of X-ray film for 24 hours at ⁇ 70° C.; subsequently the film was developed and analyzed.
  • Radiolabeled 2B8 was prepared by reaction with 125 I in the presence of lodobeads. Following removal of free iodine the radiolabeled antibody was incubated in various concentrations, in duplicate, ranging from 5000 ng per well to 35 ng/well with 10,000 SB cells. The amount of antibody binding to cells was calculated from the specific activity of the 125 1-labeled 2B8. The ratio of bound/free antibody was plotted against the molar concentration of bound antibody and the apparent affinity constant was determined from the ratio of the X and Y axis intercepts.
  • MX-DTPA carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylenetriaminepentaacetic acid
  • the murine anti-CD20 antibody 2B8 was purified initially from ascites by Protein A and QAE chromatography. For later experiments 2B8 was purified from hollow-fiber bioreactor supernatants using the same purification process. The hollow-fiber-derived antibody has now been replaced for commercialization purposes with the CHO-derived antibody described in Example 2.
  • the antibody was prepared for conjugation by transferring it into metal-free 50 mM bicine-NaOH, pH 8.6, containing 150 mM NaCl, using dialysis or repetitive buffer exchange.
  • buffer exchange was effected using repetitive ultrafiltration with Centricon 30 (Amicon) spin filters (30,OOOD MWCO).
  • 50-200 ⁇ L of protein (10 mg/mL) was added to the filter unit and 2 mL of bicine buffer added.
  • the filter was centrifuged at 4° C. in a Sorval SS-34 rotor at 6,000 rpm for 45 min. Retentate volume was approximately 50-100 ⁇ L. This process was repeated twice with the same filter.
  • Retentate was transferred to a polypropylene 1.5 mL screw cap tube, assayed for protein, diluted to 10.0 mg/mL and stored at 4° C. until used for conjugation.
  • the protein was transferred into 50 mM sodium citrate, pH 5.5 containing l5OmM NaCl and 0.05% sodium azide using the same protocol described above.
  • Conjugation of 2B8 with MX-DTPA was performed in polypropylene tubes at ambient temperature. Frozen stock solutions of MX-DTPA were thawed immediately before use. Typically, 50-200 ⁇ L of antibody at 10 mg/mL were reacted with chelate at a molar ratio of chelate-to-protein of 4:1. Reactions were initiated by adding the chelate stock solution and gently mixing; the conjugation was allowed to proceed overnight, generally for 14 to 20 h, at ambient temperature. Unreacted chelate was removed from the conjugate by dialysis or repetitive ultrafiltration, as described above, into metal-free normal saline (0.9% w/v) containing 0.05% sodium azide. The protein concentration was adjusted to 10 mg/mL and stored at 4° C. in a polypropylene tube until radiolabeled.
  • Chelate incorporation was determined by scintillation counting and comparing the value obtained with the purified conjugate to the specific activity of the carbon-[14]-labeled chelate. For later studies, in which non-radioactive chelate obtained from Coulter Immunology was used, chelate incorporation was assessed by incubating the conjugate with an excess of a radioactive carrier solution of 90 Y of known concentration and specific activity.
  • a stock solution of yttrium chloride of known concentration was prepared in metal-free 0.05 N HC1 to which carrier-free 90 Y (chloride salt) was added. An aliquot of this solution was analyzed by liquid scintillation counting to determine an accurate specific activity for this reagent.
  • a volume of the yttrium chloride reagent equal to 3-times the number of mols of chelate expected to be attached to the antibody, typically 2 mol/mol antibody, was added to a polypropylene tube, and the pH adjusted to 4.0-4.5 with 2 M sodium acetate. Conjugated antibody was subsequently added and the mixture incubated 15-30 min at ambient temperature. The reaction was quenched by adding 20 mM EDTA to a final concentration of 1 mM and the pH of the solution adjusted to approximately pH 6 with 2M sodium acetate.
  • Samples for assay were diluted in 1 ⁇ PBS/1% BSA, applied to plates and serially diluted (1:2) into the same buffer. After incubating plates for 1 h at ambient temperature, the plates were washed three times with 1 ⁇ PBS. Secondary antibody (goat anti-mouse IgGl-specific HRP conjugate) (50 ⁇ L) was added to wells (1:1500 dilution in 1 ⁇ PBS/1% BSA) and incubated 1 h at ambient temperature. Plates were washed four times with 1 ⁇ PBS followed by the addition of ABTS substrate solution (50 mM sodium citrate, pH 4.5 containing 0.01% ATBS and 0.001% H202). Plates were read at 405 nm after 15-30 min incubation.
  • ABTS substrate solution 50 mM sodium citrate, pH 4.5 containing 0.01% ATBS and 0.001% H202
  • Antigen-negative HSB cells were included in assays to monitor non-specific binding. Immunoreactivity of the conjugate was calculated by plotting the absorbance values vs. the respective dilution factor and comparing these to values obtained using native antibody (representing 100% immunoreactivity) tested on the same plate. Several values on the linear portion of the titration profile were compared and a mean value determined.
  • the radiolabeled conjugate was diluted to a final antibody concentration of 1-5 ng/mL with dilution buffer and 0.35 mL was added to each tube. Following a 75-90 min incubation period at ambient temperature the cells were pelleted by centrifugation and the supernatants collected. Radioactivity remaining in the supernatant fraction was determined with a gamma or scintillation counter. The data were plotted as the quotient of the total radioactivity added divided by the cell-associated radioactivity, versus the inverse of the cell number per tube. The y axis intercept thus represents the immunoreactive fraction.
  • radiolabeled conjugates were diluted 1:10 with normal human serum (non-heat-inactivated) and aliquots placed aseptically into sterile capped tubes; these tubes were then incubated at 37° C. for periods up to 96 hours. At selected times conjugate samples were removed and analyzed by non-reducing SDS-PAGE in 4-20% gradient gels followed by autoradiography, and by instant thin layer chromatography.
  • the 2B8-MX-DTPA conjugate was radiolabeled with 111 In and used without HPLC purification (“mix-and-shoot” protocol).
  • the radiolabeled antibody was diluted into PBS and human serum albumin (HSA) added to a final concentration of 50 mg/mL.
  • HSA human serum albumin
  • the specific activity of the formulated radiolabeled conjugate was 2.2 mCi/mg.
  • the formulated conjugate was subsequently incubated at 4° C. for 48 hours and aliquots analyzed at time 0, 24 h and 48 hours using non-reducing SDS-PAGE in 4-20% gradient gels followed by autoradiography, and by instant thin layer chromatography.
  • the immunoreactivity at each time point was assessed using the whole-cell suspension assay described in section 1 above.
  • the 2B8-MX-DTPA conjugate was radiolabeled with 90 Y and purified by size-exclusion chromatography on HPLC using 1 ⁇ PBS as an elution buffer.
  • the radiolabeled conjugate fractions were pooled and human serum albumin and DTPA were added to final concentrations of 75 mg/mL and 1 mM, respectively.
  • the specific activity of the formulated radiolabeled conjugate was 14.6 mCi/mg.
  • the formulated conjugate was subsequently incubated at 4° C. for 48 hours and aliquots analyzed at time 0, 24 h and 48 hours using non-reducing SDS-PAGE in 4-20% gradient gels followed by autoradiography, and instant thin layer chromatography. Immunoreactivity at each time point was assessed using the whole-cell suspension assay described in section 1 above.
  • Antibody 2B8 was evaluated in a high-dose pharmacology study performed under GLP regulations at White Sands Research Center (Study Number 920111).
  • Adult Macaca fascicularis (cynomolgus) monkeys were used; study groups each consisted of one male and one female.
  • the antibody was injected intravenously every 48 hours for a total of seven injections.
  • the study consisted of five groups: Group I (saline); Group II (0.6 mg/kg); Group III (2.5 mg/kg); Group IV (10 mg/kg); and, Group V (10 mg/kg on day 0 only).
  • the mean serum beta half life of 2B8 in cynomolgus monkeys was determined using Group V animals (above).
  • Goat anti-mouse IgGl (Fisher Scientific) was diluted to 2.0 ⁇ g per ml in 10 mM borate buffer, pH 9.6, and 50 ⁇ L was added to each well of a 96-well plate.
  • the antibody was allowed to bind to the plate during an overnight incubation at 4° C., or for 2 h at ambient temperature. Each plate was blocked for 30 min. at ambient temperature with 150 ⁇ L per well of PBS containing 1% BSA.
  • the plates were washed with distilled water and serum or plasma samples were applied in triplicate to individual wells at 1: 100 initial dilution followed by serial 1:2 dilutions.
  • Purified 2B8 was added to pre-bleed sera and diluted for use as a standard curve beginning with 0.5 mg/mL; samples were diluted 1:100 and then serially diluted as with the other samples.
  • the plates were incubated for 1 h at ambient temperature and washed 4 times with distilled water.
  • the secondary reagent (goat anti-mouse IgGl-HRPO) was then added at 1:4000 dilution and incubated at ambient temperature for an additional hour.
  • the plates were washed again in distilled water and 0.1 mL peroxidase substrate was added containing hydrogen peroxide. Color was allowed to develop from the reaction for 20 min.; the absorbance was subsequently determined at 405 nm using a microplate ELISA reader. The results were plotted in ⁇ g antibody per mL serum.
  • ⁇ t 1 ⁇ 2 values of 2B8 and 2B8-MX-DTPA were determined in BALB/c mice.
  • Unconjugated 2B8 stored at ⁇ 70° C. in 1 ⁇ PBS, pH 7.4/10% glycerol was thawed, diluted to 0.5 mg/mL and sterile filtered.
  • Conjugated antibody was prepared following standard protocols but with carbon-[14]-labeled chelate; chelate incorporation was 1.5 mol/mol antibody.
  • the purified conjugate was diluted to 0.5 mg/mL in normal saline (0.9%), sterile filtered, and stored at 4° C. with the native antibody until used.
  • mice Six-to-eight week old mice were injected with 100 ⁇ L of purified 2B8 antibody at a concentration of 250 ⁇ g/mL. Mice were subsequently bled by retro-orbital puncture at various times ranging from 0 to 264 hours and their sera analyzed for the presence of the native and conjugated 2B8 antibody by whole-cell enzyme immunoassay using the antigen-positive B-cell line SB as the capture. The resulting data were plotted as the concentration of 2B8 or 2B8-MX-DTPA versus time; from these results a linear regression plot was generated and the slope used to determine the ⁇ t 1 ⁇ 2 values.
  • Yttrium-[89]-bearing 2B8-MX-DTPA was prepared using the protocol described for insertion of 90 Y, except that HPLC purification was not used.
  • the non-radioactive, metal-bearing conjugate was formulated in 1 ⁇ PBS containing 75 mg/mL HSA and 1 mM DTPA and evaluated in GLP study number 920611 at White Sands Research Center.
  • One male and one female monkey were included in each of four groups. The animals were injected intravenously every 48 hours for a total of 7 injections with the following amounts of drug: group 1 (saline); group II (0.003 mg/kg); group III (0.03 mg/kg); and, group IV (0.3 mg/kg).
  • mice were evaluated during the study by determining body weights and temperatures, food and water consumption, elimination, serum chemistries, hematology, urinalysis, and physical examinations. Animals in groups I through IV were bled prior to infusion on days 0, 2, 7, 10 and 14 and the blood analyzed for circulating B-cell levels by FACS analyses.
  • 2B8-MX-DTPA was radiolabeled with 111 In to a specific activity of 2.3 mCi/mg and approximately 1.1 ⁇ Ci was injected into each of 20 BALB/c mice. Subsequently, groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their organs removed and prepared for analysis. In addition, portions of the skin, muscle and bone were removed and processed for analysis; the urine and feces were also collected and analyzed for the 24-72 hour time points.
  • 2B8-MX-DTPA was also radiolabeled with 90 Y and its biological distribution evaluated in BALB/c mice over a 72-hour time period.
  • four groups of five mice each were injected intravenously with approximately 1 ⁇ Ci of clinically-formulated conjugate (specific activity: 12.2 mCi/mg); groups were subsequently sacrificed at 1, 24, 48 and 72 hours and their organs and tissues analyzed as described above. Radioactivity associated with each tissue specimen was determined by measuring bremstrahlung energy with a gamma scintillation counter. Activity values were subsequently expressed as percent injected dose per gram tissue or percent injected dose per organ. While organs and other tissues were rinsed repeatedly to remove superficial blood, the organs were not perfused. Thus, organ activity values were not discounted for the activity contribution represented by internally associated blood.
  • B1-FITC Coulter Immunology, (IgG2a,k) 11 Leu 16-FITC (Becton Dickinson, IgGl,k) 12 2B8-FITC (EDEC, IgGl,k) 14 B72.3-FITC (IgGl,k irrelevant control) 4 anti-CD4-FITC (Coulter Immunology 37 anti-CD3-FITC (Becton Dickinson) 59 anti-CD19-RPE (Becton Dickinson) 11 anti-CD19-FITC (Becton Dickinson) 14 B.
  • Double Staining B1-FITC/anti CD19-RPE 10 Leu 16-FITC/anti CD19-RPE 10 2B8 FITC/anti CD19-RPE 9 anti-CD19 FITC/anti CD19-RPE 13 B1-FITC/anti Hu Ig RPE 10 2B8-FITC/anti Hu Ig RPE 10 B72.3-FITC/anti Hu Ig RPE 2 Leucogate Simultest 99
  • the 2B8-MX-DTPA conjugate was produced by reacting the antibody with a 4:1 molar excess of isothiocyanatobenzyl-3-methyldiethylene-triaminepentaacetic acid (4). Typically, 1-2 mol of MX-DTPA chelate were introduced per mol of 2B8 antibody. As shown by the results presented in FIG. 4, the 2B8-MX-DTPA conjugate exhibited no apparent loss in immunoreactivity, vis a vis native 2B8, as both the native and conjugated 2B8 antibodies exhibited virtually identical Bi inhibition profiles; the IC50 values for 2B8 and 2B8-MX-DTPA were approximately 3 and 4 ⁇ g/mL, respectively.
  • the headings represent: 2B8 4 SAL, 2B8 incubated at 4° C. in saline; 2B8 30 SAL, 2B8 incubated at 30° C. in saline; 2B8 4 GLY, 2B8 incubated at 4° C. in normal saline containing 10 mM glycine; 2B8 30 GLY, 2B8 incubated at 30° C. in normal saline containing 10 mM glycine; 2B8-MX 4 SAL; 2B8-MX-DTPA (conjugate) incubated at 4° C. in saline; 2B8-MX 30 SAL, # conjugate incubated at 30° C.
  • 2B8-MX 4′ GLY conjugate incubated at 4° C. in normal saline containing 10 mM glycine
  • 2B8-MX 30 GLY conjugate incubated at 30° C. in normal saline containing 10 mM glycine.
  • Desensitometric Scans of Non-Reducing SDS Gels Reference 0 100.00 0 12 wk/4° C./saline 0 95.42 4.58 12 wk/4° C./glycine 0 100.00 0 12 wk/30° C./saline 7.63 83.34 9.03 12 wk/30° C./glycine 16.70 72.11 11.18
  • Desensitometric Scans of Reducing SDS Gels Reference 0 100.00 0 12 wk/30° C./saline 0 100.00 0 12 wk/30° C./glycine 0 10.00 0
  • the radiolabeling protocols for both 111 In and 90 Y were found to be reproducible. Typically, radioincorporations of >95% for 111 In and >90% for 90 Y were obtained. Specific activities for 111 I- and 90 Y-labeled conjugates were routinely in the range of 2-3 and 10-15 mCi/mg antibody, respectively.
  • uncomplexed radioisotopes were removed from the radiolabeled 2B8-MX-DTPA using HPLC gel permeation chromatography. In later experiments, HPLC purification of the indium-labeled conjugate was eliminated because of the high radioincorporations obtained (>95%) with this isotope.
  • the 90 Y-labeled antibody was formulated in 1 ⁇ PBS, pH 7.4 containing 75 mg/mL HSA; diethylenetriaminepentaacetic acid (DTPA) was also added to a final concentration of 1 mM to insure that any 90 Y which may be lost from the antibody would be chelated.
  • Degradation of 2B8-MX-DTPA, radiolabeled to a specific activity of 14.6 mCi/mg was evaluated at 0 and 48 hours using SDS-PAGE and autoradiography.
  • FIGS. 8 and 9 show that the radiolabeled antibody exhibited no significant degradation over a period of 48 h when incubated at 4° C.
  • FIG. 10 shows photographs of the autoradiograms for time zero and 48 h incubation samples; densitometric analysis of the autoradiograms indicate that there was no degradation of the radiolabeled antibody over the course of the study (FIGS. 11, 12).
  • Instant thin-layer chromatography analysis of the samples demonstrated no loss of 125 In (Table 25); moreover, immunoreactivity was maintained at approximately 100% (Table 25).
  • the study duration was 14 days and the animals were evaluated during the study in the following categories: clinical observations, body weights, body temperature, food and water intake, fecal elimination, serum chemistries, hematology, urinalysis, and physical examinations. Additionally, the animals in each group were bled on days 0, 1, 7, and 13 and the blood analyzed for serum antibody (2B8) levels and for T- and B-cell levels. On day 13 the animals in Groups III and IV were sacrificed and selected tissues examined by light microscopy following specimen preparation. The tissues evaluated were: heart, spleen, liver, kidney, lung, cerebral cortex, spinal cord, lymph node, stomach, ileum, colon, skeletal muscle, testis/ovary, pancreas, and bone marrow.
  • the 89 Y-labeled 2B8-MX-DTPA had little, if any, effect on circulating B-cells in these animals, regardless of the dose administered.
  • the 89 Y-labeled 2B8-MX-DTPA had little, if any, effect on circulating B-cells in these animals, regardless of the dose administered.
  • other than a general depletion of lymphocytes (20-43%) no significant abnormalities were found in any clinical parameter evaluated, including serum chemistry, urinalysis, body weights and temperatures.
  • conjugated 2B8 was radiolabeled with 111 In to a specific activity of 2.3 mCi/mg and roughly 1.1 ⁇ Ci was injected into each of twenty BALB/c mice to determine biodistribution of the radiolabeled material. Subsequently, groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their organs and a portion of the skin, muscle and bone were removed and processed for analysis. In addition, the urine and feces were collected and analyzed for the 24-72 hour time-points.
  • 2B8-MX-DTPA was prepared and radiolabeled with 111 In to a specific activity of 2.7 mCi/mg.
  • One hundred microliters of labeled conjugate (approximately 24 ⁇ Ci) were subsequently injected into each of 12 athymic mice bearing Ramos B-cell tumors. Tumors ranged in weight from 0.1 to 1.0 grams.
  • 50 ⁇ L of blood was removed by retro-orbital puncture, the mice sacrificed by cervical dislocation, and the tail, heart, lungs, liver, kidney, spleen, muscle, femur, and tumor removed.
  • the radioactivity associated with each tissue specimen was determined using a gamma counter and the values expressed as percent injected dose per gram.
  • mice and guinea pigs were administered a single intra peritoneal dose of 2B8 (0.5 mL or 5.0 mL, respectively) and observed for seven days. No overt signs of toxicity were detected.
  • tissue reactivity of murine monoclonal antibody 2B8 was evaluated using a panel of 32 different human tissues fixed with acetone.
  • Antibody 2B8 reacts with the anti-CD20 antigen which had a very restricted pattern of tissue distribution, being observed only in a subset of cells in lymphoid tissues including those of hematopoietic origin.
  • lymph node In the lymph node, immunoreactivity was observed in a population of mature cortical B-lymphocytes as well as proliferating cells in the germinal centers. Positive reactivity was also observed in the peripheral blood, B-cell areas of the tonsils, white pulp of the spleen, and with 40-70% of the medullary lymphocytes found in the thymus. Positive reactivity was also seen in the follicles of the lamina limbal (Peyer's Patches) of the large intestines.
  • aggregates or scattered lymphoid cells in the stroma of various organs including the bladder, breast, cervix, esophagus, lung, parotid, prostate, small intestine, and stomach, were also positive with antibody 2B8.
  • the tissue reactivity of the 2B8-MX-DTPA conjugate was evaluated using a panel of sixteen human tissues which had been fixed with acetone. As previously demonstrated with the native antibody, the 2B8-MX-DTPA conjugate recognized the CD20 antigen which exhibited a highly restricted pattern of distribution, being found only on a subset of cells of lymphoid origin. In the lymph node, immunoreactivity was observed in the B-cell population. Strong reactivity was seen in the white pulp of the spleen and in the medullary lymphocytes of the thymus.
  • Immunoreactivity was also observed in scattered lymphocytes in the bladder, heart, large intestines, liver, lung, and uterus, and was attributed to the presence of inflammatory cells present in these tissues. As described with the native antibody (above), no reactivity was observed with neuroectodermal cells or with mesenchymal elements.
  • the murine monoclonal anti-CD20 antibody 2B8 produced by a clone with the same designation, exhibits an affinity for the B-cell CD20 antigen which may be higher than that observed for the Bi antibody, as determined by competition with antibodies of known specificity for the CD20 antigen, and by Scatchard analysis. Further, immunoprecipitation data suggest that the antigen precipitated by 2B8 appears to be the same antigen as the one precipitated by B1, as both antibodies precipitated a doublet with relative molecular weights of 33 and 35 KD.
  • a 90 Y-labeled murine monoclonal anti-CD20 antibody (2B8) has been evaluated in a Phase I clinical trial for the treatment of relapsed B-cell lymphoma.
  • the original protocol used for the preparation of the yttrium-labeled antibody used a high performance liquid chromatographic (HPLC) step for removal of non-protein bound radioisotope prior to formulation and administration to patients.
  • HPLC high performance liquid chromatographic
  • this process is particularly time consuming, resulting in a longer exposure of the antibody to radioisotope in an unprotected state. This results in increased radiolysis of the antibody with a concomitant decrease in immunoreactivity.
  • the laborious aspect of the process makes it difficult to prepare more than one dose per day at the radiopharmacy. Simplification of the process would expedite implementation at the clinical site as an alternative to using NIPI Pharmacy Services as a radiopharmacy.
  • the human lymphoblastic cell lines SB (CD20 positive) and HSB (CD20 negative; ) were obtained from the American Type Culture Collection and maintained in RPMI-1640 containing 10% fetal bovine serum and supplemented with glutamine.
  • the 2B8 antibody was purified by the Manufacturing department from hollow-fiber bioreactor supernatant using protocols previously described in the IND (BB-IND 4850/4851).
  • Yttrium-[90] chloride was obtained from Amersham. All other reagents were obtained from sources described in the appended reports cited below. Reagents used for radiolabeling protocols were processed to remove contaminating heavy metal ions which could compete with-the radioisotopes during the radiolabeling step (see Methods section). Reagents were made under GMP conditions by IDEC's—Manufacturing department following established Batch Production Records.
  • Clinical-grade MX-DTPA was obtained from Coulter Immunology as the disodium salt in water and stored at ⁇ 70° C.
  • Conjugate (2B8-MX-DTPA) was prepared by the Manufacturing department. Two different lots of conjugate were used in these studies; both were provided in normal saline at 10 mg/mL. The conjugates were filled in sterile 2 mL polypropylene syringes and stored at 2-8° C.
  • Radioincorporation was determined using instant thin-layer chromatography (ITLC) in triplicate according to SOP SP-13-008.
  • ILC instant thin-layer chromatography
  • the protocol was as follows: radiolabeled conjugate was diluted 1:20 in 1 ⁇ PBS containing 1 mM DTPA or 5 mM EDTA, then 1 ⁇ -L spotted 1.5 cm from one end of a 1 ⁇ 5 cm strip of ITLC SG paper (Gehnan Sciences). The paper was developed using 10% ammonium acetate in methanol:water (1: 1;v/v). The strips were dried, cut in half crosswise, and the radioactivity associated with each section determined by scintillation counting. The radioactivity associated with the bottom half of the strip (protein-associated radioactivity) was expressed as a percentage of the total radioactivity determined by summing the values for both top and bottom halves.
  • Antibodies were radiolabeled with carrier-free 90 Y chloride provided by Amersham in 0.04 M HCl. An aliquot of radioisotope (10-20 mCi/mg antibody) was transferred to a polypropylene tube and 0.02 ⁇ volume of metalfree 2 M sodium acetate was added to adjust the solution to pH 3.6. 2B8-NaDTPA (0.3 mg; 10.0 mg/mL in normal saline) was added immediately and the solution gently mixed. The solution was checked with pH paper to verify a pH of 3.8-4.1 and incubated for 5 min.
  • the reaction was quenched by transferring the reaction mixture to a separate polypropylene tube containing 1 ⁇ PBS with 75 mg/mL human serum albumin (HSA) and 1 mM diethylenetriaminepentaacetic acid (DTPA) and gently mixed.
  • HSA human serum albumin
  • DTPA diethylenetriaminepentaacetic acid
  • Radioactivity remaining in the supernatant fraction was determined with a scintillation counter. Data were plotted as the quotient of the total radioactivity added divided by the cell-associated radioactivity versus the inverse of the cell number per tube. The y axis intercept represents the immunoreactive fraction.
  • the 2B8-MX-DTPA conjugate was radiolabeled with 90 Y and formulated as described in the “mix & shoot” protocol provided above. Two lots of radiolabeled conjugate were prepared; one lot was used for assessing radioincorporation stability and the other lot used to assess retention of immunoreactivity. The formulated conjugates were incubated at 4° C. for 48 hours and aliquots analyzed at time 0, 24 h and 48 hours using non-reducing SDS-PAGE and autoradiography. Immunoreactivity at each time point was assessed using the assay described above.
  • the stability of 90 Y-labeled 2B8-MX-DTPA was assessed by incubation in human serum at 37° C. for up to 72 hours.
  • the conjugated antibody was radiolabeled with yttrium-[90] and formulated as described above.
  • the radiolabeled conjugate was diluted 1:10 with normal human serum (non-heatinactivated) and aliquots incubated in plastic tubes at 37° C. At selected times, samples were removed and analyzed by non-reducing SDS-PAGE and autoradiography.
  • Yttrium-[90]-labeled 2B8-MX-DTPA was evaluated for tissue biodistribution in eight to ten week old BALB/c mice.
  • the radiolabeled conjugate was prepared and formulated as described above. Mice were injected intravenously with 5 ⁇ Ci of 90 Y-labeled 2B8-MX-DTPA and groups of five mice were sacrificed at 1, 24, 48, and 72 hours. After sacrifice, the tail, heart, lungs, liver, kidney, spleen, muscle, femur were removed, washed, weighed; a sample of blood and skin were also removed for analysis. Radioactivity associated with each tissue sample was determined by measuring bremstrahlung radiation using a gamma counter and the percent injected dose per gram tissue and percent injected dose per organ determined.
  • FIGS. 2, 3, and 4 show that the radiolabeled conjugate exhibited no significant degradation over a period of 48 h when incubated at 4° C.
  • Instant thin-layer chromatography analysis showed no loss of 90 Y during the 48 h incubation; these results were corroborated by SDS-PAGE/autoradiographic analysis (Table 28).
  • the immunoreactivity also was relatively constant at >88% (Table 29).
  • radioactivity decreased from 10-12% at 1 h to 8%-10% at 72 h.
  • radioactivity was relatively constant at approximately 3% from 24 h through 72 h.
  • the radioactivity in the gastrointestinal tract was constant at 0.5-1% from 24 h to 72 h.
  • Radioactivity for muscle remained approximately 0.6% throughout the course of the study.
  • the uptake of radioactivity by femur (bone) remained less than 4% at all time points indicating that the amount free yttrium in the conjugate preparation was negligible and that little free radiometal was released during the course of the study.
  • Ref A Schema for Absorbed-dose Calculation for Biologically Distributed Radionuclides, MIRD J. of Nucl. Med./Suppl. #1, 2/68 Calculations Performed Using a Spreadsheet Template in Symphony (Lotus Development Corporation) and Created by Phillip L. Hagan, MS, Nuclear Medicine Service, VA Hospital, San Diego, Calif. 92161
  • the original radiolabeling protocol for preparing 90 Y-labeled 2B8-MX-DTPA utilized a particularly laborious and time-consuming HPLC purification step for removing non-protein bound 90 Y from the preparation.
  • HPLC step in favor of what has been termed a “mix-and-shoot” protocol.
  • the goal was to identify radiolabeling conditions which would result in a very high radioincorporation of isotope into the conjugate, thereby obviating the need for the purification step. It was discovered that >95% radioincorporation could be-obtained at pH 3.6 with a five to ten minute incubation.
  • the dosimetry results are comparable to results obtained for patients enrolled in an on-going clinical trial (IDEC study #1315), when compared per millicurie of injected dose.
  • mean values (rads +SD) for whole body, heart, liver, and spleen were 1.40 ⁇ 0.57, 10.50 ⁇ 4.68, 9.89 ⁇ 8.91, and 9.75 ⁇ 6.00, respectively.
  • the radiolabeled conjugate was stable when incubated in human serum at 37° C. for 72 hours.
  • Biodistribution studies in BALB/c mice demonstrated no unusual tissue deposition, including bone.
  • Estimates of radiation absorbed doses to a “standard” 70 Kg human were comparable to those obtained in an on-going clinical trial using 90 Y-labeled 2B8-MX-DTPA.
  • the results of these studies showed that 90 Y-labeled 2B8-MX-DTPA produced using the “mix-and-shoot” protocol was comparable to that prepared using the conventional HPLC process.
  • Validation of the scale-up protocol for preparing clinical-grade radiolabeled conjugate showed that the method was reproducible and that the product was comparable to that produced using the current HPLC method.
  • the results of these pre-clinical studies indicate that this new “mix-&-shoot” protocol can be used to prepare 90 Y-labeled 2B8-MX-DTPA suitable for use in clinical trials.
  • Radiolabeling kit protocols for preparation of 111 In and 90 Y-labeled 2B8-MX-DTPA (In2B8 and Y2B8, respectively) and to establish release specifications for clinical products.
  • the radiolabeling kit protocols are reproducible with respect to radioincorporation and binding to antigen-positive SB cells and indicate the suitability of the radiolabeling kit for use in the clinical trials. It is recommended that In2B8 and Y2B8 release specifications for radioincorporation and binding be established at ⁇ 95% and ⁇ 70%, respectively.
  • a 90 Y-labeled murine monoclonal anti-CD20 antibody (Y2B8) is currently being evaluated in clinical trials for the treatment of relapsed B-cell lymphoma.
  • the yttrium isotope lacks a gamma component making it unsuitable for imaging systems. Therefore, 111 In-labeled 2B8-MX-DTPA (In2B8) will be used to assess tumor localization and dosimetry in patients prior to or after treatment with the yttrium-labeled therapeutic.
  • the protocols used currently for the preparation of Y2B8 and In2B8, referred to as the “mix-&-shoot” methods produce radiolabeled antibodies suitable for clinical studies. However, simplification of the labeling process would expedite dose preparation in a clinical setting.
  • the new radiolabeling kit is preferably comprised of four components: 1.) 2B8-MX-DTPA in low-metal normal saline at 2 mg/mL, 2.) 50 mM sodium acetate used to adjust radioisotope solution to appropriate labeling pH, 3.) formulation buffer (1 ⁇ PBS, pH 7.4 containing 7.5% human serum albumin and 1 mM DTPA), 4.) empty 10 mL glass vial (reaction vial). All components are tested to be sterile and pyrogen-free.
  • Radiolabeling kit components should be allowed to come to room temperature before use.
  • C 0 Radioactivity concentration at time of calibration (see manufacturer's Certificate of Analysis).
  • Radiolabeling kit components should be allowed to come to room temperature before use.
  • C 0 Radioactivity concentration at time of calibration (see manufacturer's Certificate of Analysis).
  • Step 3 The septa of the reaction vial and the sodium acetate vial were wiped with alcohol. Using a 1 cc syringe, the calculated volume (Step 1a or 1b) of 50 mM sodium acetate (Step 2) was transferred to the reaction vial. The vial was mixed by inverting several times.
  • reaction mixture was calculated by adding the amount of Y-90 chloride added (Step 4), plus the amount of 50 mM sodium acetate added (Step 3), plus the amount of 2B8-MX-DTPA added (Step 5).
  • radiolabeled antibodies were formulated with 1 ⁇ PBS containing 7.5% (w/v) human serum albumin (HSA; clinical-grade; Baxter-Hyland) and 1 mM DTPA. Results of the release tests performed on each validation lot are described below.
  • HSA human serum albumin
  • the human cell lines SB (CD20-positive) and HSB (CD20-negative) were obtained from American Type Culture Collection and cultured in T-flasks using RPMI-1640 containing 10% fetal bovine serum supplemented with 2% glutamine. Cultures were maintained at 37° C. and 5% CO 2 . Cells were typically split 1:2 every other day and harvested at 0.5-2.5 ⁇ 10 6 cells/mL and viability's >80%. Cell concentrations were determined using a hemacytometer and viability determined by trypan blue exclusion.
  • Percent binding was assessed by each operator using lyophilized CD20 positive SB cells according to the following protocols for In2B8 and Y2B8, respectively. These assays provide for a fast and efficient method of confirming that the radiolabeled antibody still recognizes CD20 as an antigen. At one clinical site, CD20-negative HSB cells were also evaluated. Lyophilized cells were prepared and stored according to the above method, “Preparation of Lyophilized SB and HSB Cells”.
  • Dilution buffer (1 ⁇ PBS, pH 7.2-7.4 containing 1% Bovine Serum Albumin (BSA), and 0.02% Sodium Azide 0.2 ⁇ m filtered and stored at room temperature.
  • BSA Bovine Serum Albumin
  • SWFI sterile water for injection
  • kits were developed. The concentrations of sodium acetate and 2B8-MX-DTPA were reduced to 50 mM and 2 mg/mL, respectively, to allow accurate volume transfers using syringes. All kit components were preferably filled in glass septum vials and tested for sterility and pyrogenicity by IDEC before release, thus eliminating the need for these tests to be performed at the clinical sites. At the site, all reagent manipulations are performed using sterile syringes and needles. Therefore, adherence to aseptic technique customarily found in a radiopharmacy environment insures that the radiolabeled and formulated anti-bodies are suitable for patient administration.
  • the murine anti-CD20 monoclonal antibody designated 2B8 has been cloned in CHO cells to yield a high expression cell line. Specificity of the CHO-derived antibody for CD20-positive human cells was demonstrated by FACS analysis and competitive binding. Negligible binding was observed to human T-cells. The affinity of the antibody for CD20-positive cells was determined to be 1.3 ⁇ 10 ⁇ 10 M using a competitive binding assay. The antibody was reacted with the chelating agent MX-DTPA to form a conjugate, 2B8-MX-DTPA, with negligible loss of immunoreactivity (affinity value was 4.4 ⁇ 10 ⁇ 10 M.
  • Optimal chelator conjugation as determined by measuring radioincorporation of 111 In, was achieved after eight hours reaction. Radiolabeling protocols for 2B8-MX-DTPA were optimized for 90 Y or 111 In with respect to pH and incubation time to insure maximal radioincorporation (>95%) and retention of immunoreactivity (>70%). Release specifications for In2B8 and Y2B8 prepared using CHO-derived 2B8-MX-DTPA in clinical trials were recommended for radioincorporation (>95%) and binding to lyophilized and reconstituted CD20-positive human cells (>70%). Taken together, these results indicate the suitability of CHO-derived 2B8-MX-DTPA for use in clinical trials.
  • the 2B8 antibody previously used was produced in hollow-fiber bioreactors. To reduce the manufacturing costs of this antibody, it has been cloned and expressed in CHO cells to yield a high-expression production cell line.
  • This example describes results of the in vitro characterization of the CHO-derived 2B8 antibody, the conjugated antibody (2B8-MX-DTPA), and the 90 Y and 111 In-labeled antibody products prepared using the clinical radiolabeling kit protocols.
  • the human cell lines SB (CD20-positive) and HSB (CD20-negative) were obtained from American Type Culture Collection and cultured in T-flasks using RPMI-1640 containing 10% fetal bovine serum supplemented with 2% glutamine. Cultures were maintained at 37° C. and 5% CO 2 . Cells were typically split 1:2 every other day and harvested at 0.5-2.5 ⁇ 10 6 cells/mL and viability's >80%. Cell concentrations were determined using a hemacytometer and viability determined by trypan blue exclusion. Specific information on cell lots is recorded in Notebook# 1553 and in the binder titled “Cell Activity Logbook 1995 & 1996” authored by Ron Morena.
  • CHO-derived 2B8 was produced under GMP conditions in IDEC's manufacturing facility. The antibody was formulated in low-metal normal saline at 11.5 mg/ml. Antibodies were determined to be homogeneous by SDS-PAGE. 2B8-MX-DTPA was produced under GMP conditions according to PSBR-043 from CHO-derived 2B8 and formulated in low-metal saline at 2 mg/mL (Lot #'s 0165A and 0165B).
  • Ruthenium-labeled tracer antibody was prepared by incubating CHO-derived 2B8 (lot #165) in 1 ⁇ PBS, pH 7.2 with the N-hydroxysuccinimide ester of ruthenium (II) tris-bipyridine chelator (TAG-NHS) at a 15:1 molar ratio of TAG-NHS to antibody. After 1 h incubation at ambient temperature, protected from light, the reaction was quenched with glycine for 10 min. Unreacted TAG was removed by size exclusion chromatography using a Pharmacia PD-10 column equilibrated with 1 ⁇ PBS. Protein concentration was determined using the Bradford protein assay. TAG incorporation was determined by measuring absorbance at 455 nm. The molar ratio of TAG to protein was calculated to be 3.0.
  • Assays were performed in 12 ⁇ 75 mm polypropylene tubes. Varying amounts of competing antibody (0.002-17 ug/mL) were incubated in 1 ⁇ PBS, pH 7.2, containing 1% (w/v) BSA with 0.08 ug/mL TAG-labeled CHO 2B8, 0.08 mg/mL anti-CD19 beads, and 167,000 cells/mL. After incubation at ambient temperature with orbital mixing for 3 h, relative electrochemiluminescence (ECL) was determined using the ORIGEN instrument. Mean ECL values were determined for duplicated samples and plotted vs. competing antibody concentration using Kaleidagraph software. For some experiments, per cent inhibition was plotted. Competition curves were fitted and EC 50 values (antibody concentration giving 50% maximal binding) determined using the following 4-parameter program:
  • m4 maximum signal response in relative ECL units Average affinity values were calculated from EC50 values and the known concentration of trace antibody using the method of Muller.
  • the chelating agent, 1-isothiocyanatobenzyl-3-methyldiethylenetriaminepentaacetic acid (MX-DTPA) was provided as a dry powder (free-acid) and stored desiccated at ⁇ 20° or ⁇ 70° C.
  • Approximately 3 mg of CHO 2B8 antibody in low-metal normal saline were adjusted to pH 8.6 by adding one-tenth volume of 50 mM sodium borate, pH 8.6.
  • Antibody at 10-11 mg/mL was incubated at a 4: 1 molar ratio of MX-DTPA to protein by adding MX-DTPA dissolved in 50 mM sodium borate, pH 8.6. After incubation at ambient temperature (2 to 24 h), unreacted chelator was removed from the conjugate by repetitive diafiltration in low-metal normal saline using Centricon 30 spin-filters.
  • In2B8 was prepared using the radiolabeling kit protocol as described herein. Antibody was labeled at a specific activity of 3 mCi/mg and formulated to 0.2 mg/mL. Briefly, 0.5 to 2 mCi of 111 In chloride was transferred to a metal-free microfuge tube and adjusted to approximately pH 4.2 using a 1.2 ⁇ volume of low-metal 50 mM sodium acetate.
  • 2B8-MX-DTPA at 2 mg/mL was added to the indium acetate solution and after incubation at ambient temperature for 30 min., the labeled antibody was formulated to 0.2 mg/mL in 1 ⁇ PBS, pH 7.2 containing 7.5% (w/v) human serum albumin and 1 mM DTPA (4% to 6% final concentration of HSA). All samples were tested for radioincorporation in triplicate; values were >95%.
  • Y2B8 was also prepared using a small-scale version of the radiolabeling kit protocol described in Example 1. Antibody was labeled at a specific activity of 15 mCi/mg and formulated to 0.3 mg/mL. Briefly, 0.5 to 2 mCi of 90Y chloride was transferred to a metal-free microfuge tube and adjusted to approximately pH 4.2 using a 1.2 ⁇ volume of low-metal 50 mM sodium acetate.
  • 2B8-MX-DTPA at 2 mg/mL was added to the 90 Y acetate solution and after incubation at ambient temperature for 5 min., the labeled antibody was formulated to 0.3 mg/mL in 1 ⁇ PBS, pH 7.2 containing 7.5% (w/v) human serum albumin and 1 mM DTPA (final concentration of HSA, 4% to 6%). All samples were tested for radioincorporation in triplicate; values were >95%.
  • radioactivity concentrations of the final radiolabeled products were calculated based on the amount of radioactivity added to the reaction mixture and by reference to the Certificate of Analysis for the radioisotope.
  • Antibody concentration of the quenched reaction mixtures were calculated from the known amount of antibody added.
  • the amount of radioactivity associated with the conjugates (radioincorporation) in the final products or incubation samples was determined using a commercially available kit manufactured by Biodex (Tec-Control Radiochromatographic Kit; see Example 1). In general, 1 ⁇ L of the test samples were applied in duplicate or triplicate using a micropipetter and developed according to the Biodex instructional insert. Strip halves were counted for radioactivity in glass tubes using an Isodata gamma counter or a Packard Top Count scintillation counter as described below. The radiolabel incorporation was calculated by dividing the amount of radioactivity in the top half of the strip by the total radioactivity found in both top and bottom halves. This value was expressed as a percentage and the mean value determined.
  • In2B8 and Y2B8 were prepared and formulated as described above.
  • Assay In2B8 or Y2B8 samples were diluted with assay dilution buffer (LXPBS, pH 7.2, containing 1% (w/v) bovine serum albumin (BSA) to 40 ng/mL and 11 ng/mL, respectively.
  • LXPBS assay dilution buffer
  • BSA bovine serum albumin
  • Antigen-positive (SB) and antigen-negative (HSB) cells were maintained in RPMI 1640 supplemented with 10% fetal calf serum at 37° C. and 5% CO 2 .
  • Cells (viability >90% as determined by trypan blue exclusion) were harvested at ambient temperature at a cell density of 0.5-2 ⁇ 10 6 cells/mL by centrifugation (1300 rpm in a Sorvall centrifuge) and washed twice with 50 mL 1 ⁇ HBSS.
  • Pelleted cells were resuspended to 50 ⁇ 10 6 cells/mL in prechilled 1 ⁇ HBSS containing 1% (w/v) bovine serum albumin (BSA) and 10% (w/v/) mannitol (lyophilization buffer).
  • BSA bovine serum albumin
  • mannitol lyophilization buffer
  • Cell suspensions were dispensed into 1.5 mL polypropylene microfuge tubes with o-ring gaskets at 50 ⁇ 10 6 cells/mL (0.5 mL per tube) and lyophilized overnight at 30 to 60 millitorr. Lyophilized cells were stored desiccated at 2-8° C. and reconstituted in sterile water for assays.
  • Lyophilized SB and HSB cells in 1.5 mL polypropylene tubes were reconstituted to 50 ⁇ 10 6 cells/mL using sterile water. Diluted In2B8 or Y2B8 was added to cells, in triplicate, and incubated for 45 to 60 min with end-over-end mixing at ambient temperature, respectively. After incubation, cells were pelleted by centrifugation and cell-bound radioactivity determined by counting samples in an Isodata Gamma Counter or a Packard Top Count scintillation counter as described below. Radioactivity bound (B) to cells was calculated by subtracting the unbound radioactivity (supernatant) from the total radioactivity added. Total radioactivity was determined from the radioactivity counted in tubes without cells. Percent binding was calculated by expressing the bound counts as a percentage of the total counts.
  • Radioincorporation samples were counted for 1 min. using an Isodata gamma counter.
  • the counter was set for energy windows of 100-500 KeV and the background adjusted to zero immediately before use for samples using 111 In.
  • the Isodata gamma counter was also used for counting ITLC strips having 90 Y spotted on them.
  • the energy windows for detection of the bremstrulung radiation were 100-1000 KeV.
  • Release specifications for radioincorporation and binding to CD20-positive cells were established by preparing six doses each of In2B8 and Y2B8 using two lots of clinical-grade 2B8-MX-DTPA (lot #'s 0219 and 0220) prepared according to the present invention and filled under GMP conditions. Release assays were performed as described above.
  • Binding of CHO 2B8 to CD20-positive cells was evaluated in competition assays using the ORIGEN chemiluminescent detection system. Lyophilized and reconstituted antigen-positive SB cells were incubated with increasing amounts bf antibody in the presence of ruthenium-labeled CHO 2B8 tracer. Results showed that CHO 2B8 inhibits binding to CD20-positive cells to the same extent as the antibody derived from hollow-fiber bioreactors (2B8-49) (FIG. 35). The EC50 values were determined graphically and the method of Muller (1980) used to calculate average affinity values.
  • the affinity for CHO 2B8 was determined to be 1.3 ⁇ 10 ⁇ 10 M; the 2B8 antibody derived from hollow-fiber bioreactors gave an affinity value of 2.5 10 ⁇ 10 M. Non-specific binding was negligible as demonstrated by the lack of competition with the irrelevant isotype antibody, S004.
  • the 2B8 conjugate (2B8-MX-DTPA) was prepared using a protocol similar to that used for the previously characterized 2B8-49. Reactions were performed using approximately 3 mg of antibody and a 4:1 molar ratio of chelator to antibody. Incubations times of 2, 4, 8, 17, and 24 h were evaluated to determine the reaction time giving acceptable retention of binding to CD20 positive cells and high radioincorporation with 111 In.
  • Competitive binding curves comparing CHO 2B8 to CHO 2B8-MX-DTPA conjugate reacted for 8-24 h were similar, indicating that the conjugation process did not significantly alter the binding of the antibody to the CD20 antigen (FIG. 36). Using EC 50 values determined graphically (FIG.
  • Indium-[111]-labeled CHO 2B8-MX-DTPA (In2B8) was prepared using the small-scale radiolabeling kit protocol previously described for the hollow-fiber bioreactor-derived antibody (Example 1). Briefly, conjugated antibody (CHO-derived 2B8-MX-DTPA; lot # 0165A) was incubated with 111 In acetate at the indicated pH for 30 minutes at ambient temperature. Reaction mixtures were formulated with PBS, pH 7.2, containing 7.5% (w/v) human serum albumin and 1 mM DTPA. Formulated samples of In2B8 were assayed for radioincorporation using instant thin-layer chromatography.
  • Binding of In2B8 to CD20-positive cells was determined using lyophilized and reconstituted SB cells.
  • the conjugate prepared from hybridoma-produced antibody(2B8-49) was incubated with 111 In acetate for 30 min. at pH 4.2 (conditions previously established for this antibody).
  • Yttrium-[90]-labeled antibody was prepared by incubating conjugated antibody (CHO-derived 2B8-MX-DTPA) with 90 Y acetate at the indicated pH for 5 minutes at ambient temperature. Reaction mixtures were formulate in PBS, pH 7.2 containing 7.5%(w/v) human serum albumin and 1 mM DTPA. Formulated samples of Y2B8 were assayed for radioincorporation using instant thin-layer chromatography. Binding of Y2B8 to CD20-positive cells was determined using lyophilized and reconstituted SB cells. For comparison, the conjugate prepared from hybridoma-produced antibody (2B8-49) was incubated with 90Y acetate for 5 min. at pH 4.2 (conditions previously established for this antibody).
  • conjugated antibody CHO-derived 2B8-MX-DTPA
  • Immunoreactivities for In2B8 and Y2B8 prepared from CHO 2B8 were determined using the method of Lindmo et al. Increasing amounts of freshly harvested CD20-positive SB cells were incubated with a fixed amount of In2B8 or Y2B8 under conditions of antigen excess. Reciprocal plot analysis of the binding data allowed immunoreactivities of 80.6% and 72.2% for In2B8 and Y2B8, respectively, to be determined (FIGS. 37 and 38).
  • Binding to CD20-positive cells ranged from 81.9% to 85.1% with a mean of 83.6%; binding to CD20-negative cells was ⁇ 4%.
  • radioincorporation ranged from 97.4% to 98.7% with a mean of 98.2%.
  • Binding to CD20-positive cells ranged from 81.4% to 82.7% with a mean of 81.9%; binding to CD20-negative cells was ⁇ 8%.
  • the anti-CD20 murine monoclonal antibody (2B8) cloned and expressed in CHO cells maintains specificity for CD20-positive human cells as shown by FACS and competitive binding analysis. Binding to human T-cells was minimal. The affinity of the antibody for human CD20-positive cells was determined to be 1.3 ⁇ 10 ⁇ 10 M using a competitive binding assay. Using the same assay, the 2B8 antibody derived from hollow-fiber bioreactors gave an affinity value of 2.5 ⁇ 10 ⁇ 10 M.
  • the CHO 2B8 antibody was reacted with MX-DTPA to form a conjugate, 2B8-MX-DTPA, while maintaining suitable retention of immunoreactivity.
  • Optimal chelator incorporation was determined by measuring radioincorporation with 111 In and was achieved after eight hours incubation at ambient temperature. Radiolabeling protocols for the 2B8-MX-DTPA conjugate were optimized for 90 Y or 111 In with respect to pH and incubation time to insure maximal radioincorporation and retention of immunoreactivity.
  • the present invention discloses a labeling procedure, referred to as the “mix-and-shoot” method, for the preparation of clinical doses of radiolabeled antibodies which obviates the need for the currently used high performance liquid chromatographic (HPLC) step for removal of non-protein bound radioisotope.
  • HPLC high performance liquid chromatographic
  • the simplified protocol eliminates this laborious purification step while maintaining a high level of radioisotope incorporation (>95%) and improved retention of immunoreactivity (>70%).
  • the clinically-formulated radiolabeled conjugate was found to be stable in vitro when incubated at 4° C. for 48 hours based on retention of radioisotope and immunoreactivity. Additionally, the radiolabeled conjugate was stable when incubated in human serum at 37° C.

