[go: up one dir, main page]

WO2024034759A1 - Immuno-pet using 89zr-labeled anti-cd25 antibodies - Google Patents

Immuno-pet using 89zr-labeled anti-cd25 antibodies Download PDF

Info

Publication number
WO2024034759A1
WO2024034759A1 PCT/KR2023/002337 KR2023002337W WO2024034759A1 WO 2024034759 A1 WO2024034759 A1 WO 2024034759A1 KR 2023002337 W KR2023002337 W KR 2023002337W WO 2024034759 A1 WO2024034759 A1 WO 2024034759A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
tumor
present
cells
pet
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.)
Ceased
Application number
PCT/KR2023/002337
Other languages
French (fr)
Korean (ko)
Inventor
이경한
정경호
이진희
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.)
Sungkyunkwan University
Samsung Life Public Welfare Foundation
Original Assignee
Sungkyunkwan University
Samsung Life Public Welfare Foundation
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 Sungkyunkwan University, Samsung Life Public Welfare Foundation filed Critical Sungkyunkwan University
Publication of WO2024034759A1 publication Critical patent/WO2024034759A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody

Definitions

  • the present invention relates to a composition for medical imaging comprising 89 Zr labeled anti-CD25 antibody as an active ingredient.
  • Positron emission tomography uses radioactive pharmaceuticals that emit positrons to image biological changes in the human body caused by disease, and can provide accurate information for early diagnosis of disease and decision-making on treatment methods.
  • the radiopharmaceutical for cancer diagnosis most commonly used in clinical practice is [ 18 F]FDG.
  • 18 F-FDG is a glucose analogue with a radioactive isotope that enters the cell through the glucose transporter (Glut) and is phosphorylated. It is no longer metabolized and stays in the cell, emitting positrons. Cancer cells have an increased number of glucose transporters on the cell surface compared to normal cells, so more 18 F-FDG flows into cancer cells. Therefore, while existing radiological tests image structures within the human body to find and diagnose anatomical lesions, 18 F-FDG PET/CT can image biochemical changes before anatomical changes.
  • Lymphoma is the most common malignant blood cancer that occurs in lymphoid tissue. It is broadly classified into non-Hodgkin's lymphoma and Hodgkin's lymphoma, of which non-Hodgkin's lymphoma, which accounts for the majority, can be classified into B-cell lymphoma and T-cell lymphoma. In the treatment of lymphoma, antibody-based medicines are reported to have a significantly greater effect in eliminating cancer cells through immune responses. For example, in B-cell lymphoma, a monoclonal antibody called rituximab is known to significantly improve the course of lymphoma treatment.
  • Non-patent Document 1 In order to successfully develop antibody treatments for refractory lymphoma, imaging technology is essential to evaluate whether target expression is sufficient in the tumor and monitor whether the treatment reaches the lymphoma.
  • biological information cannot be provided by existing 18 F-FDG PET/CT.
  • Non-patent Document 1 To overcome this, immuno-PET imaging technology targeting tumor-specific cell-surface antigens is attracting attention.
  • immuno-PET imaging technology has the characteristics of shortening the preclinical trial period for new drug development, selecting patients whose lymphoma cells express a sufficient amount of target antigen, and predicting the effect of antibody treatment, it still has the potential to produce new lymphoma-specific antigens. A target is needed.
  • antibody-drug conjugates are emerging.
  • antibody conjugates loaded with beta-emitting isotopes are raising expectations as radioimmunotherapy agents for lymphoma.
  • radioimmunotherapy uses 177 Lu. Radioactive isotopes are preferred.
  • Radiation-loaded anti-CD25 antibody radioimmunotherapy for refractory lymphoma is also in the preclinical and clinical development stages, but there is still no immuno-PET imaging technology to support this technology.
  • the present inventors developed an immuno-PET imaging technology using the 89 Zr-labeled anti-CD25 antibody unique to the present invention and confirmed that it binds with high affinity to lymphoma cells and Treg cells expressing CD25, as well as to The present invention was completed by confirming the high uptake by lymphoma in the body and the excellent target specificity in which uptake was inhibited by non-radioactive anti-CD25 antibodies.
  • the purpose of the present invention is to image refractory lymphoma and Treg cells in the body as new treatment targets, so that it can be used to not only contribute to the development of antibody treatments, but also to select treatment targets and predict treatment effects, using mouse and human 89 Zr-CD25
  • the goal is to develop and provide antibodies and immuno-PET imaging technology using them.
  • the purpose of the present invention is to provide an 89 Zr labeled anti-CD25 antibody.
  • Another object of the present invention is to provide a composition for positron emission tomography imaging for tumor diagnosis, comprising 89 Zr labeled anti-CD25 antibody as an active ingredient.
  • Another object of the present invention is to provide a method for preparing 89 Zr labeled anti-CD25 antibody comprising the following steps:
  • step (b) reacting the anti-CD25 antibody reduced in step (a) with deferoxamine-maleimide (DFO-Mal) to produce a DFO-Mal conjugated anti-CD25 antibody; and
  • DFO-Mal deferoxamine-maleimide
  • step (c) reacting the DFO-Mal conjugated anti-CD25 antibody produced in step (b) with 89 Zr-oxalate to obtain an 89 Zr labeled anti-CD25 antibody.
  • Another object of the present invention is to detect tumor or tumor infiltrating regulatory T cells (regulatory T cells, Tregs);
  • a method of acquiring a PET image using the 89 Zr-labeled anti-CD25 antibody of the present invention as a contrast agent is provided.
  • Another object of the present invention is to provide a positron emission tomography method for tumor diagnosis, comprising the following steps:
  • Another object of the present invention is to provide a kit for positron emission tomography imaging for tumor diagnosis, including the composition and instructions.
  • the present invention provides an 89 Zr labeled anti-CD25 antibody.
  • the present invention provides a composition for positron emission tomography imaging for