Abstract

Antibody binding assays and radiolabeling kits are disclosed for radiolabeling and testing therapeutic antibodies in the commercial setting. In particular, the kits are designed for making and evaluating radiolabeled anti-CD20 conjugates to be used for the treatment and imaging of B cell lymphoma tumors. All kit reagents are sterile and are designed to achieve a high level of antibody radiolabeling and product stability with results which are highly reproducible.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention [0001]
  • The present invention relates to antibody binding assays and radiolabeling kits, lyophilized cell preparations, reagents and protocols for testing the clinical efficacy of therapeutic antibodies for the treatment/imaging of tumors and tumor cells. Specifically, the kits of the present invention are used for making and evaluating radiolabeled antibody conjugates that will be used for the treatment and imaging of B-cell lymphoma tumors by targeting the B cell surface antigen BP35 (“CD20”). [0002]
  • 2. Technology Background [0003]
  • All publications and patent applications herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. [0004]
  • The immune system of vertebrates (for example, primates, which include humans, apes, monkeys, etc.) consists of a number of organs and cell types which have evolved to: accurately and specifically recognize foreign microorganisms (“antigen”) which invade the vertebrate-host; specifically bind to such foreign microorganisms; and, eliminate/destroy such foreign microorganisms. Lymphocytes, as well as other types of cells, are critical to the immune system. Lymphocytes are produced in the thymus, spleen and bone marrow (adult) and represent about 30% of the total white blood cells present in the circulatory system of humans (adult). [0005]
  • There are two major sub-populations of lymphocytes: T cells and B cells. T cells are responsible for cell mediated immunity, while B cells are responsible for antibody production (humoral immunity). However, T cells and B cells can be considered as interdependent—in a typical immune response, T cells are activated when the T cell receptor binds to fragments of an antigen that are bound to major histocompatability complex (“MHC”) glycoproteins on the surface of an antigen presenting cell; such activation causes release of biological mediators (“interleukins”) which, in essence, stimulate B cells to differentiate and produce antibody (immunoglobulins”) against the antigen. [0006]
  • Each B cell within the host expresses a different antibody on its surface—thus one B cell will express antibody specific for one antigen, while another B cell will express antibody specific for a different antigen. Accordingly, B cells are quite diverse, and this diversity is critical to the immune system. In humans, each B cell can produce an enormous number of antibody molecules (i.e., about 10[0007] 7 to 108). Such antibody production most typically ceases (or substantially decreases) when the foreign antigen has been neutralized. Occasionally, however, proliferation of a particular B cell will continue unabated; such proliferation can result in a cancer referred to as “B cell lymphoma.”
  • T cells and B cells both comprise cell surface proteins which can be utilized as “markers” for differentiation and identification. One such human B cell marker is the human B lymphocyte-restricted differentiation antigen Bp35, referred to as “CD20.” CD20 is expressed during early pre-B cell development and remains until plasma cell differentiation. Specifically, the CD20 molecule may regulate a step in the activation process which is required for cell cycle initiation and differentiation and is usually expressed at very high levels on neoplastic (“tumor”) B cells. CD20, by definition, is present on both “normal” B cells as well as “malignant” B cells, i.e., those B cells whose unabated proliferation can lead to B cell lymphoma. Thus, the CD20 surface antigen has the potential of serving as a candidate for “targeting” of B cell lymphomas. [0008]
  • In essence, such targeting can be generalized as follows: antibodies specific to the CD20 surface antigen of B cells are, e.g., injected into a patient. These anti-CD20 antibodies specifically bind to the CD20 cell surface antigen of (ostensibly) both normal and malignant B cells; the anti-CD20 antibody bound to the CD20 surface antigen may lead to the destruction and depletion of neoplastic B cells. Additionally, chemical agents or radioactive labels having the potential to destroy the tumor can be conjugated to the anti-CD20 antibody such that the agent is specifically “delivered” to, e.g., the neoplastic B cells. Irrespective of the approach, a primary goal is to destroy the tumor: the specific approach can be determined by the particular anti-CD20 antibody which is utilized and, thus, the available approaches to targeting the CD20 antigen can vary considerably. [0009]
  • For example, attempts at such targeting of CD20 surface antigen have been reported. Murine (mouse) monoclonal antibody 1F5 (an anti-CD20 antibody) was reportedly administered by continuous intravenous infusion to B cell lymphoma patients. Extremely high levels (>2 grams) of 1F5 were reportedly required to deplete circulating tumor cells, and the results were described as being “transient.” Press et al., “Monoclonal Antibody 1F5 (Anti-CD20) Serotherapy of Human B-Cell Lymphomas,” Blood 69/2:584-591 (1987). [0010]
  • A potential problem with this approach is that non-human monoclonal antibodies (e.g., murine monoclonal antibodies) typically lack human effector functionality, i.e., they are unable to, inter alia, mediate complement dependent lysis or lyse human target cells through antibody dependent cellular toxicity or Fc-receptor mediated phagocytosis. Furthermore, non-human monoclonal antibodies can be recognized by the human host as a foreign protein; therefore, repeated injections of such foreign antibodies can lead to the induction of immune responses leading to harmful hypersensitivity reactions. For murine-based monoclonal antibodies, this is often referred to as a Human Anti-Mouse Antibody response, or “HAMA” response. Additionally, these “foreign” antibodies can be attacked by the immune system of the host such that they are, in effect, neutralized before they reach their target site. [0011]
  • Lymphocytes and lymphoma cells are inherently sensitive to radiotherapy. Therefore, B cell malignancies are attractive targets for radioimmunotherapy (RIT) for several reasons: the local emission of ionizing radiation of radiolabeled antibodies may kill cells with or without the target antigen (e.g., CD20) in close proximity to antibody bound to the antigen; penetrating radiation, i.e., beta emitters, may obviate the problem of limited access to the antibody in bulky or poly vascularized tumors; and, the total amount of antibody required may be reduced. The radionuclide emits radioactive particles which can damage cellular DNA to the point where the cellular repair mechanisms are unable to allow the cell to continue living; therefore, if the target cells are tumors, the radioactive label beneficially kills the tumor cells. Radiolabeled antibodies, by definition, include the use of a radioactive substance which may require the need for precautions for both the patient (i.e., possible bone marrow transplantation) as well as the health care provider (i.e., the need to exercise a high degree of caution when working with radioactivity). [0012]
  • Therefore, an approach at improving the ability of murine monoclonal antibodies to effect the treatment of B-cell disorders has been to conjugate a radioactive label to the antibody such that the label or toxin is localized at the tumor site. Toxins (i.e., chemotherapeutic agents such as doxorubicin or mitomycin C) have also been conjugated to antibodies. See, for example, PCT published application WO 92/07466 (published May 14, 1992). [0013]
  • “Chimeric” antibodies, i.e., antibodies which comprise portions from two or more different species (e.g., mouse and human) have been developed as an alternative to “conjugated” antibodies. Mouse/human chimeric antibodies have been created, and shown to exhibit the binding characteristics of the parental mouse antibody, and effector functions associated with the human constant region. See e.g., Cabilly et al., U.S. Pat. No. 4,816,567; Shoemaker et al., U.S. Pat. No. 4,978,745; Beavers et al., U.S. Pat. No. 4,975,369; and Boss et al., U.S. Pat. No. 4,816,397 all of which are incorporated by reference herein. Generally these chimeric antibodies are constructed by preparing a genomic gene library from DNA extracted from pre-existing murine hybridomas. Nishimura et al. (1987) Cancer Research 47: 999. The library is then screened for variable region genes from both heavy and light chains exhibiting the correct antibody fragment rearrangement patterns. The cloned variable region genes are then ligated into an expression vector containing cloned cassettes of the appropriate heavy or light chain human constant region gene. The chimeric genes are then expressed in a cell line of choice, usually a murine myeloma line. [0014]
  • For example, Liu, A. Y., et al., “Production of a Mouse-Human Chimeric Monoclonal Antibody to CD20 with Potent Fc-Dependent Biologic Activity”, J. Immun. 139/10:3521-3526 (1987), describes a mouse/human chimeric antibody directed against the CD20 antigen. See also, PCT Publication No. WO 88/04936. However, no information is provided as to the ability, efficacy or practicality of using Liu's chimeric antibodies for the treatment of B cell disorders in the reference. [0015]
  • It is noted that in vitro functional assays (e.g. complement dependent lysis (“CDC”); antibody dependent cellular cytotoxicity (“ADCC”), etc.) cannot inherently predict the in vivo capability of any antibody to destroy or deplete target cells expressing the specific antigen. See, for example, Robinson, R. D., et al., “Chimeric mouse-human anti-carcinoma antibodies that mediate different anti-tumor cell biological activities,” [0016] Hum. Antibod. Hybridomas, 2:84-93 (1991) (chimeric mouse-human antibody having undetectable ADCC activity). Therefore, the potential therapeutic efficacy of antibodies can only truly be assessed by in vivo experimentation.
  • To this end, copending application Ser. Nos. 08/475,813, 08/475,815 and 08/478,967, herein incorporated by reference in their entirety, disclose radiolabeled anti-CD20 conjugates for diagnostic “imaging” of B cell lymphoma tumors before administration of therapeutic antibody. “In2B8” conjugate comprises a murine monoclonal antibody, 2B8, specific to human CD20 antigen, that is attached to Indium[111] ([0017] 111In) via a bifunctional chelator, i.e., MX-DTPA (diethylenetriaminepentaacetic acid), which comprises a 1: 1 mixture of 1-isothiocyanatobenzyl-3-methyl-DTPA and 1-methyl-3-isothiocyanatobenzyl-DTPA. Indium-[111] is selected as a diagnostic radionuclide because it emits gamma radiation and finds prior usage as an imaging agent.
  • Patents relating to chelators and chelator conjugates are known in the art. For instance, U.S. Pat. No. 4,831,175 of Gansow is directed to polysubstituted diethylenetriaminepentaacetic acid chelates and protein conjugates containing the same, and methods for their preparation. U.S. Pat. Nos. 5,099,069, 5,246,692, 5,286,850, and 5,124,471 of Gansow also relate to polysubstituted DTPA chelates. These patents are incorporated herein in their entirety. [0018]
  • The specific bifunctional chelator used to facilitate chelation in application Ser. Nos. 08/475,813, 08/475,815 and 08/478,967 was selected as it possesses high affinity for trivalent metals, and provides for increased tumor-to-non-tumor ratios, decreased bone uptake, and greater in vivo retention of radionuclide at target sites, i.e., B-cell lymphoma tumor sites. However, other bifunctional chelators are known in the art and may also be beneficial in tumor therapy. [0019]
  • Also disclosed in application Ser. Nos. 08/475,813, 08/475,815 and 08/478,967 are radiolabeled therapeutic antibodies for the targeting and destruction of B cell lymphomas and tumor cells. In particular, the Y2B8 conjugate comprises the same anti-human CD20 murine monoclonal antibody, 2B8, attached to yttrium-[90] ([0020] 90Y) via the same bifunctional chelator. This radionuclide was selected for therapy for several reasons. The 64 hour half-life of 90Y is long enough to allow antibody accumulation by the tumor and, unlike e.g. 131I, it is a pure beta emitter of high energy with no accompanying gamma irradiation in its decay, with a range of 100 to 1000 cell diameters. The minimal amount of penetrating radiation allows for outpatient administration of 90Y-labeled antibodies. Furthermore, internalization of labeled antibodies is not required for cell killing, and the local emission of ionizing radiation should be lethal for adjacent tumor cells lacking the target antigen.
  • Because the [0021] 90Y radionuclide was attached to the 2B8 antibody using the same bifunctional chelator molecule MX-DTPA, the Y2B8 conjugate possesses the same advantages discussed above, e.g., increased retention of radionuclide at a target site (tumor). However, unlike 111In, it cannot be used for imaging purposes due to the lack of gamma radiation associated therewith. Thus, a diagnostic “imaging” radionuclide, such as 111In, can be used for determining the location and relative size of a tumor prior to and/or following administration of therapeutic chimeric or 90Y-labeled antibodies for the purpose of tumor reduction. Additionally, indium-labeled antibody enables dosimetric assessment to be made.
  • Depending on the intended use of the antibody, i.e., as a diagnostic or therapeutic reagent, other radiolabels are known in the art and have been used for similar purposes. For instance, radionuclides which have been used in clinical diagnosis include [0022] 131I, 125I, 123I, 99Tc, 67Ga, as well as 111In. Antibodies have also been labeled with a variety of radionuclides for potential use in targeted immunotherapy (Peirersz et al. (1987) The use of monoclonal antibody conjugates for the diagnosis and treatment of cancer. Immunol. Cell Biol. 65: 111-125). These radionuclides include 188Re and 186Re as well as 90Y, and to a lesser extent 199Au and 67Cu. I-[131] has also been used for therapeutic purposes. U.S. Pat. No. 5,460,785 provides a listing of such radioisotopes and is herein incorparted by reference.
  • As reported in copending application Ser. Nos. 08/475,813, 08/475,815 and 08/478,967 administration of the radiolabeled Y2B8 conjugate, as well as unlabeled chimeric anti-CD20 antibody, resulted in significant tumor reduction in mice harboring a B cell lymphoblastic tumor. Moreover, human clinical trials reported therein showed significant B cell depletion in lymphoma patients infused with chimeric anti-CD20 antibody. In fact, chimeric 2B8 has recently been heralded the nation's first FDA-approved anti-cancer monoclonal antibody under the name of Rituxan®. Thus, at least one chimeric anti-CD20 antibody has been shown to demonstrate therapeutic efficacy in the treatment of B cell lymphoma. [0023]
  • In addition, U.S. application Ser. No. 08/475,813, herein incorporated by reference, discloses sequential administration of Rituxan®, a chimeric anti-CD20, with both or either indium-labeled or yttrium-labeled murine monoclonal antibody. Although the radiolabeled antibodies used in these combined therapies are murine antibodies, initial treatment with chimeric anti-CD20 sufficiently depletes the B cell population such that the HAMA response is decreased, thereby facilitating a combined therapeutic and diagnostic regimen. [0024]
  • Thus, in this context of combined immunotherapy, murine antibodies may find particular utility as diagnostic reagents. Moreover, it was shown in U.S. application Ser. No. 08/475,813 that a therapeutically effective dosage of the yttrium-labeled anti-CD20 antibody following administration of Rituxan® is sufficient to (a) clear any remaining peripheral blood B cells not cleared by the chimeric anti-CD20 antibody; (b) begin B cell depletion from lymph nodes; or (c) begin B cell depletion from other tissues. [0025]
  • Thus, conjugation of radiolabels to cancer therapeutic antibodies provides a valuable clinical tool which may be used to assess the potential therapeutic efficacy of such antibodies, create diagnostic reagents to monitor the progress of treatment, and devise additional therapeutic reagents which may be used to enhance the initial tumor-killing potential of the chimeric antibody. Given the proven efficacy of an anti-CD20 antibody in the treatment of non-Hodgkin's lymphoma, and the known sensitivity of lymphocytes to radioactivity, it would be highly advantageous for such therapeutic antibodies to become commercially available in kit form whereby they may be readily modified with a radiolabel and administered directly to the patient in the clinical setting. [0026]
  • Although there exist many methods and reagents for accomplishing radiolabeling of antibodies, what is lacking in the art is a convenient vehicle for placing these reagents in the clinical setting, in a way that they may be easily produced and administered to the patient before significant decay of the radiolabel or significant destruction of the antibody due to the radiolabel occurs. The lack of such convenient means to commercialize this valuable technology could be due to the poor incorporation efficiencies demonstrated by some known labeling protocols, and the subsequent need to column purify the reagent following the radiolabeling procedure. The delay in development of such kits might also in part be due to the previously lack of accessibility to pure commercial radioisotopes which may be used to generate efficiently labeled products absent subsequent purification. Alternatively, perhaps the reason such kits are generally unavailable is the actual lack of antibodies which have been able to achieve either the approval or the efficacy that Rituxan® has achieved for the treatment of lymphoma in human patients. [0027]
  • For instance, as discussed in U.S. Pat. No. 4,636,380, herein incorporated by reference, it has been generally believed in the scientific community that for a radiopharmaceutical to find clinical utility, it must endure a long and tedious separation and purification process. Indeed, injecting unbound radiolabel into the patient would not be desirable. The need for additional purification steps renders the process of radiolabeling antibodies in the clinical setting an impossibility, particularly for doctors who have neither the equipment nor the time to purify their own therapeutics. [0028]
  • Furthermore, radiolabeled proteins may be inherently unstable, particularly those labeled with radiolytic isotopes such as [0029] 90Y, which have the tendency to cause damage to the antibody the longer they are attached to it in close proximity. In turn, such radiolysis causes unreliable efficiency of the therapeutic due to loss of radiolabel and/or reduced binding to the target antigen, and may lead to undesired immune responses directed at denatured protein. Yet without the facilities for labeling and purifying the antibodies on site, clinicians have had no choice but to order therapeutic antibodies already labeled, or have them labeled off site at a related facility and transported in following labeling for administration to the patient. All such manipulations add precious time to the period between labeling and administration, thereby contributing to the instability of the therapeutic, while in effect decreasing the utility of radiolabeling kits in the clinical setting.
  • Others have tried unsuccessfully to develop antibody radiolabeling kits that would be proficient enough to forego a separate purification step of the antibody. For instance, Cytogen has recently launched a commercial kit for radiolabeling a murine monoclonal antibody directed to tumor-associated glycoprotein TAG-72. However, Cytogen's antibody is particularly unamenable to a kit formulation due to the tendency to develop particulates during storage which must later be removed by a further filtration step. Moreover, Cytogen's antibody has caused adverse reactions in patients due to a HAMA responses. [0030]
  • Others have claimed to have developed radiolabeling protocols which would be amenable to kit format in that a separate purification step would not be required (Richardson et al. (1987) Optimization and batch production of DTPA-labeled antibody kits for routine use in [0031] 111In immunoscintography. Nuc. Med. Commun. 8: 347-356; Chinol and Hnatowich (1987) Generator-produced yttrium-[90] for radioimmunotherapy. J. Nucl. Med. 28(9): 1465-1470). However, such protocols were not able to achieve the level of incorporation that the present inventors have achieved using the protocols disclosed herein, which have resulted in incorporation efficiencies of at least 95%. Such a level of incorporation provides the added benefit of increased safety, in that virtually no unbound label will be injected into the patient as a result of low radioincorporation.
  • The protocols included in the kits of the present invention allow rapid labeling which may be affected in approximately a half an hour or as little as five minutes depending on the label. Moreover, the kit protocols of the present invention have a labeling efficiency of over 95% thereby foregoing the need for further purification. By foregoing the need for further purification, the half-life of the radiolabel and the integrity of the antibody is reserved for the therapeutic purpose for which it is labeled. [0032]
  • The present application discloses convenient kits and methods whereby diagnostic and therapeutic antibodies may be radiolabeled and administered to a patient in a reproducible, reliable and convenient manner. The kits of the present invention transform the process of radiolabeling antibodies into a hassle-free, worry-free standardized process, which greatly facilitates patient treatment protocols. The present kits provide advantages over the prior art in that the optimum parameters for labeling and administering therapeutic or diagnostic have been determined, thereby reducing the cost of goods. Since the kits described herein provide the optimum parameters according to the particular label, use of a kit designed for a particular label will also minimize cannibalization, i.e., which occurs when an inappropriate kit is used for a particular label. Avoiding cannibalization in turn also provides for optimum labeling efficiency. Moreover, the protocols and sterile, pyrogen-free ingredients included with each kit make for a more user-friendly process, since sterility, pyrogen testing and post-labeling purification of the reagents are obviated. [0033]
  • SUMMARY OF THE INVENTION
  • The present invention includes a kit for radiolabeling a diagnostic or therapeutic antibody before administration to a patient comprising at least (i) a vial containing a chelator-conjugated antibody, (ii) a vial containing formulation buffer for stabilizing and administering the radiolabeled antibody, and (iii) instructions for radiolabeling the antibody, wherein said vial components are supplied in such an amount and at such a concentration that when they are combined with a radiolabel of sufficient purity and activity according to the kit instructions, no further purification of the labeled antibody is required before administration to said patient. Moreover, when labeled according to the kit instructions and with a radioisotope of sufficient purity and activity, such isotope incorporation may reach levels higher than 95%, and even as high as 98% or higher. [0034]
  • The antibody included in the kit is most preferably an anti-CD20 antibody. The antibody is supplied in a form whereby it is attached to a bifunctional chelator. Preferably, the antibody is conjugated to MX-DTPA, but other chelators such as phenyl- or benzyl-conjugated DTPA, cyclohexyl-DTPA, EDTA derivatives and DOTA may be used. A chelator according to the present invention may be any chelator that is at least bifunctional, i.e., which possesses at least two binding sites (at least one site for chelating a metallic ion and at least one site for coupling to a protein ligand). [0035]
  • Depending on the antibody used, the conjugated antibody is typically supplied at a concentration of 0.5 to 30 mg/ml, more preferably 2 mg/ml. The volume of conjugated antibody will depend on the concentration and the amount required for optimum labeling depending on the radiolabel. However, the conjugated antibody is to be supplied in such a volume and concentration that the entire volume will be added to the reaction vial using a sterile syringe and aseptic technique. This will allow for increased reproducibility and ease of use. All reagents of the kits disclosed herein are sterile and pyrogen-free, and specifically designed for simplicity and speed in advancing directly from antibody testing to administration. With some labels, the need for testing labeling efficiency may not be required. [0036]
  • A particularly advantageous component of the kit is the formulation buffer for stabilizing against the effects of radiolysis and administering the radiolabeled conjugated antibody to a patient. The formulation buffer is a pharmaceutically acceptable carrier which serves as both a diluent for the labeled antibody and an administration buffer. Although any pharmaceutically acceptable diluent may be used for administering therapeutic or diagnostic antibodies to patient, the formulation buffer of the present invention is particularly suited for administering radiolabeled antibodies. [0037]
  • For instance, the formulation buffer of the present invention comprises a radioprotectant such as human serum albumin (HSA) or ascorbate, which minimize radiolysis due to yttrium, and to a lesser degree, indium. Other radioprotectants are known in the art and could also be used in the formulation buffer of the present invention, i.e., free radical scavengers (phenol, sulfites, glutathione, cysteine, gentisic acid, nicotinic acid, ascorbyl palmitate, HOP(:O)H[0038] 2, glycerol, sodium formaldehyde sulfoxylate, Na2S2O5, Na2S2O3, and SO2, etc.).
  • It should be noted that, while radioprotectants are generally employed in the formulation buffer to protect the antibody from radiolysis, it may be possible to affect further protection by including the radioprotectant in the reaction buffer as well. This has generally not been done before, i.e., with HSA, due to the presence of metals which would interfere with the labeling process. However, it may be possible to “clean” the HSA using a chelating resin such that it could be included in the reaction buffer as well. Ascorbate or other radioprotectants may also need to be treated to remove contaminating metals. [0039]
  • The formulation buffer of the present invention also comprises excess unconjugated chelator. The purpose for including unconjugated chelator is that this chelator serves to scavenge any non-protein bound radiolabel in the patient, and effects excretion of the radiolabel thereby reducing uptake of “bone-seeking” isotopes, i.e., [0040] 90Y, by the bones of the patient. For instance, when the antibody of the kit is conjugated to a DTPA chelator, excess DTPA or any other chelator may be included in the formulation buffer. The formation buffer is also preferably supplied in a volume such that the entire contents are transferred to the reaction vial. As discussed above, this results in increased ease of use and reproducibility because exact volumes do not have to be measured and transferred.
  • A preferred formulation buffer comprises phosphate buffered or physiological saline, human serum albumin and DTPA. The human serum albumin is preferably at a concentration of between about 1 to 25% (w/v), and more preferably at a concentration of about 7.5% (w/v). The concentration of DTPA is preferably about 1 mM. Ascorbate may be used as an alternative to human serum albumin, and is typically used at a concentration of about 1 to 100 mg/ml. Although a wider range of concentrations may be used without compromising patient safety. [0041]
  • The antibody of the radiolabeling kit is readily labeled with a radioisotope of choice via a bifunctional chelator according to the methods of the present invention. For further simplicity in this regard, the kit of the present invention may also include a vial containing a buffer for adjusting the pH of the radioisotope solution, and a sterile glass reaction vial for performing the labeling and subsequently for resuspending the final radiolabeled antibody in formulation buffer. A 10 ml reaction vial is typically sufficient, but vials capable of holding 5 to 20 mls may also be used. The buffer is preferably a low-metal sodium acetate solution at a concentration of 10 to 1000 mM, most preferably 50 mM. [0042]
  • A specific kit of the present invention comprises the MX-DTPA conjugated antibody, 2B8-MX-DTPA. 2B8 is an anti-CD20 antibody shown to affect B cell depletion upon administration to lymphoma patients. However, it should be apparent to those skilled in the art that the radiolabeling kit of the present invention may be optimized for the radiolabeling of other anti-CD20 antibodies, or any other antibody which has been conjugated to DTPA or other polyvalent chelator. The preferred kit of the present invention may comprise at least (i) a vial containing the MX-DTPA conjugated 2B8 antibody, either in solution or lyophilized (requiring reconstitution); and (ii) a vial containing formulation buffer for administering the radiolabeled antibody to a patient. The preferred kit will also contain (iii) a buffer for adjusting isotope pH, and (iv) a reaction vial. Alternatively, and more preferably, the buffer is supplied in the reaction vial, thereby eliminating the steps of measuring and transferring the buffer and increasing the simplicity, consistency and sterility of the kit components, However, other embodiments are also envisioned, i.e., whereby the buffer is added to the isotope vial first, and the buffered isotope is then transferred to the reaction vial. In this case, the reaction vial could be supplied with the required antibody volume. Alternatively, the isotope/buffer vial could be made large enough to accommodate addition of the antibody conjugate, i.e., directly to the supplier's vial. This would eliminate the need for the reaction vial. [0043]
  • As described above, another preferred kit configuration is encompassed whereby the reaction vial itself contains the required volume of conjugated antibody (i.e., 1 or 1.5 mL for [0044] 111In and 90Y, respectively). The antibody may be supplied in a buffer that provides the appropriate radiolabeling pH according to the specific desired isotope (i.e., pH 3-6 for 111In, pH 3-5 for 90Y). Different buffers may be used, depending on the isotope (i.e., sodium acetate for 90Y, sodium citrate for 111In). The pH and composition of the buffer may also vary depending on the nature of the binding ligand to be labeled (i.e., labeling peptides may permit <pH 3 to be used). Essentially then, the isotope would be transferred directly to the reaction vial, as would the formulation buffer. Limiting use of the kit to two transfer steps would further increase reproducibility and simplicity, and further decrease the chance for contamination of sterility during manipulation of the kit components.
  • The radiolabeling kits of the present invention may further comprise a vial of radioisotope, or radioisotope may be ordered separately from an appropriate supplier. Preferred radioisotopes of the present invention are [0045] 111In chloride and 90Y chloride in HCl although the disclosed methods are not limited to these isotopes. Other radionuclides that have been used for imaging applications are known in the art, i.e., as described in U.S. Pat. Nos. 4,634,586, 5,460,785 and 5,766,571, which are herein incorporated by reference. Indium-[111] is particularly advantageous for imaging B cell tumors and beta emitters such as 90Y are particularly useful as radiotherapeutic agents. Although other radioisotopes suitable for these or other purposes, i.e., alpha emitters, may be used depending on the chelator used for antibody conjugation.
  • Given the proven efficacy of the combined therapeutic regimens disclosed in U.S. application Ser. No. 08/475,813, a further kit embodiment will also include a separate vial of chimeric antibody, i.e., Rituxan®, to be administered before or after the radiolabeled anti-CD20 antibody. When the chimeric antibody is administered before the radiolabeled antibody, the HAMA response which might generally occur in response to administration of a murine anti-CD20 antibody may be significantly decreased, thereby increasing the therapeutic utility of radiolabeled murine antibodies. Moreover, when chimeric anti-CD20 is employed to clear circulating B cells, subsequent diagnostic images achieved with [0046] 111In-labeled antibodies may be much clearer.
  • It should also be apparent that both a diagnostic radiolabeled antibody and a therapeutic radiolabeled antibody may be used together in a combined therapeutic regimen. In this regard, the diagnostic antibody may be used either before or after the therapeutic antibody to visualize tumor size before and after treatment. In this case, the kit of the present invention may include separate, perhaps color-coded, buffer vials specifically formulated according to the optimum pH requirements for radiolabeling antibodies with the specific radioisotopes to be used. Such a system would ensure that the appropriate buffer was used for each label, and would allow the clinician the same ease in radiolabeling the two antibodies as if two kits had been purchased. Such a kit in effect combines the components from two radiolabeling kits into one. [0047]
  • The components of the radiolabeling kit of the present invention are supplied at the appropriate concentration and pH so that sterility is readily maintained before antibody administration and there is little need for additional buffers or media. However, it should be apparent to those of skill in the art that some of the reagents can be prepared, sterilized and tested for sterility on site. Thus, variations of the kit of the invention are envisioned depending on the budget and preference of the consumer. [0048]
  • The radiolabeling kit of the present invention may be used in a method for radiolabeling a chelator-conjugated antibody for administration to a patient. According to the present invention, such a method comprises, in general, (i) mixing a chelator-conjugated antibody with a solution containing a radioisotope; (ii) incubating the mixture for an appropriate amount of time at appropriate temperature; and (iii) diluting the labeled antibody to an appropriate concentration in formulation buffer, such that the radiolabeled antibody may be administered directly to a patient without further purification. [0049]
  • Most preferably the antibody is an anti-CD20 antibody, and in particular, the anti-CD20 antibody may be 2B8. The antibody may be conjugated to any appropriate chelator, i.e., MX-DTPA, CHX-DTPA, phenyl- or benzyl-DTPA, DOTA, EDTA derivatives, etc. MX-DTPA is preferred. Methods for affecting antibody conjugation are known in the art (Kozak et al. (1989); Mirzadeh et al. (1990), Brachbiel et al. (1986)). [0050]
  • The present inventors have found that the method of radiolabeling a chelator-conjugated antibody works best wherein the solution containing the radiolabel is adjusted to a pH of between about 3.0 and 6.0, and more preferably to about 4.2 before it is mixed with the chelator-conjugated antibody. Low-metal sodium acetate is particularly preferred for adjusting the pH, although other buffers may be used. Preferably, the sodium acetate is at a concentration of between about 10 and 1000 mM, and more preferably 50 mM. [0051]
  • When the radioisotope is [0052] 111In chloride, the volume quantity of 111In chloride which should be used to prepare a single administrative dose is typically about 5.5 mCi divided by the radioactivity concentration at the time of labeling. For patient administration, a typical diagnostic dose of 111In is about 2 to 10 mCi. The quantity of sodium acetate used for adjusting the pH varies depending on the sodium acetate concentration and the isotope carrier solution, and may therefor be quite broad. When the concentration of sodium acetate is 50 mM, the amount required for adjusting the pH is typically about 1.2 times the volume quantity of 111In chloride used although larger volumes may be used. It should be appreciated that the ratio of sodium acetate to HCl is what is important, and the amount of sodium acetate used would change depending on the amount and concentration of HCl in the buffer. About 1 ml of a chelator-conjugated antibody at a concentration of about 2 mg/ml is then mixed with the radiolabel acetate solution, and the mixture is incubated for about 30 minutes, or for a time sufficient to achieve optimal labeling of the antibody. Such time may range from about 30 seconds to about 60 minutes. Formulation buffer is then added in an amount necessary to achieve a total final volume of about 10 ml.
  • The optimum time required for labeling the antibody may vary depending on the antibody, the particular radiolabel and the particular conjugate employed. An underlying factor in the optimization of the time allotted for radiolabeling is the chelator to antibody ratio of the reagent which is to be labeled. For instance, the chelator to antibody ratio must be high enough to achieve a therapeutically useful level of incorporation, i.e., 90 to 95% depending on the radioisotope, but must also not be too high such that the structural integrity or immunoreactivity of the antibody is compromised. This requires a certain balancing process that in some cases may lead to a lower level of conjugated chelator and longer labeling time. [0053]
  • For instance, for 2B8 and MX-DTPA, it has been discovered that labeling may be accomplished in under five minutes for [0054] 90Y and in about thirty minutes for 111In to achieve the desired level of radioincorporation, with only about a 1½ to 1 molar ratio of chelator to antibody. It was not necessary, therefor, to increase the chelator to antibody ratio, because a desirable level of radioincorporation was achieved. Moreover, it was not advantageous to increase the quantity of conjugated chelator because this could effect antibody immunoreactivity. Such parameters could be empirically determined for other antibodies for the design of kits such as those described in the present invention.
  • When the radioisotope is [0055] 90Y chloride, the volume quantity of 90Y chloride used for preparing a single administrative dose typically ranges from about 10 to 50 mCi, and is preferably about 45 mCi, divided by the radioactivity concentration at the time of labeling. The quantity of sodium acetate used for adjusting the pH varies depending on the sodium acetate concentration and the concentration of isotope carrier, and may therefor be quite broad. When the concentration of sodium acetate is 50 mM and the 90Y is supplied in 50 mM HCl, the amount required for adjusting the pH is typically about 1.2 times the volume quantity of 90Y chloride used. About 1.5 ml of a chelator-conjugated antibody at a concentration of about 2 mg/ml is then mixed with the radiolabel acetate solution, and incubated for about 5 minutes, or for a time sufficient to achieve optimal labeling of the antibody. Such time may range from about 30 seconds to about 60 minutes. Formulation buffer is added in an amount necessary to achieve a total final volume of about 10 ml.
  • Preferably, the radiolabeling method of the invention is performed using the radiolabeling kit described herein. However, it should be apparent to those of skill in the art that the preferred components and conditions are merely acceptable guidelines for practicing the method of the invention, and may be altered to some degree with appropriate optimization. Conditions which depart from those preferred but still accomplish the purpose of the method are considered to be within the scope of the invention. [0056]
  • The radiolabeling kit of the present invention may also be supplied with reagents suitable for conveniently verifying the binding affinity of the antibody following radiolabeling. In such a case, the kit of the invention may also be used for determining the percent binding of a radiolabeled antibody to its target cell before administering the antibody to a patient. The present inventors have also found that the particular binding assay kit disclosed may be useful for testing the affinity of any antibody generally for which purified antigen is not available. Accordingly, the binding assay components may also be sold as a separate kit. [0057]
  • In general, a binding assay and radiolabeling kit comprises (i) at least one vial of lyophilized cells which express the antigen which is recognized by the antibody in the kit; (ii) a vial containing chelator-conjugated antibody; (iii) a vial containing formulation buffer, and (iv) instructions for radiolabeling the antibody such that the radiolabeled antibody may be administered directly to a patient without the need for subsequent purification. As described above for the radiolabeling kit, this kit may also comprise a vial containing a buffer for adjusting the pH of the radioisotope, and a sterile glass reaction vial. Preferably the buffer is a low-metal sodium acetate solution at a concentration of between about 10 and 1000 mM, and the glass reaction vial holds a volume of at least 5 ml. The antibody is preferably an anti-CD20 antibody, and the chelator is preferably MX-DTPA. Other chelators may be used as described previously. The preferred conjugated antibody is 2B8-MX-DTPA, although any chelator-conjugated antibody may be labeled and its affinity assessed. The formulation buffer is phosphate buffered saline comprising a radioprotectant and unconjugated chelator as described above, and radioisotope may or may not be included and is preferably [0058] 111In chloride or 90Y chloride. Other radioisotopes may be used depending on the chelator.
  • The difference between the binding assay/radiolabeling kit and the radiolabeling kit described above is the inclusion of antigen-positive cells to serve as a substrate target for testing antibody affinity. When the antigen is CD20, preferred CD20-positive cells are SB cells (ATCC # CCL 120) but any CD20-positive cells may be used. The binding assay and radiolabeling kit may further include antigen-negative cells for use as a negative control. Preferred CD20-negative cells are HSB cells (ATCC # CCL 120.1) but any CD20-negative cells may be used. [0059]
  • Of course, the combined radiolabeling and binding assay kit may further comprise a vial of chimeric anti-CD20 antibody in addition to the antibody to be labeled for the purposes of affecting a combined therapeutic regimen, or for clearing peripheral B cells prior to diagnostic imagery. Such separate antibody is preferably Rituxan®, but may be any antibody shown to effectuate tumor cell killing. In fact, two different types of antibodies may be combined in one kit, i.e., antibodies directed to two different B cell antigens, so long as the combined therapeutic regimen serves to target the same type of cell, i.e., the B cell lymphoma. [0060]
  • Just as the components of the kit may be used to label other antibodies, other cells for testing antibody affinity may be prepared depending on the target antigen. However, for anti-CD20 antibodies, the binding assay and radiolabeling kit of the present invention is particularly suited for the commercial setting in that the target cells are provided in lyophilized form. This allows the verification of antibody efficacy to proceed simply and systematically, and negates the hassle and expense involved in maintaining tissue culture facilities. The lyophilized cells are generally supplied in aliquots of between 0.5 and 500×10[0061] 6 cells per vial according to the methods of the invention.
  • It is possible that particular facilities will prefer to order antibody which has already been radiolabeled, in which case such a facility might desire the binding assay reagents in order to ensure that the antibodies retain target affinity. In this case, the present invention also provides for a binding assay kit for determining the percent binding of a radiolabeled antibody to its target cell. Such a kit includes at least one vial of fixed and/or lyophilized antigen-positive cells, and may optionally contain antigen-negative cells as described above for the binding assay and radiolabeling kit. Moreover, it should be apparent that variations of such a kit may include an unlabeled control antibody for verifying the binding specificity of the consumer's antibody via a competitive assay. [0062]
  • Again, when the antigen is CD20, the CD20-positive cells are preferably SB cells (ATCC # CCL 120) and the CD20-negative cells are preferably HSB cells (ATCC # CCL120. 1), which are supplied in lyophilized form in aliquots of between 0.5 and 50×10[0063] 6 cells. In this case, the antibody is preferably an MX-DTPA conjugate of 2B8 labeled with 111In or 90Y.
  • In view of the additional kit embodiments disclosed herein, it should be stressed that one of the advantages of the radiolabeling kit and method of the present invention is that no further purification step is necessary, and the radiolabeled antibody may be administered directly to the patient, thereby saving valuable time and increasing antibody stability. Therefor, it is emphasized that, while it might be desirable for the clinician to test or verify the binding specificity and affinity of the radiolabeled antibody prior to administration, such test may be foregone with particular radioisotopes if antibody stability and the inhibition of radiolysis are particular concerns, i.e., as with yttrium. By providing kit embodiments whereby the binding affinity and specificity may be tested, the present inventors are in no way suggesting that such tests are absolutely required in the methods or kits of the invention. The option to test such antibody validity is purely at the option of the clinician. [0064]
  • The present inventors have also found that the method used for preparing fixed and lyophilized cells for the binding assay kits of the present invention is particularly suitable for preparing cells for commercial kits. Cells may be fixed prior to lyophilization to improve structure/stability. In particular, the cells of the present invention demonstrate high reproducibility when used for antibody binding assays. [0065]
  • In particular, the present invention includes a method of preparing lyophilized cells comprising (i) harvesting cells at a cell density of 0.5 to 2×10[0066] 6 cells per ml by centrifugation; (ii) washing cells at least one time in a balanced salt solution, i.e., HBSS; (iii) resuspending pelleted cells in a lyophilization buffer comprising a balanced salt solution containing carrier protein and at least one type of sugar; (iv) dispensing an aliquot of resuspended cells into a microfuge tube or a glass vial; and (v) lyophilizing the cells 12-96 h and more preferably 24-72 h at about 30-60 millitor. The method is particularly suitable for preparing lyophilized cells wherein said cells are SB cells (ATCC # CCL 120) or HSB cells (ATCC # CCL 120.1), but is likely applicable to other cell types as well.
  • Preferably, the buffer generally contains bovine serum albumin as the carrier protein at a concentration of 1% (w/v) and mannitol at a concentration of 10%. However, conceivably other carrier proteins, i.e., HSA, and other sugars may be used. The cells are harvested by centrifugation at a speed of about 1300 rpm, and the salt solution HBSS (Hank's balanced salt solution) is added. The cells are generally resuspended at a concentration of 50×10[0067] 6 cells per ml. However, it should be apparent to those of skill in the art that the above conditions may be modified slightly without significantly compromising cell viability. Moreover, the above conditions may be supplemented by additional procedures designed to optimize the process for larger quantities of cells, e.g., tangential flow diafiltration to exchange cells into the lyophilization buffer.
  • The binding assay kits of the present invention may be used in an assay for assessing the binding affinity of a radiolabeled antibody. Such an assay is also a subject of the present invention. A binding assay for determining the percent binding of a radiolabeled antibody to its target cell comprises in general the following steps: (i) mixing at least one aliquot of a radiolabeled antibody with at least one aliquot of antigen positive cells; (ii) mixing at least one aliquot of a radiolabeled antibody identical to the aliquot of step (i) with at least one aliquot of dilution buffer of the same volume as the aliquot of antigen-positive cells in step (i) as a control; (iii) pelleting the cells by centrifugation; (iv) measuring the radioactivity in the supernatant of the pelleted cells and the control; and (v) comparing the quantity of radioactivity in the cell supernatant to the quantity of radioactivity in the control. [0068]
  • Just as the radiolabeling kits of the present invention optionally contain [0069] 111In chloride or 90Y chloride, the binding assay of the present invention is typically performed with antibodies labeled with 111In or 90Y. When 111In is the radiolabel, radioactivity in the assay tubes is measure using a gamma counter. When 90Y is the label, radioactivity is measured using a scintillation counter, although a gamma counter could be used.
  • For the binding assay of the present invention, the preferred antibody is an anti-CD20 antibody, and the anti-CD20 antibody is preferably 2B8, wherein the 2B8 antibody is labeled using the radiolabeling kit of the present invention. However, any radiolabeled antibody may be tested provided cells expressing the particular antigen are available. When CD20 is the antigen the preferred cells for performing the assay are SB cells (ATCC # CCL 120), however, the assay may also be optimized and performed with any radiolabeled antibody and appropriate target cell. [0070]
  • The dilution buffer used for the assay should maintain binding of the antibody, i.e., physiological buffer, possibly containing a carrier protein, e.g. BSA, to minimize non-specific binding to cells. Although the tube with dilution buffer serves as a control, a further control may be included in the assay by using antigen-negative cells. In this case, the binding assay further comprises the following steps: (i) mixing at least one aliquot of a radiolabeled antibody with at least one aliquot of antigen-negative cells; (ii) pelleting the antigen-negative cells by centrifugation; (iv) measuring the radioactivity in the supernatant of the antigen-negative pelleted cells; and (v) comparing the quantity of radioactivity in the antigen-negative cell supernatant to the quantity of radioactivity in the supernatant of the antigen-positive cell supernatant and the control. Comparing the radioactivity obtained from this tube to the dilution buffer control will serve as a measure of the amount of non-specific binding to antigen-positive cells. When CD20 is the antigen, and the CD20-positive cells are SB cells, CD20 negative cells are preferably HSB cells (ATCC # CCL 120.1). [0071]
  • As described above, the lyophilized cells of the present invention provide a simple, efficient and reproducible standard for testing the binding efficacy of a radiolabeled antibody. Therefore, the binding assay of the present invention is preferably performed using the lyophilized cells included in the binding assay kits of the present invention. In addition, the radiolabeling assays of the present invention may be combined with the binding assays of the present invention, wherein the antibody is first labeled by the method of labeling an chelator-conjugated antibody as described in the present invention. Most preferably, the binding assay of the present invention is performed using one of the binding assay and radiolabeling kits described herein. [0072]
  • There may be some instances where the affinity of an antibody should be tested or verified but a radiolabel has not been attached. For instance, under certain circumstances, i.e., trouble-shooting, it may be advantageous to test the binding affinity of an antibody before radiolabeling. For such a case, the present invention also encompasses a competitive binding assay for assessing affinity of a test antibody to a target cell, comprising (i) preparing a ruthenium-labeled control antibody using a known antibody specific for the same antigen; (ii) incubating increasing amounts of test antibody and increasing amounts of unlabeled control antibody with a fixed concentration of target cells and a trace amount of ruthenium-labeled antibody wherein each separate concentration of test antibody and each separate concentration of control antibody are in separate tubes, respectively; (iii) determining the quantity of binding in each reaction tube based on relative electrochemiluminescence (ECL) using ORIGEN instrumentation; and (iv) calculating the average affinity value of the test antibody. The average affinity value may be calculated from the EC50 values and the known concentration of trace antibody using the method of Muller (J. Immunological Methods (1980) 34:345) or any other appropriate method. It should be noted that this assay may also be used to test the affinity of radiolabeled antibodies, or any antibody for which antigen cannot be purified and cells are required as an antigen source. The fixed, lyophilized cells of the present invention may be used as target cells. [0073]
  • When the competitive binding assay of the present invention is performed to test the affinity of anti-CD20 antibodies, the control antibody may be 2B8, or any other unconjugated anti-CD20 antibody. The control antibody may be a chelator-conjugated antibody. The test antibody may also be a chelator-conjugate of the control antibody. Alternatively, the test antibody may be another anti-CD20 antibody whose binding affinity to CD20 as compared to 2B8 is of interest. However, the assay may be adapted for use with antibodies having other specificities so long as an appropriate target cell is available. [0074]
  • In the competitive binding assay of the present invention, the preferred target cells are CD20-positive cells, more preferably SB cells (ATCC # CCL 120), and are more preferably resuspended lyophilized SB cells prepared according to the method of the present invention. Cells lyophilized using other methods or fixed cells may also be used. The ruthenium-labeled antibody is typically prepared by a process comprising incubating the control antibody with N-hydroxysuccinimide ester of ruthenium (II) tris-bypyridine chelator (TAG-NHS), although other known method of labeling antibodies are also envisioned. For labeling, the control antibody and TAG-NHS are preferably incubated at about a 1:15 molar ratio. [0075]
  • These and other aspects of the present invention will become clearly understood by reference to the following figures, examples and description of the invention.[0076]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Immunoreactivity of native 2B8 was compared to commercially available anti-CD20 antibodies Bi (Coulter) and Leu 16 (Becton Dickinson) by direct competition in a radioimmunoassay using [0077] 125I-labeled B1. Antigen-positive SB cells (100,000) were added to each well of V&P filter plates; 10 ng of radiolabeled B1 was mixed with various concentrations of unlabeled competition and the mixture added to the cells. The antibodies were incubated with the cells for one hour at ambient temperature; determinations were performed in triplicate. Subsequently, the wells were washed, dried and the filter-associated radioactivity determined. The data shown were corrected for background radioactivity and are the means of triplicate determinations.
  • FIG. 2. Increasing amounts of unconjugated 2B8 were analyzed for binding to human B-cells (SB) using FACS analysis. Comparisons were made with a commercially available anti-CD20 monoclonal antibody (B1) and with two irrelevant isotype antibodies. Goat anti-mouse IgG-FITC F(ab)′[0078] 2 was used as the secondary reagent. The results show that 2B8 is specific for the CD20 antigen and that it exhibits greater binding than B1.
  • FIG. 3. Human B-cells (SB) were incubated with increasing amounts of [0079] 125I-labeled 2B8. Triplicate samples were incubated for one hour and cell-bound radioactivity was determined after filtration to collect cells. Scatchard analysis allowed calculation of an apparent affinity constant of 4.3×10−9 M.
  • FIG. 4. Immunoreactivity of native 2B8, 2B8-MX-DTPA, and B1. The B1 antibody was radiolabeled as described in the Methods section. Ten nanograms of radiolabeled B1 were mixed with increasing concentrations of the competitor and the mixture added to wells of V&P filter plates containing 100,000 antigen-positive SB cells each; all determinations were performed in triplicate. Following a one hour incubation at ambient temperature, the wells were washed extensively. Subsequently, the filters were dried and the associated radioactivity determined by gamma counting; all values were corrected for background. Values shown are the means of triplicate determinations. [0080]
  • FIG. 5. Antibody 2B8 was formulated at a final concentration of 10 mg/mL in normal saline or normal saline containing 10 mM glycine-HCl, pH 6.8. Duplicate sets of samples were then placed in screw-capped vials, the vials purged with nitrogen, and then capped. The samples were then incubated at 4° C. or 30° C. for 12 weeks; the immunoreactivity of the samples was evaluated weekly. No loss of immunoreactivity was observed with any of the 2B8 samples throughout the 12-week study. Immunoreactivities at week 1 (FIG. 5A), week 6 (FIG. 5B) and week 12 (FIG. 5C) are depicted. [0081]
  • FIG. 6. Binding assay for determination of immunoreactivity of [0082] 111In-labeled 2B8-MX-DTPA incubated in PBS, pH 7.4 containing 50 mg/mL human serum albumin (48 h incubation).
  • FIG. 6A) A constant amount of radiolabeled antibody (5 ng/mL) was incubated with increasing volumes of SB cells (20×106 cells/mL). The amount of radioactivity (cpm) bound to cells was plotted against the volume of cell suspension added. [0083]
  • FIG. 6B) Total applied radioactivity over bound radioactivity (AT/B) was plotted. Linear extrapolation allowed calculation of the y-intercept (0.997). The reciprocal of the y-intercept×100 yielded an immunoreactivity value of 100% at infinite antigen excess. [0084]
  • FIG. 7. Autoradiograms obtained from SDS-PAGE analysis of [0085] 90Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and lmM DTPA. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions, denaturing conditions (SDS). The samples were loaded at 5 μL (lanes 1,2), 10 μL (lanes 5,6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • FIG. 8. Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of [0086] 90Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Trib-glycine gel using non-reducing conditions. Samples were loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 96.2%.
  • FIG. 9. Densitometric scan of 48 h autoradiogram obtained from SDS-PAGE analysis of [0087] 90Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. Samples were loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 95.5%.
  • FIG. 10. Autoradiograms obtained from SDS-PAGE analysis of [0088] 111In-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 50 mg/mL human serum albumin. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions. The samples were loaded at 5 μL (lanes 1, 2), 10 μL (lanes 3, 4), and 20 μL (lanes 5, 6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed. (Note: The 48 h autoradiogram was obtained using intensifying screens resulting in a more intense signal compared to the time zero autoradiogram).
  • FIG. 11. Densitometry scan of time zero autoradiogram obtained from SDS-PAGE analysis of [0089] 111In-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 50 mg/mL human serum albumin. The sample was electrophoresed on a 4-20% Tris-glycine gel under non-reducing conditions. The sample was loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#3) was 95.9%.
  • FIG. 12. Densitometry scan of 48 h autoradiogram obtained from SDS-PAGE analysis of [0090] 111In-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 50 mg/mL human serum albumin. The sample was electrophoresed on a 4-20% Tris-glycine gel under non-reducing conditions. The sample was loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes scanned using a densitometer. The relative area of the radiolabeled conjugate was 97.0% (combined areas of peaks # 2, 3, and 4).
  • FIG. 13. Autoradiograms obtained from SDS-PAGE analysis of [0091] 90Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non- reducing conditions. The samples were loaded at 5 μL (lanes 1, 2), 10 μL (lanes 3, 4), and 20 μL (lanes 5, 6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • FIG. 14. Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of [0092] 90Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 μL, 10 ,L, and 20 μL in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 97.9%.
  • FIG. 15. Densitometric scan of 98 h autoradiogram obtained from SDS-PAGE analysis of [0093] 90Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 94.7%.
  • FIG. 16. Autoradiograms obtained from SDS-PAGE analysis of [0094] 111In-labeled 2B8-MX-DTPA incubate at 37° C. in human serum. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions. The samples were loaded at 5 μL (lanes 1, 2), 10 μL (lanes 3, 4), and 20 μL (lanes 5, 6). The gels were exposed to x-ray film for approximately 16-20 h at ambient temperature and photographed.
  • FIG. 17. Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of [0095] 111In-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 16-20 h at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#3) was 95.3%.
  • FIG. 18. Densitometric scan of the 96 h autoradiogram obtained from SDS-PAGE analysis of [0096] 111In-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 16-20 h at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#3) was 94.0%.
  • FIG. 19. Cynomolgus monkeys were injected intravenously every 48 hours for a total of seven injections; the amounts administered are shown. Circulating T- and B-cell levels were determined by FACS analysis using anti-CD2 (T-cell), anti-Mo-IgG (2B8), anti-CD20 (Leu 16), and anti-human-IgG (B-cell). No effect was observed on circulating T-cell levels. (Group V animals were given a single dose). [0097]
  • FIG. 20. The recovery of circulating B-cell levels in animals receiving 2B8 was followed by FACS analysis using the fluorescently-labeled antibodies described in the brief description of FIG. 19. The animals in Groups III and IV were not monitored as they were sacrificed on day 13. [0098]
  • FIG. 21. Cynomolgus monkeys were injected intravenously with 89y 2B8-MX-DTPA which had been prepared using clinical-grade 2B8-MX-DTPA. The animals were dosed every 48 hours with the amounts shown above for a total of seven doses. On [0099] days 0, 2, 7, 10 and 14 the monkeys were bled and evaluated for serum chemistries hematology and circulating B-cell levels (day 10 sera were not analyzed for B-cell content). Other than decreased total lymphocyte count in all animals, except one individual in groups II, no significant abnormalities were noted during the course of the study.
  • FIG. 22. The clearance of murine anti-CD20 antibody 2B8 from cynomolgus monkeys was determined by ELISA following a single injection of 10 mg/kg on day zero. As shown in panel A, the antibody exhibited at βt[0100] ½ value of approximately 4.5 days. The clearance of the 2B8 antibody and its MX-DTPA conjugate from the circulation of BALB/c mice are shown in panel B. Mice were injected intravenously with 25 μg of native or conjugated 2B8 and blood samples taken at various times up to 264 hours following injection; sera was subsequently analyzed by enzyme immunoassay using SB cells as the capture agent. Both the native and conjugated antibodies exhibited clearance values of 8.75 days.
  • FIG. 23. Twenty BALB/c mice were each injected with 1.1 μCi of radiolabeled conjugate (100 μL) formulated in PBS, pH 7.4, containing 50 mg/mL HSA. Groups of five mice each were sacrificed at 1, 24, 48, and 72 hours and then blood and various tissues prepared and analyzed for associated radioactivity. [0101]
  • FIG. 24. Twenty BALB/c mice were each injected intravenously with approximately 1.0 μCi (in 100 μl) of radiolabeled conjugate formulated in 1× PBS, pH 7.4, containing 75 mg/mL human serum albumin and 1 m MDPA. Groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their blood and various tissues prepared and analyzed for associated radioactivity. [0102]
  • FIG. 25. Athymic mice bearing Ramos B-cell tumors were injected intravenously with 24 μCi of [0103] 111-In-2B8-MX-DTPA and groups of three mice each were sacrificed at 0, 24, 48 and 72 hours. Following tissue preparation and determination of associated radioactivity, the percent injected dose per gram tissue values were determined and plotted as shown.
  • FIG. 26. Binding assay for determination of immunoreactivity of “mix-&-shoot” [0104] 90Y-labeled 2B8-MX-DTPA incubated in PBS, pH 7.4 containing 50-75 mg/mL human serum albumin (48 h incubation). Panel A) A constant amount of 90Y-labeled antibody (approximately 1 ng/ml) was incubated with increasing amounts of SB cells. The amount of radioactivity (cpm) bound to cells was plotted against the cell concentration. Panel B) Total applied 90Y radioactivity over bound radioactivity (AT/B) was plotted. Linear extrapolation allowed calculation of the y-intercept (1.139). The reciprocal of the y-intercept×100 yielded an immunoreactivity value of 87.9% at infinite antigen excess. No binding was observed with CD20-negative cells (HSB).
  • FIG. 27. Autoradiograms obtained from SDS-PAGE analysis of [0105] 90Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions, denaturing conditions (SDS). The samples were loaded at 5 μL (lanes 1,2), 10 μL (lanes 5,6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • FIG. 28. Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of [0106] 90Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Trib-glycine gel using non-reducing conditions. Samples were loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 96.1%.
  • FIG. 29. Densitometric scan of 48 h autoradiogram obtained from SDS-PAGE analysis of [0107] 90Y-labeled 2B8-MX-DTPA incubated at 4° C. in PBS, pH 7.4 containing 75 mg/mL human serum albumin and 1 mM DTPA. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. Samples were loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. The gel was exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 94.1%.
  • FIG. 30. Autoradiograms obtained from SDS-PAGE analysis of “mix-&-shoot” [0108] 90Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. At the indicated times, samples were electrophoresed on 4-20% Tris-glycine gels using non-reducing conditions. The samples were loaded at 5 μL (lanes 1, 2), 10 μL (lanes 3, 4), and 20 μL (lanes 5, 6). The gels were exposed to x-ray film for approximately 15 min at ambient temperature and photographed.
  • FIG. 31. Densitometric scan of time zero autoradiogram obtained from SDS-PAGE analysis of “mix-&-shoot” [0109] 90Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 89.1%.
  • FIG. 32. Densitometric scan of 72 h autoradiogram obtained from SDS-PAGE analysis of “mix-&-shoot” [0110] 90Y-labeled 2B8-MX-DTPA incubated at 37° C. in human serum. The sample was electrophoresed on a 4-20% Tris-glycine gel using non-reducing conditions. The sample was loaded at 5 μL, 10 μL, and 20 μL in duplicate wells. Gels were exposed to x-ray film for approximately 15 min at ambient temperature and one of the lanes was scanned using a densitometer. The relative area of the radiolabeled conjugate peak (#2) was 88.8%.
  • FIG. 33. Twenty BALB/c mice were each injected intravenously with 5 μCi [0111] 90Y-labeled 2B8-MX-DTPA formulated in 1× PBS, pH 7.4, containing 75 mg/mL human serum albumin and 1 mM DTPA. Groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their blood and various tissues prepared and analyzed for associated radioactivity.
  • FIG. 34. Increasing amounts of CHO-derived 2B8 antibody labeled were incubated with a fixed concentration of freshly harvested CD20-positive B-cells (SB) or CD20-negative T-cells (HSB). Antibody binding to cells was quantified using FACS analysis using goat anti-mouse IgG-FITC F(ab)′[0112] 2 as described herein. Comparison was made to an irrelevant isotype antibody (S004). Only the CHO-derived 2B8 antibody showed any appreciable binding to CD20-positive SB cells.
  • FIG. 35. The immunoreactivity of CHO-derived 2B8 was compared to the 2B8-49 parent antibody produced in a hybridoma cell line by direct competition in an ORIGEN assay. Increasing amounts of antibody was incubated with a fixed concentration of CD20-positive B-cells (SB) and a trace amount of ruthenium-labeled CHO 2B8. After incubation for three hours at ambient temperature, binding, expressed as relative electrochemiluminescence (ECL), was determined using the ORIGEN instrument as described in the Materials and Methods. Values represent the means of duplicate determinations. Average affinity constants for CHO 2B8 and 2B8-49 were calculated to be 1.3×10[0113] −10 M and 2.5×10−10 M, respectively. An irrelevant isotype antibody (S004), was included for comparison.
  • FIG. 36. The binding of 2B8-MX-DTPA conjugates prepared from CHO-derived 2B8 was compared to the unconjugated antibody by direct competition in an ORIGEN assay. Conjugates were prepared by incubation of 2B8 with MX-DTPA for 8, 17, and 24 h before removal of unreacted chelate. For binding assessment, antibodies were incubated with a fixed concentration of CD20-positive B-cells (SB) and a trace amount of ruthenium-labeled CHO 2B8. After incubation for three hours at ambient temperature, binding, expressed as relative electrochemiluminescence (ECL), was determined using the ORIGEN instrument as described in the Materials and Methods. Values represent the means of duplicate determinations. Conjugate preparations exhibited similar binding compared to unconjugated 2B8 antibody. [0114]
  • FIG. 37. A) SB cells were washed and resuspended to 90×10[0115] 6 cells/mL with dilution buffer (1× PBS, pH 7.4 containing 1% (w/v) bovine serum albumin. Increasing concentrations of cells were incubated for 3 h with 7.5 ng/mL In2B8 prepared using 2B8-MX-DTPA lot 0165A. B) Double-inverse plot of cell concentration vs. bound radioactivity/total radioactivity (B/AT). Immunoreactivity was calculated as 1/y-intercept×100. Immunoreactivity and correlation coefficient (R) values were 80.6% and 0.981, respectively.
  • FIG. 38. A) SB cells were washed and resuspended to 90×10[0116] 6 cells/mL with dilution buffer (1× PBS, pH 7.4 containing 1% (w/v) bovine serum albumin. Increasing concentrations of cells were incubated for 3 h with 2 ng/mL Y2B8 prepared using 2B8-MX-DTPA lot # 0165A. B) Double-inverse plot of cell concentration vs. bound radioactivity/total radioactivity (B/AT). Immunoreactivity was calculated as 1/y-intercept x 100. Immunoreactivity and correlation coefficient (R) values were 72.2% and 0.999, respectively.
  • DETAILED DESCRIPTION OF THE INVENTION DEFINITIONS
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are described. For purposes of the present invention, the following terms are defined below. [0117]
  • low metal—refers to reagents treated to reduce metal contamination to a level which does not impact radioincorporation [0118]
  • antigen positive—means expresses antigen that is recognized by particular antibody of the invention in such a way that the antibody is capable of binding. [0119]
  • % radioincorporation—refers to the amount of radiolabel from a radiolabeling reaction that is conjugated to the antibody relative to the total amount of radiolabel initially added to the reaction. [0120]
  • % binding—refers to the amount of antibody from a sample which binds to the target antigen, with or without specificity. [0121]
  • % immunoreactivity or binding specificity—refers to that amount of an antibody sample which binds to the target antigen with specificity. diagnostic antibody—refers to an antibody conjugated to a radiolabel such as [0122] 111In which can effect diagnostic imaging of tumors and antigen positive cells.
  • therapeutic antibody—refers to an antibody conjugated to a alpha or beta emitting radiolabel (such as [0123] 90Y) which can effect cell killing when bound to the targeted antigen.
  • DESCRIPTION OF THE INVENTION
  • Pre-clinical Development of Murine Monoclonal Anti-CD20 Antibody 2B8, Conjugated 2B8, [0124] 111In and 90Y-Labeled 2B8
  • I. Materials and Methods for Development of Murine Monoclonal Anti-CD20 Antibody 2B8, Conjugate 2B8-MX-DTPA, [0125] 111In-Labeled 2B8-MX-DTPA and HPLC-Purified 90Y-MX-DTPA
  • A. Materials. [0126]
  • 1. Cells. [0127]
  • The human cell lines SB and HSB were obtained from the American Type Culture Collection and cultured in RPMI-1640 containing 10% fetal bovine serum. The CD20-positive SB cell line is a B lymphoblastoid cell line derived from the peripheral blood buffy coat of a patient with acute lymphoblastic leukemia (1). The antigen-negative cell line HSB is a T lymphoblastoid cell line developed from tumors induced in newborn Syrian hamsters (2). The murine myeloma cell line SP2/0 was similarly maintained in RPMI-1640 containing 10% fetal bovine serum. [0128]
  • 2. Antibodies. [0129]
  • The anti-CD20 antibodies Bi and [0130] Leu 16 were purchased from Coulter Immunology and Becton/Dickinson, respectively. The 125I-labeled goat anti-mouse IgG and goat anti-human IgG antibodies were obtained from ICN. Goat F(ab′)2 anti-mouse IgG was obtained from Cappel.
  • 3. Reagents. [0131]
  • Freund's complete and incomplete adjuvants were purchased from Sigma Chemical Company. Polyethylene glycol, HAT concentrate, and HT concentrate were all obtained from Boehringer Mannheim. Fluorescein isothiocyanate (FITC) was purchased from Sigma Chemical Company. Indium-[[0132] 111] chloride and 90Y chloride were obtained from Amersham or NEN Dupont. Yttrium-[89] chloride was purchased from Aldrich Chemical Company. All other reagents were obtained from standard sources.
  • Reagents used for conjugation and radiolabeling protocols were processed to remove contaminating heavy metal ions which could compete with the radioisotopes during the radiolabeling step. Reagents were typically processed by passing the solutions through a column of [0133] Chelex 100 ion exchange resin (BioRad Industries) or batch processing by addition of Chelex 100 to a prepared solution. Low metal-containing water, either Milli-Q-purified or Water for Irrigation (WFIr) was used for all preparations and dilutions. The metal-free solutions were sterile-filtered and collected in sterile plastic containers.
  • B. Methods. [0134]
  • 1. Production and Screening of 2B8 Hybridoma Supernatants by RIA. [0135]
  • Ten BALB/c mice were immunized with 20 million SB cells suspended in PBS containing Freunds complete adjuvant. The cells were injected both s.c and i.p at multiple sites on the animal. After a 2 week rest period the mice were injected a second time with SB cells emulsified in Freund′ s incomplete adjuvant. Subsequent immunization boosters were performed on a weekly schedule with SB cells suspended in PBS. Mice were immunized for a period of 6 weeks to 4 months. [0136]
  • Two animals at a time were sacrificed by cervical dislocation and their spleens removed for fusion with the murine myeloma SP2/0. Animals were chosen based on the ability of post-immune sera to effectively inhibit the binding of radiolabeled Coulter B1 anti-CD20 antibody to human SB cells. Three days prior to each fusion the selected animals were given one last intravenous (tail vein) injection of 20 million SB cells in PBS. Upon sacrifice the spleens were removed under aseptic conditions and the splenocytes fused with SP2/0 cells at a ratio of 5:1 (splenocytes:SP2/0). Fused cells were washed in tissue culture media and distributed into 96 well plates containing HAT selection media. Hybridomas were screened by inhibition radioimmunoassay using Coulter B1 antibody after 10-14 days. [0137]
  • Screening of hybrids secreting anti-CD20 antibody was accomplished using established radioimmunoassay methods. Briefly, Coulter B1 anti-CD20 antibody was purified by Protein A affinity chromatography. Fifty micrograms of purified antibody was coupled to [0138] 125I by brief oxidation in the presence of lodobeads (Pierce Chemical Co.), following the manufacturer's procedure. The radiolabeled antibody was desalted on amberlite resin and stored in dilution buffer (PBS, pH 7.4, containing 0.2% gelatin, 0.02% sodium azide, and 1.0% BSA). Ten nanograms of radiolabeled antibody was placed in each well of a previously blocked filter assay plate (blocking buffer: dilution buffer containing 10% FBS) along with 50 μL of hybridoma supernatant from test wells and 100,000 SB cells suspended in 50 μL dilution buffer. The suspension was incubated for one hour at ambient temperature. The plates were washed thoroughly with wash buffer (PBS, pH 7.4, containing 0.2% gelatin and 0.02% sodium azide) on a V&P Scientific vacuum manifold and filter bottoms containing trapped SB cells were transferred to a gamma counter. Wells containing only HAT media and labeled B1 antibody were used as background controls and identical wells containing SB cells were used as positive controls. Inhibition controls contained radiolabeled B1 and various amounts of unlabeled B1 antibody ranging from 2 μg to 2 ng.
  • 2. Flow Cytometry Studies. [0139]
  • a. Cell Lines [0140]
  • Preliminary flow cytometry studies were performed with supernatants from 2B8 hybridoma cultures. One hundred microliters of hybridoma supernatant was incubated with SB cells for one hour at ambient temperature; a secondary antibody (goat F(ab′)2 anti-mouse IgG; Cappel), used at a 1/400 dilution, was added subsequently and the incubation continued for 1 hour in the dark. The cells were washed for 5 times. Controls included cells only (no primary or secondary antibody) from which autofluorescence was determined, cells with secondary antibody only to determine non-specific binding and commercially available fluorescein isothiocyanate-conjugated B1 (B1-FITC) for a CD20 population control. [0141]
  • For some experiments, fluorescein was coupled to purified 2B8 antibody through the reaction of amino groups with fluorescein isothiocyanate (FITC). Briefly, 2B8 antibody (1.2 mg/mL) was incubated in pH 9.5, 0. 1M sodium carbonate buffer with 150-200 μg FITC per mg protein. The solution was incubated at room temperature for 2 hours and the resulting 2B8-FITC conjugate was purified on a Sephadex G-25 column. Other reagents used in these studies such as B1 and [0142] Leu 16 were purchased as fluorescein conjugates directly from Coulter or Becton Dickinson.
  • Cells to be analyzed were harvested and washed three times with PBS containing 0.2% BSA and 0.1% sodium azide. Viability was determined by trypan blue exclusion with a viability requirement of >90%. Cell concentrations were adjusted to 3 million per ml with 50 μL added per well into 96 well U-bottom plates. Primary antibody (50 μL) was added to appropriate wells and the mixture incubated for 30 min. to 1 h. at ambient temperature; subsequently the cells were washed 5 times with 200 μL/well of PBS containing 0.2% BSA and 0.02% sodium azide. Cells were centrifuged in the plates at 1300 RPM for 1 min. in a Sorvall centrifuge and the supernatants removed by gently “flicking” the plates. Secondary antibody, if needed, was added and incubated for an additional 30 min to 1 h at ambient temperature in the dark; wells were then washed as above. Finally, 200 μL of fixing buffer (0.15 M sodium chloride containing 1% paraformaldehyde, pH 7.4) was added to each sample and the treated cells transferred to 12×75 mm tubes for analysis. [0143]
  • b. Whole Blood From Cynomolgus Monkeys. [0144]
  • After removal of plasma, the cells were washed twice by centrifugation and resuspension in HBSS. Fetal bovine serum (2 mL) was added and the cells resuspended. One hundred microliters of the resuspended cells were then distributed to each of 6, 15 ml conical centrifuge tubes. Fluorescently-labeled monoclonal antibodies were added as follows: [0145]
    Tube #1: Murine anti-CD2-FITC (AMAC), 2.5 μg/mL, 5 μg;
    Tube #2: Goat anti-Human IGM-FITC (Fisher) 2.5 μg/mL, 5 μg;
    Tube#3: Goat anti-mouse IgG-RPE (Fisher) 2.5 μg/mL, 5 μg;
    Tube #4: Goat anti-Human IgM-FITC + Goat anti-mouse IgG-RPE
    (absorbed), 2.5 μg/mL, 5 μg;
    Tube #5: anti-human CD20-FITC (anti-Leu 16, Becton Dickinson),
    5 μg;
    Tube #6: Cells only (auto-fluorescence).
  • Labeled antibodies and cells were centrifuged for 2 min at 1500 rpm to mix cells and antibodies and all 6 samples were then placed on ice and incubated for 30 min. Subsequently the tubes were removed from the ice and lysing buffer (prewarmed to 37° C.) was added to a volume of 12 mL. The samples were then incubated for 15 min at room temperature, centrifuged for 5 min at 40° C. at 1500 rpm, and the supernatants removed. Cell pellets were then washed twice in labeling buffer (PBS containing 1% BSA and 0. 05% sodium azide). [0146]
  • Subsequently the cells were fixed by the addition of 0.5 mL of fixation buffer (0.15 M sodium chloride, pH 7.4, containing 1% paraformaldehyde) per tube and analyzed on a Becton Dickinson FACScan instrument using autocompensation and precalibration with Calibrite beads. Green fluorescence from fluorescein was measured in FL1 mode and red fluorescence from phycoeretherin was measured in FL2 mode. Data were expressed in log form. Viable lymphocyte populations were initially identified by forward vs. right angle light scatter in a dot plot bitmap. The total lymphocyte population was then isolated by gating out all other events. Subsequent fluorescence measurements reflected only those specific events which occurred within the gated area. [0147]
  • For high-dose pharmacology/toxicology studies the pre-study lymphocyte levels were determined for each cynomolgus monkey and used as baseline values. The percentage of T- and B-cells and T:B ratios were calculated and used as depletion references. The pre-study B cell population was enumerated with [0148] Leu 16 and anti-human IgM antibodies.
  • After injection of 2B8 into the monkeys, when the CD20 antigen was saturated with 2B8, the percentage of B cells in the total population was approximated using goat anti-human IgM-FITC, anti-mouse IgG-RPE and the double staining population containing these two markers. The double staining population was used for quantitation until all of the 2B8 was cleared from the peripheral blood of the animals. The percentage of T cells in the total lymphocyte population was estimated using anti-CD2-FITC. Data were averaged from three, 10,000 event measurements made with each sample. Cells from each of the designated blood samples were evaluated subsequently, enumerating in each case the T- and B-cell subpopulations within the total lymphocyte population. The T:B ratios were also examined. Depletion of B-cells was calculated as the percent of reduction of B-cells relative to original B-cell levels for each individual monkey. [0149]
  • 3. Radioiodination and Immunoprecipitation of CD20. [0150]
  • One hundred million SB cells were divided into two equal parts after surface iodination with 1251 and lodobeads (Pierce Chemical Co.). The cells were washed repeatedly by centrifugation until radioactivity levels in the supernatant returned to background. One hundred micrograms of 2B8 or B1 (Coulter Immunology) antibody were added to either of the two samples of labeled B cells. The antibodies and SB cells were incubated overnight and then washed three times by centrifugation until all of the unbound antibody was removed. The cell pellets containing bound 2B8 and B1 were then lysed and extracted by addition of 1% NP-40 detergent in 0.1 M Tris-HC1, pH 8.0, followed by incubation at room temperature for 1 h. The extract was centrifuged in a microfuge at high speed for 30 min and the supernatants were transferred to new tubes. Protein A-Sepharose (300 μL) was added to each tube and the resin pelleted by centrifugation. The protein A-Sepharose was then washed 20 times to remove non specifically bound iodinated protein. When the bead-to-supernatant radioactivity ratio reached a value of 100, the pellet was extracted with SDS PAGE sample buffer and heated to boiling. After cooling, approximately 15,000 cpm of each of the extracts were added to wells of a 10% polyacrylamide gel. A low molecular weight pre-stained standard (BioRad Inc.) was added to a separate well and used for molecular weight estimation. The proteins were resolved by electrophoresis and the gel was dried and exposed to a sheet of X-ray film for 24 hours at −70° C.; subsequently the film was developed and analyzed. [0151]
  • 4. Scatchard Analysis of 2B8 Binding. [0152]
  • Purified 2B8 was evaluated for apparent affinity by Scatchard analysis. Radiolabeled 2B8 was prepared by reaction with [0153] 125I in the presence of lodobeads. Following removal of free iodine the radiolabeled antibody was incubated in various concentrations, in duplicate, ranging from 5000 ng per well to 35 ng/well with 10,000 SB cells. The amount of antibody binding to cells was calculated from the specific activity of the 1251-labeled 2B8. The ratio of bound/free antibody was plotted against the molar concentration of bound antibody and the apparent affinity constant was determined from the ratio of the X and Y axis intercepts.
  • 5. Preparation of 2B8-MX-DTPA [0154]
  • a. Source of MX-DTPA [0155]
  • For some pre-clinical studies, carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylenetriaminepentaacetic acid (MX-DTPA) was provided as a dry solid by Dr. Otto Gansow at the National Institute of Health and stored desiccated at 4° C. protected from light. Stock solutions of the chelate were prepared in Milli-Q water and the concentration determined by assessing the radioactivity and using the specific activity of the compound. Stock solutions were generally 2-5 mM and were stored at −70° C. in polypropylene tubes. For other studies, MX-DTPA was obtained from Coulter Immunology as the disodium salt in water and stored at −70° C. [0156]
  • b. Maintenance of Metal-Free Conditions [0157]
  • In addition to using metal-free reagents, all manipulations of reagents were performed so as to minimize the possibility of metal contamination. When possible, polypropylene plastic containers such as flasks, beakers and graduated cylinders were used. These were washed with Alconox and exhaustively rinsed with Milli-Q water before use. In addition, metal-free pipette tips (BioRad) were used for accurately manipulating small volumes. For manipulating larger volumes of reagents, sterile, plastic serological pipettes (available in 1 to 25 mL sizes) were used. Reactions were conveniently performed in screw-top, polypropylene microfuge tubes (Sardstedt Industries; 1.5 μL capacity) or polypropylene conical tubes (Costar; 15 mL and 50 μL). When dialysis tubing was manipulated, disposable surgical gloves, previously rinsed with Milli-Q water, were worn. [0158]
  • c. Preparation of Antibody [0159]
  • The murine anti-CD20 antibody 2B8 was purified initially from ascites by Protein A and QAE chromatography. For later experiments 2B8 was purified from hollow-fiber bioreactor supernatants using the same purification process. The hollow-fiber-derived antibody has now been replaced for commercialization purposes with the CHO-derived antibody described in Example 2. [0160]
  • The antibody was prepared for conjugation by transferring it into metal-free 50 mM bicine-NaOH, pH 8.6, containing 150 mM NaCl, using dialysis or repetitive buffer exchange. In some studies, buffer exchange was effected using repetitive ultrafiltration with Centricon 30 (Amicon) spin filters (30,OOOD MWCO). In general, 50-200 μL of protein (10 mg/mL) was added to the filter unit and 2 mL of bicine buffer added. The filter was centrifuged at 4° C. in a Sorval SS-34 rotor at 6,000 rpm for 45 min. Retentate volume was approximately 50-100 μL. This process was repeated twice with the same filter. Retentate was transferred to a polypropylene 1.5 mL screw cap tube, assayed for protein, diluted to 10.0 mg/mL and stored at 4° C. until used for conjugation. For some studies, the protein was transferred into 50 mM sodium citrate, pH 5.5 containing l5OmM NaCl and 0.05% sodium azide using the same protocol described above. [0161]
  • d. Conjugation Protocol [0162]
  • Conjugation of 2B8 with MX-DTPA was performed in polypropylene tubes at ambient temperature. Frozen stock solutions of MX-DTPA were thawed immediately before use. Typically, 50-200 μL of antibody at 10 mg/mL were reacted with chelate at a molar ratio of chelate-to-protein of 4:1. Reactions were initiated by adding the chelate stock solution and gently mixing; the conjugation was allowed to proceed overnight, generally for 14 to 20 h, at ambient temperature. Unreacted chelate was removed from the conjugate by dialysis or repetitive ultrafiltration, as described above, into metal-free normal saline (0.9% w/v) containing 0.05% sodium azide. The protein concentration was adjusted to 10 mg/mL and stored at 4° C. in a polypropylene tube until radiolabeled. [0163]
  • e. Determination of Chelate Incorporation [0164]
  • Chelate incorporation was determined by scintillation counting and comparing the value obtained with the purified conjugate to the specific activity of the carbon-[14]-labeled chelate. For later studies, in which non-radioactive chelate obtained from Coulter Immunology was used, chelate incorporation was assessed by incubating the conjugate with an excess of a radioactive carrier solution of [0165] 90Y of known concentration and specific activity.
  • Briefly, a stock solution of yttrium chloride of known concentration was prepared in metal-free 0.05 N HC1 to which carrier-free [0166] 90Y (chloride salt) was added. An aliquot of this solution was analyzed by liquid scintillation counting to determine an accurate specific activity for this reagent. A volume of the yttrium chloride reagent equal to 3-times the number of mols of chelate expected to be attached to the antibody, typically 2 mol/mol antibody, was added to a polypropylene tube, and the pH adjusted to 4.0-4.5 with 2 M sodium acetate. Conjugated antibody was subsequently added and the mixture incubated 15-30 min at ambient temperature. The reaction was quenched by adding 20 mM EDTA to a final concentration of 1 mM and the pH of the solution adjusted to approximately pH 6 with 2M sodium acetate.
  • After a 5 min incubation the entire volume was purified by high-performance size-exclusion chromatography as described below. The eluted protein-containing fractions were combined, the protein concentration determined, and an aliquot assayed for radioactivity. The chelate incorporation was calculated using the specific activity of the [0167] 90Y chloride preparation and the protein concentration.
  • f. Immunoreactivity of 2B8-MX-DTPA [0168]
  • The immunoreactivity of conjugated 2B8 was assessed using whole-cell ELISA. Mid-log phase SB cells were harvested from culture by centrifugation and washed two times with 1× HBSS. Cells were diluted to 1-2×10[0169] 6 cells/mL in HBSS and aliquoted into 96-well polystyrene microliter plates at 50,000-100,000 cells/well. The plates were dried under vacuum for 2 h at 40-45° C. to fix the cells to the plastic. The plates were stored dry at −20° C. until used. For assay, the plates were warmed to ambient temperature immediately before use, then blocked with 1× PBS, pH 7.2-7.4 containing 1% BSA (2 h). Samples for assay were diluted in 1× PBS/1% BSA, applied to plates and serially diluted (1:2) into the same buffer. After incubating plates for 1 h at ambient temperature, the plates were washed three times with 1× PBS. Secondary antibody (goat anti-mouse IgGl-specific HRP conjugate) (50 μL) was added to wells (1:1500 dilution in 1× PBS/1% BSA) and incubated 1 h at ambient temperature. Plates were washed four times with 1× PBS followed by the addition of ABTS substrate solution (50 mM sodium citrate, pH 4.5 containing 0.01% ATBS and 0.001% H202). Plates were read at 405 nm after 15-30 min incubation. Antigen-negative HSB cells were included in assays to monitor non-specific binding. Immunoreactivity of the conjugate was calculated by plotting the absorbance values vs. the respective dilution factor and comparing these to values obtained using native antibody (representing 100% immunoreactivity) tested on the same plate. Several values on the linear portion of the titration profile were compared and a mean value determined.
  • g. In Vitro Stability of Native 2B8 and 2B8-MX-DTPA [0170]
  • For this 12-week assessment of antibody and conjugate stability, aliquots of 2B8 antibody and 2B8-MX-DTPA were formulated in either normal saline or normal saline containing 10 mM glycine-HCl, pH 6.8. Duplicate sets of samples were incubated at both 4° and 30° C. and samples assayed weekly using the following methods: SDS-PAGE (both reducing and nonreducing), immunoreactivity by whole-cell enzyme immunoassay using either SB (antigen-positive) or HSB (antigen-negative) cells as capture, and isoelectric focusing gel electrophoresis (pH range, 3-10). In addition, the radiolabeling efficiency of the conjugate was assessed at [0171] weeks 4, 8, and 12 by radiolabeling the conjugate with 90Y and analyzing the product by SDS-PAGE and autoradiographic analysis. Finally, in a separate study, aliquots of 2B8-MX-DTPA incubated at 4° and 30° C. for 10 weeks were radiolabeled with 111In and evaluated in a biodistribution study in BALB/c mice as described below.
  • h. Immunohistology Studies. [0172]
  • Immunohistology studies with both the native and conjugated (2B8-MX-DTPA) antibodies were performed by IMPATH Laboratories using sections of human tissues fixed with acetone. The antibody was purified from hollow-fiber bioreactor supernatants by chromatography on protein A and Q Sepharose. Clinical-grade conjugate was prepared using MX-DTPA from Coulter Immunology according to the protocol described above. [0173]
  • i. In Vitro Immunoreactivity of Radiolabeled 2B8-MX-DTPA. [0174]
  • For some experiments, the whole-cell ELISA protocol used for unlabeled 2B8-MX-DTPA was used. In later experiments, immunoreactivity of [0175] 111In and 90Y-labeled conjugates (each prepared at IDEC Pharmaceuticals or, alternatively, at MPI Pharmacy Services, Inc.) was determined using a modified version of the whole-cell binding assay described by Lindmo (3). Briefly, increasing concentrations of mid-log phase, antigen-positive SB cells or antigen-negative HSB cells [20-30×106 cells/mL in dilution buffer (PBS, pH 7.4 containing 1% BSA, 0.1% gelatin, and 0.02% sodium azide)] were added to duplicate sets of tubes. The radiolabeled conjugate was diluted to a final antibody concentration of 1-5 ng/mL with dilution buffer and 0.35 mL was added to each tube. Following a 75-90 min incubation period at ambient temperature the cells were pelleted by centrifugation and the supernatants collected. Radioactivity remaining in the supernatant fraction was determined with a gamma or scintillation counter. The data were plotted as the quotient of the total radioactivity added divided by the cell-associated radioactivity, versus the inverse of the cell number per tube. The y axis intercept thus represents the immunoreactive fraction.
  • j. In Vitro Stability of Radiolabeled 2B8-MX-DTPA in Human Serum. [0176]
  • The in vitro stability of [0177] 111In- and 90Y-labeled 2B8-MX-DTPA was assessed by incubation in human serum at 37° C. for 96 hours. The conjugated antibody was prepared and radiolabeled with 111In (“mix-and-shoot” protocol) or 90Y as described above. The specific activities of the 111In and 90Y-labeled conjugates were 2.5 and 14.6 mCi/mg, respectively; the radiolabeled conjugates were suspended in buffer containing 75 mg/mL human serum albumin (HSA) and 1 mM DTPA (yttrium-labeled conjugate) or buffer containing 50 mg/mL HSA (indium-labeled conjugate). The radiolabeled conjugates were diluted 1:10 with normal human serum (non-heat-inactivated) and aliquots placed aseptically into sterile capped tubes; these tubes were then incubated at 37° C. for periods up to 96 hours. At selected times conjugate samples were removed and analyzed by non-reducing SDS-PAGE in 4-20% gradient gels followed by autoradiography, and by instant thin layer chromatography.
  • k. In Vitro Stabilitv of Clinically-Formulated [0178] 111In-2B8-MX-DTPA.
  • The 2B8-MX-DTPA conjugate was radiolabeled with [0179] 111In and used without HPLC purification (“mix-and-shoot” protocol). The radiolabeled antibody was diluted into PBS and human serum albumin (HSA) added to a final concentration of 50 mg/mL. The specific activity of the formulated radiolabeled conjugate was 2.2 mCi/mg. The formulated conjugate was subsequently incubated at 4° C. for 48 hours and aliquots analyzed at time 0, 24 h and 48 hours using non-reducing SDS-PAGE in 4-20% gradient gels followed by autoradiography, and by instant thin layer chromatography. The immunoreactivity at each time point was assessed using the whole-cell suspension assay described in section 1 above.
  • 1. In Vitro Stability of Clinically-Formulated [0180] 90Y-2B8-MX-DTPA.
  • The 2B8-MX-DTPA conjugate was radiolabeled with [0181] 90Y and purified by size-exclusion chromatography on HPLC using 1× PBS as an elution buffer. The radiolabeled conjugate fractions were pooled and human serum albumin and DTPA were added to final concentrations of 75 mg/mL and 1 mM, respectively. The specific activity of the formulated radiolabeled conjugate was 14.6 mCi/mg. The formulated conjugate was subsequently incubated at 4° C. for 48 hours and aliquots analyzed at time 0, 24 h and 48 hours using non-reducing SDS-PAGE in 4-20% gradient gels followed by autoradiography, and instant thin layer chromatography. Immunoreactivity at each time point was assessed using the whole-cell suspension assay described in section 1 above.
  • 2. Animal Studies. [0182]
  • a. Primate High Dose Pharmacology/Toxicology Study Using 2B8. [0183]
  • Antibody 2B8 was evaluated in a high-dose pharmacology study performed under GLP regulations at White Sands Research Center (Study Number 920111). Adult [0184] Macaca fascicularis (cynomolgus) monkeys were used; study groups each consisted of one male and one female. The antibody was injected intravenously every 48 hours for a total of seven injections. The study consisted of five groups: Group I (saline); Group II (0.6 mg/kg); Group III (2.5 mg/kg); Group IV (10 mg/kg); and, Group V (10 mg/kg on day 0 only).
  • Prior to initiation of the study, blood was obtained from all 10 animals and used to determine reagent backgrounds and initial B cell populations. All subsequent blood samples were drawn prior to each antibody injection. Groups III and IV were sacrificed at day 13 for complete necropsy and histopathology. [0185]
  • Animals in groups I, II, and V were bled on [0186] days 0, 1, 3, 7, 13, 21, 37 and 52; approximately 5 mL whole blood was drawn in heparinized tubes. Whole blood was kept at 4° C. and analyzed within 24 hours. Blood from each animal was centrifuged at 2000 rpm for 5 min. and the supernatant plasma was removed for assay of serum 2B8 levels by RIA (see RIA procedure for specific assay methods). The pelleted material containing PBLs and RBCs was resuspended in FCS for FACS analysis.
  • b. Pharmacokinetic Studies with 2B8 and 2B8-MX-DTPA. [0187]
  • The mean serum beta half life of 2B8 in cynomolgus monkeys was determined using Group V animals (above). Goat anti-mouse IgGl (Fisher Scientific) was diluted to 2.0 μg per ml in 10 mM borate buffer, pH 9.6, and 50 μL was added to each well of a 96-well plate. The antibody was allowed to bind to the plate during an overnight incubation at 4° C., or for 2 h at ambient temperature. Each plate was blocked for 30 min. at ambient temperature with 150 μL per well of PBS containing 1% BSA. The plates were washed with distilled water and serum or plasma samples were applied in triplicate to individual wells at 1: 100 initial dilution followed by serial 1:2 dilutions. Purified 2B8 was added to pre-bleed sera and diluted for use as a standard curve beginning with 0.5 mg/mL; samples were diluted 1:100 and then serially diluted as with the other samples. The plates were incubated for 1 h at ambient temperature and washed 4 times with distilled water. The secondary reagent (goat anti-mouse IgGl-HRPO) was then added at 1:4000 dilution and incubated at ambient temperature for an additional hour. The plates were washed again in distilled water and 0.1 mL peroxidase substrate was added containing hydrogen peroxide. Color was allowed to develop from the reaction for 20 min.; the absorbance was subsequently determined at 405 nm using a microplate ELISA reader. The results were plotted in μg antibody per mL serum. [0188]
  • In addition, the βt[0189] ½ values of 2B8 and 2B8-MX-DTPA were determined in BALB/c mice. Unconjugated 2B8 stored at −70° C. in 1× PBS, pH 7.4/10% glycerol was thawed, diluted to 0.5 mg/mL and sterile filtered. Conjugated antibody was prepared following standard protocols but with carbon-[14]-labeled chelate; chelate incorporation was 1.5 mol/mol antibody. The purified conjugate was diluted to 0.5 mg/mL in normal saline (0.9%), sterile filtered, and stored at 4° C. with the native antibody until used.
  • Six-to-eight week old mice were injected with 100 μL of purified 2B8 antibody at a concentration of 250 μg/mL. Mice were subsequently bled by retro-orbital puncture at various times ranging from 0 to 264 hours and their sera analyzed for the presence of the native and conjugated 2B8 antibody by whole-cell enzyme immunoassay using the antigen-positive B-cell line SB as the capture. The resulting data were plotted as the concentration of 2B8 or 2B8-MX-DTPA versus time; from these results a linear regression plot was generated and the slope used to determine the βt[0190] ½ values.
  • c. Pharmacology/Toxicology Study of [891-Y-2B8-MX-DTPA in Cynomolgus Monkeys. [0191]
  • Yttrium-[89]-bearing 2B8-MX-DTPA was prepared using the protocol described for insertion of [0192] 90Y, except that HPLC purification was not used. The non-radioactive, metal-bearing conjugate was formulated in 1× PBS containing 75 mg/mL HSA and 1 mM DTPA and evaluated in GLP study number 920611 at White Sands Research Center. One male and one female monkey were included in each of four groups. The animals were injected intravenously every 48 hours for a total of 7 injections with the following amounts of drug: group 1 (saline); group II (0.003 mg/kg); group III (0.03 mg/kg); and, group IV (0.3 mg/kg). The animals were evaluated during the study by determining body weights and temperatures, food and water consumption, elimination, serum chemistries, hematology, urinalysis, and physical examinations. Animals in groups I through IV were bled prior to infusion on days 0, 2, 7, 10 and 14 and the blood analyzed for circulating B-cell levels by FACS analyses.
  • d. Biodistribution of Radiolabeled 2B8-MX-DTPA [0193]
  • In a preliminary study [0194] 111In-labeled 2B8-MX-DTPA was evaluated for tissue biodistribution in six-to-eight week old BALB/c mice. The radiolabeled conjugate was prepared using clinical-grade 2B8-MX-DTPA following the “mix and shoot” protocol described above. The specific activity of the conjugate was 2.3 mCi/mg and the conjugate was formulated in PBS, pH 7.4 containing 50 mg/mL HSA. Mice were injected intravenously with 100 μL of 111In-labeled 2B8-MX-DTPA (approximately 21 μCi) and groups of three mice were sacrificed by cervical dislocation at 0, 24, 48, and 72 hours. After sacrifice, the tail, heart, lungs, liver, kidney, spleen, muscle, and femur were removed, washed, weighed; a sample of blood was also removed for analysis. Radioactivity associated with each specimen was determined by gamma counting and the percent injected dose per gram tissue subsequently determined. No attempt was made to discount the activity contribution represented by the blood associated with individual organs.
  • In a separate protocol, aliquots of 2B8-MX-DTPA incubated at 40 and 30° C. for 10 weeks were radiolabeled with [0195] 111In to a specific activity of 2.1 mCi/mg for both preparations. These conjugates were then used in biodistribution studies in mice as described above.
  • For dosimetry determinations, 2B8-MX-DTPA was radiolabeled with [0196] 111In to a specific activity of 2.3 mCi/mg and approximately 1.1 μCi was injected into each of 20 BALB/c mice. Subsequently, groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their organs removed and prepared for analysis. In addition, portions of the skin, muscle and bone were removed and processed for analysis; the urine and feces were also collected and analyzed for the 24-72 hour time points.
  • Using a similar approach, 2B8-MX-DTPA was also radiolabeled with [0197] 90Y and its biological distribution evaluated in BALB/c mice over a 72-hour time period. Following purification by HPLC size exclusion chromatography, four groups of five mice each were injected intravenously with approximately 1 μCi of clinically-formulated conjugate (specific activity: 12.2 mCi/mg); groups were subsequently sacrificed at 1, 24, 48 and 72 hours and their organs and tissues analyzed as described above. Radioactivity associated with each tissue specimen was determined by measuring bremstrahlung energy with a gamma scintillation counter. Activity values were subsequently expressed as percent injected dose per gram tissue or percent injected dose per organ. While organs and other tissues were rinsed repeatedly to remove superficial blood, the organs were not perfused. Thus, organ activity values were not discounted for the activity contribution represented by internally associated blood.
  • e. Tumor Localization of [0198] 111In-Labeled 2B8-MX-DTPA.
  • The localization of radiolabeled 2B8-MX-DTPA was determined in athymic mice bearing Ramos B-cell tumors. Six-to-eight week old athymic mice were injected subcutaneously (left-rear flank) with 0.1 mL of RPMI-1640 containing 1.2×10[0199] 7 Ramos tumor cells which had been previously adapted for growth in athymic mice. Tumors arose within two weeks and ranged in weight from 0.07 to 1.1 grams. Mice were injected intravenously with 100 μL of 111In-labeled 2B8-MX-DTPA (16.7 μCi) and groups of three mice were sacrificed by cervical dislocation at 0, 24, 48, and 72 hours. After sacrifice the tail, heart, lungs, liver, kidney, spleen, muscle, femur, and tumor were removed, washed, weighed; a sample of blood was also removed for analysis. Radioactivity associated with each specimen was determined by gamma counting and the percent injected dose per gram tissue determined.
  • 3. Dosimetry Calculations [0200]
  • Using the biodistribution data obtained using BALB/c mice injected with either the [0201] 111In or 90Y-labeled 2B8-MX-DTPA (Tables 1-4 and 5-8), estimates of the radiation dose absorbed from a 1.0 mCi dose administered to a 70 Kg patient were calculated using the approach formalized by Medical Internal Radiation Dose (MIRD) Committee of the Society of Nuclear Medicine. The biological half-lives of the radiolabeled conjugates were determined from the injected dose per organ values determined from the biodistribution data for each radioimmunoconjugate. For some tissues, e.g. blood, it was assumed that the biological decay of the radioconjugate followed a two-compartrnent model with an exponential decay from these compartments. For other tissues, e.g. the liver, whose activity levels remained roughly constant throughout the 72-hour biodistribution study, it was assumed that the biological half-life was very long and assigned a value of 1000 hours.
    TABLE 1
    Distribution of Activity 1.0 Hour Following I.V. Injection
    of 111In-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.47 ± 0.17 40.3 ± 5.32 58.4 ± 3.1 
    Heart 0.087 ± 0.01  5.88 ± 0.76 0.51 ± 0.05
    Lung (2) 0.149 ± 0.01  14.2 ± 1.4  2.10 ± 0.17
    Kidney (1) 0.127 ± 0.02  9.82 ± 0.86 1.22 ± 0.12
    Liver 1.06 ± 0.20 10.32 ± 1.58  10.76 ± 1.93 
    Spleen 0.090 ± 0.01  6.94 ± 1.17 0.61 ± 0.03
    Muscle 8.39 ± 0.98 0.70 ± 0.25 5.67 ± 1.35
    Bone 3.15 ± 0.35 2.97 ± 0.71 9.10 ± 1.09
    Skin 3.15 ± 0.35 0.96 ± 0.29  3.0 ± 1.12
    GI Tract 2.58 ± 0.31 6.10 ± 2.00 7.80 ± 1.80
    Urine
    Feces
    TOTAL 99.04 ± 4.8 
  • [0202]
    TABLE 2
    Distribution of Activity 24 Hours Following I.V. Injection
    of 111In-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.47 ± 0.07 21.97 ± 1.87  32.22 ± 1.35 
    Heart 0.128 ± 0.03  4.02 ± 0.23 0.38 ± 0.01
    Lung (2) 0.152 ± 0.02  7.90 ± 1.61 1.20 ± 0.18
    Kidney (1) 0.128 ± 0.01  5.94 ± 0.40 0.76 ± 0.04
    Liver 1.11 ± 0.10 10.08 ± 1.83  11.20 ± 2.23 
    Spleen 0.082 ± 0.01  5.04 ± 0.75 0.40 ± 0.02
    Muscle 8.41 ± 0.38 1.24 ± 0.05 10.44 ± 0.76 
    Bone 3.15 ± 0.14 2.02 ± 0.33 6.31 ± 0.81
    Skin 3.15 ± 0.14 3.75 ± 0.39 11.77 ± 1.09 
    GI Tract 2.91 ± 0.27 4.50 ± 0.52 6.65 ± 0.56
    Urine 0.98
    Feces 2.54
    TOTAL 87.10 ± 1.68 
  • [0203]
    TABLE 3
    Distribution of Activity 48 Hours Following I.V. Injection
    of 111In-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.45 ± 0.13 22.41 ± 3.95  31.90 ± 2.89 
    Heart 0.090 ± 0.01  4.05 ± 0.94 0.36 ± 0.06
    Lung (2) 0.155 ± 0.02  8.45 ± 0.53 1.31 ± 0.19
    Kidney (1) 0.125 ± 0.01  6.16 ± 1.15 0.76 ± 0.07
    Liver 1.040 ± 0.11  9.41 ± 2.33 9.84 ± 3.18
    Spleen 0.082 ± 0.01  5.32 ± 0.71 0.48 ± 0.11
    Muscle 8.26 ± 0.77 1.42 ± 0.58 11.62 ± 4.67 
    Bone 3.10 ± 0.29 2.08 ± 0.16 6.41 ± 0.44
    Skin 3.10 ± 0.29 3.43 ± 0.59 10.54 ± 1.69 
    GI Tract 2.96 ± 0.20 5.05 ± 0.63 7.46 ± 0.60
    Urine 1.46
    Feces 6.41
    TOTAL 88.49 ± 6.87
  • [0204]
    TABLE 4
    Distribution of Activity 72 Hours Following I.V. Injection
    of 111In-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.52 ± 0.06 18.97 ± 1.31  28.51 ± 2.03 
    Heart 0.094 ± 0.01  3.71 ± 0.31 0.35 ± 0.04
    Lung (2) 0.161 ± 0.01  7.60 ± 0.30 1.18 ± 0.09
    Kidney (1) 0.135 ± 0.01  5.55 ± 0.53 0.76 ± 0.09
    Liver 1.11 ± 0.11 9.90 ± 1.77 11.00 ± 2.03 
    Spleen 0.095 ± 0.01  5.12 ± 0.75 0.48 ± 0.04
    Muscle 8.58 ± 0.34 1.04 ± 0.09 8.95 ± 0.68
    Bone 3.22 ± 0.12 1.73 ± 0.34 6.04 ± 0.51
    Skin 3.22 ± 0.12 3.16 ± 0.60 10.19 ± 2.03 
    GI Tract 2.79 ± 0.19 4.53 ± 0.83 6.37 ± 1.38
    Urine 2.49
    Feces 11.50
    TOTAL 87.80 ± 4.79 
  • [0205]
    TABLE 5
    Distribution of Activity 1.0 Hour Following I.V. Injection
    of 90Y-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.27 ± 0.06 39.23 ± 2.45  49.77 ± 1.72 
    Heart 0.086 ± 0.01  5.80 ± 0.84 0.50 ± 0.09
    Lung (2) 0.137 ± 0.01  12.11 ± 1.08  1.66 ± 0.17
    Kidney (1) 0.120 ± 0.01  10.23 ± 1.30  1.15 ± 0.12
    Liver 0.921 ± 0.05  12.12 ± 1.72  11.17 ± 1.66 
    Spleen 0.080 ± 0.01  9.27 ± 0.46 0.74 ± 0.07
    Muscle 7.27 ± 0.32 0.78 ± 0.13 5.72 ± 1.05
    Bone 2.73 ± 0.12 4.35 ± 0.39 11.89 ± 1.47 
    Skin 2.73 ± 0.12 2.12 ± 0.78 5.82 ± 2.24
    GI Tract 2.22 ± 0.06 3.52 ± 1.12 4.22 ± 0.84
    Urine
    Feces
    TOTAL 94.85 ± 3.47 
  • [0206]
    TABLE 6
    Distribution of Activity at 24 Hours Following I.V. Injection
    of 90Y-2B8-MX-DTA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.517 ± 0.090 8.35 ± 2.547 12.83 ± 4.60 
    Heart 0.092 ± 0.005 2.63 ± 0.142 0.240 ± 0.006
    Lung 0.141 ± 0.005 4.56 ± 0.393 0.644 ± 0.047
    Kidney 0.138 ± 0.007 5.63 ± 0.222 0.779 ± 0.040
    Liver 0.438 ± 0.098 5.22 ± 0.335 2.259 ± 0.399
    Spleen 0.081 ± 0.003 4.23 ± 0.180 0.345 ± 0.011
    Muscle 8.668 ± 0.514 0.976 ± 0.164   8.55 ± 1.945
    Bone 3.246 ± 0.186 1.326 ± 0.102  4.289 ± 0.154
  • [0207]
    TABLE 7
    Distribution of Activity at 48 Hours Following I.V. Injection
    of 90Y-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.33 ± 0.06 17.34 ± 2.0  23.03 ± 1.95 
    Heart 0.088 ± 0.01  3.56 ± 0.31 0.31 ± 0.04
    Lung (2) 0.139 ± 0.01  7.54 ± 0.88 1.05 ± 0.15
    Kidney (1) 0.122 ± 0.01  6.53 ± 0.42 0.79 ± 0.01
    Liver 0.968 ± 0.04  9.05 ± 1.70 8.92 ± 1.57
    Spleen 0.087 ± 0.01  6.52 ± 1.13 0.57 ± 0.07
    Muscle 7.26 ± 0.36 1.05 ± 0.18 8.01 ± 1.17
    Bone 2.86 ± 0.14 3.34 ± 0.42 9.53 ± 1.08
    Skin 2.86 ± 0.14 4.13 ± 0.76 11.75 ± 1.82 
    GiTract 2.84 ± 0.19 2.74 ± 0.34 3.80 ± 0.30
    Urine 4.29
    Feces 7.67
    TOTAL 79.72 ± 3.23 
  • [0208]
    TABLE 8
    Distribution of Activity at 72 Hours Following I.V. Injection
    of 90Y-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.35 ± 0.02 15.40 ± 1.63  20.71 ± 2.13 
    Heart 0.088 ± 0.01  3.12 ± 0.24 0.28 ± 0.01
    Lung (2) 0.142 ± 0.01  8.23 ± 1.05 1.17 ± 0.20
    Kidney (1) 0.123 ± 0.01  6.45 ± 0.57 0.79 ± 0.07
    Liver 0.02 ± 0.06 8.39 ± 1.04 8.58 ± 1.31
    Spleen 0.103 ± 0.01  5.90 ± 1.19 0.59 ± 0.08
    Muscle 7.68 ± 0.11 1.01 ± 0.15 7.73 ± 1.05
    Bone 2.88 ± 0.05 3.20 ± 0.25 9.20 ± 0.61
    Skin 2.88 ± 0.05 3.97 ± 0.49 11.42 ± 1.36 
    GI Tract 2.86 ± 0.18 2.90 ± 0.65 4.06 ± 0.93
    Urine 3.00
    Feces 11.08
    TOTAL 78.62 ± 2.63 
  • In a similar manner the other biological half-life values were assigned or calculated using the standard equation for calculating the t[0209] ½ for an exponential decay. Once these values had been determined, the variables for Tue, Te1, Te2, A1, A2, and A, listed in Tables 9 and 10, were determined for each radiolabeled conjugate using the equations provided at the top of these tables (output variables). These values, as well as those shown in the subsequent tables, were calculated using a program written in the Symphony spreadsheet (Lotus Development Corp.) by Mr. Phillip Hagan, MS, Nuclear Medicine Service, VA Medical Center, La Jolla, Calif. 92161.
    TABLE 9
    INPUT VARIABLES OUTPUT VARIABLES
    A0 = Administered dose Tue = Effective uptake half-time
    Tp = Physical half-life of radionuclide Te1 = Effective disappearance half-time of first component
    Tu = Biologic uptake half-time Te2 = Effective disappearance half-time of second component
    Tb1 = Biological disappearance half-time of first component A1 = Cumulated activity of first component
    Tb2 = Biological disappearance half-time of second component A2 = Cumulated activity of second component
    f1 = Fraction of A0 with biological half-time of Tb1 A = Total cumulated activity
    f2 = Fraction of A0 with biological half-time of Tb2 Tue = Tu * Tp/(Tu + Tp) A1 - 1.44*f1*A0*Te1*(Tue/Tu)
    S = Mean Dose/Unit Cumulated Activity Te1 = Tb1 * Tp/(Tb1 + Tp) A2 - 1.44*f2*A0*Te2
    Te2 = Tb2 * Tp/(Tb2 + Tp) A - A1 + A2
    Example: A1 FOR LIVER - 1.44 * 11.000% * 1000 * 63.2 * 1.00 = 10007.5 microcuries cumulated activity
    TABLE OF INPUT AND OUTPUT VALUES USED TO EVALUATE CUMULATED ACTIVITY (A)
    Tu Tb1 Tb2 Tue Te1 Te2 A1 A2 A
    (hr) f1 f2 (hr) (hr) (hr) (hr) (hr) (uCi-hr) (uCi-hr) (uCi-hr)
    ADRENALS 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    BLAD CONTENTS 2.78E−04 1.000% 0.00% 4 0 2.78E−04 3.8 0.0 54.4 0.0 54
    STOMACH CONTENTS 2.78E−04 6.650% 0.00% 1.5 0 2.78E−04 1.5 0.0 140.5 0.0 141
    SM. INT. CONTENTS 2.78E−04 6.650% 0.00% 3.5 0 2.78E−04 3.3 0.0 318.6 0.0 319
    ULI_CONTENTS 2.78E−04 6.650% 0.00% 4.5 0 2.78E−04 4.2 0.0 404.0 0.0 404
    LLI_CONTENTS 2.78E−04 6.650% 0.00% 4.2 0 2.78E−04 4.0 0.0 378.6 0.0 379
    KIDNEYS 2.78E−04 1.220% 0.00% 35 0 2.78E−04 23.0 0.0 404.8 0.0 405
    LIVER 2.78E−04 11.000% 0.00% 1000 0 2.78E−04 63.2 0.0 10007.5 0.0 10008
    LUNGS 2.78E−04 2.100% 1.20% 30 1000 2.78E−04 20.8 63.2 627.9 1091.7 1720
    OTH TISS (TOTAL) 2.78E−04 0.000%
    MUSCLE 2.78E−04 10.400% 0.00% 1000 0 2.78E−04 63.2 0.0 9461.7 0.0 9462
    ADIPOSE 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    BLOOD 2.78E−04 58.400% 32.22% 15 1000 2.78E−04 12.3 63.2 10319.2 29313.1 39632
    BRAIN 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    HEART 2.78E−04 0.510% 0.38% 57 1000 2.78E−04 30.9 63.2 226.9 345.7 573
    PVARIES 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    PANCREAS 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    SKELETON (TOTAL) 2.78E−04 0.000%
    CORTICAL BONE 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    TRABECULAR BONE 2.78E−04 9.100% 6.30% 45 1000 2.78E−04 27.0 63.2 3536.8 5731.6 9268
    NARROW (RED) 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    MARROW (YELLOW) 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    CARTILAGE 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    OTHER CONSTIT. 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    SKIN 2.78E−04 11.770% 0.00% 1000 0 2.78E−04 63.2 0.0 10708.1 0.0 10708
    SPLEEN 2.78E−04 0.610% 0.40% 39 1000 2.78E−04 24.7 63.2 217.1 363.9 581
    TESTES 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    THYROID 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    TOTAL BODY 2.78E−04 0.000% 0.00% 1000
  • [0210]
    TABLE 10
    INPUT VARIABLES OUTPUT VARIABLES
    A0 = Administered dose Tue = Effective uptake half-time
    Tp = Physical half-life of radionuclide Te1 = Effective disappearance half-time of first component
    Tu = Biologic uptake half-time Te2 = Effective disappearance half-time of second component
    Tb1 = Biological disappearance half-time of first component A1 = Cumulated activity of first component
    Tb2 = Biological disappearance half-time of second component A2 = Cumulated activity of second component
    f1 = Fraction of A0 with biological half-time of Tb1 A = Total cumulated activity
    f2 = Fraction of A0 with biological half-time of Tb2 Tue = Tu * Tp/(Tu + Tp) A1 - 1.44*f1*A0*Te1*(Tue/Tu)
    S = Mean Dose/Unit Cumulated Activity Te1 = Tb1 * Tp/(Tb1 + Tp) A2 - 1.44*f2*A0*Te2
    Te2 = Tb2 * Tp/(Tb2 + Tp) A - A1 + A2
    Example: A1 FOR LIVER - 1.44 * 9.000% * 1000 * 60.2 * 1.00 = 7795.5 microcuries cumulated activity
    TABLE OF INPUT AND OUTPUT VALUES USED TO EVALUATE CUMULATED ACTIVITY (A)
    Tu Tb1 Tb2 Tue Te1 Te2 A1 A2 A
    (hr) f1 f2 (hr) (hr) (hr) (hr) (hr) (uCi-hr) (uCi-hr) (uCi-hr)
    ADRENALS 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 63.2 0.0 0.0 0.0 0
    BLAD CONTENTS 2.78E−04 1.000% 0.00% 4 0 2.78E−04 3.8 0.0 54.2 0.0 54
    STOMACH CONTENTS 2.78E−04 4.220% 0.00% 1.5 0 2.78E−04 1.5 0.0 89.1 0.0 89
    SM. INT. CONTENTS 2.78E−04 4.220% 0.00% 3.5 0 2.78E−04 3.3 0.0 201.7 0.0 202
    ULI_CONTENTS 2.78E−04 4.220% 0.00% 4.5 0 2.78E−04 4.2 0.0 255.5 0.0 255
    LLI_CONTENTS 2.78E−04 4.220% 0.00% 4.2 0 2.78E−04 3.9 0.0 239.5 0.0 240
    KIDNEY 2.78E−04 1.150% 0.87% 70 1000 2.78E−04 33.4 60.2 553.6 753.6 1307
    LIVER 2.78E−04 9.000% 0.00% 1000 0 2.78E−04 60.2 0.0 7795.5 0.0 7795
    LUNGS 2.78E−04 1.200% 0.00% 1000 0 2.78E−04 60.2 0.0 1039.4 0.0 1039
    OTH TISS (TOTAL) 2.78E−04 0.000%
    MUSCLE 2.78E−04 8.720% 0.00% 1000 0 2.78E−04 60.2 0.0 7552.9 0.0 7553
    ADIPOSE 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    BLOOD 2.78E−04 49.770% 25.90% 13 1000 2.78E−04 10.8 60.2 7743.9 22433.7 30178
    BRAIN 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    HEART 2.78E−04 0.500% 0.36% 51 1000 2.78E−04 28.4 60.2 204.4 311.8 516
    OVARIES 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    PANCREAS 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    SKELETON (TOTAL) 2.78E−04 0.000%
    CORTICAL BONE 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    TRABECULAR BONE 2.78E−04 11.890% 9.28% 67 1000 2.78E−04 32.7 60.2 5604.4 8038.0 13642
    MARROW (RED) 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    MARROW (YELLOW) 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    CARTILAGE 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    OTHER CONSTIT. 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    SKIN 2.78E−04 15.600% 0.00% 1000 0 2.78E−04 60.2 0.0 13512.1 0.0 13512
    SPLEEN 2.78E−04 0.740% 0.56% 60 1000 2.78E−04 31.0 60.2 330.0 485.1 815
    TESTES 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    THYROID 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
    TOTAL BODY 2.78E−04 0.000% 0.00% 1000 0 2.78E−04 60.2 0.0 0.0 0.0 0
  • Using the Total Cumulated Activity (A) values from Tables 9 and 10, and the S values provided from MIRD Pamphlet Number 11 (Tables 11 and 12, and 13 and 14), the radiation absorbed dose estimates were determined for each of the radiolabeled conjugates for the listed tissues (Tables 15, 16, 17 and 18). In determining the summary radiation dose estimates for the indium-labeled conjugate provided in Table 19, the self-dose of a given organ was summed with the absorbed dose produced by activity in adjacent organs or tissues. However, in calculating the radiation dose estimate values attributed to the yttrium-labeled conjugate (Table 20), certain of the values are absent for the listed tissues (e.g. adrenals). This is due to the shorter path length of the released 13 particle, relative to the path-length of the emitted g particle, hence providing a negligible activity contribution from adjacent tissues, and to the absence of primary biodistribution data for these tissues. [0211]
    TABLE 11
    S. ABSORBED DOSE PER UNIT CUMULATED ACTIVITY, (RAD/UCI-H)
    INDIUM-[111]  HALF-LIFE  67.44 HOURS
    SOURCE ORGANS
    Intestinal Tract Other
    Target Bladder Stomach Si Uli Lli Tissue
    Organs Adrenals Contents Contents Contents Contents Contents Kidneys Liver Lungs (Muscle)
    ADRENALS 7.4E−03 5.7E−07 7.3E−06 4.4E−06 2.8E−06 1.3E−06 3.4E−05 1.5E−05 7.6E−06 4.8E−06
    BLADDER WALL 3.6E−07 4.5E−04 7.5E−07 8.0E−06 6.4E−06 2.0E−05 9.3E−07 5.2E−07 1.5E−07 5.5E−06
    BONE 5.2E−06 2.3E−06 2.3E−06 3.2E−06 2.9E−06 4.2E−06 3.7E−06 2.9E−06 3.8E−06 3.2E−06
    GI (STOM WALL) 8.8E−06 8.5E−07 3.4E−04 1.1E−05 1.2E−05 5.4E−06 1.0E−05 5.8E−06 5.7E−06 4.3E−06
    GI (SI) 2.5E−06 8.6E−06 7.9E−06 2.1E−04 5.4E−05 3.0E−05 8.6E−06 5.0E−06 6.1E−07 4.8E−06
    GI (ULI WALL) 2.8E−06 6.9E−06 1.1E−05 8.3E−05 3.3E−04 1.4E−05 8.6E−06 7.5E−06 7.4E−07 5.0E−06
    GI (LLI WALL) 7.1E−07 2.2E−05 3.8E−06 2.4E−05 9.5E−06 4.7E−04 2.5E−06 7.3E−07 3.0E−07 5.2E−06
    KIDNEYS 3.7E−05 8.5E−07 1.1E−05 9.2E−06 8.3E−06 2.8E−06 5.2E−04 1.2E−05 2.7E−06 4.4E−06
    LIVER 1.5E−05 6.3E−07 5.9E−06 5.6E−06 7.8E−06 8.4E−07 1.2E−05 1.3E−04 7.7E−06 3.4E−06
    LUNGS 7.6E−06 8.2E−08 5.2E−06 7.5E−07 8.3E−07 2.6E−07 2.5E−06 7.8E−06 1.4E−04 4.2E−06
    MARROW (RED) 9.4E−06 5.3E−06 4.0E−06 1.1E−05 9.1E−06 1.3E−05 9.6E−06 4.1E−06 4.8E−06 5.3E−06
    OTH TISS (MUSC) 4.8E−06 5.5E−06 4.3E−06 4.8E−06 4.5E−06 5.2E−06 4.4E−06 3.4E−06 4.4E−06 7.5E−06
    OVARIES 1.8E−06 2.3E−05 1.3E−06 3.3E−05 3.7E−05 6.4E−05 3.6E−06 1.4E−06 3.6E−07 6.3E−06
    PANCREAS 2.6E−05 8.6E−07 5.7E−05 6.1E−06 7.1E−06 2.1E−06 2.0E−05 1.2E−05 7.7E−06 5.7E−06
    SKIN 1.8E−06 1.7E−06 1.4E−06 1.4E−06 1.4E−06 1.6E−06 1.8E−06 1.6E−06 1.8E−06 2.5E−06
    SPLEEN 2.0E−05 7.6E−07 3.1E−05 4.6E−06 4.2E−06 2.4E−06 2.8E−05 2.8E−06 7.1E−06 4.6E−06
    TESTES 1.4E−07 1.4E−05 1.8E−07 I.0E−06 9.8E−07 5.9E−06 3.4E−07 2.5E−07 3.9E−08 3.6E−06
    THYROID 4.7E−07 1.2E−08 3.5E−07 6.9E−08 7.5E−08 2.7E−08 2.0E−07 6.2E−07 2.6E−06 4.3E−06
    UTERUS (NONGRVD) 5.8E−06 4.9E−05 2.4E−06 2.9E−05 1.5E−05 2.1E−05 3.1E−06 1.2E−06 2.8E−07 7.4E−06
    TOTAL BODY 6.6E−06 6.2E−06 6.1E−06 7.3E−06 6.8E−06 6.9E−06 6.6E−06 6.6E−06 5.9E−06 5.6E−06
  • [0212]
    TABLE 12
    S. ABSORBED DOSE PER UNIT CUMULATED ACTIVITY, (RAD/UCI-H)
    INDIUM-[111]  HALF-LIFE  67.44 HOURS
    SOURCE ORGANS
    Target Skeleton
    Organs Ovaries Pancreas R Marrow Cort Bone TRA Bone Skin Spleen Testes Thyroid Total Body
    ADRENALS 1.1E−06 2.6E−05 7.9E−06 3.9E−06 3.9E−06 2.4E−06 2.0E−05 1.4E−07 4.7E−07 7.0E−06
    BLADDER WALL 2.1E−05 4.7E−07 2.4E−06 1.5E−06 1.5E−06 1.6E−06 4.7E−07 1.5E−05 1.2E−08 6.9E−06
    BONE 3-8E−06 3.6E−06 1.2E−05 3.0E−05 2.6E−05 2.9E−06 2.9E−06 2.4E−06 2.6E−06 6.9E−06
    GI (STOM WALL) 2.4E−06 5.9E−05 3.2E−06 1.7E−06 1.1E−06 1.7E−06 3.0E−05 1.5E−07 1.5E−07 7.1E−06
    GI (SI) 3.8E−05 5.5E−06 7.9E−06 2.3E−06 2.3E−06 1.5E−06 4.2E−06 1.2E−06 4.2E−08 7.5E−06
    GI (ULI WALL) 3.7E−05 6.6E−06 6.4E−06 2.2E−06 2.2E−06 1.4E−06 3.8E−06 1.1E−06 3.3E−08 7.0E−06
    GI (LLI WALL) 4.8E−05 1.7E−06 9.0E−06 3.2E−06 3.2E−06 1.5E−06 1.9E−06 8.3E−06 2.2E−08 6.7E−06
    KIDNEYS 2.9E−06 1.9E−05 6.8E−06 2.7E−06 2.7E−06 2.0E−06 2.8E−05 1.7E−07 1.2E−07 6.6E−06
    LIVER 1.7E−06 1.3E−05 2.9E−06 2.0E−06 2.0E−06 1.7E−06 3.0E−06 1.2E−07 3.5E−07 6.5E−06
    LUNGS 2.2E−07 7.6E−06 3.7E−06 3.0E−06 3.0E−06 1.9E−06 6.9E−06 3.4E−08 2.9E−06 5.9E−06
    MARROW (RED) 1.3E−05 6.8E−06 7.5E−05 1.3E−05 2.6E−05 2.7E−06 4.4E−06 1.9E−06 2.9E−06 7.7E−06
    OTH TISS (MUSC) 6.3E−06 5.7E−06 3.8E−06 3.2E−06 3.2E−06 2.5E−06 4.6E−06 .3.6E−06  4.3E−06 5.6E−06
    OVARIES 1.0E−02 1.0E−06 7.7E−06 2.2E−06 2.2E−06 1.4E−06 1.7E−06 0.0E+00 2.5E−08 7.0E−06
    PANCREAS 1.5E−06 1.6E−03 4.9E−06 3.IE−06 3.1E−06 1.7E−06 6.IE−05 2.IE−07 3.0E−07 7.8E−06
    SKIN 1.4E−06 1.3E−06 2.0E−06 2.3E−06 2.3E−06 3.7E−05 1.5E−06 4.9E−06 2.5E−06 3.7E−06
    SPLEEN 1.6E−06 6.2E−05 2.7E−06 2.0E−06 2.0E−06 1.7E−06 9.1E−04 8.9E−08 3.5E−07 6.8E−06
    TESTES 0.0E+00 2.1E−07 1.0E−06 1.9E−06 1.9E−06 3.4E−06 2.0E−07 3.6E−03 3.3E−09 4.9E−06
    THYROID 2.5E−08 4.5E−07 2.2E−06 2.8E−06 2.8E−06 2.4E−06 3.4E−07 3.3E−09 5.8E−03 5.2E−06
    UTERUS (NONGRVO) 6.5E−05 1.9E−06 6.7E−06 1.8E−06 1.8E−06 1.2E−06 1.2E−06 0.0E+00 2.4E−08 7.8E−06
    TOTAL BODY 7.7E−06 7.5E−06 6.4E−06 5.9E−06 5.9E−06 3.8E−06 6.6E−06 5.6E−06 5.3E−06 5.8E−06
  • [0213]
    TABLE 13
    S. ABSORBED DOSE PER UNIT CUMULATED ACTIVITY, (RAD/UCI-H)
    YTTRIUM-[90]  HALF-LIFE  64 HOURS
    SOURCE ORGANS
    Intestinal Tract
    Target Bladder Stomach SI ULI LLI Other Tissue
    Organs Adrenals Contents Contents Contents Contents Contents Kidneys Liver Lungs (Muscle)
    ADRENALS 1.4E−01 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    BLADDER WALL 0.0 5.0E−03 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    BONE 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    GI (STOM WALL) 0.0 0.0 4.0E−03 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    GI (SI) 0.0 0.0 0.0 2.5E−03 0.0 0.0 0.0 0.0 0.0 0.0
    GI (ULI WALL) 0.0 0.0 0.0 0.0 4.5E−03 0.0 0.0 0.0 0.0 0.0
    GI (LLI WALL) 0.0 0.0 0.0 0.0 0.0 7.4E−03 0.0 0.0 0.0 0.0
    KIDNEYS 0.0 0.0 0.0 0.0 0.0 0.0 6.4E−03 0.0 0.0 0.0
    LIVER 0.0 0.0 0.0 0.0 0.0 0.0 0.0 1.1E−03 0.0 0.0
    LUNGS 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.0E−03 0.0
    MARROW (RED) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    OTH TISS (MUSC) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 7.1E−05
    OVARIES 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    PANCREAS 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    SKIN 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    SPLEEN 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    TESTES 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    THYROID 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    UTERUS (NONGRVD) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0
    TOTAL BODY 2.8E−05 3.2E−06 8.5E−05 2.3E−05 1.4E−05 1.7E−05 2.8E−05 2.8E−05 2.8E−05 2.8E−05
  • [0214]
    TABLE 14
    S. ABSORBED DOSE PER UNIT CUMULATED ACTIVITY, (RAD/UCI-H)
    YTTRIUM-[90]  HALF-LIFE  64 HOURS
    SOURCE ORGANS
    Target Skeleton
    Organs Ovaries Pancreas R Marrow Cort Bone TRA Bone Skin Spleen Testes Thyroid Total Body
    ADRENALS 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    BLADDER WALL 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    BONE 0.0 0.0 1.1E−04 4.0E−04 2.3E−04 0.0 0.0 0.0 0.0 2.8E−05
    GI (STOM WALL) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    GI (SI) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    GI (ULI WALL) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    GI (LLI WALL) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    KIDNEYS 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    LIVER 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    LUNGS 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    MARROW (RED) 0.0 0.0 8.6E−04 3.3E−05 5.7E−04 0.0 0.0 0.0 0.0 2.8E−05
    OTH TISS (MUSC) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    OVARIES 1.8E−01 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    PANCREAS 0.0 2.0E−02 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    SKIN 0.0 0.0 0.0 0.0 0.0 7.6E−04 0.0 0.0 0.0 2.8E−05
    SPLEEN 0.0 0.0 0.0 0.0 0.0 0.0 1.1E−02 0.0 0.0 2.8E−05
    TESTES 0.0 0.0 0.0 0.0 0.0 0.0 0.0 5.7E−02 0.0 2.8E−05
    THYROID 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 9.9E−02 2.8E−05
    UTERUS (NONGRVO) 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 2.8E−05
    TOTAL BODY 2.8E−05 2.8E−05 2.8E−05 2.8E−05 2.8E−05 2.8E−05 2.8E−05 2.8E−05 2.8E−05 2.8E−05
  • [0215]
    TABLE 15
    RADIATION ABSORBED DOSE (RAD = A * S)
    INDIUM-[111]  HALF-LIFE  67.44 HOURS
    SOURCE ORGANS
    Intestinal Tract
    Target Bladder Stomach SI ULI LLI Other
    Organs Adrenals Contents Contents Contents Contents Contents Kidneys Liver Lungs Tissue
    ADRENALS 0.0E+00 3.1E−05 1.0E−03 1.4E−03 1.1E−03 4.9E−04 1.4E−02 1.5E−01 1.3E−02 2.4E−01
    BLADDER WALL 0.0E+00 2.4E−02 1.1E−04 2.5E−03 2.6E−03 7.6E−03 3.8E−04 5.2E−03 2.6E−04 2.7E−01
    GI (STOM WALL) 0.0E+00 4.6E−05 4.8E−02 3.5E−03 2.2E−02 2.0E−03 4.0E−03 5.8E−02 9.8E−03 2.1E−01
    GI (SI) 0.0E+00 4.7E−04 1.1E−03 6.7E−03 2.2E−02 1.1E−02 3.5E−03 5.0E−02 1.0E−03 2.4E−01
    GI (ULI WALL) 0.0E+00 3.8E−04 1.5E−03 2.6E−02 1.3E−01 5.3E−03 3.5E−03 7.5E−02 1.3E−03 2.5E−01
    GI (LLI WALL) 0.0E+00 1.2E−03 5.3E−04 7.6E−03 3.8E−03 1.8E−01 1.0E−03 7.3E−03 5.2E−04 2.6E−01
    KIDNEYS 0.0E+00 4.6E−05 1.5E−03 2.9E−03 3.4E−03 1.1E−03 2.1E−01 1.2E−01 4.6E−03 2.2E−01
    LIVER 0.0E+00 3.4E−05 8.3E−04 1.8E−03 3.2E−03 3.2E−04 4.9E−03 1.3E+00 1.3E−02 1.7E−01
    LUNGS 0.0E+00 4.5E−06 7.3E−04 2.4E−04 3.4E−04 9.8E−05 1.0E−03 7.8E−02 2.4E−01 2.1E−01
    OTHER TISSUES
    MUSCLE 0.0E+00 3.0E−04 6.0E−04 1.5E−03 1.8E−03 2.0E−03 1.8E−03 3.4E−02 7.6E−03 3.7E−01
    ADIPOSE 0.0E+00 3.0E−04 6.0E−04 1.5E−03 1.8E−03 2.0E−03 1.8E−03 3.4E−02 7.6E−03 3.7E−01
    BLOOD 0.0E+00 3.0E−04 6.0E−04 1.5E−03 1.8E−03 2.0E−03 1.8E−03 3.4E−02 7.6E−03 3.7E−01
    BRAIN 0.0E+00 3.0E−04 6.0E−04 1.5E−03 1.8E−03 2.0E−03 1.8E−03 3.4E−02 7.6E−03 3.7E−01
    HEART 0.0E+00 4.1E−05 4.4E−03 1.5E−03 1.7E−03 9.1E−04 1.1E−02 2.8E−02 1.2E−02 3.7E−01
    OVARIES 0.0E+00 1.3E−03 1.8E−04 1.1E−02 1.5E−02 2.4E−02 1.5E−03 1.4E−02 6.2E−04 2.8E−01
    PANCREAS 0.0E+00 4.7E−05 8.0E−03 1.9E−03 2.9E−03 8.0E−04 8.1E−03 1.2E−01 1.3E−02 1.2E−01
    SKELETON
    CORTICAL BONE 0.0E+00 1.3E−04 3.2E−04 1.0E−03 1.2E−03 1.6E−03 1.5E−03 2.9E−02 6.5E−03 1.6E−01
    TRABECULAR BONE 0.0E+00 1.3E−04 3.2E−04 1.0E−03 1.2E−03 1.6E−03 1.5E−03 2.9E−02 6.5E−03 1.6E−01
    MARROW (RED) 0.0E+00 2.9E−04 5.6E−04 3.5E−03 3.7E−03 4.6E−03 3.9E−03 4.1E−02 8.3E−03 2.6E−01
    MARROW (YELLOW) 0.0E+00 2.9E−04 5.6E−04 3.5E−03 3.7E−03 4.9E−03 3.9E−03 4.1E−02 8.3E−03 2.6E−01
    CARTILAGE 0.0E+00 1.3E−04 3.2E−04 1.0E−03 1.2E−03 1.6E−03 1.5E−03 2.9E−02 6.5E−03 1.6E−01
    OTHER CONSTIT. 0.0E+00 1.3E−04 3.2E−04 1.0E−03 1.2E−03 1.6E−03 1.5E−03 2.9E−02 6.5E−03 1.6E−01
    SKIN 0.0E+00 9.2E−05 2.0E−04 4.5E−04 5.7E−04 6.1E−04 7.3E−04 1.6E−02 3.1E−03 1.2E−01
    SPLEEN 0.0E+00 4.1E−05 4.4E−03 1.5E−03 1.7E−03 9.1E−04 1.1E−02 2.8E−02 1.2E−02 2.3E−01
    TESTES 0.0E+00 7.6E−04 2.5E−05 3.2E−04 4.0E−04 2.2E−03 1.4E−04 2.5E−03 6.7E−05 1.8E−01
    THYROID 0.0E+00 6.SE−07 4.9E−05 2.2E−05 3.0E−05 1.0E−05 8.1E−05 6.2E−03 4.5E−03 2.1E−01
    UTERUS (NONGRVD) 0.0E+00 3.4E−04 8.6E−04 2.3E−03 2.7E−03 2.6E−03 2.7E−03 6.6E.02 1.0E−02 3.7E−01
    TOTAL BODY 0.0E+00 2.7E−03 3.4E.04 9.2E−03 6.1E−03 8.0E.03 1.3E−03 1.2E−02 4.8E−04 2.8E−01
  • [0216]
    TABLE 16
    RADIATION ABSORBED DOSE (RAD = A * S)
    INDIUM-[111]  HALF-LIFE  67.44 HOURS
    SOURCE ORGANS
    Target Skeleton
    Organs Ovaries Pancreas R Marrow Cort Bone TRA Bone Skin Spleen Testes Thyroid Total Body
    ADRENALS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.6E−02 2.6E−02 1.2E−02 0.0E+00 0.0E+00 0.0E+00
    BLADDER WALL 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.4E−02 1.7E−02 2.7E−04 0.0E+00 0.0E+00 0.0E+00
    GI (STOM WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.6E−02 1.8E−02 1.7E−02 0.0E+00 0.0E+00 0.0E+00
    GI (SI) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.1E−02 1.6E−02 2.4E−03 0.0E+00 0.0E+00 0.0E+00
    GI (ULI WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.0E−02 1.5E−02 2.2E−03 0.0E+00 0.0E+00 0.0E+00
    GI (LLI WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.0E−02 1.6E−02 1.1E−03 0.0E+00 0.0E+00 0.0E+00
    KIDNEYS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.5E−02 2.1E−02 1.6E−02 0.0E+00 0.0E+00 0.0E+00
    LIVER 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.9E−02 1.8E−02 1.7E−03 0.0E+00 0.0E+00 0.0E+00
    LUNGS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.8E−02 2.0E−02 4.0E−03 0.0E+00 0.0E+00 0.0E+00
    OTHER TISSUES
    MUSCLE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.0E−02 2.7E−02 2.7E−03 0.0E+00 0.0E+00 0.0E+00
    ADIPOSE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.0E−02 2.7E−02 2.7E−03 0.0E+00 0.0E+00 0.0E+00
    BLOOD 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.0E−02 2.7E−02 2.7E−03 0.0E+00 0.0E+00 0.0E+00
    BRAIN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.0E−02 2.7E−02 2.7E−03 0.0E+00 0.0E+00 0.0E+00
    HEART 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.0E−02 2.7E−03 2.7E−03 0.0E+00 0.0E+00 0.0E+00
    OVARIES 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.0E−02 1.5E−02 9.9E−04 0.0E+00 0.0E+00 0.0E+00
    PANCREAS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.9E−02 1.8E−02 3.5E−02 0.0E+00 0.0E+00 0.0E+00
    SKELETON
    CORTICAL BONE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.4E−01 3.1E−02 1.7E−03 0.0E+00 0.0E+00 0.0E+00
    TRABECULAR 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.4E−01 3.1E−02 1.7E−03 0.0E+00 0.0E+00 0.0E+00
    BONE
    MARROW (RED) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.4E−01 2.9E−02 2.6E−03 0.0E+00 0.0E+00 0.0E+00
    MARROW 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.4E−01 2.9E−02 2.6E−03 0.0E+00 0.0E+00 0.0E+00
    (YELLOW)
    CARTILAGE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.4E−01 3.1E−02 1.7E−03 0.0E+00 0.0E+00 0.0E+00
    OTHER CONSTIT. 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.4E−01 3.1E−02 1.7E−03 0.0E+00 0.0E+00 0.0E+00
    SKIN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.1E−02 4.0E−01 8.7E−04 0.0E+00 0.0E+00 0.0E+00
    SPLEEN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.9E−02 1.8E−02 5.3E−01 0.0E+00 0.0E+00 0.0E+00
    TESTES 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.8E−02 3.6E−02 1.2E−04 0.0E+00 0.0E+00 0.0E+00
    THYROID 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.6E−02 2.6E−02 2.0E−04 0.0E+00 0.0E+00 0.0E+00
    UTERUS (NONGRVO) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.7E−02 0.0E+00 7.0E−04 0.0E+00 0.0E+00 0.0E+00
    TOTAL BODY 0.0E+00 0.0E+00 0.0E+00 0.0E+00 5.5E−02 4.1E−02 3.8E−03 0.0E+00 0.0E+00 0.0E+00
  • [0217]
    TABLE 17
    RADIATION ABSORBED DOSE (RAD = A *S)
    YTTRIUM-[90]  HALF-LIFE  64 HOURS
    SOURCE ORGANS
    Intestinal Tract
    Target Bladder Stomach SI ULI LLI Other
    Organs Adrenals Contents Contents Contents Contents Contents Kidneys Liver Lungs Tissue
    ADRENALS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    BLADDER WALL 0.0E+00 2.7E−01 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (STOM WALL) 0.0E+00 0.0E+00 3.6E−01 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (SI) 0.0E+00 0.0E+00 0.0E+00 5.0E−01 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (ULI WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.1E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (LLI WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    KIDNEYS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 8.4E+00 0.0E+00 0.0E+00 0.0E+00
    LIVER 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E−00 0.0E+00 0.0E+00 8.6E+00 0.0E+00 0.0E+00
    LUNGS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.1E+00 0.0E+00
    OTHER TISSUES
    MUSCLE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.7E+00
    ADIPOSE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.7E+00
    BLOOD 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.7E+00
    BRAIN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.7E+00
    HEART 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 2.7E+00
    OVARIES 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    PANCREAS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    SKELETON
    CORTICAL BONE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    TRABECULAR BONE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    MARROW (RED) 0.0E+00 0.0E−00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    MARROW (YELLOW) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    CARTILAGE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    OTHER CONSTIT. 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    SKIN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    SPLEEN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    TESTES 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    THYROID 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    UTERUS (NONGVO) 0.0E+00 1.7E−04 7.6E−03 4.6E−03 3.6E−03 4.1E−03 3.7E−02 2.2E−01 2.9E−02 0.0E+00
    TOTAL BODY 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 1.1E+00
  • [0218]
    TABLE 18
    RADIATION ABSORBED DOSE (RAD = A * S)
    INDIUM-[111]  HALF-LIFE  64.00 HOURS
    SOURCE ORGANS
    Target Skeleton
    Organs Ovaries Pancreas R Marrow Cort Bone TRA Bone Skin Spleen Testes Thyroid Total Body
    ADRENALS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    BLADDER WALL 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (STOM WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (SI) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (ULI WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    GI (LLI WALL) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    KIDNEYS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    LIVER 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    LUNGS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    OTHER TISSUES
    MUSCLE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    ADIPOSE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    BLOOD 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    BRAIN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    HEART 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    OVARIES 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    PANCREAS 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    SKELETON
    CORTICAL BONE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.1E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    TRABECULAR BONE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.1E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    MARROW (RED) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 7.8E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    MARROW (YELLOW) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 7.8E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    CARTILAGE 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.1E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    OTHER CONSTIT. 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.1E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    SKIN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    SPLEEN 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 9.0E+00 0.0E+00 0.0E+00 0.0E+00
    TESTES 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    THYROID 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    UTERUS (NONGRVO) 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00
    TOTAL BODY 0.0E+00 0.0E+00 0.0E+00 0.0E+00 3.8E−01 3.8E+01 2.3E−02 0.0E+00 0.0E+00 0.0E+00
  • [0219]
    TABLE 19
    Radiation Dosimetry Estimates Resulting from the Administration of
    Indium-[111] Labeled 2B8-MX Uniformly Distributed
    in Standard Man(70 Kg) and Based on Animal Distribution Data
    Over 72 Hours after Injection
    1000
    AMOUNT OF ACTIVITY = MICROCIJRIES/PATIENT DOSE
    RADS RADS
    ADRENALS 0.493 OVARIES 0.387
    BLADDER WALL 0.348 PANCREAS 0.362
    STOMACH WALL 0.412 SKELETON
    SMALL INTESTINE 0.434 CORTICAL BONE 0.474
    UL INTEST. WALL 0.533 TRABECULAR BONE 0.474
    LL INTEST. WALL 0.505 MARROW (RED) 0.602
    KIDNEYS 0.625 MARROW (YELLOW) 0.602
    LIVER 1.533 CARTILAGE 0.474
    LUNGS 0.582 OTHER CONSTIT. 0.474
    OTHER TISSUES SKIN 0.564
    MUSCLE SPLEEN 0.854
    ADIPOSE TESTES 0.239
    BLOOD THYROID 0.276
    BRAIN UTERUS (NONGRVD) 0.473
    HEART TOTAL BODY 0.417
  • Calculations Performed Using a Spreadsheet Template in Symphony (Lotus Development Corporation) and Created by [0220]
  • Phillip L. Hagan, MS [0221]
  • Nuclear Medicine Service [0222]
  • VA Hospital [0223]
  • San Diego, Calif. 92161 [0224]
    TABLE 20
    Radiation Dosimetry Estimates Resulting from the
    Administration of Yttrium-[90] Labeled 2B8-MX Uniformly Distributed
    in Standard Man(70 Kg) and Based on Animal
    Distribution Data Over 72 Hours After Injection
    1000
    AMOUNT OF ACTIVITY = MICROCURIES/PATIENT DOSE
    RADS RADS
    ADRENALS 0.000 OVARIES 0.000
    BLADDER WALL 0.271 PANCREAS 0.000
    STOMACH WALL 0.356 SKELETON
    SMALL INTESTINE 0.504 CORTICAL BONE 3.138
    UL INTEST. WALL 1.150 TRABECULAR BONE 3.138
    LL INTEST. WALL 1.772 MARROW (RED) 7.776
    KIDNEYS 8.366 MARROW (YELLOW) 7.776
    LIVER 8.575 CARTILAGE 3.138
    LUNGS 2.079 OTHER CONSTIT. 3.138
    OTHER TISSUES SKIN 10.269
    MUSCLE 2.716 SPLEEN 8.965
    ADIPOSE 2.716 TESTES 0.000
    BLOOD 2.716 THYROID 0.000
    BRAIN 2.716 UTERUS (NONGRYD) 0.304
    HEART 2.176 TOTAL BODY 1.854
  • Calculations Performed Using a Spreadsheet Template in Symphony (Lotus Development Corporation) and Created by [0225]
  • Phillip L. Hagan, MS [0226]
  • Nuclear Medicine Service [0227]
  • VA Hospital [0228]
  • San Diego, Calif. 92161 [0229]
  • II. Results [0230]
  • A. In Vitro Studies With 2B8 and 2B8-MX-DTPA [0231]
  • 1. Production and Characterization of the Anti-CD20 Antibody 2B8 [0232]
  • A total of nine fusions resulted in three hybridomas producing antibodies which effectively competed with radiolabeled Coulter B1 antibody. In each case, the hybridoma was expanded into a 24 well plate. The first two antibodies isolated from [0233] fusions 3 and 4, were isotyped and both identified as IgM. The third antibody, produced in fusion 5 and designated 2B8, was determined to be an IgGl kappa isotype and was selected for continuation studies. Clone 2B8.H11 was expanded and placed in long term storage in liquid nitrogen. Clone 2B8.H11 was subcloned to produce clone 2B8.H11.G3 and again to produce clone 2B8.H11.G3.G9. This clone was expanded for further study and the antibody was purified by protein A affinity chromatography.
  • Competition assays using unlabeled 2B8, B1 and [0234] Leu 16 and radiolabeled Coulter B1 demonstrated that 2B8 was able to inhibit B1 binding to CD20 more effectively than equal concentrations of either B1 or Leu 16 (FIG. 1). Similar results were obtained (data not shown) in a competition study using FITC-conjugated 2B8, native B1 and the irrelevant antibodies UPC-10 and S-003 (IgG 2a and 1 isotypes, respectively).
  • Direct binding to cellular CD20 antigen by 2B8 and B1 antibodies was compared by FACS analysis using CD20-positive SB cells and CD20-negative HSB cells. The results shown in FIG. 2 indicate that for comparable amounts of antibody, more 2B8 than B1 was bound to the SB cells. No significant binding to SB cells was observed with the irrelevant antibodies. Only background fluorescence was observed with any reagent used with HSB cells. These results confirm the specificity of interaction of 2B8 with the CD20 antigen and suggest that 2B8 may have higher affinity for the cell-surface antigen than B1. [0235]
  • To determine the apparent affinity of 2B8, purified antibody was radiolabeled with [0236] 125I and increasing concentrations of the labeled antibody were incubated with antigen-positive SB cells; cell-associated radioactivity was determined following a 1 hour incubation period (FIG. 3). The results suggest that the 2B8 antibody binds to the CD20 antigen with an apparent affinity constant of 4.3×10−9M.
  • Flow cytometry studies with human normal peripheral blood lymphocytes indicated that 2B8 was specific for B-cells and did not react with other types of lymphocytes (e.g. T-cells, monocytes, macrophages). FITC-labeled 2B8 was compared to B1-FITC and Leu 16-FITC using the same population of human lymphocytes. The results shown in Table 21 indicate that 2B8 reacted with approximately 14 percent of the peripheral blood lymphocytes versus approximately 12 percent for [0237] Leu 16 and 11 percent for B1. The lymphocyte population based on another B lymphocyte marker (CD-19) was between 11 and 14 percent. Finally, when human peripheral blood lymphocytes were incubated with 2B8 and either B1 or Leu 16 and then counterstained with the CD19 marker (Becton/Dickinson) the double staining population of B lymphocytes was 9 percent with 2B8, and 10 percent with either B1 or Leu 16. These results confirm the similarity of these reagents.
    TABLE 21
    Comparison of Binding of 2B8 to Human Peripheral Blood
    Lymphocytes with other B- and T-Lymphocyte Specific Reagents
    Antibody Marker
    Percent of CD45 Gated
    Lymphocytes
    A. Single Staining:
    None (autofluorescence) 0
    B1-FITC (Coulter Immunology, (IgG2a,k) 11
    Leu 16-FITC (Becton Dickinson, IgGl,k) 12
    2B8-FITC (EDEC, IgGl,k) 14
    B72.3-FITC (IgGl,k irrelevant control) 4
    anti-CD4-FITC (Coulter Immunology 37
    anti-CD3-FITC (Becton Dickinson) 59
    anti-CD19-RPE (Becton Dickinson) 11
    anti-CD19-FITC (Becton Dickinson) 14
    B. Double Staining:
    B1-FITC/anti CD19-RPE 10
    Leu 16-FITC/anti CD19-RPE 10
    2B8 FITC/anti CD19-RPE 9
    anti-CD19 FITC/anti CD19-RPE 13
    B1-FITC/anti Hu Ig RPE 10
    2B8-FITC/anti Hu Ig RPE 10
    B72.3-FITC/anti Hu Ig RPE 2
    Leucogate Simultest 99
  • Immunoprecipitation of radiolabeled cellular CD20 antigen by either 2B8 or Bi resulted in the precipitation of indistinguishable doublet protein species with molecular weights of approximately 33 and 35 KD (data not shown). [0238]
  • 2. Production and Characterization of 2B8-.MX-DTPA [0239]
  • The 2B8-MX-DTPA conjugate was produced by reacting the antibody with a 4:1 molar excess of isothiocyanatobenzyl-3-methyldiethylene-triaminepentaacetic acid (4). Typically, 1-2 mol of MX-DTPA chelate were introduced per mol of 2B8 antibody. As shown by the results presented in FIG. 4, the 2B8-MX-DTPA conjugate exhibited no apparent loss in immunoreactivity, vis a vis native 2B8, as both the native and conjugated 2B8 antibodies exhibited virtually identical Bi inhibition profiles; the IC50 values for 2B8 and 2B8-MX-DTPA were approximately 3 and 4 μg/mL, respectively. These results were obtained using [0240] 125I-labeled B1 antibody in a whole-cell radioimmunoassay performed using SB cells. Similar results were obtained using 2B8 or 2B8-MX-DTPA as inhibitors of 125I-labeled 2B8 binding to SB cells; both 2B8 and its MX-DTPA conjugate inhibited 125I-2B8 binding to SB cells at concentrations of approximately 3-4 μg/mL (data not shown).
  • To assess the in vitro stability of the native 2B8 antibody and the 2B8-.MX-DTPA conjugate, samples in normal saline or saline containing 10 mM glycine-HCl, pH 6.8, were incubated at 40 and 30° C. for 12 weeks and aliquots were assayed weekly using the following assays: immunoreactivity by whole-cell enzyme immunoassay, SDS-PAGE under reducing and non-reducing conditions, and isoelectric focusing gel electrophoresis. While immunoreactivity assays detected no loss of antigen recognition by antibody samples incubated at either temperature (FIG. 5), the isoelectric focusing range for the antibody (pH 7.30-8.40 at week zero), which was stable at 4° C., did exhibit a decrease of 0.2 pH unit at 30° C. after week six (Table 22). This result may be equivocal, however, as it is at the limit of experimental error for the assay. [0241]
    TABLE 22
    2B8/2B8-MX-DTPA pI SUMMARY
    2B8 2B8 2B8 2B8 2B8-MX 2B8-MX 2B8-MX 2B8-MX
    WEEK 4 SAL 30 SAL 4 GLY 30 GLY 4 SAL 30 SAL 4 GLY 30 GLY
    0 7.46-8.37 7.46-8.37 6.30-8.21 6.30-8.21
    1 7.39-8.24 7.42-8.27 7.46-8.31 7.46-8.24 6.39-8.26 6.39-8.26 6.32-8.24 6.25-8.24
    2 7.38-8.27 7.45-8.34 7.45-8.40 7.45-8.34 6.02-8.40 6.02-8.34 6.02-8.40 5.95-8.27
    3 7.47-8.35 7.33-8.35 7.40-8.29 7.33-8.29  6.0-8.29  6.0-8.29  6.0-8.22  6.0-8.15
    4 7.38-8.24 7.38-8.24 7.38-8.35 7.38-8.28 5.99-8.28 5.99-8.35 5.99-8.35 5.99-8.35
    5 7.29-8.25 7.29-8.25 7.37-8.32 7.37-8.32 5.90-8.32 5.90-8.27 5.90-8.32 5.90-8.27
    6 7.24-8.12 7.20-8.27 7.27-8.27 7.20-8.12 5.85-8.27 5.85-8.27 5.85-8.27 5.85-7.95
    7 7.39-8.32 7.17-8.32 7.35-8.25 7.17-8.47 6.02-8.25 5.95-8.32 5.95-8.32
    8 7.33-8.29 7.26-8.36 7.40-8.36 5.86-8.36 5.86-8.36 5.86-8.36 5.86-8.21
    9 7.49-8.53 7.26-8.45 7.41-8.45 7.34-8.30 5.93-8.45 5.93-8.45 5.93-8.45 5.93-8.23
    10  7.26-8.27 7.19-8.27 7.26-8.27 7.19-8.27 5.95-8.35 5.95-8.35 5.88-8.35 5.95-8.13
    11  7.40-8.27 7.18-8.27 7.40-8.35 7.18-8.13 5.93-8.35 5.93-8.27 5.93-8.27 5.93-8.13
    12  7.26-8.18 7.04-8.18 7.26-8.18 7.19-8.11 5.90-8.26 5.90-8.18 5.90-8.26 5.90-8.18
    # during the stability study. The headings represent: 2B8 4 SAL, 2B8 incubated at 4° C. in saline; 2B8 30 SAL, 2B8 incubated at 30° C. in saline; 2B8 4 GLY, 2B8 incubated at 4° C. in normal saline containing 10 mM glycine; 2B8 30 GLY, 2B8 incubated at 30° C. in normal saline containing 10 mM glycine; 2B8-MX 4 SAL; 2B8-MX-DTPA (conjugate) incubated at 4° C. in saline; 2B8-MX 30 SAL,
    # conjugate incubated at 30° C. in saline; 2B8-MX 4′ GLY, conjugate incubated at 4° C. in normal saline containing 10 mM glycine; and, 2B8-MX 30 GLY, conjugate incubated at 30° C. in normal saline containing 10 mM glycine.
  • Finally, using non-reducing SDS-PAGE, the 30° C. antibody samples exhibited high molecular weight aggregates after week 1 (Table 23). Densitometric analyses of the gels indicated that the aggregates represented between 8 and 17% of the samples (Table 23). However, when these samples were analyzed by reducing SDS-PAGE, no evidence of the high molecular weight species was found, suggesting the formation of covalent antibody aggregates at 30° C. Again, no loss of immunoreactivity was observed. [0242]
    TABLE 23
    In Vitro Stability of 2B8
    Percentage
    Sample High MW Monomer Low MW
    A. Desensitometric Scans of Non-Reducing SDS Gels
    Reference
    0 100.00 0
    12 wk/4° C./saline 0 95.42 4.58
    12 wk/4° C./glycine 0 100.00 0
    12 wk/30° C./saline 7.63 83.34 9.03
    12 wk/30° C./glycine 16.70 72.11 11.18
    B. Desensitometric Scans of Reducing SDS Gels
    Reference
    0 100.00 0
    12 wk/30° C./saline 0 100.00 0
    12 wk/30° C./glycine 0 10.00 0
  • During the course of this stability study, samples of 2B8-MX-DTPA, incubated at both 4° and 30° C. were also tested for radiometal incorporation using 90Y. Samples assayed at [0243] weeks 4, 8, and 12 incorporated >90% of the 90Y, regardless of the incubation temperature.
  • Finally, in a separate study, aliquots of 2B8-MX-DTPA incubated at 40 and 30° C. for 10 weeks were radiolabeled with [0244] 125In and their tissue biodistribution assessed in BALB/c mice. Conjugate from both incubation temperatures produced similar biodistributions (data not shown). Moreover, the results obtained were similar to biodistribution results obtained in BALB/c mice using 125I-labeled conjugate stored at 4° C. (see below).
  • The radiolabeling protocols for both [0245] 111In and 90Y were found to be reproducible. Typically, radioincorporations of >95% for 111In and >90% for 90Y were obtained. Specific activities for 111I- and 90Y-labeled conjugates were routinely in the range of 2-3 and 10-15 mCi/mg antibody, respectively. In initial development of the 111I- and 90Y radiolabeling protocols, uncomplexed radioisotopes were removed from the radiolabeled 2B8-MX-DTPA using HPLC gel permeation chromatography. In later experiments, HPLC purification of the indium-labeled conjugate was eliminated because of the high radioincorporations obtained (>95%) with this isotope.
  • The immunoreactivity of [0246] 111In and 90Y-labeled preparations of 2B8-MX-DTPA were analyzed by the method of Lindmo (3). The 111In labeled 2B8-MX-DTPA was found to be 100% immunoreactive (FIG. 6), and the 90Y-labeled conjugate was determined to be 60% immunoreactive (data not shown).
  • 3. Characterization of [0247] 111I- and 90Y-Labeled 2B8-MX-DTPA
  • Preliminary experiments with the [0248] 90Y-labeled conjugate demonstrated that significant antibody degradation and loss of immunoreactivity occurred at specific activities >10 mCi/mg antibody. Therefore, a formulation was developed to minimize the effects of radiolysis. While a number of low molecular weight free-radical scavengers were evaluated and found to be effective, high concentrations of human serum albumin (HSA) were the most effective in preserving antibody integrity and immunoreactivity (FIGS. 7-9).
  • The [0249] 90Y-labeled antibody was formulated in 1× PBS, pH 7.4 containing 75 mg/mL HSA; diethylenetriaminepentaacetic acid (DTPA) was also added to a final concentration of 1 mM to insure that any 90Y which may be lost from the antibody would be chelated. Degradation of 2B8-MX-DTPA, radiolabeled to a specific activity of 14.6 mCi/mg was evaluated at 0 and 48 hours using SDS-PAGE and autoradiography. FIGS. 8 and 9 show that the radiolabeled antibody exhibited no significant degradation over a period of 48 h when incubated at 4° C. Analysis using instant thin layer chromatography showed that the loss of 90Y was less than 2% during the 48 h incubation (Table 24). The immunoreactivity was also relatively constant at 60% (Table 24).
    TABLE 24
    Stability of Clinically-Formulated 90Y-2B8-MX-DTPA
    Percent Conjugate- Percent
    Time (Hours at 4° C.) Associated Radioactivity Immunoreactivity
    0 97.2 62
    24 96.2 60
    48 96.2 60
    #immunoreactivity remained constant at approximately 60%.
  • Formulation studies were also performed with the [0250] 111In-labeled conjugate; the specific activity was 2.2 mCi/mg. The radiolabeled antibody was evaluated in 1× PBS, pH 7.4 containing 50 mg/mL HSA. FIG. 10 shows photographs of the autoradiograms for time zero and 48 h incubation samples; densitometric analysis of the autoradiograms indicate that there was no degradation of the radiolabeled antibody over the course of the study (FIGS. 11, 12). Instant thin-layer chromatography analysis of the samples demonstrated no loss of 125In (Table 25); moreover, immunoreactivity was maintained at approximately 100% (Table 25).
    TABLE 25
    Stability of Clinically-Formulated 111In-2B8-MX-DTPA
    Percent Conjugate- Percent
    Time (Hours at 4° C.) Associated Radioactivity Immunoreactivity
    0 94.0 105
    24 96.5 104
    48 96.0 100
    # at approximately 100%.
  • When a clinically-formulated preparation of 2B8-MX-DTPA, radiolabeled with 90Y to a specific activity 14.6 mCi/mg, was incubated for 96 hours at 37° C. in human serum and analyzed by non-reducing SDS-PAGE and autoradiography, less than 4% of the radioisotope was lost during the course of the incubation period. Densitometric scans of the autoradiograms at time zero and 96 h indicated no significant degradation of the radiolabeled conjugate (FIGS. [0251] 13-15). These results were corroborated by analytical thin-layer chromatographic analyses of the time zero and 96-hour samples (Table 26). Taken together these results suggest that the yttrium-labeled conjugate is stable under the conditions used in this study. Similar results were obtained with the 111In labeled 2B8-MX-DTPA conjugate (FIGS. 16-18).
    TABLE 26
    Analytical Thin-Layer Chromatographic Analysis of
    90Y-2B8-MX-DTPA Conjugate Incubated in Human
    Serum for 96 Hours at 37° C.
    Percent Conjugate-
    Time (Hours at 37° C.) Associated Radioactivity
    0 95.1
    24 95.2
    48 93.2
    72 92.0
    96 91.4
    # each strip was then determined and the percent conjugate-associated radioactivity calculated. (Free radiometal migrates with the solvent front while protein-associated radioactivity remains at the origin.) The means of each determination of conjugate-associated radioactivity are shown.
  • B. Animal Studies. [0252]
  • 1. High-Dose Pharmacology/Toxicology Studies with 2B8 and 2B8-MX-DTPA [0253]
  • In a GLP study performed at White Sands Research Center (Study Number 920111), cynomolgus monkeys were given intravenous injections of various doses of 2B8. Blood samples were taken before each new injection and the blood was processed for flow cytometric evaluation of the lymphocyte populations (Table 27). [0254]
    TABLE 27
    Primate B cell Populations Determined by Flow Cytometry,
    Following Infusion of Anti-CD20
    Murine Monoclonal Antibody 2B8
    Animal # Dose Day B cellsa,b % Depletion
    Group I 452 saline 0 20.1 0
    1 18.3 9
    7 21.6 0
    13 14.6 27
    38 15.5 23
    52 18.6 7
    424 saline 0 12.4 0
    1 11.6 6
    7 11.2 10
    13 8.4 32
    38 7.7 38
    52 13.1 0
    Group II 540 0.6 mg/kg 0 16.1 0
    1 7.1 54
    7 6.0 63
    13 5.7 65
    38 10.8 33
    52 14.4 11
    804 0.6 mg/kg 0 17.6 0
    1 8.3 53
    7 6.1 65
    13 6.6 62
    38 5.1 71
    52 5.2 68
    Group III 701 2.5 mg/kg 0 21.6 0
    1 10.7 50
    7 3.0 86
    13 10.7 50
    754 2.5 mg/kg 0 19.9 0
    1 11.2 44
    7 10.5 47
    13 9.0 55
    Group IV 782  10 mg/kg 0 15.9 0
    1 3.0 81
    7 3.5 78
    13 6.5 59
    164  10 mg/kg 0 17.7 0
    1 8.4 47
    7 7.9 50
    13 7.7 42
    Group V 705  10 mg/kg 0 17.2 0
    1 5.2 70
    7 1.3 69
    13 8.2 52
    38 17.1 1
    52 13.3 22
    716  10 mg/kg 0 34.7 0
    1 18.6 46
    7 8.1 77
    13 3.5 90
    38 6.9 80
    52 9.2 61
  • No significant pharmacotoxic effects related to the administration of the anti-CD20 antibody 2B8 were noted in any clinical parameter evaluated during or following the study. Similarly, no abnormalities were noted during analysis of the various histopathology specimens obtained from animals in groups III and IV. [0255]
  • The study duration was 14 days and the animals were evaluated during the study in the following categories: clinical observations, body weights, body temperature, food and water intake, fecal elimination, serum chemistries, hematology, urinalysis, and physical examinations. Additionally, the animals in each group were bled on [0256] days 0, 1, 7, and 13 and the blood analyzed for serum antibody (2B8) levels and for T- and B-cell levels. On day 13 the animals in Groups III and IV were sacrificed and selected tissues examined by light microscopy following specimen preparation. The tissues evaluated were: heart, spleen, liver, kidney, lung, cerebral cortex, spinal cord, lymph node, stomach, ileum, colon, skeletal muscle, testis/ovary, pancreas, and bone marrow.
  • When the blood from the treated animals was analyzed for levels of circulating T- and B-cells, animals in groups II through V exhibited >50% loss of circulating B-cells through day 13 (FIG. 19); administration of the antibody had no effect on T-cell levels (data not shown). All groups receiving 2B8 showed saturation of B-cells and excess antibody in the plasma (not shown). The animals in group V, which received a single 10.0 mg/Kg dose of 2B8 also exhibited reduction in circulating B-cell levels equivalent to that observed in animals in the other groups. [0257]
  • The animals in groups I, II, and V were examined through day 52 (FIG. 20). The levels of B-cells returned to >70% of normal by day 38, except for one animal in Group II (PRO804) and one animal in Group V (PRO716). The levels of circulating B-cells in these animals remained at approximately 40% of normal levels after 52 days. [0258]
  • In addition to this study, the pharmacotoxic effects of [0259] 89Y-2B8-MX-DTPA were assessed in cynomolgus monkeys in a GLP study performed at White Sands Research Center (Study No. 920611). Clinical-grade conjugate was loaded with non-radioactive 89Y. The yttrium-bearing conjugate was formulated in PBS pH 6.8, containing 75 mg/mL human serum albumin and 1 mM DTPA (clinical formulation) and administered intravenously as described in the Methods Section.
  • As shown by the results in FIG. 21, the [0260] 89Y-labeled 2B8-MX-DTPA had little, if any, effect on circulating B-cells in these animals, regardless of the dose administered. In addition, other than a general depletion of lymphocytes (20-43%), no significant abnormalities were found in any clinical parameter evaluated, including serum chemistry, urinalysis, body weights and temperatures.
  • 2. Pharmacokinetic Studies with 2B8 and 2B8-MX-DTPA [0261]
  • As described above, the animals in group V of the GLP study received a single dose of 10.0 mg/Kg of 2B8. Linear regression analysis of the data suggest that the native antibody was cleared from the circulation of these monkeys with a 13 t½ value of approximately 4.5 days. In a similar study using BALB/c mice, the β t[0262] ½ values for native and conjugated 2B8 were determined by linear regression analysis (not shown) to be 8.75 days (FIG. 22). These results suggest that conjugation of 2B8 had no effect on its clearance from BALB/c mice.
  • 3. Biodistribution and Tumor Localization Studies with Radiolabeled 2B8-MX-DTPA [0263]
  • Building on the preliminary biodistribution experiment described above (Section 2d), conjugated 2B8 was radiolabeled with [0264] 111In to a specific activity of 2.3 mCi/mg and roughly 1.1 μCi was injected into each of twenty BALB/c mice to determine biodistribution of the radiolabeled material. Subsequently, groups of five mice each were sacrificed at 1, 24, 48 and 72 hours and their organs and a portion of the skin, muscle and bone were removed and processed for analysis. In addition, the urine and feces were collected and analyzed for the 24-72 hour time-points. The level of radioactivity in the blood dropped from 40.3% of the injected dose per gram at 1 hour to 18.9% at 72 hours (Tables 1-4; FIG. 23). Values for the heart, kidney, muscle and spleen remained in the range of 0.7-9.8% throughout the experiment. Levels of radioactivity found in the lungs decreased from 14.2% at 1 hour to 7.6% at 72 hours; similarly the respective liver injected dose per gram values were 10.3% and 9.9%. These data were used in determining radiation absorbed dose estimates 111In-2B8-MX-DTPA (Table 19).
  • The biodistribution of [0265] 90Y-labeled conjugate, having a specific activity of 12.2 mCi/mg antibody, was evaluated in BALB/c mice. Radioincorporations of >90% were obtained and the radiolabeled antibody was purified by HPLC. Tissue deposition of radioactivity was evaluated in the major organs, and the skin, muscle, bone, and urine and feces over 72 hours and expressed as percent injected dose/g tissue. The results shown in Tables 5-8 and FIG. 24 demonstrate that while the levels of radioactivity associated with the blood dropped from approximately 39.2% injected dose per gram at 1 hour to roughly 15.4% after 72 hours; the levels of radioactivity associated with tail, heart, kidney, muscle and spleen remained fairly constant at 10.2% or less throughout the course of the experiment. Importantly, the radioactivity associated with the bone ranged from 4.4% of the injected dose per gram bone at 1 hour to 3.2% at 72 hours. Taken together, these results suggest that little free yttrium was associated with the conjugate and that little free radiometal was released during the course of the study. These data were used in determining radiation absorbed dose estimates for 90Y-2B8-MX-DTPA (Table 20).
  • For tumor localization studies, 2B8-MX-DTPA was prepared and radiolabeled with [0266] 111In to a specific activity of 2.7 mCi/mg. One hundred microliters of labeled conjugate (approximately 24 μCi) were subsequently injected into each of 12 athymic mice bearing Ramos B-cell tumors. Tumors ranged in weight from 0.1 to 1.0 grams. At time points of 0, 24, 48, and 72 hours following injection, 50 μL of blood was removed by retro-orbital puncture, the mice sacrificed by cervical dislocation, and the tail, heart, lungs, liver, kidney, spleen, muscle, femur, and tumor removed. After processing and weighing the tissues, the radioactivity associated with each tissue specimen was determined using a gamma counter and the values expressed as percent injected dose per gram.
  • The results (FIG. 25) demonstrate that the tumor concentrations of the [0267] 111In-2B8-MX-DTPA increased steadily throughout the course of the experiment. Thirteen percent of the injected dose was accumulated in the tumor after 72 hours. The blood levels, by contrast, dropped during the experiment from over 30% at time zero to 13% at 72 hours. All other tissues (except muscle) contained between 1.3 and 6.0% of the injected dose per gram tissue by the end of the experiment; muscle tissue contained approximately 13% of the injected dose per gram.
  • C. Dosimetry [0268]
  • The summary dosimetry data derived from biodistribution studies in normal BALB/c mice and presented in Tables 19 and 20, for the indium- and yttrium-labeled conjugates, respectively, are in agreement with data presented in the literature when compared per millicurie of injected dose (5) and suggest that both the yttrium- and indium-labeled conjugates of 2B8 may be safely evaluated for clinical efficacy in lymphoma patients. [0269]
  • D. Toxicology. [0270]
  • 1. 2B8: Single Dose General Safety Test. [0271]
  • Mice and guinea pigs were administered a single intra peritoneal dose of 2B8 (0.5 mL or 5.0 mL, respectively) and observed for seven days. No overt signs of toxicity were detected. [0272]
  • 2. 2B8 and 2B8-MX-DTPA: Immunohistology Studies with Human Tissues. [0273]
  • The tissue reactivity of murine monoclonal antibody 2B8 was evaluated using a panel of 32 different human tissues fixed with acetone. Antibody 2B8 reacts with the anti-CD20 antigen which had a very restricted pattern of tissue distribution, being observed only in a subset of cells in lymphoid tissues including those of hematopoietic origin. [0274]
  • In the lymph node, immunoreactivity was observed in a population of mature cortical B-lymphocytes as well as proliferating cells in the germinal centers. Positive reactivity was also observed in the peripheral blood, B-cell areas of the tonsils, white pulp of the spleen, and with 40-70% of the medullary lymphocytes found in the thymus. Positive reactivity was also seen in the follicles of the lamina propria (Peyer's Patches) of the large intestines. Finally, aggregates or scattered lymphoid cells in the stroma of various organs, including the bladder, breast, cervix, esophagus, lung, parotid, prostate, small intestine, and stomach, were also positive with antibody 2B8. [0275]
  • All simple epithelial cells, as well as the stratified epithelia and squamous epithelia of different organs, were found to be unreactive. Similarly, no reactivity was seen with neuroectodermal cells, including those in the brain, spinal cord and peripheral nerves. Mesenchymal elements, such as skeletal and smooth muscle cells, fibroblasts, endothelial cells, and polymorphonuclear inflammatory cells were also found to be negative. [0276]
  • The tissue reactivity of the 2B8-MX-DTPA conjugate was evaluated using a panel of sixteen human tissues which had been fixed with acetone. As previously demonstrated with the native antibody, the 2B8-MX-DTPA conjugate recognized the CD20 antigen which exhibited a highly restricted pattern of distribution, being found only on a subset of cells of lymphoid origin. In the lymph node, immunoreactivity was observed in the B-cell population. Strong reactivity was seen in the white pulp of the spleen and in the medullary lymphocytes of the thymus. Immunoreactivity was also observed in scattered lymphocytes in the bladder, heart, large intestines, liver, lung, and uterus, and was attributed to the presence of inflammatory cells present in these tissues. As described with the native antibody (above), no reactivity was observed with neuroectodermal cells or with mesenchymal elements. [0277]
  • III. Discussion [0278]
  • The murine monoclonal anti-CD20 antibody 2B8, produced by a clone with the same designation, exhibits an affinity for the B-cell CD20 antigen which may be higher than that observed for the Bi antibody, as determined by competition with antibodies of known specificity for the CD20 antigen, and by Scatchard analysis. Further, immunoprecipitation data suggest that the antigen precipitated by 2B8 appears to be the same antigen as the one precipitated by B1, as both antibodies precipitated a doublet with relative molecular weights of 33 and 35 KD. Cytofluorographic analysis of the specificity of the 2B8 antibody for peripheral blood lymphocytes demonstrates that the antibody reacts specifically with B-cells and has no demonstrated reactivity with T-cells or other types of lymphocytes. Finally, preliminary stability data suggest that the antibody is stable at 30° C. for 12 weeks with no loss of immunoreactivity. [0279]
  • When the 2B8 antibody was conjugated to methylbenzyl diethylenetriamine-pentaacetic acid (MX-DTPA), virtually no reduction in immunoreactivity, relative to the native antibody, was observed. Further, radiolabeling the conjugate with either [0280] 111In or 90Y produced labeled conjugates with immunoreactivities of 100% and 60%, respectively. Stability studies of 111In or 90Y-labeled conjugates incubated in human serum for 96 hours at 37° C. indicated negligible loss of the radiometal during the course of the study, suggesting that the conjugates will be stable when used clinically.
  • Tumor localization studies in athymic mice using an indium-labeled preparation of 2B8-MX-DTPA demonstrated that increasing amounts of the conjugate bound to the tumor cells during the course of the experiment without unusual accumulations in other tissues. Moreover, dosimetry estimates derived from the biodistribution. Moreover, dosimetry estimates derived from the biodistribution studies are in agreement with data published in the literature. Finally, human tissue cross-reactivity studies with the native and conjugated antibodies indicated that both antibodies recognize an antigen with highly restricted tissue distribution, reacting only with a subset of cells in lymphoid tissues, including those of hematopoietic origin. Taken together, these results suggest that conjugation did not alter the tissue specificity of the antibody, and that the radiolabeled conjugates are stable in vivo and recognize the CD20 antigen present on the surface of tumors produced experimentally in athymic mice. [0281]
  • When 2B8 was used in a high-dose pharmacology/toxicology study, the antibody produced no significant pharmacotoxic effects in any parameter evaluated, either during or following the study. Similarly, no abnormalities were noted during analysis of the various histopathology specimens examined by light microscopy. Surprisingly, all doses of the antibody used produced marked depletion of circulating B-cells. Circulating B-cell levels did, however, return to roughly normal levels once administration of the antibody ceased. In the single-dose group of monkeys (Group V) the native antibody was cleared from the circulation with an apparent βt[0282] ½ value of approximately 4.5 days. Predictably, when this pharmacokinetic study was performed in BALB/c mice the 2B8 antibody was cleared with a βt½ value of 8.75 days. Thus, taken together, these data suggest that the native antibody may also provide some clinical effect when administered as an adjunct to the radiolabeled conjugates.
  • Overall our data indicate that the high affinity 2B8 antibody and its MX-DTPA conjugate exhibit a restricted pattern of human tissue reactivity. Moreover, in primates, the native antibody is non-toxic and produces transient clearance of B-cells; however, once the antibody is cleared from the circulation the B-cell levels return reasonably rapidly. Additionally, the indium- and yttrium-labeled 2B8-MX-DTPA conjugates appeared stable in vitro, exhibiting no loss of radiometal during prolonged incubation in human serum. Finally, radiation dose estimates derived from the biodistribution of [0283] 90Y- or 111In-labeled 2B8-MX-DTPA in BALB/c mice are in agreement, per millicurie of injected dose, with dose estimates derived from human clinical studies using conjugated anti-shared idiotype antibodies radiolabeled with these isotopes.
  • IV. Summary of Pre-clinical Development of “Mix-&-shoot” Radiolabeling Protocol for Preparation of [0284] 90Y-2b8-MX-DTPA
  • A. Introduction [0285]
  • A [0286] 90Y-labeled murine monoclonal anti-CD20 antibody (2B8) has been evaluated in a Phase I clinical trial for the treatment of relapsed B-cell lymphoma. The original protocol used for the preparation of the yttrium-labeled antibody used a high performance liquid chromatographic (HPLC) step for removal of non-protein bound radioisotope prior to formulation and administration to patients. Unfortunately, this process is particularly time consuming, resulting in a longer exposure of the antibody to radioisotope in an unprotected state. This results in increased radiolysis of the antibody with a concomitant decrease in immunoreactivity. Additionally, the laborious aspect of the process makes it difficult to prepare more than one dose per day at the radiopharmacy. Simplification of the process would expedite implementation at the clinical site as an alternative to using NIPI Pharmacy Services as a radiopharmacy.
  • Accordingly, a revised radiolabeling procedure was developed, referred to as the “mix-and-shoot” method, which obviates the need for HPLC purification while maintaining a high radioincorporation and improved retention of immunoreactivity. In vitro stability studies as well as biodistribution studies in mice showed that radiolabeled antibody prepared using the “mix-and-shoot” method is comparable to material produced using the current HPLC process. The results of these pre-clinical studies indicate that this new “mix-&-shoot” protocol can be used to prepare [0287] 90Y-labeled 2B8-MX-DTPA suitable for use in clinical trials.
  • B. Materials and Methods [0288]
  • Materials
  • 1. Cells [0289]
  • The human lymphoblastic cell lines SB (CD20 positive) and HSB (CD20 negative; ) were obtained from the American Type Culture Collection and maintained in RPMI-1640 containing 10% fetal bovine serum and supplemented with glutamine. [0290]
  • 2. Antibodies [0291]
  • The 2B8 antibody was purified by the Manufacturing department from hollow-fiber bioreactor supernatant using protocols previously described in the IND (BB-IND 4850/4851). [0292]
  • 3. Additional Reagents [0293]
  • Yttrium-[90] chloride was obtained from Amersham. All other reagents were obtained from sources described in the appended reports cited below. Reagents used for radiolabeling protocols were processed to remove contaminating heavy metal ions which could compete with-the radioisotopes during the radiolabeling step (see Methods section). Reagents were made under GMP conditions by IDEC's—Manufacturing department following established Batch Production Records. [0294]
  • Methods [0295]
  • 1. Preparation of 2B8-D4X-DTPA [0296]
  • Clinical-grade MX-DTPA was obtained from Coulter Immunology as the disodium salt in water and stored at −70° C. Conjugate (2B8-MX-DTPA) was prepared by the Manufacturing department. Two different lots of conjugate were used in these studies; both were provided in normal saline at 10 mg/mL. The conjugates were filled in sterile 2 mL polypropylene syringes and stored at 2-8° C. [0297]
  • 2. Maintenance of Metal-free Conditions [0298]
  • All manipulations of reagents were performed to minimize the possibility of metal contamination. Polypropylene or polystyrene plastic containers such as flasks, beakers and graduated cylinders were used. These were washed with Alconox and exhaustively rinsed with Milli-Q water or Water for Irrigation (WFIr) before use. Metal-free pipette tips (BioRad) were used for accurately manipulating small volumes. Larger volumes of reagents were manipulated using sterile, plastic serological pipettes. Reactions were conveniently performed in 1.8 mL screw-top microfuge tubes made from polypropylene. [0299]
  • 3. Determination of Radioincorporation Radioincorporation was determined using instant thin-layer chromatography (ITLC) in triplicate according to SOP SP-13-008. In general, the protocol was as follows: radiolabeled conjugate was diluted 1:20 in 1× PBS containing 1 mM DTPA or 5 mM EDTA, then 1μ-L spotted 1.5 cm from one end of a 1×5 cm strip of ITLC SG paper (Gehnan Sciences). The paper was developed using 10% ammonium acetate in methanol:water (1: 1;v/v). The strips were dried, cut in half crosswise, and the radioactivity associated with each section determined by scintillation counting. The radioactivity associated with the bottom half of the strip (protein-associated radioactivity) was expressed as a percentage of the total radioactivity determined by summing the values for both top and bottom halves. [0300]
  • 4. “Mix and-Shoot” Protocol for Yttrium-[90]-Labeled 2B8-MX-DTPA [0301]
  • Antibodies were radiolabeled with carrier-free [0302] 90Y chloride provided by Amersham in 0.04 M HCl. An aliquot of radioisotope (10-20 mCi/mg antibody) was transferred to a polypropylene tube and 0.02× volume of metalfree 2 M sodium acetate was added to adjust the solution to pH 3.6. 2B8-NaDTPA (0.3 mg; 10.0 mg/mL in normal saline) was added immediately and the solution gently mixed. The solution was checked with pH paper to verify a pH of 3.8-4.1 and incubated for 5 min. The reaction was quenched by transferring the reaction mixture to a separate polypropylene tube containing 1× PBS with 75 mg/mL human serum albumin (HSA) and 1 mM diethylenetriaminepentaacetic acid (DTPA) and gently mixed. The radiolabeled antibody was stored at 2-8° C.
  • Specific activities were determined by measuring the radioactivity of an appropriate aliquot of the radiolabeled conjugate. This value was corrected for the counter efficiency, related to the protein concentration of the conjugate, determined by absorbance at 280 nm and expressed as mCi/mg proteins. [0303]
  • 5. In Vitro Immunoreactivity of Yttrium-[90]-2B8-MX-DTPA Immunoreactivity of [0304] 90Y-labeled conjugate was determined using SOP #SP13-009 based on a modified version of the whole-cell binding assay described by Lindmo. Increasing concentrations of log phase, CD20-positive SB cells or CD20-negative HSB cells were added to duplicate sets of 1.5 mL polypropylene tubes; final volume of cells, 0.40 mL. The radiolabeled conjugate was diluted to a final antibody concentration of 1-2.5 ng/nL and 0.35 mL was added to each tube. Following a 90 min incubation, the cells were pelleted by centrifugation and the supernatants collected. Radioactivity remaining in the supernatant fraction was determined with a scintillation counter. Data were plotted as the quotient of the total radioactivity added divided by the cell-associated radioactivity versus the inverse of the cell number per tube. The y axis intercept represents the immunoreactive fraction.
  • 6. In Vitro Stability of Clinically-Formulated Yttrium-F90]-2B8-MX-DTPA [0305]
  • The 2B8-MX-DTPA conjugate was radiolabeled with [0306] 90Y and formulated as described in the “mix & shoot” protocol provided above. Two lots of radiolabeled conjugate were prepared; one lot was used for assessing radioincorporation stability and the other lot used to assess retention of immunoreactivity. The formulated conjugates were incubated at 4° C. for 48 hours and aliquots analyzed at time 0, 24 h and 48 hours using non-reducing SDS-PAGE and autoradiography. Immunoreactivity at each time point was assessed using the assay described above.
  • 7. In Vitro Stability of Yttrium-[90]-2B8-NTX-DTPA in Human Serum [0307]
  • The stability of [0308] 90Y-labeled 2B8-MX-DTPA was assessed by incubation in human serum at 37° C. for up to 72 hours. The conjugated antibody was radiolabeled with yttrium-[90] and formulated as described above. The radiolabeled conjugate was diluted 1:10 with normal human serum (non-heatinactivated) and aliquots incubated in plastic tubes at 37° C. At selected times, samples were removed and analyzed by non-reducing SDS-PAGE and autoradiography.
  • 8. Biodistribution of Yttrium-[90]-2B8-MX-DTPA [0309]
  • Yttrium-[90]-labeled 2B8-MX-DTPA was evaluated for tissue biodistribution in eight to ten week old BALB/c mice. The radiolabeled conjugate was prepared and formulated as described above. Mice were injected intravenously with 5 μCi of [0310] 90Y-labeled 2B8-MX-DTPA and groups of five mice were sacrificed at 1, 24, 48, and 72 hours. After sacrifice, the tail, heart, lungs, liver, kidney, spleen, muscle, femur were removed, washed, weighed; a sample of blood and skin were also removed for analysis. Radioactivity associated with each tissue sample was determined by measuring bremstrahlung radiation using a gamma counter and the percent injected dose per gram tissue and percent injected dose per organ determined.
  • 9. Dosimetry [0311]
  • Biodistribution data obtained using mice injected with [0312] 90Y-labeled 2B8-MX-DTPA were used to calculate estimates of the radiation doses absorbed from a 1.0 mCi dose administered to a 70 Kg patient. Estimates were made according to methods adopted by the Medical Internal Radiation Dose (MIRD) Committee of the Society of Nuclear Medicine. These calculations were performed Mr. Phillip Hagan, Nuclear Medicine Service, VA Medical Center, La Jolla, Calif. 92161. 10. Validation of Protocol for Preparation of Clinical Doses of Yttrium-[90]-2B8-MX-DTPA
  • (Reference R&D report titled “Validation of “Mix-and-Shoot” Radiolabeling Protocol for the Preparation of Clinical Doses of [0313] 90Y-2B8-MX-DTPA; author, P. Chinn; dated Apr. 22, 1994).
  • C. Results [0314]
  • 1. Preparation of Yttrium-[90]-Labeled 2B8-MX-DTPA Using “Mix-&-Shoot” Protocol [0315]
  • Preliminary experiments evaluating the kinetics of the radiolabeling reaction with 2B8-MX-DTPA and 90Y showed that at pH 3.6-4.0, 95% of the radioisotope was incorporated for a reaction time of 5 to 10 min. The reproducibility of this radioincorporation (95.7%±1.7%) was subsequently confirmed in a validation study for the scale-up protocol (Reference R&D report titled “Validation of “Mix-and-Shoot” Radiolabeling Protocol for the Preparation of Clinical Doses of [0316] 90Y-2B8-MX-DTPA; author, P. Chinn; dated Apr. 22, 1994). The preparation of 90Y-labeled 2B8-MX-DTPA using this “mix-&-shoot” protocol gave a product comparable to that produced with the, HPLC method (see BB-IND 4850/4851). The radiolabeling protocol was found to be reproducible with specific activities typically ranging from 10 to 15 mCi/mg antibody.
  • The immunoreactivity of the [0317] 90Y-labeled 2B8-MX-DTPA prepared using this protocol was typically greater than 70%, compared with the 55-60% observed for the validations runs for the HPLC protocol (FIG. 26). This difference is probably due to the reduced effects of radiolysis because of the reduced incubation time with the “mix-and-shoot” protocol. This result was typical, and, as discussed below, was representative of the validation runs for the scale-up protocol for preparing clinical doses of the radiolabeled conjugate.
  • 2. In vitro Stability of [0318] 90Y-Labeled 2B8-MX-DTPA
  • Preliminary experiments with unprotected [0319] 90Y-labeled antibody conjugate prepared using the HPLC process demonstrated that radiolysis caused significant antibody degradation and loss of immunoreactivity. Therefore, a formulation buffer was developed to minimize the effects of radiolysis. Human serum albumin (HSA) was shown to be effective in minimizing. antibody degradation due to radiolysis. An evaluation was made with the radiolabeled conjugate prepared with the “mix-&-shoot” method to confirm the efficacy of the formulation in minimizing radiolysis. The 90Y-labeled antibody, radiolabeled to a specific activity of 14.5 mCi/mg antibody, was formulated in 1× PBS, pH 7.4 containing 75 mg/mL HSA and 1 mM DTPA. Degradation of the conjugate 2B8-MX-DTPA was evaluated at 0, 24, and 48 hours using SDS-PAGE and autoradiography. FIGS. 2, 3, and 4 show that the radiolabeled conjugate exhibited no significant degradation over a period of 48 h when incubated at 4° C. Instant thin-layer chromatography analysis showed no loss of 90Y during the 48 h incubation; these results were corroborated by SDS-PAGE/autoradiographic analysis (Table 28). The immunoreactivity also was relatively constant at >88% (Table 29).
    TABLE 28
    Stability of “Mix-&-Shoot” 90Y-2B8-MX-DTPA
    in PBS Containing Human Serum Albumin and DTPA
    Percent of Conjugate-
    Associated Radioactivity
    Time (h) ITLC SDS/PAGE
    0 92.9 96.0
    24 95.5 95.4
    48 91.3 94.6
  • [0320]
    TABLE 29
    Immunoreactivity of “Mix-&-Shoot” 90Y-2B8-MX-DTPA
    in PBS Containing Human Serum Albumin and DTPA
    Percent
    Time (Hours at 4° C.) Immunoreactivity
    0 87.9
    24 88.5
    48 90.4
  • A clinically-formulated [0321] 90Y-labeled 2B8-MX-DTPA at a specific activity 15.7 mCi/mg was incubated for 72 hours at 37° C. in human serum. Samples analyzed by non-reducing SDS-PAGE and autoradiography (FIG. 30) showed no loss of radioisotope during the course of the incubation period (Table 30). Densitometric scans of the autoradiograms at time zero and 72 h indicated no significant degradation of the radiolabeled conjugate (FIGS. 31 and 32). These results were corroborated by thin-layer chromatographic analyses (Table 30). It should be noted that the radioincorporation for the antibody used in this study was lower than that obtained in the validation studies of the labeling protocol. This lower radioincorporation was due to the reduced quality of the batch of 90Y chloride used for this particular preparation of radiolabeled antibody. The lower radioincorporation did not alter the conclusion that the yttrium-labeled conjugate prepared with the “mix-and-shoot” method is stable under these incubation conditions.
    TABLE 30
    Stability of 90Y-2B8-MX-DTPA Conjugate Incubated in Human Serum
    Percent Conjugate-
    Associated Radioactivity
    Time (hours at 37° C.) ITLC SDS-PAGE/Autoradiography
    0 85.7 88.8
    24 76.4 90.0
    72 87.6 88.7
  • Human serum samples containing [[0322] 90] Y-2B8-MX-DTPA (specific activity 15.7 mCi/mg) were analyzed for non-protein bound 90Y at the times shown using instant thin-layer chromatography strips and SDS-PAGE/autoradiography.
  • 3. Biodistribution Studies with Yttrium-[90] 2B8-MX-DTPA [0323]
  • The biodistribution of the [0324] 90Y-labeled conjugate, with a specific activity of 11.5 mCi/mg antibody and a radioincorporation of >95%, was evaluated in BALB/c mice. Deposition of radioactivity in tissues was evaluated for major organs, skin, muscle, bone, urine and feces over 72 hours and expressed as percent injected dose per g tissue and as percent injected dose per organ. The results shown in Tables 31-34 and FIG. 33 show that the levels of radioactivity associated with the blood decreased from approximately 43% injected dose per gram (%ID/g) at 1 hour to approximately 16% after 72 hours; at 24 h and later, the levels of radioactivity associated with heart, kidney, and spleen remained fairly constant at 4-8%. For lung and liver, radioactivity decreased from 10-12% at 1 h to 8%-10% at 72 h. For the skin, radioactivity was relatively constant at approximately 3% from 24 h through 72 h. The radioactivity in the gastrointestinal tract was constant at 0.5-1% from 24 h to 72 h. Radioactivity for muscle remained approximately 0.6% throughout the course of the study. The uptake of radioactivity by femur (bone) remained less than 4% at all time points indicating that the amount free yttrium in the conjugate preparation was negligible and that little free radiometal was released during the course of the study.
    TABLE 31
    Distribution of Activity 1.0 Hour Following I.V. Injection
    of 90Y-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood  1.37 ± 0.053 42.74 ± 0.78  58.52 ± 1.74 
    Heart 0.101 ± 0.01  8.03 ± 3.33 0.82 ± 0.37
    Lung (2) 0.126 ± 0.01  12.44 ± 0.94  1.56 ± 0.05
    Kidney (1) 0.129 ± 0.01  7.81 ± 1.24 0.997 ± 0.10 
    Liver 0.899 ± 0.07  10.08 ± 1.28  9.01 ± 0.52
    Spleen 0.077 ± 0.004 10.74 ± 0.96  0.823 ± 0.04 
    Muscle 7.83 ± 0.28 0.44 ± 0.08 3.43 ± 0.51
    Bone 2.94 ± 0.11 3.44 ± 0.57 10.11 ± 1.80 
    Skin 2.94 ± 0.11 1.46 ± 0.58 4.24 ± 1.57
    GI Tract 2.33 ± 0.08 1.02 ± 0.19 2.36 ± 0.35
    Urine
    Feces
    TOTAL 94.66 ± 3.47 
  • [0325]
    TABLE 32
    Distribution of Activity at 24 Hours Following I.V. Injection
    of 90Y-2B8-MX-DTA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.55 ± 0.12 19.77 ± 2.42  30.77 ± 6.04 
    Heart 0.105 ± 0.01  4.44 ± 0.55 0.47 ± 0.08
    Lung (2) 0.127 ± 0.02  8.78 ± 1.61 1.11 ± 0.21
    Kidney (1) 0.139 ± 0.01  5.02 ± 0.52 0.69 ± 0.05
    Liver 0.966 ± 0.09  8.62 ± 2.73 8.20 ± 1.97
    Spleen 0.083 ± 0.01  6.75 ± 1.27  0.55 ± 0.064
    Muscle 8.83 ± 0.69 0.692 ± 0.01  6.12 ± 0.52
    Bone 3.31 ± 0.26 2.24 ± 0.31 7.47 ± 1.53
    Skin 3.31 ± 0.26 3.33 ± 0.76 10.88 ± 1.76 
    GI Tract 2.89 ± 0.43 0.73 ± 0.09 1.02 ± 0.05
    Urine 2.31
    Feces 1.23
    Total: 73.52 ± 6.18%
  • [0326]
    TABLE 33
    Distribution of Activity at 48 Hours Following I.V. Injection
    of 90Y-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.50 ± 0.14 14.97 ± 5.77  22.53 ± 8.48 
    Heart 0.104 ± 0.01  3.99 ± 1.43 0.415 ± 0.16 
    Lung (2) 0.122 ± 0.02  8.41 ± 1.57 1.04 ± 0.31
    Kidney (1) 0.124 ± 0.01  3.99 ± 1.62 0.49 ± 0.19
    Liver 0.966 ± 0.13  6.12 ± 3.21 5.69 ± 2.25
    Spleen 0.079 ± 0.01  6.05 ± 2.38 0.46 ± 0.16
    Muscle 8.59 ± 0.82 0.54 ± 0.19 4.67 ± 1.67
    Bone 3.22 ± 0.31 2.07 ± 0.84 6.65 ± 2.56
    Skin 3.22 ± 0.31 2.30 ± 0.70 7.34 ± 1.95
    GI Tract 2.63 ± 0.40 0.652 ± 0.30  1.67 ± 0.64
    Urine 2.83
    Feces 2.06
    TOTAL 57.28 ± 17.60
  • [0327]
    TABLE 34
    Distribution of Activity at 72 Hours Following I.V. Injection
    of 90Y-2B8-MX-DTPA Into Normal BALB/c Mice
    Mean Values ± SD
    Organ Weight % ID/ % ID per
    Sample Gram Gram Organ
    Blood 1.45 ± 0.07 15.87 ± 4.81  23.14 ± 7.26 
    Heart 0.093 ± 0.01  4.16 ± 1.27 0.392 ± 0.13 
    Lung (2) 0.123 ± 0.02  10.67 ± 3.79  1.30 ± 0.45
    Kidney (1) 0.123 ± 0.01  4.79 ± 1.03 0.596 ± 0.16 
    Liver 0.876 ± 0.07  7.26 ± 1.79 6.39 ± 1.76
    Spleen 0.081 ± 0.01  7.37 ± 2.34 0.584 ± 0.16 
    Muscle 8.30 ± 0.39 0.67 ± 0.13 5.58 ± 1.22
    Bone 3.11 ± 0.15 2.58 ± 0.51 8.05 ± 1.76
    Skin 3.11 ± 0.15 3.09 ± 0.82 9.66 ± 2.68
    GI Tract 2.59 ± 0.20 0.79 ± 0.18 2.05 ± 0.53
    Urine 3.56
    Feces 2.82
    TOTAL 65.47 ± 14.0 
  • 4. Dosimetry [0328]
  • The radiation absorbed doses for a “standard” 70 Kg human calculated for the [0329] 90Y-labeled conjugate using the mouse biodistribution data (%ID/organ values in Tables 31-34) are presented in Table 35. These results are comparable to results obtained previously using 90Y-labeled 2B8-MX-DTPA prepared using the HPLC radiolabeling method.
    TABLE 35
    Radiation Dosimetry Estimates Resulting from the
    Administration of Yttrium-[90] Labeled 2B8-MX Uniformly Distributed
    in Standard Man(70 Kg) and Based on Animal
    Distribution Data Over 72 Hours After Injection
    1000
    AMOUNT OF ACTIVITY = MICROCURIES/PATIENT DOSE
    RADS RADS
    ADRENALS 0.534 OVARIES 0.534
    BLADDER WALL 0.534 PANCREAS 0.534
    STOMACH WALL 0.534 SKELETON
    SMALL INTESTINE 1.158 CORTICAL BONE 1.466
    UL INTEST. WALL 1.657 TRABECULAR BONE 1.466
    LL INTEST. WALL 2.380 MARROW (RED) 4.452
    KIDNEYS 7.015 MARROW (YELLOW) 2.096
    LIVER 7.149 CARTILAGE 1.466
    LUNGS 2.157 OTHER CONSTIT. 1.466
    OTHER TISSUES SKIN 6.603
    MUSCLE 2.646 SPLEEN 4.973
    ADIPOSE 2.646 TESTES 0.534
    BLOOD 2.646 THYROID 0.534
    BRAIN 2.112 UTERUS (NONGRVD) 0.767
    HEART 2.646 TOTAL BODY 1.755
  • Ref: A Schema for Absorbed-dose Calculation for Biologically Distributed Radionuclides, MIRD J. of Nucl. Med./[0330] Suppl. # 1, 2/68 Calculations Performed Using a Spreadsheet Template in Symphony (Lotus Development Corporation) and Created by Phillip L. Hagan, MS, Nuclear Medicine Service, VA Hospital, San Diego, Calif. 92161
  • 5. Validation of Protocol for Preparation of Clinical Doses of Yttrium-[90]-2B8-MX-DTPA [0331]
  • A total of ten validation lots were prepared at MPI Pharmacy Services, Inc. The results of testing on each lot are summarized in Table 36. The mean value for each test result was calculated and standard deviations noted where appropriate. To evaluate the variability of the process due to different labeling times, [0332] lot #1 through #8 were prepared using a 10 min labeling time; lot #9 and #10 were prepared using a reaction time of 5 min. Based on the test results for the ten validation lots, release specifications were established. Release specifications are summarized in Table 37.
    TABLE 36
    Assay Results for the Ten Validation Lots of 90Y-Labeled
    2B8-MX-DTPA Prepared Using “Mix-&-Shoot”
    #1 #2 #3 #4 #5 #6 #7 #8 #9 #10 mean
    % imunoreactivity 72.8 93.3 71.7 70.2 60.6 68.2 79.5 72.4 88.2 68.5 74.5 ± 9.8 
    endotoxin (Eu/ml) <0.12 <0.12 <0.12 <0.12 <0.12 <0.25 <0.25 <0.25 <0.12 <0.12 <0.162 ± 0.06   
    5 5 5 5 5 5 5
    % radioincorporation 97.5 97.0 93.5 96.0 94.7 94.9 95.9 96.5 97.5 93.5 95.7 ± 1.4 
    antibody conc. (mg/ml) 0.122 0.102 0.088 0.128 0.134 0.119 0.093 0.088 0.111 0.096 0.108 ± 0.017
    radioactivity (mCi/ml) 1.22 1.22 0.98 1.26 1.51 1.55 1.06 0.98 1.28 1.02 1.21 ± 0.21
    specific act. (mCi/mg) 10.0 12.0 11.2 9.8 11.3 13.0 11.3 11.1 11.5 10.7 11.2 ± 0.9 
    vial radioactivity (mCi) 8.16 8.68 8.26 8.52 8.43 8.45 8.56 8.45 8.45 8.45 8.44 ± 0.15
    protein conc. (mg/ml) 76.2 76.1 73.6 73.1 72.4 76.1 76.4 74.3 71.2 74.8 74.4 ± 1.8 
    pH 6.4 6.4 6.4 6.4 6.4 6.4 6.4 6.4 6.4 6.4 6.4 ± 0.0
    sterility pass pass pass pass pass pass pass pass pass pass
  • [0333]
    TABLE 37
    Release Specifications for 90Y-Labeled 2B8-MX-DTPA
    Prepared Using “Mix-&-Shoot” Protocol
    Test Specification Method
    Immunoreactivity ≧60% RIA
    Endotoxin <5 EU/ml LAL
    Radiolabel incorporation ≧90% ITLC
    Antibody conc. 0.075-0.150 mg/ml A280
    Radioact. conc.1 ≧6.0 mCi/ml dose calibration
    Specific activity1 ≧9.0 mCi/mg antibody A280/dose calib.
    Total vial radioact.1 ≧6.0 mCi dose calibration
    pH 6.0-8.0 pH paper
    total protein conc. 65-85 mg/ml A280
    sterility testing passes CFR 610.12
  • D. Discussion [0334]
  • The original radiolabeling protocol for preparing [0335] 90Y-labeled 2B8-MX-DTPA utilized a particularly laborious and time-consuming HPLC purification step for removing non-protein bound 90Y from the preparation. In order to simplify this process and make it more amenable to use at the clinical site, efforts were directed at eliminating the HPLC step in favor of what has been termed a “mix-and-shoot” protocol. The goal was to identify radiolabeling conditions which would result in a very high radioincorporation of isotope into the conjugate, thereby obviating the need for the purification step. It was discovered that >95% radioincorporation could be-obtained at pH 3.6 with a five to ten minute incubation. An additional benefit of this protocol was increased retention of immunoreactivity (>70%), presumably due to the shorter exposure time of the antibody to the high energy radioisotope before the addition of human serum albumin which provides protection against radiolysis. This retention of immunoreactivity is superior to that previously observed using the HPLC method.
  • Stability studies with [0336] 90Y-labeled conjugate prepared using the “mix-and-shoot” protocol incubated in formulation buffer (1× PBS containing 75 mg/mL human serum albumin and 1 mM DTPA) for up to 48 h at 4° C. showed no loss of radioisotope and complete retention of immunoreactivity. Stability studies conducted with human serum for 72 hours at 37° C. also indicated minimal radioisotope loss. These stability results are comparable to those previously seen with conjugate radiolabeled using the HPLC protocol.
  • Biodistribution in BALB/c mice using [0337] 90Y-labeled conjugate prepared with the “mix-and-shoot” method indicated no unusual tissue deposition. These results suggested that the radiolabeled antibody was not altered significantly so as to dramatically affect the in vivo characteristics of the antibody. Also, these results are comparable to those obtained previously with the radiolabeled conjugated prepared using the HPLC method of radiolabeling (see BB-IND 4850/4851). Dosimetry estimates for a “standard” 70 Kg human calculated from the biodistribution data for mice are in agreement with those obtained with conjugate radiolabeled using the HPLC procedure (see BB-IND 4850/4851). Additionally, the dosimetry results are comparable to results obtained for patients enrolled in an on-going clinical trial (IDEC study #1315), when compared per millicurie of injected dose. For six patients in the study, mean values (rads +SD) for whole body, heart, liver, and spleen were 1.40±0.57, 10.50±4.68, 9.89±8.91, and 9.75±6.00, respectively.
  • Before implementing the “mix-and-shoot” labeling protocol for preparing clinical-grade [0338] 90Y-2B8-MX-DTPA, it was necessary to assess the reproducibility of the protocol. Therefore, ten validation lots were prepared using different lots of 90Y chloride. For the ten lots prepared, the immunoreactivity values obtained using the “mix-and-shoot” method were in the range of 60.6% to 93.3% with a mean of 74.5% and a median of 72.1%. This retention of immunoreactivity is significantly better than the approximately 60% previously obtained using the current HPLC method (range of 54.9% to 65.1%; mean of 60.2%). The average radioincorporation for the ten lots was 95.7% (range of 93.5% to 97.5%). This value is comparable to that previously seen with the HPLC method (range of 91.7% to 93.7% and a mean of 93.1%). Also, results for endotoxin, antibody concentration, radioactivity concentration, specific activity, total vial radioactivity, total protein concentration, pH, and sterility were comparable for the ten lots. Together, these results confirmed the reproducibility of the “mix-and-shoot” method. In addition, we evaluated the variability of the process due to different labeling times by performing reactions for 5 and 10 minutes. Since there were no significant differences noted for the two reaction times, it was decided that the shorter incubation time would be used in the final protocol.
  • E. Summary [0339]
  • We have developed a labeling procedure, referred to as the “mix-and-shoot” method, for the preparation of clinical doses of [0340] 90Y-labeled 2B8-MX-DTPA which obviates the need for the currently used high performance liquid chromatographic (HPLC) step for removal of non-protein bound radioisotope. The simplified protocol eliminates this laborious purification step while maintaining a high level of radioisotope incorporation (>95%) and improved retention of immunoreactivity (>70%). The clinically-formulated radiolabeled conjugate was found to be stable in vitro when incubated at 4° C. for 48 hours based on retention of radioisotope and immunoreactivity. Additionally, the radiolabeled conjugate was stable when incubated in human serum at 37° C. for 72 hours. Biodistribution studies in BALB/c mice demonstrated no unusual tissue deposition, including bone. Estimates of radiation absorbed doses to a “standard” 70 Kg human were comparable to those obtained in an on-going clinical trial using 90Y-labeled 2B8-MX-DTPA. The results of these studies showed that 90Y-labeled 2B8-MX-DTPA produced using the “mix-and-shoot” protocol was comparable to that prepared using the conventional HPLC process. Validation of the scale-up protocol for preparing clinical-grade radiolabeled conjugate showed that the method was reproducible and that the product was comparable to that produced using the current HPLC method. The results of these pre-clinical studies indicate that this new “mix-&-shoot” protocol can be used to prepare 90Y-labeled 2B8-MX-DTPA suitable for use in clinical trials.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS EXAMPLE 1. Radioincorporation—Kits and Assays
  • I. Summary One objective of the present invention was to devise radiolabeling kit protocols for preparation of [0341] 111In and 90Y-labeled 2B8-MX-DTPA (In2B8 and Y2B8, respectively) and to establish release specifications for clinical products. The radiolabeling kit protocols are reproducible with respect to radioincorporation and binding to antigen-positive SB cells and indicate the suitability of the radiolabeling kit for use in the clinical trials. It is recommended that In2B8 and Y2B8 release specifications for radioincorporation and binding be established at ≧95% and ≧70%, respectively.
  • II. Introduction [0342]
  • A [0343] 90Y-labeled murine monoclonal anti-CD20 antibody (Y2B8) is currently being evaluated in clinical trials for the treatment of relapsed B-cell lymphoma. The yttrium isotope lacks a gamma component making it unsuitable for imaging systems. Therefore, 111In-labeled 2B8-MX-DTPA (In2B8) will be used to assess tumor localization and dosimetry in patients prior to or after treatment with the yttrium-labeled therapeutic. The protocols used currently for the preparation of Y2B8 and In2B8, referred to as the “mix-&-shoot” methods, produce radiolabeled antibodies suitable for clinical studies. However, simplification of the labeling process would expedite dose preparation in a clinical setting.
  • The new radiolabeling kit is preferably comprised of four components: 1.) 2B8-MX-DTPA in low-metal normal saline at 2 mg/mL, 2.) 50 mM sodium acetate used to adjust radioisotope solution to appropriate labeling pH, 3.) formulation buffer (1× PBS, pH 7.4 containing 7.5% human serum albumin and 1 mM DTPA), 4.) empty 10 mL glass vial (reaction vial). All components are tested to be sterile and pyrogen-free. [0344]
  • This report summarizes the validation of this radiolabeling kit which is simple and easy to use and which yields radiolabeled antibodies with ≧95% radioincorporation and acceptable retention of binding to antigen-positive cells. Release testing specifications are recommended for the clinical products. [0345]
  • III. Materials and Methods for Radioincorporation [0346]
  • A. Reagents in Radiolabeling Kit [0347]
  • 1. 2B8-MX-DTPA, IDEC; Lot# 082395RM2 [0348]
  • 2. 50 mM Sodium Acetate, low-metal, IDEC; Lot# 082395RM3 [0349]
  • 3. Formulation Buffer (1× PBS, pH 7.4 containing 7.5% (w/v) human serum albumin and 1 mM DTPA), IDEC, Lot# 082395RM1 [0350]
  • 4. Reaction vial, 10 mL, IDEC [0351]
  • B. Materials and Equipment [0352]
  • 1. Biodex Tec-Control Radioincorporation Kit, Cat.#151-770 [0353]
  • 2. Gloves: powder-free [0354]
  • 3. Sterile polypropylene syringes [0355]
  • 4. Sterile syringe needles [0356]
  • 5. Small tubes with closure; 1.5 ml [0357]
  • C. Methods [0358]
  • 1. Preparation of Y2B8 and In2B8 Using Radiolabeling Kit Kit reagents were prepared and filled into glass septum vials. Type I borosilicate vials (2 or 10 mL) were rinsed with sterile water for injection (WFI) and autoclaved before filling. Butyl rubber septa were rinsed with sterile WFI and autoclaved before use. Reagents were manually filled and crimped in a [0359] Class 100 room and tested for pyrogenicity and sterility using USP methods.
  • a. Preparation of In2B8 [0360]
  • Additional Reagents: [0361]
  • 1. Indium-[ 1 1]: chloride salt, carrier-free, in HCl. [0362]
  • Precautions: [0363]
  • 1. All steps are preferably performed using aseptic technique. [0364]
  • 2. Radiolabeling kit components should be allowed to come to room temperature before use. [0365]
  • 3. Final product should be administered to patient within 8 hours of completing step 9 below. [0366]
  • In2B8 Radiolabeling protocol [0367]
  • Procedure [0368]
  • 1. The volume of [0369] 111InCl3 to add to the reaction vial was calculated as follows:
  • a. Radioactivity concentration at the time of radiolabeling in mCi/ml: [0370]
  • C[0371] 0=Radioactivity concentration at time of calibration (see manufacturer's Certificate of Analysis).
  • Δt=Change in time (positive number is post calibration, negative number is pre calibration). [0372] Radioactivity Concentration at time of labeling = C 0 e 0.0103 ( Δ t )
    Figure US20020102208A1-20020801-M00001
  • b. Volume of [0373] 111InCl3 to add to the reaction vial: 5.5 mCi Radioactivity Concentration at time of labeling = Volume to add to reaction vial
    Figure US20020102208A1-20020801-M00002
  • 2. The volume of 50 mM sodium acetate to be added to the reaction vial was calculated as follows: [0374]
  • Volume of [0375] 111InCl3 added (Step 1b)×(1.2)=Volume of 50 mM sodium acetate to be added.
  • 3. The septa of the reaction vial and the 50 mM sodium acetate vial were wiped with alcohol. Using a icc syringe, the calculated volume of 50 mM sodium acetate (Step 2) was transferred to the reaction vial. [0376]
  • 4. The septum of [0377] 111InCl3 source was wiped with alcohol. The vial was vented with a needle fitted with sterile 0.2 μm filter. Using a lcc sterile syringe, the required volume (Step 1b) of 111InCl3 was transferred to the reaction vial. The vial was mixed by inverting several times.
  • 5. The septum of the 2B8-MX-DTPA vial was wiped with alcohol. Using a 1 cc syringe, 1.0 mL of 2B8-MX-DTPA was slowly transferred to the reaction vial. The vial was mixed by inverting several times. [0378]
  • 6. The reaction was allowed to proceed for 30 minutes±5 minutes at ambient temperature. [0379]
  • 7. The total volume of reaction mixture was calculated by adding together the volume of [0380] 111InCl3 added (Step 4), the volume of 50 mM sodium acetate added (Step 3) and the volume of 2B8-MX-DTPA added (Step 5).
  • 8. The volume of Formulation Buffer to add to the Reaction Vial to obtain a final volume of 10 mL was calculated by subtracting the total amount calculated in step 7 from 10. [0381]
  • 9. The Formulation Buffer vial was wiped with alcohol and the vial was vented. Due to the viscosity of the Formulation Buffer, the reaction vial was vented using a needle fitted with a 0.2 μm syringe filter. Using a 10 cc sterile syringe fitted with an appropriate gauge needle, the volume of Formulation Buffer calculated in [0382] Step 8 was transferred to the reaction vial. The vent needle was removed from the reaction vial and the vial was mixed by inverting several times (Final Product). This vial was incubated at least 5 minutes prior to doing the “Radioincorporation Assay”. The color of the solution was amber and the vial was full, confirming that Formulation Buffer was added.
  • 10. Total radioactivity of the Final Product vial was measured using the appropriate instrumentation set for [0383] 111In measurement.
  • 11. The Final Product was stored immediately at 2°-8° C. for “Binding Assay” and “Radioincorporation Assay”. [0384]
  • b. Preparation of Y2B8 [0385]
  • Additional Reagents: [0386]
  • 1. Yttrium-[90]: chloride salt, carrier-free, in HCl. [0387]
  • Precautions: [0388]
  • 1. All steps should be performed using aseptic technique. [0389]
  • 2. Radiolabeling kit components should be allowed to come to room temperature before use. [0390]
  • 3. The product should be administered to the patient within 8 hours of completing [0391] step 8 below.
  • Y2B8 Radiolabeling Protocol [0392]
  • 1. The volume of [0393] 90YCl3 to add to the reaction vial was calculated as follows:
  • a. The radioactivity concentration at the time of radiolabeling: [0394]
  • C[0395] 0=Radioactivity concentration at time of calibration (see manufacturer's Certificate of Analysis).
  • Δt =Change in time (positive number is post calibration, negative number is pre calibration). [0396] Radioactivity Concentration at time of labeling = C 0 e 0.0108 ( Δ t )
    Figure US20020102208A1-20020801-M00003
  • b. The volume of [0397] 90YCl3 to add to the reaction vial: 45 mCi Radioactivity Concentration time of labeling = Volume added to reaction vial
    Figure US20020102208A1-20020801-M00004
  • 2. The volume of 50 mM sodium acetate to add to the reaction vial was calculated as follows: [0398]
  • a. For [0399] 90YCl3 in 0.040 M HCl (Amersham):
  • Volume [0400] 90YCl3 (Step 1b)×(0.8)=volume of sodium acetate to add
  • b. For [0401] 90YCl3 in 0.050 M HCl(Nordion):
  • Volume [0402] 90YCl3 (Step 1b)×(1.0)=volume of sodium acetate to add
  • 3. The septa of the reaction vial and the sodium acetate vial were wiped with alcohol. Using a 1 cc syringe, the calculated volume (Step 1a or 1b) of 50 mM sodium acetate (Step 2) was transferred to the reaction vial. The vial was mixed by inverting several times. [0403]
  • 4. The septum of the [0404] 90YCl3 source vial was wiped with alcohol. The vial with a needle fitted with sterile 0.2 μm filter. Using a lcc sterile syringe, was vented the required volume (Step 1b) of 90YCl3 was transferred to the reaction vial. The vial was mixed by inverting several times.
  • 5. The septum of the 2B8-MX-DTPA vial was wiped with alcohol. Using a 3 cc sterile syringe, 1.5 mL of 2B8-MX-DTPA was transferred to the reaction vial. The vial was mixed by inverting several times. [0405]
  • 6. The total volume of reaction mixture was calculated by adding the amount of Y-90 chloride added (Step 4), plus the amount of 50 mM sodium acetate added (Step 3), plus the amount of 2B8-MX-DTPA added (Step 5). [0406]
  • 7. The volume of Formulation Buffer to add to the Reaction Vial to obtain a final volume of 10 mL was calculated by subtracting the total reaction volume calculated in [0407] step 6 from 10.
  • [0408] 8. The Formulation Buffer vial was wiped with alcohol and the vial was vented. Due to the viscosity of the Formulation Buffer, the reaction vial using a needle fitted with a 0.20 μm syringe filter. Using a 10 cc sterile syringe fitted with an appropriate gauge needle, the volume of Formulation Buffer calculated in Step 7 was transferred to the reaction vial. The vent needle was removed from the reaction vial and the vial was mixed by inverting several times (Final Product). The vial was incubated at least 5 minutes prior to doing the “Radioincorporation Assay”. The color of the solution was amber and the reaction vial was full thereby confirming that Formulation Buffer was added.
  • 9. The total radioactivity of the Final Product vial was measured using the appropriate instrumentation set for measurement of [0409] 90Y.
  • 10. The Final Product was immediately stored at 2°-8° C. until required for patient administration. [0410]
  • 11. Immunoreactivity testing: [0411]
  • Using a 1 mL syringe, 0.1 mL was aseptically removed from the reaction vial and transferred to a separate 1.5 mL screwcap tube. The tube was immediately stored at 2°-8° C. for “Binding Assay” and “Radioincorporation Assay”. [0412]
  • Validation of the radiolabeling kit protocols was performed at IDEC Pharmaceuticals (San Diego, Calif.), MD Anderson Health Center (Houston, Tex.), Mayo Clinic (Rochester, MN), and City of Hope (Duarte, Calif.). All kit components, including clinical-grade 2B8-MX-DTPA, were prepared by IDEC Pharmaceuticals under GMP conditions (Good Manufacturing Conditions according to the Code of Federal Regulations) and determined to be sterile and pyrogen-free. [0413]
  • The radiolabeled antibodies were formulated with 1× PBS containing 7.5% (w/v) human serum albumin (HSA; clinical-grade; Baxter-Hyland) and 1 mM DTPA. Results of the release tests performed on each validation lot are described below. [0414]
  • Six validation lots each of In2B8 and Y2B8 were prepared by five operators. These lots were designated as follows and performed at the following facilities: [0415]
  • In2B8: [0416]
  • #1: IDEC Pharmaceuticals [0417]
  • #2: IDEC Pharmaceuticals [0418]
  • #3: IDEC Pharmaceuticals [0419]
  • #4: MD Anderson Health Center [0420]
  • #5: Mayo Clinic [0421]
  • #6: City of Hope [0422]
  • Y2B8: [0423]
  • #1: IDEC Pharmaceuticals [0424]
  • #2: IDEC Pharmaceuticals [0425]
  • #3: IDEC Pharmaceuticals [0426]
  • #4: MD Anderson Health Center [0427]
  • #5: Mayo Clinic [0428]
  • #6: City of Hope [0429]
  • 2. Preparation of Lyophilized SB and HSB Cells [0430]
  • The human cell lines SB (CD20-positive) and HSB (CD20-negative) were obtained from American Type Culture Collection and cultured in T-flasks using RPMI-1640 containing 10% fetal bovine serum supplemented with 2% glutamine. Cultures were maintained at 37° C. and 5% CO[0431] 2. Cells were typically split 1:2 every other day and harvested at 0.5-2.5×106 cells/mL and viability's >80%. Cell concentrations were determined using a hemacytometer and viability determined by trypan blue exclusion.
  • Cells were harvested at ambient temperature at a cell density of 0.5-2×10[0432] 6 cells/mL by centrifugation (1300 rpm in a Sorvall centrifuge) and washed twice with 1× HBSS. Pelleted cells were resuspended to 50×106 cells/mL in 1× HBSS containing 1% (w/v) bovine serum albumin (BSA) and 10% (w/v/) mannitol (lyophilization buffer), 0.5 mL dispensed into 1.5 mL polypropylene microfuge tubes with o-ring gaskets and stored at −70° C., and lyophilized overnight at 30-60 millitorr. Tubes of lyophilized cells were stored desiccated at 2-8° C. and reconstituted in sterile water for assays; tubes of cells lyophilized in microfuge tubes were stored with desiccant.
  • 3. Analytical Assays [0433]
  • The analytical methods used to test the validation lots of In2B8 and Y2B8 are described below. The following assays were performed for each validation lot: [0434]
  • 1. Binding Assay using lyophilized SB cells [0435]
  • 2. Y2B8/In2B8 Radioincorporation Assay using Biodex Kit [0436]
  • a. Binding Assays [0437]
  • Percent binding was assessed by each operator using lyophilized CD20 positive SB cells according to the following protocols for In2B8 and Y2B8, respectively. These assays provide for a fast and efficient method of confirming that the radiolabeled antibody still recognizes CD20 as an antigen. At one clinical site, CD20-negative HSB cells were also evaluated. Lyophilized cells were prepared and stored according to the above method, “Preparation of Lyophilized SB and HSB Cells”. [0438]
  • i. In2B8 Binding Assay [0439]
  • Additional Reagents: [0440]
  • 1. Indium-[111]-2B8-MX-DTPA [0441]
  • 2. Lyophilized SB cells; three tubes containing 25×10[0442] 6 cells/tube.
  • 3. Lyophilized HSB cells; three tubes containing 25×10[0443] 6 cells/tube.
  • 4. Sterile water for irrigation or sterile water for injection. [0444]
  • 5. Dilution buffer (1× PBS, pH 7.2-7.4 containing 1% Bovine Serum Albumin (BSA), and 0.02% Sodium Azide 0.2 μm filtered and stored at room temperature. [0445]
  • 6. Glass or plastic test tubes for counting radioactivity. [0446]
  • Procedure: [0447]
  • Assay set-up (Non-radioactive portion) [0448]
  • 1. Three tubes of lyophilized SB and HSB cells were obtained. [0449]
  • 2. A volume of 0.50 mL of SWFI (sterile water for injection) was added to each tube, and the tubes were vortexed until homogenous suspensions were obtained. [0450]
  • 3. Four empty 1.5 mL microfuge tubes. To three of the tubes 0.50 mL of Dilution buffer was added, representing a control with no cells. [0451]
  • 4. To the other 1.5 mL microfuge tube, 0.99 mL of Dilution buffer was added; this tube was labeled 1:100. [0452]
  • 5. A 50 mL sterile polypropylene tube with cap was obtained and 10 mL of Dilution buffer was added to the tube. [0453]
  • Assay Set-Up (Radioactive Portion) [0454]
  • 1. The radiolabeled antibody stored at 2°-8° C. was obtained. [0455]
  • 2. A volume of 0.01 mL was withdrawn with a P20 and added to the 1.5 mL microfuge tube containing 0.99 mL of Dilution buffer (1:100 dilution). The tip was rinsed and the tube vortexed gently to mix. [0456]
  • 3. A volume 0.20 mL was withdrawn with a P200 from the 1:100 dilution tube and added to the conical tube containing 10 mL of Dilution buffer. The tube was mixed thoroughly. [0457]
  • Assay Protocol [0458]
  • 1. A volume of 0.50 mL of the diluted [0459] 111In2B8-MX-DTPA was added to all tubes.
  • 2. The caps were securely tightened on all tubes, and the tubes mixed continuously for 60 minutes. [0460]
  • 3. After 60 minutes incubation at ambient temperature, all tubes were centrifuged for 5 minutes at minimum of 2000 g. [0461]
  • 4. A volume of 0.75 mL of each supernatant was transferred to tubes appropriate for the counting instrument. [0462]
  • 5. The radioactivity in tubes was counted using a gamma counter, adjusting for background. [0463]
  • ii. Y2B8 Binding Assay [0464]
  • Additional Reagents [0465]
  • 1. [0466] 90Y2B8-MX-DTPA
  • 2. Lyophilized SB cells [0467]
  • 3. Sterile water for irrigation or sterile water for injection [0468]
  • 4. Dilution buffer (1× PBS, pH 7.2-7.4 containing 1% Bovine Serum Albumin (BSA), and 0.02% Sodium Azide) [0469]
  • Procedure: [0470]
  • Radiolabeled Antibody Sample Prep [0471]
  • 1. The radiolabeled antibody stored at 2°-8° C. was obtained. [0472]
  • 2. A volume of 10 μL was withdrawn with a P20 and added to a 1.5 mL microfuge tube containing 990 μL of Dilution buffer (1:100 dilution). The tip was rinsed and the tube was vortexed slightly. [0473]
  • 3. A 50 mL sterile polypropylene tube with cap was obtained and 10 mL of Dilution buffer to the tube, using a 10 mL serological pipette. [0474]
  • 4. A volume of 35 μL was withdrawn with a P200 from the 1:100 dilution tube and added to the conical tube containing 10 mL of Dilution buffer. Mix thoroughly. [0475]
  • Lyophilized Cell Prep [0476]
  • 1. Three tubes of lyophilized SB Cells were obtained. [0477]
  • 2. A volume of 0.5 mL of SWFI was added to each tube, and the tubes were vortexed until single cell suspensions were obtained. [0478]
  • 3. Three empty 1.5 mL microfuge tubes were obtained; to three of the tubes, 0.5 mL of Dilution buffer was added, representing a control with no cells. [0479]
  • Assay Protocol [0480]
  • 1. A volume of 0.5 mL of the diluted [0481] 90Y2B8-MX-DTPA was added to each tube.
  • 2. The tubes were placed on end over mixer for 45 minutes, after making sure caps are securely tightened. [0482]
  • 3. After 45 minutes incubation at ambient temperature, the cells were pelleted by microcentrifugation for 5 minutes. [0483]
  • 4. A volume of 0.8 mL of the supernatant was transferred to scintillation vials. [0484]
  • 5. Scintillation cocktail was added to each vial. [0485]
  • 6. The amount of radioactivity in each vial was determined using a scintillation counter, adjusting for background. [0486]
  • b. Radioincorporation Assay [0487]
  • Percent radioincorporation was determined by instant thin-layer chromatography (ITLC) using the Biodex Tec-Control Radiochromatographic Kit according to the following protocol: [0488]
  • Additional Materials and Equipment: [0489]
  • 1. [0490] 111In- or 90Y-radiolabeled 2B8-MX-DTPA
  • 2. Tubes for counting radioactive TLC strips [0491]
  • 3. Scissors [0492]
  • 4. Sterile syringe, 1 cc [0493]
  • 5. Sterile needles, 26 G [0494]
  • 6. Gamma counter or scintillation counter [0495]
  • 7. Pipettor [0496]
  • Procedure: [0497]
  • 1. The entire Biodex Operation Manual should be read first. [0498]
  • 2. Each radiolabeled sample in triplicate was tested according to kit instructions; one strip per vial was developed. [0499]
  • 3. To spot the radiolabeled sample on the chromatography strip, a pipettor was used to spot 1 μl on the origin line. Alternatively, one small drop dispensed from a 26 G needle attached to a sterile 1 cc syringe may be spotted. [0500]
  • 4. Each section was counted for activity using the appropriate counter, i.e., gamma counter for [0501] 111In and a scintillation counter for 90Y, adjusting for background.
  • 5. The Biodex instructions for calculating the percentage of radiolabeled antibody were followed. [0502]
  • IV. Results [0503]
  • The results of testing on each validation lot of In2B8 or Y2B8 are summarized in Tables 38 and 39. [0504]
    TABLE 38
    Release Assay Results for Y2B8 Validation
    Lot Number % Radioincorporation % Binding
    1 99.5 78.6
    2 99.3 87.0
    3 99.4 85.9
    4 99.2 81.8
    5 99.2 79.6
    6 96.3 80.8
    Mean = 98.8   Mean = 82.3
    Standard Deviation = 1.24    Standard Deviation = 3.4
     % CV = 1.25%    CV = 4.2%
  • [0505]
    TABLE 39
    Release Assay Results for In2B8 Validation Lots
    Lot Number % Radioincorporation % Binding
    1 99.4 86.2
    2 98.7 86.8
    3 99.3 85.8
    4 98.3 86.7
    5 99.0 82.1
    6 99.3 83.0
    Mean = 99.0   Mean = 85.2
    Standard Deviation = 0.43    Standard Deviation = 2.06
     % CV = 0.45%    CV = 2.42%
  • V. Discussion and Conclusions [0506]
  • To simplify the current radiolabeling methods for In2B8 and Y2B8, a four-component kit was developed. The concentrations of sodium acetate and 2B8-MX-DTPA were reduced to 50 mM and 2 mg/mL, respectively, to allow accurate volume transfers using syringes. All kit components were preferably filled in glass septum vials and tested for sterility and pyrogenicity by IDEC before release, thus eliminating the need for these tests to be performed at the clinical sites. At the site, all reagent manipulations are performed using sterile syringes and needles. Therefore, adherence to aseptic technique customarily found in a radiopharmacy environment insures that the radiolabeled and formulated anti-bodies are suitable for patient administration. [0507]
  • Reproducibility and ruggedness of the radiolabeling protocols for In2B8 and Y2B8 was evaluated by performing several validation runs using different lots of each radioisotope. For the six validation lots of In2B8 prepared, binding ranged from 82.1% to 86.8% with a mean of 85.1%; radioincorporation values for were approximately 99% (range of 98.3% to 99.4%). For the six validation lots of Y2B8 prepared, the percent binding obtained was in the ranged from 78.6% to 87.0% with a mean of 82.3%. Radioincorporation values for Y2B8 averaged 98.8% (range of 96.3% to 99.5%). Together, these results confirm the reproducibility and ruggedness of the radiolabeling kit methods for preparation of both In2B8 and Y2B8. Based on these validation results, it is recommended that release specifications for radioincorporation and binding be established at ≧95% and ≧70%, respectively, for both In2B8 and Y2B8. Additionally, because of the increased ease of use and reduced potential for mistakes during preparation, it is recommended that percent binding using lyophilized CD20-positive cells and radioincorporation be used to release test In2B8 and Y2B8 at the clinical sites. [0508]
  • To summarize, these results together indicate that In2B8 and Y2B8 prepared using the radiolabeling kit are suitable for use in the clinical setting. Additionally, for both radiolabeled antibodies, release specifications are established reflecting the results of several validation runs by the five different operators. [0509]
  • EXAMPLE 2 Radioincorporation and Binding—Kits and Assays
  • I. Summary [0510]
  • The murine anti-CD20 monoclonal antibody designated 2B8 has been cloned in CHO cells to yield a high expression cell line. Specificity of the CHO-derived antibody for CD20-positive human cells was demonstrated by FACS analysis and competitive binding. Negligible binding was observed to human T-cells. The affinity of the antibody for CD20-positive cells was determined to be 1.3×10[0511] −10 M using a competitive binding assay. The antibody was reacted with the chelating agent MX-DTPA to form a conjugate, 2B8-MX-DTPA, with negligible loss of immunoreactivity (affinity value was 4.4×10−10 M. Optimal chelator conjugation, as determined by measuring radioincorporation of 111In, was achieved after eight hours reaction. Radiolabeling protocols for 2B8-MX-DTPA were optimized for 90Y or 111In with respect to pH and incubation time to insure maximal radioincorporation (>95%) and retention of immunoreactivity (>70%). Release specifications for In2B8 and Y2B8 prepared using CHO-derived 2B8-MX-DTPA in clinical trials were recommended for radioincorporation (>95%) and binding to lyophilized and reconstituted CD20-positive human cells (>70%). Taken together, these results indicate the suitability of CHO-derived 2B8-MX-DTPA for use in clinical trials.
  • II. Introduction [0512]
  • The 2B8 antibody previously used was produced in hollow-fiber bioreactors. To reduce the manufacturing costs of this antibody, it has been cloned and expressed in CHO cells to yield a high-expression production cell line. This example describes results of the in vitro characterization of the CHO-derived 2B8 antibody, the conjugated antibody (2B8-MX-DTPA), and the [0513] 90Y and 111In-labeled antibody products prepared using the clinical radiolabeling kit protocols.
  • III. Materials and Methods [0514]
  • A. Reagents The human cell lines SB (CD20-positive) and HSB (CD20-negative) were obtained from American Type Culture Collection and cultured in T-flasks using RPMI-1640 containing 10% fetal bovine serum supplemented with 2% glutamine. Cultures were maintained at 37° C. and 5% CO[0515] 2. Cells were typically split 1:2 every other day and harvested at 0.5-2.5×106 cells/mL and viability's >80%. Cell concentrations were determined using a hemacytometer and viability determined by trypan blue exclusion. Specific information on cell lots is recorded in Notebook# 1553 and in the binder titled “Cell Activity Logbook 1995 & 1996” authored by Ron Morena. CHO-derived 2B8 was produced under GMP conditions in IDEC's manufacturing facility. The antibody was formulated in low-metal normal saline at 11.5 mg/ml. Antibodies were determined to be homogeneous by SDS-PAGE. 2B8-MX-DTPA was produced under GMP conditions according to PSBR-043 from CHO-derived 2B8 and formulated in low-metal saline at 2 mg/mL (Lot #'s 0165A and 0165B).
  • Pharmaceutical-grade [0516] 111In chloride was purchased from Amersham (U.K.) or Cyclotron Products Inc. (Coral Gables, Fla.). Yttrium[90] chloride was obtained from Amersham (U.K.), Nordion International (Kanatta, Canada), or Pacific Northwest National Laboratory (Richland, Wash.). MX-DTPA prepared under GMP was obtained from Hauser Chemical (Boulder, Colo.). Clinical-grade calcium trisodium diethylenetriaminepentaacetic acid (DTPA) was obtained from Heyl (Berlin, Germany). TAG-NHS was obtained from IGEN Inc. (Rockville, Md.). Murine anti-CD19 beads were purchased from Dynal Inc. (Lake Success, N.Y.). Goat anti-mouse FITC-labeled F(ab′)2 was purchased from Jackson ImmunoResearch.
  • Reagents requiring removal of contaminating heavy metals were batch treated with Chelex 100 (BioRad Industries) or with Chelating Sepharose (Pharmacia) by passing solutions through a column. Low-metal stock solutions were diluted with Sterile Water for Irrigation (SWFIr). Solutions were stored in sterile plastic containers. [0517]
  • Additional reagents are described below for specific methods. [0518]
  • B. Materials and Equipment [0519]
  • 1. Origen Analyzer; IGEN Inc. Model #1100-1000; IDEC #1492 [0520]
  • 2. Top-Count scintillation counter; Packard, Model #A9912; IDEC #1329 [0521]
  • 3. Gamma counter; Isodata, Model # 20-10; IDEC #0628 [0522]
  • 4. Tec-Control Radiochromatographic Kit, Biodex, Model #151-770 [0523]
  • 5. Lyophilizer; Virtis, [0524] Model Freezemobile 12; IDEC #0458
  • Additional materials and equipment are described for specific methods. [0525]
  • C. Methods [0526]
  • 1. Preparation of Lyophilized SB and HSB Cell [0527]
  • Cells were cultured as described above and harvested at ambient temperature at a cell density of 0.5-2×10[0528] 6 cells/mL by centrifugation (1300 rpm in a Sorvall centrifuge) and washed twice with 1× HBSS. Pelleted cells were resuspended to 50×106 cells/mL in 1× HBSS containing 1% (w/v) bovine serum albumin (BSA) and 10% (w/v/) mannitol (lyophilization buffer), 0.5 mL dispensed into 1.5 mL polypropylene microfuge tubes with o-ring gaskets and stored at −70° C., and lyophilized overnight at 30-60 millitorr. Tubes of lyophilized cells were stored desiccated at 2-8° C. and reconstituted in sterile water for assays; tubes of cells lyophilized in microfuge tubes were stored with desiccant.
  • 2. FACS Binding Analysis [0529]
  • Direct binding of antibodies to human B-cells was determined by flow cytometry. Increasing concentrations of antibody were incubated in 1× PBS, pH 7.2, containing 1% (w/v) BSA (binding buffer) with 5×10[0530] 6 CD20-positive (SB) or CD20-negative (HSB) cells for 30 min. on ice. Cells were washed by centrifugation, resuspended in binding buffer, and incubated with FITC-labeled goat anti-mouse F(ab′)2 for 30 min. on ice. After incubation with the secondary reagent, cells were washed by centrifugation and resuspended in 1× PBS containing 1.1% (v/v) formaldehyde to fix cells. Mean fluorescence intensity was determined using flow cytometry.
  • 3. Competitive Binding Assays [0531]
  • Immunoreactivity of 2B8 and 2B8-MX-DTPA was determined by competitive binding to CD20-positive SB cells using the ORIGEN electrochemiluminescent method (Leland and Powell). Log-phase SB cells were harvested from culture and washed twice with 1× HBSS. Cells were diluted in 1× PBS pH 7.2 containing 1% (w/v) bovine serum albumin. In some experiments, lyophilized cells were used after reconstitution with sterile water. Ruthenium-labeled tracer antibody was prepared by incubating CHO-derived 2B8 (lot #165) in 1× PBS, pH 7.2 with the N-hydroxysuccinimide ester of ruthenium (II) tris-bipyridine chelator (TAG-NHS) at a 15:1 molar ratio of TAG-NHS to antibody. After 1 h incubation at ambient temperature, protected from light, the reaction was quenched with glycine for 10 min. Unreacted TAG was removed by size exclusion chromatography using a Pharmacia PD-10 column equilibrated with 1× PBS. Protein concentration was determined using the Bradford protein assay. TAG incorporation was determined by measuring absorbance at 455 nm. The molar ratio of TAG to protein was calculated to be 3.0. [0532]
  • Assays were performed in 12×75 mm polypropylene tubes. Varying amounts of competing antibody (0.002-17 ug/mL) were incubated in 1× PBS, pH 7.2, containing 1% (w/v) BSA with 0.08 ug/mL TAG-labeled CHO 2B8, 0.08 mg/mL anti-CD19 beads, and 167,000 cells/mL. After incubation at ambient temperature with orbital mixing for 3 h, relative electrochemiluminescence (ECL) was determined using the ORIGEN instrument. Mean ECL values were determined for duplicated samples and plotted vs. competing antibody concentration using Kaleidagraph software. For some experiments, per cent inhibition was plotted. Competition curves were fitted and [0533] EC 50 values (antibody concentration giving 50% maximal binding) determined using the following 4-parameter program:
  • y=((m1−m4)/(1+(m0/m3)^ m2))+m4;m1=;m2=;m3=;m4=
  • m0=independent variable [0534]
  • m1=zero signal response in relative ECL units [0535]
  • m2=curvature parameter [0536]
  • m3=EC50 in ug/mL [0537]
  • m4=maximum signal response in relative ECL units Average affinity values were calculated from EC50 values and the known concentration of trace antibody using the method of Muller. [0538]
  • 4. Preparation of 2B8-MX-DTPA [0539]
  • The chelating agent, 1-isothiocyanatobenzyl-3-methyldiethylenetriaminepentaacetic acid (MX-DTPA) was provided as a dry powder (free-acid) and stored desiccated at −20° or −70° C. Approximately 3 mg of CHO 2B8 antibody in low-metal normal saline were adjusted to pH 8.6 by adding one-tenth volume of 50 mM sodium borate, pH 8.6. Antibody at 10-11 mg/mL was incubated at a 4: 1 molar ratio of MX-DTPA to protein by adding MX-DTPA dissolved in 50 mM sodium borate, pH 8.6. After incubation at ambient temperature (2 to 24 h), unreacted chelator was removed from the conjugate by repetitive diafiltration in low-metal normal [0540] saline using Centricon 30 spin-filters.
  • 5. Preparation of In2B8 and Y2B8 [0541]
  • In2B8 was prepared using the radiolabeling kit protocol as described herein. Antibody was labeled at a specific activity of 3 mCi/mg and formulated to 0.2 mg/mL. Briefly, 0.5 to 2 mCi of [0542] 111In chloride was transferred to a metal-free microfuge tube and adjusted to approximately pH 4.2 using a 1.2× volume of low-metal 50 mM sodium acetate. 2B8-MX-DTPA at 2 mg/mL was added to the indium acetate solution and after incubation at ambient temperature for 30 min., the labeled antibody was formulated to 0.2 mg/mL in 1× PBS, pH 7.2 containing 7.5% (w/v) human serum albumin and 1 mM DTPA (4% to 6% final concentration of HSA). All samples were tested for radioincorporation in triplicate; values were >95%.
  • Y2B8 was also prepared using a small-scale version of the radiolabeling kit protocol described in Example 1. Antibody was labeled at a specific activity of 15 mCi/mg and formulated to 0.3 mg/mL. Briefly, 0.5 to 2 mCi of 90Y chloride was transferred to a metal-free microfuge tube and adjusted to approximately pH 4.2 using a 1.2× volume of low-[0543] metal 50 mM sodium acetate. 2B8-MX-DTPA at 2 mg/mL was added to the 90Y acetate solution and after incubation at ambient temperature for 5 min., the labeled antibody was formulated to 0.3 mg/mL in 1× PBS, pH 7.2 containing 7.5% (w/v) human serum albumin and 1 mM DTPA (final concentration of HSA, 4% to 6%). All samples were tested for radioincorporation in triplicate; values were >95%.
  • The radioactivity concentrations of the final radiolabeled products were calculated based on the amount of radioactivity added to the reaction mixture and by reference to the Certificate of Analysis for the radioisotope. Antibody concentration of the quenched reaction mixtures were calculated from the known amount of antibody added. [0544]
  • For radiolabeling kinetic studies evaluating the effect of pH on radioincorporation and binding, the pH of the reaction mixtures was adjusted. by adding varying amounts of low-[0545] metal 50 mM sodium acetate (0.8 to 2.2× volume of radioisotope solution).
  • 6. Determination of Radioincorporation for In2B8 and Y2B8 [0546]
  • The amount of radioactivity associated with the conjugates (radioincorporation) in the final products or incubation samples was determined using a commercially available kit manufactured by Biodex (Tec-Control Radiochromatographic Kit; see Example 1). In general, 1 μL of the test samples were applied in duplicate or triplicate using a micropipetter and developed according to the Biodex instructional insert. Strip halves were counted for radioactivity in glass tubes using an Isodata gamma counter or a Packard Top Count scintillation counter as described below. The radiolabel incorporation was calculated by dividing the amount of radioactivity in the top half of the strip by the total radioactivity found in both top and bottom halves. This value was expressed as a percentage and the mean value determined. [0547]
  • 7. Determination of Immunoreactivity of In2B8 and Y2B8 Immunoreactivity was assessed using the method of Lindmo et al and as described above in Example 1. [0548]
  • 8. Direct Binding Assay for In2B8 and Y2B8 [0549]
  • The same protocols as described herein were used to determine binding to CD20-positive SB cells for In2B8 and Y2B8, respectively. In2B8 and Y2B8 were prepared and formulated as described above. For assay, In2B8 or Y2B8 samples were diluted with assay dilution buffer (LXPBS, pH 7.2, containing 1% (w/v) bovine serum albumin (BSA) to 40 ng/mL and 11 ng/mL, respectively. [0550]
  • Antigen-positive (SB) and antigen-negative (HSB) cells were maintained in RPMI 1640 supplemented with 10% fetal calf serum at 37° C. and 5% CO[0551] 2. Cells (viability >90% as determined by trypan blue exclusion) were harvested at ambient temperature at a cell density of 0.5-2×106 cells/mL by centrifugation (1300 rpm in a Sorvall centrifuge) and washed twice with 50 mL 1× HBSS. Pelleted cells were resuspended to 50×106 cells/mL in prechilled 1× HBSS containing 1% (w/v) bovine serum albumin (BSA) and 10% (w/v/) mannitol (lyophilization buffer). Cell suspensions were dispensed into 1.5 mL polypropylene microfuge tubes with o-ring gaskets at 50×106 cells/mL (0.5 mL per tube) and lyophilized overnight at 30 to 60 millitorr. Lyophilized cells were stored desiccated at 2-8° C. and reconstituted in sterile water for assays. Lyophilized SB and HSB cells in 1.5 mL polypropylene tubes were reconstituted to 50×106 cells/mL using sterile water. Diluted In2B8 or Y2B8 was added to cells, in triplicate, and incubated for 45 to 60 min with end-over-end mixing at ambient temperature, respectively. After incubation, cells were pelleted by centrifugation and cell-bound radioactivity determined by counting samples in an Isodata Gamma Counter or a Packard Top Count scintillation counter as described below. Radioactivity bound (B) to cells was calculated by subtracting the unbound radioactivity (supernatant) from the total radioactivity added. Total radioactivity was determined from the radioactivity counted in tubes without cells. Percent binding was calculated by expressing the bound counts as a percentage of the total counts.
  • 9. Radioactivity Measurement [0552]
  • Radioincorporation samples were counted for 1 min. using an Isodata gamma counter. The counter was set for energy windows of 100-500 KeV and the background adjusted to zero immediately before use for samples using [0553] 111In. The Isodata gamma counter was also used for counting ITLC strips having 90Y spotted on them. The energy windows for detection of the bremstrulung radiation were 100-1000 KeV.
  • For the binding assays, 90Y samples were transferred to 24-well plates and [0554] MicroScint 40 cocktail and counted in a Packard TopCount for 1 min using minimum and maximum energy settings. Indium-[111] samples were counted for 1 min. using an Isodata gamma counter. The counter was set for energy windows of 100-500 KeV and the background adjusted to zero immediately before use.
  • 10. Release Specifications for Clinical Doses of In2B8 and Y2B8 [0555]
  • Release specifications for radioincorporation and binding to CD20-positive cells were established by preparing six doses each of In2B8 and Y2B8 using two lots of clinical-grade 2B8-MX-DTPA (lot #'s 0219 and 0220) prepared according to the present invention and filled under GMP conditions. Release assays were performed as described above. [0556]
  • IV. Results [0557]
  • A. Characterization of CHO-Derived 2B8 [0558]
  • Using flow cytometric analysis, it was demonstrated that CHO 2B8 binds directly to CD20-positive SB cells without binding to CD20-negative HSB cells (FIG. 34). No significant binding to SB or HSB cells was noted for art irrelevant isotype (γ1κ) antibody (S004). [0559]
  • Binding of CHO 2B8 to CD20-positive cells was evaluated in competition assays using the ORIGEN chemiluminescent detection system. Lyophilized and reconstituted antigen-positive SB cells were incubated with increasing amounts bf antibody in the presence of ruthenium-labeled CHO 2B8 tracer. Results showed that CHO 2B8 inhibits binding to CD20-positive cells to the same extent as the antibody derived from hollow-fiber bioreactors (2B8-49) (FIG. 35). The EC50 values were determined graphically and the method of Muller (1980) used to calculate average affinity values. The affinity for CHO 2B8 was determined to be 1.3×10[0560] −10 M; the 2B8 antibody derived from hollow-fiber bioreactors gave an affinity value of 2.5 10−10 M. Non-specific binding was negligible as demonstrated by the lack of competition with the irrelevant isotype antibody, S004.
  • B. Characterization of CHO-Derived 2B8-MX-DTPA [0561]
  • The 2B8 conjugate (2B8-MX-DTPA) was prepared using a protocol similar to that used for the previously characterized 2B8-49. Reactions were performed using approximately 3 mg of antibody and a 4:1 molar ratio of chelator to antibody. Incubations times of 2, 4, 8, 17, and 24 h were evaluated to determine the reaction time giving acceptable retention of binding to CD20 positive cells and high radioincorporation with [0562] 111In. Competitive binding curves comparing CHO 2B8 to CHO 2B8-MX-DTPA conjugate reacted for 8-24 h were similar, indicating that the conjugation process did not significantly alter the binding of the antibody to the CD20 antigen (FIG. 36). Using EC 50 values determined graphically (FIG. 36), affinity constants for the unconjugated and conjugated antibodies ranged from 2.3×10−10 M to 5.9×10−10 M (Table 40). Radioincorporation was >95% for conjugation times of 8 to 24 h (Table 30).
    TABLE 40
    Effect of Conjugation Reaction Time on Radioincorporation and
    Immunoreactivity of CHO 2B8-MX-DTPA
    Incubation Time (h) Radioincorporation (%) Affinity (M)
    0 ND 2.3 × 10−10
    2 83.5 ND
    4 90.5 ND
    8 96.1 5.9 × 10−10
    17 97.3 5.9 × 10−10
    24 98.8 4.4 × 10−10
  • C. Characterization of In2B8 and Y2B8 Prepared From CHO-Derived 2B8-MX-DTPA [0563]
  • Indium-[111]-labeled CHO 2B8-MX-DTPA (In2B8) was prepared using the small-scale radiolabeling kit protocol previously described for the hollow-fiber bioreactor-derived antibody (Example 1). Briefly, conjugated antibody (CHO-derived 2B8-MX-DTPA; lot # 0165A) was incubated with [0564] 111In acetate at the indicated pH for 30 minutes at ambient temperature. Reaction mixtures were formulated with PBS, pH 7.2, containing 7.5% (w/v) human serum albumin and 1 mM DTPA. Formulated samples of In2B8 were assayed for radioincorporation using instant thin-layer chromatography. Binding of In2B8 to CD20-positive cells was determined using lyophilized and reconstituted SB cells. For comparison, the conjugate prepared from hybridoma-produced antibody(2B8-49) was incubated with 111In acetate for 30 min. at pH 4.2 (conditions previously established for this antibody).
  • Kinetics studies were performed to determine labeling conditions providing maximal retention of binding to CD20-positive cells and high radioincorporation (Tables 41 and 42). Conjugated antibody (CHO-derived 2B8-MX-DTPA) was incubated at ambient temperature with [0565] 111In acetate at pH 4.2 for the times indicated (Table 42).
    TABLE 41
    In2B8 Radiolabeling Kinetics: Effect of pH on Radioincorporation
    and Binding to CD20-Positive Cells
    Reaction pH Radioincorporation (%) Binding (%)
    3.0 97.7 85.3
    3.7 98.5 83.9
    4.0 98.6 84.1
    4.3 98.0 84.0
    4.6 98.9 83.4
    Control (2B8-49) 99.3 86.5
  • [0566]
    TABLE 42
    In2B8 Radiolabeling Kinetics: Effect of Incubation Time on
    Radioincorporation and Binding to CD20-Positive Cells
    Incubation Time (min) Radioincorporation (%) Binding (%)
    pH 2.9: 15 97.2 85.3
    30 99.1 85.2
    45 97.2 84.8
    pH 4.6: 15 99.0 87.2
    30 97.2 86.8
    45 99.4 86.3
    Control (2B8-49) 99.4 87.8
  • Results demonstrated that for the range of pH 3.0 to 4.6 and an incubation time of 30 min, >97% radioincorporation of the radioisotope was attained while maintaining binding at approximately 84%. Radioincorporation and binding values were invariant for incubation times of 15 to 45 min for reactions at pH 2.9 to 4.6 (Table 42). Results were comparable to those obtained using the 2B8-49 antibody (Tables 41 and 42). [0567]
  • Yttrium-[90]-labeled antibody was prepared by incubating conjugated antibody (CHO-derived 2B8-MX-DTPA) with [0568] 90Y acetate at the indicated pH for 5 minutes at ambient temperature. Reaction mixtures were formulate in PBS, pH 7.2 containing 7.5%(w/v) human serum albumin and 1 mM DTPA. Formulated samples of Y2B8 were assayed for radioincorporation using instant thin-layer chromatography. Binding of Y2B8 to CD20-positive cells was determined using lyophilized and reconstituted SB cells. For comparison, the conjugate prepared from hybridoma-produced antibody (2B8-49) was incubated with 90Y acetate for 5 min. at pH 4.2 (conditions previously established for this antibody).
  • Similar kinetic studies were performed to evaluate the preparation of the [0569] 90Y-labeled antibody (Y2B8). For radiolabeling reactions in the range of pH 3.9 to 4.7 at an incubation time of 5 min, radioincorporation was >96% with >80% retention of binding to CD20-positive cells (Table 43). Similar results were obtained for incubation times of 3, 5, and 10 min for the range of pH 2.9 to 4.6 (Table 44). Then, conjugated antibody (CHO-derived 2B8-MX-DTPA) was incubated at ambient temperature with 90Y acetate at pH 4.2 for the times indicated (Table 44). Results were comparable to those obtained using the 2B8-49 antibody (Tables 43 and 44).
    TABLE 43
    Y2B8 Radiolabeling Kinetics: Effect of pH on Radioincorporation
    and Binding to CD20-Positive Cells
    Reaction pH Radioincorporation (%) Binding (%)
    3.9 98.4 80.7
    4.2 97.8 81.0
    4.4 96.1 80.0
    4.6 97.0 80.2
    4.7 97.4 80.6
    Control (2B8-49) 99.3 82.6
  • [0570]
    TABLE 44
    Y2B8 Radiolabeling Kinetics: Effect of Incubation Time on
    Radioincorporation and Binding to CD20-Positive Cells
    Incubation Time (min) Radioincorporation (%) Binding (%)
    pH 3.9: 3 97.0 82.0
    5 98.9 82.1
    10 99.2 82.3
    pH 4.7: 3 97.2 82.5
    5 96.7 81.8
    10 97.6 81.5
    Control (2B8-49) 99.2 84.2
  • Immunoreactivities for In2B8 and Y2B8 prepared from CHO 2B8 were determined using the method of Lindmo et al. Increasing amounts of freshly harvested CD20-positive SB cells were incubated with a fixed amount of In2B8 or Y2B8 under conditions of antigen excess. Reciprocal plot analysis of the binding data allowed immunoreactivities of 80.6% and 72.2% for In2B8 and Y2B8, respectively, to be determined (FIGS. 37 and 38). [0571]
  • D. Release Specifications for CHO-Derived In2B8 and Y2B8 [0572]
  • Two lots of clinical-grade In2B8/Y2B8 radiolabeling kits were used to prepare six lots each of In2B8 and Y2B8. In2B8 and Y2B8 were prepared using small-scale versions of the radiolabeling protocols currently used in the clinical trials. Each lot of radiolabeled 2B8-MX-DTPA was tested for radioincorporation and binding to CD20-positive (SB) and CD20-negative (HSB) human cells. These results are summarized in Tables 45 and 46. For the six lots of In2B8 prepared, radioincorporation ranged from 98.9% to 99.3% with a mean of 99. 1%. Binding to CD20-positive cells ranged from 81.9% to 85.1% with a mean of 83.6%; binding to CD20-negative cells was <4%. For the six lots of Y2B8 prepared, radioincorporation ranged from 97.4% to 98.7% with a mean of 98.2%. Binding to CD20-positive cells ranged from 81.4% to 82.7% with a mean of 81.9%; binding to CD20-negative cells was <8%. [0573]
    TABLE 45
    Release Assay Results for CHO-Derived In2B8 Prepared
    Using the Radiolabeling Kit Protocol
    Radio- Binding (%)
    Run # incorporation (%) SB Cells HSB Cells
    #1 (Lot #0219) 99.1 81.9 2.8
    #2 (Lot #0219) 99.3 83.2 2.8
    #3 (Lot #0219) 99.2 83.6 3.7
    #4 (Lot #0220) 99.0 83.8 2.6
    #5 (Lot #0220) 98.9 84.1 2.6
    #6 (Lot #0220) 98.9 85.1 3.3
    Mean = 99.1% Mean = 83.6% Mean = 2.9%
    SD = 0.2% SD = 1.1% SD = 0.4%
  • [0574]
    TABLE 46
    Release Assay Results for CHO-Derived Y2B8 Prepared
    Using the Radiolabeling Kit Protocol
    Radio- Binding (%)
    Run # incorporation (%) SB Cells HSB Cells
    #1 (Lot #0219) 98.7 82.1 7.4
    #2 (Lot #0219) 98..6 82.7 0.7
    #3 (Lot #0219) 98.3 82.2 7.2
    #4 (Lot #0220) 97.4 81.8 1.7
    #5 (Lot #0220) 97.6 81.4 2.2
    #6 (Lot #0220) 98.4 81.4 1.1
    Mean = 98.2% Mean = 81.9% Mean = 3.4%
    SD = 0.5% SD = 0.5% SD = 3.1%
  • V. Discussion and Conclusions [0575]
  • The anti-CD20 murine monoclonal antibody (2B8) cloned and expressed in CHO cells (CHO-derived 2B8) maintains specificity for CD20-positive human cells as shown by FACS and competitive binding analysis. Binding to human T-cells was minimal. The affinity of the antibody for human CD20-positive cells was determined to be 1.3×10[0576] −10 M using a competitive binding assay. Using the same assay, the 2B8 antibody derived from hollow-fiber bioreactors gave an affinity value of 2.5×10−10 M.
  • The CHO 2B8 antibody was reacted with MX-DTPA to form a conjugate, 2B8-MX-DTPA, while maintaining suitable retention of immunoreactivity. Optimal chelator incorporation was determined by measuring radioincorporation with [0577] 111In and was achieved after eight hours incubation at ambient temperature. Radiolabeling protocols for the 2B8-MX-DTPA conjugate were optimized for 90Y or 111In with respect to pH and incubation time to insure maximal radioincorporation and retention of immunoreactivity.
  • The results of several preparations of In2B8 and Y2B8 demonstrate the reproducibility of the radiolabeling protocol used to prepare clinical doses. Based on these radiolabeling results, it is suggested that release specifications for radioincorporation and binding, using lyophilized CD20-positive cells, be established at ≧95% and ≧70%, respectively. Taken together, these results demonstrate the comparability of CHO-derived 2B8 and hollow-fiber-derived 2B8-49, and indicate the suitability of the CHO-derived 2B8-MX-DTPA for use in clinical trials. [0578]
  • Finally, the present invention discloses a labeling procedure, referred to as the “mix-and-shoot” method, for the preparation of clinical doses of radiolabeled antibodies which obviates the need for the currently used high performance liquid chromatographic (HPLC) step for removal of non-protein bound radioisotope. The simplified protocol eliminates this laborious purification step while maintaining a high level of radioisotope incorporation (>95%) and improved retention of immunoreactivity (>70%). The clinically-formulated radiolabeled conjugate was found to be stable in vitro when incubated at 4° C. for 48 hours based on retention of radioisotope and immunoreactivity. Additionally, the radiolabeled conjugate was stable when incubated in human serum at 37° C. for 72 hours. Biodistribution studies in -BALB/c mice demonstrated no unusual tissue deposition, and no significant accumulation in the bone. Estimates of radiation absorbed doses to a “standard” 70 Kg human were comparable to those obtained in an on-going clinical trial using [0579] 90Y-labeled 2B8-MX-DTPA. The results of these studies showed that 90Y-labeled 2B8-MX-DTPA produced using the “mix-and-shoot” protocol was comparable to that prepared using the conventional HPLC process. Validation of the scale-up protocol for preparing clinical-grade radiolabeled conjugate showed that the method was reproducible and that the product was comparable to that produced using the current HPLC method. The results of these pre-clinical studies indicate that this new “mix-&-shoot” protocol can be used to prepare 90Y-labeled 2B8-MX-DTPA suitable for use in clinical trials.
  • REFERENCES
  • Each of the following citations is herein incorporated by reference: [0580]
  • 1. Adams, R. A., Flowers, A., and Davis, B. J. Direct Implantation and Transplantation of Human Acute Lymphoblastic Leukemia in Hamsters, SB-2. CancerResearch28: 1121-1125, 1968. [0581]
  • 2. Adams, R. A. Formal Discussion: The Role of Transplantation in the Experimental Investigation of Human Leukemia and Lymphoma. Cancer Res. 27(1): 2479-2482, 1967. [0582]
  • 3. Lindmo, T., Boven, E., Cuttitta, F., Fedoroko, J., and Bunn, P. A., J. Immunol. Methods, 72: 77-1984. [0583]
  • 4. Kozak, R. W., Raubitschek, A., Mirzadeh, S., Brechbiel, M. W., Junghaus, R., Gansow, O. A., and Waldmann, T. A. Cancer Res. (1989) 49:2639-2644. [0584]
  • 5. Parker, B. A., Halpern, S. E., Miller, R. A., Hupf, H., Shawler, D. L., Collins, H. A., Amox, D., White, C. A. and Royston, I. N. Eng. J. Med., submitted. [0585]
  • 6. Leland, J. K. and Powell, M. J. J. (1990) Electrochem. Soc. 137, 3127. [0586]
  • 7. Muller, R. J. Immunological Methods (1980) 34, 345. [0587]
  • 8. Mirzadeh, S., Brechbiel, M. W., Atcher, R. W. and Gansow, O. A. (1990) Bioconjugate Chemistry 1(1), 59. [0588]
  • 9. Brechbiel, M. W., Gansow, O. A., Atcher, R. W., Sclom, J., Esteban, J., Simpson, D. E. and Colcher, D. (1986) 25, 2772. [0589]

Claims (82)

What is claimed:
1. A kit for radiolabeling an anti-CD20 antibody before administration to a patient comprising:
(i) a vial containing a chelator-conjugated antibody,
(ii) a vial containing formulation buffer for stabilizing and administering the radiolabeled antibody to a patient, and
(iii) instructions for radiolabeling the antibody,
wherein said vial components are supplied in such an amount and at such a concentration that when they are combined with a radiolabel according to the kit instructions, no further purification of the labeled antibody is required before administration to said patient.
2. The radiolabeling kit of claim 1, wherein the antibody is a chimeric anti-CD20 antibody.
3. The radiolabeling kit of claim 1, wherein the chelator is selected from the group consisting of MX-DTPA, phenyl-DTPA, benzyl-DTPA, CHX-DTPA and DOTA.
4. The radiolabeling kit of claim 1 further comprising a vial containing a buffer for adjusting the pH of the radioisotope.
5. The radiolabeling kit of claim 4, wherein the buffer is a sodium acetate solution.
6. The radiolabeling kit of claim 1 further comprising a reaction vial.
7. The radiolabeling kit of claim 3, wherein the chelator is MX-DTPA.
8. The radiolabeling kit of claim 1, wherein the antibody is 2B8.
9. The radiolabeling kit of claim 8, wherein the chelator-conjugated antibody is 2B8-MX-DTPA.
10. The kit of claim 1 wherein the antibody conjugate is supplied at a concentration of about 0.5 to 30 mg/ml.
11. The radiolabeling kit of claim 5 wherein the sodium acetate solution is at a concentration of 10 to 1000 mM.
12. The radiolabeling kit of claim 11 wherein the sodium acetate solution is at a concentration of 50 mM.
13. The radiolabeling kit of claim 1 wherein the formulation buffer comprises a radioprotectant and non-protein-conjugated chelator.
14. The radiolabeling kit of claim 13 wherein the radioprotectant is selected from the group consisting of human serum albumin (HSA), ascorbate, ascorbic acid, phenol, sulfites, glutathione, cysteine, gentisic acid, nicotinic acid, ascorbyl palmitate, HOP(:O)H2, glycerol, sodium formaldehyde sulfoxylate, Na2S2O5, Na2S2O3, and SO2.
15. The radiolabeling kit of claim 14 wherein the radioprotectant is HSA.
16. The radiolabeling kit of claim 13 wherein the unconjugated chelator is DTPA or EDTA.
17. The radiolabeling kit of claim 15 wherein the HSA is at a concentration of about 1 to 25% (w/v).
18. The radiolabeling kit of claim 17 wherein the concentration of HSA is about 7.5% (w/v).
19. The radiolabeling kit of claim 14 wherein the radioprotectant is ascorbate.
20. The radiolabeling kit of claim 19 wherein the ascorbate is at a concentration of about 1 to 100 mg/ml.
21. The radiolabeling kit of claim 1 wherein the radioisotope is 111In chloride.
22. The radiolabeling kit of claim 1 wherein the radioisotope is 90Y chloride.
23. A formulation buffer for administering a radiolabeled chelator-conjugated antibody to a patient comprising:
(i) a physiological salt solution;
(ii) a radioprotectant; and
(iii) non-protein conjugated chelator.
24. The formulation buffer of claim 23 wherein the radioprotectant is selected from the group consisting of HSA, ascorbate, ascorbic acid, phenol, sulfites, glutathione, cysteine, gentisic acid, nicotinic acid, ascorbyl palmitate, HOP(:O)H2, glycerol, sodium formaldehyde sulfoxylate, Na2S2O5, Na2S2O3, and SO2.
25. The formulation buffer of claim 23 wherein the chelator is DTPA.
26. The formulation buffer of claim 24 wherein the radioprotectant is HSA.
27. The formulation buffer of claim 24 wherein the radioprotectant is ascorbate.
28. The formulation buffer of claim 26 wherein the concentration of human serum albumin is about 1 to 25% (w/v).
29. The formulation buffer of claim 28 wherein the HSA concentration is about 7.5%.
30. The formulation buffer of claim 25 wherein the concentration of DTPA is about 1 mM.
31. The formulation buffer of claim 27 wherein the concentration of ascorbate is about 1 to 100 mg/mL.
32. A method for radiolabeling a chelator-conjugated anti-CD20 antibody for administration to a patient comprising
(i) mixing chelator-conjugated antibody with a solution containing a radiolabel;
(ii) incubating the mixture for an appropriate amount of time at appropriate temperature; and
(iii) diluting the labeled antibody to an appropriate concentration in formulation buffer, such that said radiolabeled antibody may be administered directly to the patient without further purification.
33. The method of claim 32 wherein the antibody is a chimeric anti-CD20 antibody.
34. The method of claim 32 wherein the anti-CD20 antibody is 2B8-MX-DTPA.
35. The method of claim 32 wherein the formulation buffer contains physiological saline, a radioprotectant, and unconjugated chelator.
36. The method of claim 35 wherein the radioprotectant is selected from the group consisting of human serum albumin (HSA), ascorbate, ascorbic acid, phenol, sulfites, glutathione, cysteine, gentisic acid, nicotinic acid, ascorbyl palmitate, HOP(:O)H2, glycerol, sodium formaldehyde sulfoxylate, Na2S2O5, Na2S2O3, and SO2.
37. The method of claim 35 wherein the unconjugated chelator is DTPAor EDTA.
38. The method of claim 32 wherein the solution containing the radiolabel is adjusted to a pH of about 3 to 6 before it is mixed with the chelator-conjugated antibody.
39. The method of claim 38 wherein the pH is adjusted with a low metal sodium acetate solution.
40. The method of claim 39 wherein the sodium acetate is at a concentration of about 10 to 1000 mM.
41. The method of claim 32 wherein the radiolabel is 111In chloride.
42. The method of claim 41 wherein the volume quantity of 111In chloride used is about 4-6 mCi divided by the radioactivity concentration at the time of labeling.
43. The method of claim 41 wherein about 1 ml of chelator-conjugated antibody at a concentration of about 0.5 to 30 mg/ml is mixed with the radiolabel solution.
44. The method of claim 43 wherein the mixture is incubated for between about 30 seconds and 60 minutes.
45. The method of claim 44 wherein formulation buffer is added in an amount necessary to achieve a total final volume of about 10 ml to about 50 ml.
46. The method of claim 32 wherein the radiolabel is 90Y chloride.
47. The method of claim 46 wherein the volume quantity of 90Y chloride used is between about 5 to 100 mCi divided by the radioactivity concentration at the time of labeling.
48. The method of claim 47 wherein the volume quantity of 90Y chloride used is about 45 mCi divided by the radioactivity concentration at the time of labeling.
49. The method of claim 46 wherein about 1 to 2 ml of MX-DTPA-conjugated antibody at a concentration of about 0.5 to 30 mg/ml is mixed with the radiolabel solution.
50. The method of claim 49 wherein the mixture is incubated for a time between about 30 seconds to 60 minutes.
51. The method of claim 50 wherein formulation buffer is added in an amount necessary to achieve a total final volume of about 10 ml to about 50 ml.
52. A binding assay and radiolabeling kit for radiolabeling an anti-CD20 antibody and determining the percent binding of the radiolabeled antibody to its target cell before administering the antibody to a patient, comprising the following components:
(i) at least one vial of fixed or lyophilized antigen-positive cells;
(ii) a vial containing a chelator-conjugated antibody;
(iii) a vial containing formulation buffer; and
(iv) instructions for radiolabeling the antibody,
wherein said vial components are supplied in such an amount and at such a concentration that when they are combined with a radiolabel according to the kit instructions, no further purification of the labeled antibody is required before administration to said patient.
53. The binding assay and radiolabeling kit of claim 52, wherein said antibody is a chimeric anti-CD20 antibody.
54. The binding assay and radiolabeling kit of claim 52 further comprising a vial containing a buffer for adjusting the pH of the radiolabel.
55. The binding assay and radiolabeling kit of claim 52 wherein the formulation buffer is phosphate buffered saline comprising a radioprotectant and unconjugated chelator.
56. The binding assay and radiolabeling kit of claim 52, wherein the antigen positive cells are CD20-positive cells.
57. The binding assay and radiolabeling kit of claim 56 wherein the CD20-positive cells are SB cells (ATCC # CCL 120).
58. The binding assay and radiolabeling kit of claim 52 further comprising at least one vial of antigen-negative cells.
59. The binding assay and radiolabeling kit of claim 58, wherein said antigen-negative cells are CD20-negative cells.
60. The binding assay and radiolabeling kit of claim 59 wherein said CD20-negative cells are HSB cells (ATCC # CCL120.1).
61. A binding assay kit for determining the percent binding of a radiolabeled anti-CD20 antibody to its target cell comprising at least one vial of fixed or lyophilized antigen-positive cells.
62. The binding assay kit of claim 61, further comprising a control anti-CD20 antibody.
63. The binding assay kit of claim 62 wherein the CD20-positive cells are SB cells (ATCC # CCL 120).
64. The binding assay kit of claim 61 further comprising antigen-negative cells.
65. The binding assay kit of claim 64, wherein said cells are CD20-negative.
66. The binding assay kit of claim 65 wherein said CD20-negative cells are HSB cells (ATCC # CCL120.1).
67. A binding assay for determining the percent binding of a radiolabeled antibody to its target cell, comprising:
(i) mixing and incubating at least one aliquot of a radiolabeled antibody with at least one aliquot of antigen-positive cells;
(ii) mixing and incubating at least one aliquot of a radiolabeled antibody identical to the aliquot of step (i) with at least one aliquot of dilution buffer of the same volume as the aliquot of antigen-positive cells in step (i) as a control;
(iii) pelleting the cells by centrifugation;
(iv) measuring the radioactivity in the supernatant of the pelleted cells and the control; and
(v) comparing the quantity of radioactivity in the cell supernatant to the quantity of radioactivity in the control.
68. The binding assay of claim 67 wherein said antibody is a CD20 antibody.
69. The binding assay of claim 68 wherein the anti-CD20 antibody is 2B8.
70. The binding assay of claim 67 wherein said antigen is CD20.
71. The binding assay of claim 70 wherein said CD20 positive cells are SB cells (ATCC # CCL 120).
72. The binding assay of claim 67 further comprising
(i) mixing at least one aliquot of the radiolabeled antibody with at least one aliquot of antigen-negative cells;
(ii) pelleting the antigen-negative cells by centrifugation;
(iv) measuring the radioactivity in the supernatant of the antigen-negative pelleted cells; and
(v) comparing the quantity of radioactivity in the antigen-negative cell supernatant to the quantity of radioactivity in the supernatant of the antigen-positive cell supernatant and the control.
73. The binding assay of claim 72 wherein said antigen negative cells are CD20-negative.
74. The binding assay of claim 73 wherein said CD20 negative cells are HSB cells (ATCC # CCL 120.1).
75. A binding assay for determining the percent binding of a radiolabeled antibody to its target cell, comprising:
(i) mixing at least one aliquot of a radiolabeled antibody with at least one aliquot of antigen-positive cells;
(ii) mixing at least one aliquot of the radiolabeled antibody identical to the aliquot of step (i) with at least one aliquot of dilution buffer of the same volume as the aliquot of antigen-positive cells in step (i) as a control;
(iii) pelleting the cells by centrifugation;
(iv) measuring the radioactivity in the supernatant of the pelleted cells and the control; and
(v) comparing the quantity of radioactivity in the cell supernatant to the quantity of radioactivity in the control;
wherein said assay is performed using the binding assay and radiolabeling kit of claim 52.
76. A binding assay for determining the percent binding of a radiolabeled antibody to its target cell , comprising:
(i) mixing at least one aliquot of a radiolabeled antibody with at least one aliquot of antigen-positive cells;
(ii) mixing at least one aliquot of a radiolabeled antibody identical to the aliquot of step (i) with at least one aliquot of dilution buffer of the same volume as the aliquot of antigen-positive cells in step (i) as a control;
(iii) pelleting the cells by centrifugation;
(iv) measuring the radioactivity in the supernatant of the pelleted cells and the control; and
(v) comparing the quantity of radioactivity in the cell supernatant to the quantity of radioactivity in the control;
wherein said assay is performed using the binding assay kit of claim 61.
77. A competitive binding assay for assessing affinity of a test antibody to a target cell, comprising
(i) preparing a ruthenium-labeled control antibody;
(ii) incubating increasing amounts of test antibody and increasing amounts of unlabeled control antibody with a fixed concentration of fixed, fresh or lyophilized antigen-positive cells and a trace amount of ruthenium-labeled antibody wherein each separate concentration of test antibody and each separate concentration of control antibody are in separate tubes, respectively;
(iii) determining the quantity of binding in each reaction tube based on relative electrochemiluminescence (ECL) using ORIGEN instrumentation; and
(iv) calculating the average affinity value of the test antibody.
78. The competitive binding assay of claim 77 wherein the control and test antibodies are anti-CD20 antibodies.
79. A competitive binding assay of claim 77 wherein the antigen-positive cells as CD20-positive.
80. The competitive binding assay of claim 79 wherein the antigen-positive cells are SB cells (ATCC # CCL 120).
81. The radiolabeling kit of claim 1 further comprising a vial of chimeric anti-CD20 antibody to be administered in a combined therapeutic regimen prior to or subsequent to the radiolabeled antibody.
82. The binding assay and radiolabeling kit of claim 52, further comprising a vial of chimeric anti-CD20 antibody to be administered in a combined therapeutic regimen prior to or subsequent to the radiolabeled antibody.
US09/259,337 1999-03-01 1999-03-01 Radiolabeling kit and binding assay Abandoned US20020102208A1 (en)

Priority Applications (50)

Application Number Priority Date Filing Date Title
US09/259,337 US20020102208A1 (en) 1999-03-01 1999-03-01 Radiolabeling kit and binding assay
MYPI20000754A MY138674A (en) 1999-03-01 2000-02-28 Radiolabeling kit and binding assay
SK91-2010A SK288096B6 (en) 1999-03-01 2000-02-29 Method for radiolabeling chelator-conjugated antibody and binding assay
SK1216-2001A SK287745B6 (en) 1999-03-01 2000-02-29 Method for radiolabeling chelator-conjugated antibody and binding assay
MEP-2009-21A ME00782B (en) 1999-03-01 2000-02-29 Antibody radiolabeling kit and method and cell-binding assay method for evaluating the lmmunoreactivity of radiolabeling antibodies
NZ530868A NZ530868A (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
AU35052/00A AU776747B2 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
YU61601A RS51778B (en) 1999-03-01 2000-02-29 PROCESS FOR RADIOLABELING A CHELATOR-CONJUGATED ANTIBODY WITH 111In
AT00913645T ATE433108T1 (en) 1999-03-01 2000-02-29 RADIOM MARKING SET AND BINDING TEST
SI200031085T SI2112512T1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
SI200031036T SI1192463T1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
MXPA01008837A MXPA01008837A (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay.
CZ20013126A CZ303262B6 (en) 1999-03-01 2000-02-29 Method for radiolabeling a chelator-conjugated antibody and antibody binding assay
CNB008057397A CN1235639C (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
KR1020017011093A KR100729247B1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
DE60042326T DE60042326D1 (en) 1999-03-01 2000-02-29 RADIOMARKING KIT AND BINDING TEST
BRPI0017596A BRPI0017596B8 (en) 1999-03-01 2000-02-29 method for radiolabeling an antibody conjugated to a 111in chelator for administration to a patient
UA2001096636A UA78484C2 (en) 1999-03-01 2000-02-29 Method of radioactive marking of cd 20 antibody
IL14510900A IL145109A0 (en) 1999-03-01 2000-02-29 A radiolabeling kit and binding assay for antibodies
EEP200100465A EE05650B1 (en) 1999-03-01 2000-02-29 Antibody radioactive labeling kit and binding assay
PT00913645T PT1192463E (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
NZ513788A NZ513788A (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
PL352531A PL205780B1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
CZ2010-746A CZ306959B6 (en) 1999-03-01 2000-02-29 A kit for radionuclide and immunoassay labelling
EP00913645A EP1192463B1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
PT91585141T PT2112512E (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
DK09158514.1T DK2112512T3 (en) 1999-03-01 2000-02-29 Kit for radiolabeling and binding assay
JP2000602634A JP4704570B2 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
BRPI0008719-0 BRPI0008719B8 (en) 1999-03-01 2000-02-29 Method for radiolabeling an antibody conjugated to a chelator with 90y and binding assay for determining the percentage binding of a radiolabeled antibody to its target cell
CA2742153A CA2742153C (en) 1999-03-01 2000-02-29 Method for radiolabeling a chelator-conjugated antibody with 111in
DK00913645T DK1192463T3 (en) 1999-03-01 2000-02-29 Kit and binding assay two radioactive labeling
EP09158514.1A EP2112512B1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
ES00913645T ES2328100T3 (en) 1999-03-01 2000-02-29 RADIOOMARCAJE KIT AND UNION TEST.
ES09158514.1T ES2526723T3 (en) 1999-03-01 2000-02-29 Radiolabeling and binding test kit
HU1300275A HU230516B1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
CA2362119A CA2362119C (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
HU0202522A HU229627B1 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
PCT/US2000/005061 WO2000052473A2 (en) 1999-03-01 2000-02-29 Radiolabeling kit and binding assay
RU2001126400/15A RU2251110C2 (en) 1999-03-01 2000-02-29 Radioactive labeling set and binding analysis
TW089103582A TWI294969B (en) 1999-03-01 2000-03-01
ZA200106943A ZA200106943B (en) 1999-03-01 2001-08-22 Radiolabeling kit and binding assay.
NO20014245A NO329142B1 (en) 1999-03-01 2001-08-31 Method for radiolabeling a 90Y chelator-conjugated antibody and binding assay
BG105957A BG65690B1 (en) 1999-03-01 2001-09-27 Radiolabeled kit and method of testing the conjugation
HR20010714A HRP20010714B1 (en) 1999-03-01 2001-10-01 Radiolabeling kit and binding assay
HK02107258.0A HK1045730B (en) 1999-03-01 2002-10-03 Radiolabeling kit and binding assay
US11/033,439 US7608241B2 (en) 1999-03-01 2005-01-12 Radiolabeling method
CY20091100912T CY1109335T1 (en) 1999-03-01 2009-09-01 RADIO LABELING KIT AND COMMITMENT TEST
HK09110560.0A HK1131208A1 (en) 1999-03-01 2009-11-12 Radiolabeling kit and binding assay
NO20100717A NO330705B1 (en) 1999-03-01 2010-05-18 Method of radiolabeling antibody with 111In and binding assay for determination of such antibody
CY20151100160T CY1116265T1 (en) 1999-03-01 2015-02-16 RADIO-LABELING KIT AND DETERMINATION IDENTIFICATION

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09/259,337 US20020102208A1 (en) 1999-03-01 1999-03-01 Radiolabeling kit and binding assay

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/033,439 Division US7608241B2 (en) 1999-03-01 2005-01-12 Radiolabeling method

Publications (1)

Publication Number Publication Date
US20020102208A1 true US20020102208A1 (en) 2002-08-01

Family

ID=22984524

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/259,337 Abandoned US20020102208A1 (en) 1999-03-01 1999-03-01 Radiolabeling kit and binding assay
US11/033,439 Expired - Fee Related US7608241B2 (en) 1999-03-01 2005-01-12 Radiolabeling method

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/033,439 Expired - Fee Related US7608241B2 (en) 1999-03-01 2005-01-12 Radiolabeling method

Country Status (35)

Country Link
US (2) US20020102208A1 (en)
EP (2) EP2112512B1 (en)
JP (1) JP4704570B2 (en)
KR (1) KR100729247B1 (en)
CN (1) CN1235639C (en)
AT (1) ATE433108T1 (en)
AU (1) AU776747B2 (en)
BG (1) BG65690B1 (en)
BR (2) BRPI0008719B8 (en)
CA (2) CA2742153C (en)
CY (2) CY1109335T1 (en)
CZ (2) CZ303262B6 (en)
DE (1) DE60042326D1 (en)
DK (2) DK1192463T3 (en)
EE (1) EE05650B1 (en)
ES (2) ES2526723T3 (en)
HK (2) HK1045730B (en)
HR (1) HRP20010714B1 (en)
HU (2) HU230516B1 (en)
IL (1) IL145109A0 (en)
ME (1) ME00782B (en)
MX (1) MXPA01008837A (en)
MY (1) MY138674A (en)
NO (2) NO329142B1 (en)
NZ (2) NZ530868A (en)
PL (1) PL205780B1 (en)
PT (2) PT1192463E (en)
RS (1) RS51778B (en)
RU (1) RU2251110C2 (en)
SI (2) SI1192463T1 (en)
SK (2) SK288096B6 (en)
TW (1) TWI294969B (en)
UA (1) UA78484C2 (en)
WO (1) WO2000052473A2 (en)
ZA (1) ZA200106943B (en)

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000052031A2 (en) 1999-03-01 2000-09-08 Idec Pharmaceuticals Corporation Kit for radiolabeling proteins with yttrium-90
US20030003097A1 (en) * 2001-04-02 2003-01-02 Idec Pharmaceutical Corporation Recombinant antibodies coexpressed with GnTIII
US20050123474A1 (en) * 2003-06-25 2005-06-09 Peregrine Pharmaceuticals Methods and apparatus for continuous large-scale radiolabeling of proteins
US20080089885A1 (en) * 2006-10-10 2008-04-17 Vaccinex, Inc. Anti-cd20 antibodies and methods of use
EP1918305A1 (en) 1999-08-11 2008-05-07 Biogen Idec Inc. New clinical parameters for determining hematologic toxicity prior to radioimmunotherapy
WO2010078526A1 (en) 2008-12-31 2010-07-08 Biogen Idec Ma Inc. Anti-lymphotoxin antibodies
EP2238986A2 (en) 2005-07-08 2010-10-13 Biogen Idec MA Inc. Sp35 antibodies and uses thereof
WO2012019128A1 (en) 2010-08-06 2012-02-09 Genzyme Corporation Vegf antagonist compositions and uses thereof
WO2012125408A1 (en) 2011-03-11 2012-09-20 Genzyme Corporation Pegylated apelin and uses thereof
WO2014009426A3 (en) * 2012-07-13 2014-04-03 Innate Pharma Screening of conjugated antibodies
EP2740744A2 (en) 2007-01-09 2014-06-11 Biogen Idec MA Inc. SP35 antibodies and uses thereof
EP2835053A1 (en) 2010-03-12 2015-02-11 Genzyme Corporation Combination therapy for treating breast cancer
WO2015131153A1 (en) * 2014-02-27 2015-09-03 Board Of Regents, The University Of Texas System Methods and compositions for isolating exosomes
EP2982695A1 (en) 2008-07-09 2016-02-10 Biogen MA Inc. Compositions comprising antibodies to lingo or fragments thereof
US9427478B2 (en) 2013-06-21 2016-08-30 Innate Pharma Enzymatic conjugation of polypeptides
WO2017019858A1 (en) 2015-07-30 2017-02-02 The Trustees Of The University Of Pennsylvania Single nucleotide polymorphic alleles of human dp-2 gene for detection of susceptibility to hair growth inhibition by pgd2
WO2017048902A1 (en) 2015-09-15 2017-03-23 Board Of Regents, The University Of Texas System T-cell receptor (tcr)-binding antibodies and uses thereof
US9717803B2 (en) 2011-12-23 2017-08-01 Innate Pharma Enzymatic conjugation of polypeptides
US9845363B2 (en) 2013-08-13 2017-12-19 Sanofi Antibodies to plasminogen activator inhibitor-1 (PAI-1) and uses thereof
US9856468B2 (en) 2010-07-09 2018-01-02 Bioverativ Therapeutics Inc. Processable single chain molecules and polypeptides made using same
US9889082B2 (en) 2006-06-16 2018-02-13 The Trustees Of The University Of Pennsylvania Methods and compositions for inhibiting or reducing hair loss, acne, rosacea, prostate cancer, and BPH
US10036010B2 (en) 2012-11-09 2018-07-31 Innate Pharma Recognition tags for TGase-mediated conjugation
US10064952B2 (en) 2014-10-09 2018-09-04 Genzyme Corporation Glycoengineered antibody drug conjugates
US10071169B2 (en) 2013-06-20 2018-09-11 Innate Pharma Enzymatic conjugation of polypeptides
US10183995B2 (en) 2012-05-23 2019-01-22 Argen-X N.V. IL-6 binding molecules
WO2019018633A1 (en) 2017-07-19 2019-01-24 Hoffman Technologies, Inc. Compositions for treating stress-related disorders
US10214589B2 (en) 2013-03-11 2019-02-26 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
US10278438B2 (en) 2015-08-31 2019-05-07 Grabbies Media, Llc Consumer usable device with redeemable member
US10308712B2 (en) 2014-03-27 2019-06-04 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
US10611824B2 (en) 2013-03-15 2020-04-07 Innate Pharma Solid phase TGase-mediated conjugation of antibodies
US10836813B2 (en) 2012-09-12 2020-11-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
US10995148B2 (en) 2014-03-19 2021-05-04 Genzyme Corporation Site-specific glycoengineering of targeting moieties
US11052177B2 (en) 2013-09-06 2021-07-06 The Trustees Of The University Of Pennsylvania Antimicrobial polymer layers
US11168125B2 (en) 2003-05-06 2021-11-09 Bioverativ Therapeutics Inc. Immunoglobulin chimeric monomer-dimer hybrids
US11186638B2 (en) 2011-09-12 2021-11-30 Genzyme Corporation Anti-αβTCR antibody
US11421026B2 (en) 2015-09-30 2022-08-23 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
US11427634B2 (en) 2017-05-05 2022-08-30 Vaccinex, Inc. Human anti-semaphorin 4D antibody
US11702470B2 (en) 2020-06-11 2023-07-18 Imperial College Innovations Limited Use of CXCL13 binding molecules to promote peripheral nerve regeneration
US11813338B2 (en) 2013-03-12 2023-11-14 The Trustees Of The University Of Pennsylvania Diagnosing and treating cancer
US11879004B2 (en) 2020-02-28 2024-01-23 Genzyme Corporation Modified binding polypeptides for optimized drug conjugation

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136311A (en) 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US6107090A (en) 1996-05-06 2000-08-22 Cornell Research Foundation, Inc. Treatment and diagnosis of prostate cancer with antibodies to extracellur PSMA domains
US20020102208A1 (en) * 1999-03-01 2002-08-01 Paul Chinn Radiolabeling kit and binding assay
US6207858B1 (en) * 1999-03-03 2001-03-27 Idec Pharmaceuticals Corporation Regioselective synthesis of DTPA derivatives
ES2357051T3 (en) 2000-10-02 2011-04-15 Novartis Vaccines And Diagnostics, Inc. ANTI-CD40 HUMAN ANTIBODIES.
JP2005527474A (en) 2001-09-20 2005-09-15 コーネル リサーチ ファンデーション インコーポレーテッド Methods and compositions for treating or preventing skin disorders using binding agents specific for prostate specific membrane antigen
DE60323677D1 (en) 2003-01-10 2008-10-30 Millennium Pharm Inc METHOD FOR DETERMINING THE REPRODUCTION OF PROSTATE CANCER
PL1684805T3 (en) 2003-11-04 2010-12-31 Novartis Vaccines & Diagnostics Inc Use of antagonist anti-cd40 monoclonal antibodies for treatment of multiple myeloma
CA2544368C (en) 2003-11-04 2014-04-01 Chiron Corporation Methods of therapy for b cell-related cancers
DK1694360T3 (en) 2003-11-04 2010-10-18 Novartis Vaccines & Diagnostic Use of antagonist anti-CD40 antibodies to treat autoimmune and inflammatory diseases and organ transplant rejection
AU2004287480B2 (en) 2003-11-04 2011-09-15 Novartis Vaccines And Diagnostics, Inc. Use of antagonist anti-CD40 antibodies for treatment of chronic lymphocytic leukemia
ATE476991T1 (en) 2003-11-04 2010-08-15 Novartis Vaccines & Diagnostic METHOD FOR TREATING SOLID TUMORS WITH EXPRESSION OF CD40 CELL SURFACE ANTIGEN
ES2429564T3 (en) 2005-05-18 2013-11-15 Novartis Ag Procedures for the diagnosis and treatment of diseases that have an autoimmune and / or inflammatory component
EP1921982A2 (en) * 2005-08-25 2008-05-21 Koninklijke Philips Electronics N.V. 4d image-based planning methods and apparatus for targeted therapy
ES2548518T3 (en) 2005-11-23 2015-10-19 Ventana Medical Systems, Inc. Molecular conjugate
US8945564B2 (en) 2006-04-21 2015-02-03 Novartis Ag Antagonist anti-CD40 antibody pharmaceutical compositions
DK2089401T3 (en) * 2006-11-10 2012-09-17 Radioprotect Alphaptose Ug Haftungsbeschraenkt & Co Kg Use of trisubstituted glycerol compounds for the treatment of radiation damage
CZ300074B6 (en) * 2006-12-20 2009-01-21 Azacycles S. R. O. Smart tree-functional macro-conjugates and pharmaceutical composition in which the conjugates are comprised
GB0700133D0 (en) 2007-01-04 2007-02-14 Humabs Llc Human cytomegalovirus neutralising antibodies and use thereof
NZ585516A (en) * 2007-12-21 2012-07-27 Hoffmann La Roche Anti-cd20 antibody formulation
ES2548014T3 (en) 2008-07-16 2015-10-13 Institute For Research In Biomedicine Neutralizing antibodies to human cytomegalovirus and their use
PE20141432A1 (en) 2008-07-16 2014-10-18 Inst Research In Biomedicine NEUTRALIZING ANTIBODIES OF HUMAN CYTOMEGALOVIRUS
JP5607620B2 (en) 2008-07-25 2014-10-15 インスティテュート・フォー・リサーチ・イン・バイオメディシン Anti-influenza A virus neutralizing antibody and use thereof
US8871207B2 (en) 2008-07-25 2014-10-28 Humabs, LLC Neutralizing anti-influenza A virus antibodies and uses thereof
JP5814790B2 (en) 2008-10-13 2015-11-17 インスティテュート・フォー・リサーチ・イン・バイオメディシンInstitute For Research In Biomedicine Dengue virus neutralizing antibody and use thereof
US20110212106A1 (en) 2010-01-20 2011-09-01 Institute For Research In Bioscience Hiv-1 neutralizing antibodies and uses thereof
MX339772B (en) * 2010-08-13 2016-06-09 Rdm Empreendimentos E Participações S A * Method and composition for reducing the color of sugar.
EP3418300B1 (en) 2011-07-18 2020-10-28 Institute for Research in Biomedicine Neutralizing anti-influenza a virus antibodies and uses thereof
CN104350069B (en) 2012-03-20 2017-12-01 胡默波斯生物医学公司 Neutralize RSV, MPV and PVM antibody and its application
RU2522203C2 (en) * 2012-06-26 2014-07-10 Василий Александрович Ишутин Method of controlling sterilisation of materials and products
RU2558929C2 (en) * 2013-08-08 2015-08-10 Федеральное государственное бюджетное учреждение "Научно-исследовательский институт кардиологии" (НИИ Кардиологии) METHOD FOR DETECTING 99mTc-CIPROFLOXACIN RADIOPHARMACEUTICAL FIXATION TO BACTERIA
WO2015051010A1 (en) 2013-10-02 2015-04-09 Medimmune, Llc Neutralizing anti-influenza a antibodies and uses thereof
CN113563462A (en) 2014-07-15 2021-10-29 免疫医疗有限责任公司 Neutralizing anti-influenza b antibodies and uses thereof
WO2016055950A1 (en) 2014-10-08 2016-04-14 Novartis Ag Combination of human cytomegalovirus neutralizing antibodies
WO2016075546A2 (en) 2014-11-14 2016-05-19 Antonio Lanzavecchia Antibodies that neutralize ebola virus and uses thereof
BR112017010298B1 (en) 2014-11-18 2023-10-31 Humabs Biomed Sa ANTIBODIES THAT POTENTLY NEUTRALIZE RABIES VIRUS AND OTHER LYSAVIRUS, USES THEREOF, NUCLEIC ACID MOLECULE, VECTOR, ISOLATED CELL, PHARMACEUTICAL COMPOSITION, AND KIT OF PARTS
AU2016271323B2 (en) 2015-06-01 2022-08-25 Medimmune, Llc Neutralizing anti-influenza binding molecules and uses thereof
WO2017059878A1 (en) 2015-10-07 2017-04-13 Humabs Biomed Sa Antibodies that potently neutralize hepatitis b virus and uses thereof
CN108697789A (en) 2016-01-13 2018-10-23 免疫医疗有限责任公司 The method for treating Flu-A
WO2018010789A1 (en) * 2016-07-13 2018-01-18 Humabs Biomed Sa Novel antibodies specifically binding to zika virus epitopes and uses thereof
KR102595238B1 (en) 2017-04-19 2023-10-26 인스티튜트 포 리서치 인 바이오메드슨 Novel malaria vaccine and antibody that binds to malaria parasite spores.
WO2019042555A1 (en) 2017-08-31 2019-03-07 Humabs Biomed Sa Multispecific antibodies specifically binding to zika virus epitopes and uses thereof
CN109550061A (en) * 2017-09-26 2019-04-02 南京江原安迪科正电子研究发展有限公司 A kind of kit and application for radioisotope labeling antibody
RU2700584C1 (en) * 2018-12-13 2019-09-18 Общество С Ограниченной Ответственностью "Апто-Фарм" Method for assessing the affinity of an oligonucleotide
EP3898668B1 (en) 2018-12-19 2023-08-30 Humabs Biomed SA Antibodies that neutralize hepatitis b virus and uses thereof
KR20220063188A (en) 2019-08-29 2022-05-17 비르 바이오테크놀로지, 인코포레이티드 Antibody compositions and methods for treating hepatitis B virus infection
TW202245838A (en) 2021-01-26 2022-12-01 美商維爾生物科技股份有限公司 Compositions and methods for treating hepatitis b virus infection

Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3994966A (en) * 1972-09-28 1976-11-30 The Board Of Trustees Of The Leland Stanford Junior University Chelating agents
US4043998A (en) * 1974-10-09 1977-08-23 The Board Of Trustees Of The Leland Stanford Junior University 1-(P-benzenediazonium)-ethylenediamine tetraacetic acid
US4315851A (en) * 1978-12-29 1982-02-16 Kureha Kagaku Kogyo Kabushiki Kaisha Pharmaceutical composition having antitumor activity
US4331647A (en) * 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4348376A (en) * 1980-03-03 1982-09-07 Goldenberg Milton David Tumor localization and therapy with labeled anti-CEA antibody
US4361544A (en) * 1980-03-03 1982-11-30 Goldenberg Milton David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4444744A (en) * 1980-03-03 1984-04-24 Goldenberg Milton David Tumor localization and therapy with labeled antibodies to cell surface antigens
US4454106A (en) * 1982-06-07 1984-06-12 Gansow Otto A Use of metal chelate conjugated monoclonal antibodies
US4460559A (en) * 1980-03-03 1984-07-17 Goldenberg Milton David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4460561A (en) * 1980-03-03 1984-07-17 Goldenberg M David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4472509A (en) * 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4622420A (en) * 1980-03-18 1986-11-11 The Regents Of The University Of California Chelating agents and method
US4634586A (en) * 1984-05-21 1987-01-06 The Board Of Trustees Of The Leland Stanford Junior University Reagent and method for radioimaging leukocytes
US4676609A (en) * 1982-11-26 1987-06-30 Saburo Matsui Adjustable bows for loupe frame
US4707352A (en) * 1984-01-30 1987-11-17 Enzo Biochem, Inc. Method of radioactively labeling diagnostic and therapeutic agents containing a chelating group
US4722892A (en) * 1984-08-31 1988-02-02 Meares Claude F Monoclonal antibodies against metal chelates
US4735210A (en) * 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US4824986A (en) * 1985-04-26 1989-04-25 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Metal chelate protein conjugate
US4831175A (en) * 1986-09-05 1989-05-16 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
US4855353A (en) * 1986-02-14 1989-08-08 Nihon Medi-Physics Co., Ltd. High molecular compounds having amino groups, and their utilization
US4861579A (en) * 1988-03-17 1989-08-29 American Cyanamid Company Suppression of B-lymphocytes in mammals by administration of anti-B-lymphocyte antibodies
US4921690A (en) * 1986-12-29 1990-05-01 City Of Hope Method of enhancing the biodistribution of antibody for localization in lesions
US4923985A (en) * 1988-05-25 1990-05-08 The United States Of America As Represented By The Department Of Health & Human Services Process for synthesizing macrocyclic chelates
US5009069A (en) * 1990-08-24 1991-04-23 Molini Alberto E Method of recovering energy from ocean water
US5034223A (en) * 1986-10-09 1991-07-23 Neorx Corporation Methods for improved targeting of antibody, antibody fragments, hormones and other targeting agents, and conjugates thereof
US5059518A (en) * 1988-10-20 1991-10-22 Coulter Corporation Stabilized lyophilized mammalian cells and method of making same
US5101827A (en) * 1985-07-05 1992-04-07 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US5124471A (en) * 1990-03-26 1992-06-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Bifunctional dtpa-type ligand
US5162115A (en) * 1989-05-09 1992-11-10 Pietronigro Dennis D Antineoplastic solution and method for treating neoplasms
US5208008A (en) * 1990-11-14 1993-05-04 University Of Pittsburgh Regioselective chemical modification of monoclonal antibodies
US5219556A (en) * 1990-07-09 1993-06-15 Mallinckrodt Medical, Inc. Stabilized therapeutic radiopharmaceutical complexes
US5246692A (en) * 1986-09-05 1993-09-21 The United States Of America As Represented By The Secretary Of Health And Human Services Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
US5363846A (en) * 1986-01-16 1994-11-15 The General Hospital Corporation Method for the diagnosis and treatment of inflammation
US5376356A (en) * 1989-03-14 1994-12-27 Neorx Corporation Imaging tissue sites of inflammation
US5403573A (en) * 1992-04-23 1995-04-04 The Curators Of The University Of Missouri Radiolabeled protein composition and method for radiation synovectomy
US5428154A (en) * 1988-05-25 1995-06-27 The United States Of America As Represented By The Department Of Health And Human Services Complexes of functionalized tetraazacyclododecane chelates with bismuth, lead, ytriium, actinium, or lanthanide metal ions
US5500362A (en) * 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5545403A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal by administering a CHO-glycosylated antibody
US5595721A (en) * 1993-09-16 1997-01-21 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20
US5641637A (en) * 1991-01-11 1997-06-24 Cobe Laboratories, Inc. Method of preparing lyophilized and frozen cell standards
US5976492A (en) * 1994-10-05 1999-11-02 Immunomedics, Inc. Radioactive phosphorus labeled proteins for targeted radiotherapy
US6010680A (en) * 1994-06-03 2000-01-04 Immunomedics, Inc. Thiolation of proteins for radionuclide-based radioimmunodetection and radioimmunotherapy

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4636380A (en) 1984-04-23 1987-01-13 Wong Dennis W Novel physiologic chemical method of labeling protein substances with the radionuclides of indium
US5099069A (en) 1986-09-05 1992-03-24 Gansow Otto A Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
EP0484989B1 (en) * 1986-09-05 1996-04-24 GANSOW, Otto A. Polysubstituted diethylenetriamine chelates for forming a metal-chelate-protein conjugate
AU619218B2 (en) 1987-11-06 1992-01-23 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US5217704A (en) 1987-11-06 1993-06-08 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US5130118A (en) 1987-11-06 1992-07-14 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US4978745A (en) 1987-11-23 1990-12-18 Centocor, Inc. Immunoreactive heterochain antibodies
US4975369A (en) 1988-04-21 1990-12-04 Eli Lilly And Company Recombinant and chimeric KS1/4 antibodies directed against a human adenocarcinoma antigen
CA2010511A1 (en) 1989-03-01 1990-09-01 Roberto L. Ceriani Method of enhancing cancer therapy by administration of unsaturated fatty acids
US5460785A (en) 1989-08-09 1995-10-24 Rhomed Incorporated Direct labeling of antibodies and other protein with metal ions
EP0556285A4 (en) 1990-11-05 1993-10-27 Bristol-Myers Squibb Company Synergistic therapy with combinations of anti-tumor antibodies and biologically active agents
US5541287A (en) 1992-06-09 1996-07-30 Neorx Corporation Pretargeting methods and compounds
US5716596A (en) 1992-06-23 1998-02-10 Diatide, Inc. Radioactively labeled somatostatin-derived peptides for imaging and therapeutic uses
US5620675A (en) 1992-06-23 1997-04-15 Diatech, Inc. Radioactive peptides
PL174721B1 (en) * 1992-11-13 1998-09-30 Idec Pharma Corp Monoclonal antibody anty-cd2
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5650134A (en) 1993-01-12 1997-07-22 Novartis Ag (Formerly Sandoz Ltd.) Peptides
US5686578A (en) 1994-08-05 1997-11-11 Immunomedics, Inc. Polyspecific immunoconjugates and antibody composites for targeting the multidrug resistant phenotype
US5874540A (en) 1994-10-05 1999-02-23 Immunomedics, Inc. CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
US5942210A (en) 1994-11-15 1999-08-24 Cytogen Corporation Methods for lyoprotecting a macromolecule using tricine
US5830431A (en) 1995-06-07 1998-11-03 Mallinckrodt Medical, Inc. Radiolabeled peptide compositions for site-specific targeting
US5961955A (en) * 1997-06-03 1999-10-05 Coulter Pharmaceutical, Inc. Radioprotectant for peptides labeled with radioisotope
US20020102208A1 (en) * 1999-03-01 2002-08-01 Paul Chinn Radiolabeling kit and binding assay
MY133346A (en) * 1999-03-01 2007-11-30 Biogen Inc Kit for radiolabeling ligands with yttrium-90

Patent Citations (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3994966A (en) * 1972-09-28 1976-11-30 The Board Of Trustees Of The Leland Stanford Junior University Chelating agents
US4043998A (en) * 1974-10-09 1977-08-23 The Board Of Trustees Of The Leland Stanford Junior University 1-(P-benzenediazonium)-ethylenediamine tetraacetic acid
US4315851A (en) * 1978-12-29 1982-02-16 Kureha Kagaku Kogyo Kabushiki Kaisha Pharmaceutical composition having antitumor activity
US4401592A (en) * 1978-12-29 1983-08-30 Kureha Kagaku Kogyo Kabushiki Kaisha Pharmaceutical composition having antitumor activity
US4460561A (en) * 1980-03-03 1984-07-17 Goldenberg M David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4331647A (en) * 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4348376A (en) * 1980-03-03 1982-09-07 Goldenberg Milton David Tumor localization and therapy with labeled anti-CEA antibody
US4361544A (en) * 1980-03-03 1982-11-30 Goldenberg Milton David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4444744A (en) * 1980-03-03 1984-04-24 Goldenberg Milton David Tumor localization and therapy with labeled antibodies to cell surface antigens
US4460559A (en) * 1980-03-03 1984-07-17 Goldenberg Milton David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4622420A (en) * 1980-03-18 1986-11-11 The Regents Of The University Of California Chelating agents and method
US4454106A (en) * 1982-06-07 1984-06-12 Gansow Otto A Use of metal chelate conjugated monoclonal antibodies
US4472509A (en) * 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4676609A (en) * 1982-11-26 1987-06-30 Saburo Matsui Adjustable bows for loupe frame
US4707352A (en) * 1984-01-30 1987-11-17 Enzo Biochem, Inc. Method of radioactively labeling diagnostic and therapeutic agents containing a chelating group
US4634586A (en) * 1984-05-21 1987-01-06 The Board Of Trustees Of The Leland Stanford Junior University Reagent and method for radioimaging leukocytes
US4722892A (en) * 1984-08-31 1988-02-02 Meares Claude F Monoclonal antibodies against metal chelates
US4824986A (en) * 1985-04-26 1989-04-25 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Metal chelate protein conjugate
US4735210A (en) * 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US5101827A (en) * 1985-07-05 1992-04-07 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US5363846A (en) * 1986-01-16 1994-11-15 The General Hospital Corporation Method for the diagnosis and treatment of inflammation
US4855353A (en) * 1986-02-14 1989-08-08 Nihon Medi-Physics Co., Ltd. High molecular compounds having amino groups, and their utilization
US4831175A (en) * 1986-09-05 1989-05-16 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
US5246692A (en) * 1986-09-05 1993-09-21 The United States Of America As Represented By The Secretary Of Health And Human Services Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
US5034223A (en) * 1986-10-09 1991-07-23 Neorx Corporation Methods for improved targeting of antibody, antibody fragments, hormones and other targeting agents, and conjugates thereof
US4921690A (en) * 1986-12-29 1990-05-01 City Of Hope Method of enhancing the biodistribution of antibody for localization in lesions
US5500362A (en) * 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US4861579A (en) * 1988-03-17 1989-08-29 American Cyanamid Company Suppression of B-lymphocytes in mammals by administration of anti-B-lymphocyte antibodies
US4923985A (en) * 1988-05-25 1990-05-08 The United States Of America As Represented By The Department Of Health & Human Services Process for synthesizing macrocyclic chelates
US5428154A (en) * 1988-05-25 1995-06-27 The United States Of America As Represented By The Department Of Health And Human Services Complexes of functionalized tetraazacyclododecane chelates with bismuth, lead, ytriium, actinium, or lanthanide metal ions
US5059518A (en) * 1988-10-20 1991-10-22 Coulter Corporation Stabilized lyophilized mammalian cells and method of making same
US5376356A (en) * 1989-03-14 1994-12-27 Neorx Corporation Imaging tissue sites of inflammation
US5162115A (en) * 1989-05-09 1992-11-10 Pietronigro Dennis D Antineoplastic solution and method for treating neoplasms
US5434287A (en) * 1990-03-26 1995-07-18 The United States Of America As Represented By The Department Of Health And Human Services Bifunctional DTPA-type ligand
US5124471A (en) * 1990-03-26 1992-06-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Bifunctional dtpa-type ligand
US5286850A (en) * 1990-03-26 1994-02-15 The United States Of America As Represented By The Department Of Health And Human Services Antibody DTPA-type ligand conjugates
US5219556A (en) * 1990-07-09 1993-06-15 Mallinckrodt Medical, Inc. Stabilized therapeutic radiopharmaceutical complexes
US5009069A (en) * 1990-08-24 1991-04-23 Molini Alberto E Method of recovering energy from ocean water
US5545405A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal suffering from cancer with a cho-glycosylated antibody
US5545403A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal by administering a CHO-glycosylated antibody
US5208008A (en) * 1990-11-14 1993-05-04 University Of Pittsburgh Regioselective chemical modification of monoclonal antibodies
US5641637A (en) * 1991-01-11 1997-06-24 Cobe Laboratories, Inc. Method of preparing lyophilized and frozen cell standards
US5403573A (en) * 1992-04-23 1995-04-04 The Curators Of The University Of Missouri Radiolabeled protein composition and method for radiation synovectomy
US5595721A (en) * 1993-09-16 1997-01-21 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20
US6010680A (en) * 1994-06-03 2000-01-04 Immunomedics, Inc. Thiolation of proteins for radionuclide-based radioimmunodetection and radioimmunotherapy
US5976492A (en) * 1994-10-05 1999-11-02 Immunomedics, Inc. Radioactive phosphorus labeled proteins for targeted radiotherapy

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000052031A2 (en) 1999-03-01 2000-09-08 Idec Pharmaceuticals Corporation Kit for radiolabeling proteins with yttrium-90
EP1918305A1 (en) 1999-08-11 2008-05-07 Biogen Idec Inc. New clinical parameters for determining hematologic toxicity prior to radioimmunotherapy
US20030003097A1 (en) * 2001-04-02 2003-01-02 Idec Pharmaceutical Corporation Recombinant antibodies coexpressed with GnTIII
US11168125B2 (en) 2003-05-06 2021-11-09 Bioverativ Therapeutics Inc. Immunoglobulin chimeric monomer-dimer hybrids
US20050123474A1 (en) * 2003-06-25 2005-06-09 Peregrine Pharmaceuticals Methods and apparatus for continuous large-scale radiolabeling of proteins
US8137540B2 (en) 2003-06-25 2012-03-20 Peregrine Pharmaceuticals, Inc. Methods and apparatus for continuous large-scale radiolabeling
AU2004253924B2 (en) * 2003-06-25 2009-07-30 Peregrine Pharmaceuticals, Inc. Method and apparatus for continuous large-scale radiolabeling of proteins
US7591953B2 (en) * 2003-06-25 2009-09-22 Peregrine Pharmaceuticals, Inc. Methods and apparatus for continuous large-scale radiolabeling of proteins
US20090312523A1 (en) * 2003-06-25 2009-12-17 Peregrine Pharmaceuticals, Inc. Methods and apparatus for continuous large-scale radiolabeling
EP2238986A2 (en) 2005-07-08 2010-10-13 Biogen Idec MA Inc. Sp35 antibodies and uses thereof
EP2478917A1 (en) 2005-07-08 2012-07-25 Biogen Idec MA Inc. SP35 antibodies and uses thereof
EP2394661A1 (en) 2005-07-08 2011-12-14 Biogen Idec MA Inc. Sp35 antibodies and uses thereof
US10849841B2 (en) 2006-06-16 2020-12-01 The Trustees Of The University Of Pennsylvania Methods and compositions for inhibiting or reducing hair loss, acne, rosacea, prostate cancer, and BPH
US9889082B2 (en) 2006-06-16 2018-02-13 The Trustees Of The University Of Pennsylvania Methods and compositions for inhibiting or reducing hair loss, acne, rosacea, prostate cancer, and BPH
US20090130089A9 (en) * 2006-10-10 2009-05-21 Vaccinex, Inc. Anti-cd20 antibodies and methods of use
US20080089885A1 (en) * 2006-10-10 2008-04-17 Vaccinex, Inc. Anti-cd20 antibodies and methods of use
US10301393B2 (en) 2006-10-10 2019-05-28 Vaccinex, Inc. Methods of treating multiple sclerosis using anti-CD20 antibodies
US9382327B2 (en) 2006-10-10 2016-07-05 Vaccinex, Inc. Anti-CD20 antibodies and methods of use
EP2740744A2 (en) 2007-01-09 2014-06-11 Biogen Idec MA Inc. SP35 antibodies and uses thereof
EP2982695A1 (en) 2008-07-09 2016-02-10 Biogen MA Inc. Compositions comprising antibodies to lingo or fragments thereof
WO2010078526A1 (en) 2008-12-31 2010-07-08 Biogen Idec Ma Inc. Anti-lymphotoxin antibodies
EP2835053A1 (en) 2010-03-12 2015-02-11 Genzyme Corporation Combination therapy for treating breast cancer
EP3406141A1 (en) 2010-03-12 2018-11-28 Genzyme Corporation Combination therapy for treating cancer
US10927362B2 (en) 2010-07-09 2021-02-23 Bioverativ Therapeutics Inc. Processable single chain molecules and polypeptides made using same
US10968442B2 (en) 2010-07-09 2021-04-06 Bioverativ Therapeutics Inc. Chimeric clotting factors
US9856468B2 (en) 2010-07-09 2018-01-02 Bioverativ Therapeutics Inc. Processable single chain molecules and polypeptides made using same
WO2012019128A1 (en) 2010-08-06 2012-02-09 Genzyme Corporation Vegf antagonist compositions and uses thereof
EP3327032A1 (en) 2010-08-06 2018-05-30 Genzyme Corporation Vegf antagonist compositions and uses thereof
EP3045183A1 (en) 2011-03-11 2016-07-20 Genzyme Corporation Pegylated apelin and uses thereof
WO2012125408A1 (en) 2011-03-11 2012-09-20 Genzyme Corporation Pegylated apelin and uses thereof
US11186638B2 (en) 2011-09-12 2021-11-30 Genzyme Corporation Anti-αβTCR antibody
US9717803B2 (en) 2011-12-23 2017-08-01 Innate Pharma Enzymatic conjugation of polypeptides
US9764038B2 (en) 2011-12-23 2017-09-19 Innate Pharma Enzymatic conjugation of antibodies
US10675359B2 (en) 2011-12-23 2020-06-09 Innate Pharma Enzymatic conjugation of antibodies
US11117959B2 (en) 2012-05-23 2021-09-14 Argenx Bvba IL-6 binding molecules
US11827701B2 (en) 2012-05-23 2023-11-28 argenx BV IL-6 binding molecules
US10183995B2 (en) 2012-05-23 2019-01-22 Argen-X N.V. IL-6 binding molecules
WO2014009426A3 (en) * 2012-07-13 2014-04-03 Innate Pharma Screening of conjugated antibodies
US10132799B2 (en) 2012-07-13 2018-11-20 Innate Pharma Screening of conjugated antibodies
US10836813B2 (en) 2012-09-12 2020-11-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
US10036010B2 (en) 2012-11-09 2018-07-31 Innate Pharma Recognition tags for TGase-mediated conjugation
US11130816B2 (en) 2013-03-11 2021-09-28 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
US10214589B2 (en) 2013-03-11 2019-02-26 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
US11807690B2 (en) 2013-03-11 2023-11-07 Genzyme Corporation Hyperglycosylated binding polypeptides
US10494439B2 (en) 2013-03-11 2019-12-03 Genzyme Corporation Hyperglycosylated binding polypeptides
US11813338B2 (en) 2013-03-12 2023-11-14 The Trustees Of The University Of Pennsylvania Diagnosing and treating cancer
US10611824B2 (en) 2013-03-15 2020-04-07 Innate Pharma Solid phase TGase-mediated conjugation of antibodies
US10071169B2 (en) 2013-06-20 2018-09-11 Innate Pharma Enzymatic conjugation of polypeptides
US10434180B2 (en) 2013-06-21 2019-10-08 Innate Pharma Enzymatic conjugation of polypeptides
US9427478B2 (en) 2013-06-21 2016-08-30 Innate Pharma Enzymatic conjugation of polypeptides
US9845363B2 (en) 2013-08-13 2017-12-19 Sanofi Antibodies to plasminogen activator inhibitor-1 (PAI-1) and uses thereof
US11052177B2 (en) 2013-09-06 2021-07-06 The Trustees Of The University Of Pennsylvania Antimicrobial polymer layers
KR101738117B1 (en) 2014-02-27 2017-05-19 보드 오브 리전츠, 더 유니버시티 오브 텍사스 시스템 Methods and compositions for isolating exosomes
WO2015131153A1 (en) * 2014-02-27 2015-09-03 Board Of Regents, The University Of Texas System Methods and compositions for isolating exosomes
US9835626B2 (en) 2014-02-27 2017-12-05 Board Of Regents, The University Of Texas System Methods and compositions for isolating exosomes
CN106030304A (en) * 2014-02-27 2016-10-12 得克萨斯大学体系董事会 Methods and compositions for isolating exosomes
US11697690B2 (en) 2014-03-19 2023-07-11 Genzyme Corporation Site-specific glycoengineering of targeting moieties
US10995148B2 (en) 2014-03-19 2021-05-04 Genzyme Corporation Site-specific glycoengineering of targeting moieties
US11566069B2 (en) 2014-03-27 2023-01-31 Bird Rock Bio, Inc. Treatment of disease responsive to modulation of cannabanoid 1(CB1) receptor signaling
US10308712B2 (en) 2014-03-27 2019-06-04 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
US11160874B2 (en) 2014-10-09 2021-11-02 Genzyme Corporation Glycoengineered antibody drug conjugates
US10064952B2 (en) 2014-10-09 2018-09-04 Genzyme Corporation Glycoengineered antibody drug conjugates
WO2017019858A1 (en) 2015-07-30 2017-02-02 The Trustees Of The University Of Pennsylvania Single nucleotide polymorphic alleles of human dp-2 gene for detection of susceptibility to hair growth inhibition by pgd2
US10278438B2 (en) 2015-08-31 2019-05-07 Grabbies Media, Llc Consumer usable device with redeemable member
WO2017048902A1 (en) 2015-09-15 2017-03-23 Board Of Regents, The University Of Texas System T-cell receptor (tcr)-binding antibodies and uses thereof
US11421026B2 (en) 2015-09-30 2022-08-23 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
US11427634B2 (en) 2017-05-05 2022-08-30 Vaccinex, Inc. Human anti-semaphorin 4D antibody
WO2019018633A1 (en) 2017-07-19 2019-01-24 Hoffman Technologies, Inc. Compositions for treating stress-related disorders
US11879004B2 (en) 2020-02-28 2024-01-23 Genzyme Corporation Modified binding polypeptides for optimized drug conjugation
US11702470B2 (en) 2020-06-11 2023-07-18 Imperial College Innovations Limited Use of CXCL13 binding molecules to promote peripheral nerve regeneration

Also Published As

Publication number Publication date
CZ20013126A3 (en) 2002-07-17
RS51778B (en) 2011-12-31
US20050169838A1 (en) 2005-08-04
NO330705B1 (en) 2011-06-20
DE60042326D1 (en) 2009-07-16
HK1045730A1 (en) 2002-12-06
BRPI0017596A2 (en) 2011-09-27
KR100729247B1 (en) 2007-06-15
PT2112512E (en) 2015-01-13
AU3505200A (en) 2000-09-21
NO20014245L (en) 2001-10-31
KR20020021772A (en) 2002-03-22
JP2002538179A (en) 2002-11-12
US7608241B2 (en) 2009-10-27
SI1192463T1 (en) 2009-10-31
CZ303262B6 (en) 2012-07-04
CN1345417A (en) 2002-04-17
EP1192463A2 (en) 2002-04-03
DK1192463T3 (en) 2009-10-05
ES2526723T3 (en) 2015-01-14
BRPI0008719A (en) 2001-12-26
MXPA01008837A (en) 2002-05-14
UA78484C2 (en) 2007-04-10
HK1045730B (en) 2006-08-18
TWI294969B (en) 2008-03-21
CN1235639C (en) 2006-01-11
PL205780B1 (en) 2010-05-31
CA2742153A1 (en) 2000-09-08
YU61601A (en) 2004-07-15
BG105957A (en) 2002-08-30
BRPI0008719B8 (en) 2021-07-06
IL145109A0 (en) 2002-06-30
NZ530868A (en) 2005-12-23
EE200100465A (en) 2002-10-15
EE05650B1 (en) 2013-04-15
WO2000052473A3 (en) 2002-01-31
EP2112512B1 (en) 2014-11-19
RU2251110C2 (en) 2005-04-27
CY1116265T1 (en) 2017-02-08
HU230516B1 (en) 2016-10-28
NO20014245D0 (en) 2001-08-31
HK1131208A1 (en) 2010-01-15
NZ513788A (en) 2004-03-26
CY1109335T1 (en) 2014-07-02
BRPI0017596B8 (en) 2023-04-25
BRPI0008719B1 (en) 2015-07-21
EP2112512A3 (en) 2009-12-16
ATE433108T1 (en) 2009-06-15
PT1192463E (en) 2009-09-10
WO2000052473A9 (en) 2001-03-08
ES2328100T3 (en) 2009-11-10
ME00782B (en) 2012-03-20
HU229627B1 (en) 2014-03-28
SK288096B6 (en) 2013-07-02
PL352531A1 (en) 2003-08-25
HUP1300275A2 (en) 2002-11-28
HRP20010714A2 (en) 2003-02-28
HUP0202522A3 (en) 2011-03-28
MY138674A (en) 2009-07-31
CA2362119A1 (en) 2000-09-08
EP2112512A2 (en) 2009-10-28
BRPI0017596B1 (en) 2015-09-01
ZA200106943B (en) 2002-11-22
SK12162001A3 (en) 2002-12-03
JP4704570B2 (en) 2011-06-15
HUP0202522A2 (en) 2002-11-28
HRP20010714B1 (en) 2009-01-31
CZ306959B6 (en) 2017-10-11
CA2362119C (en) 2012-12-11
NO20100717L (en) 2001-10-31
WO2000052473A2 (en) 2000-09-08
BG65690B1 (en) 2009-06-30
SI2112512T1 (en) 2015-02-27
AU776747B2 (en) 2004-09-23
DK2112512T3 (en) 2014-12-15
EP1192463B1 (en) 2009-06-03
SK287745B6 (en) 2011-08-04
CA2742153C (en) 2015-04-14
NO329142B1 (en) 2010-08-30

Similar Documents

Publication Publication Date Title
US7608241B2 (en) Radiolabeling method
US7618613B2 (en) Method for radiolabeling antibodies with yttrium-90

Legal Events

Date Code Title Description
AS Assignment

Owner name: IDEC PHARMACEUTICALS CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHINN, PAUL;MORENA, RONALD;LABARRE, MICHAEL;AND OTHERS;REEL/FRAME:010230/0330;SIGNING DATES FROM 19990902 TO 19990907

AS Assignment

Owner name: BIOGEN IDEC INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:IDEC PHARMACEUTICALS CORPORATION;REEL/FRAME:015044/0873

Effective date: 20031112

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION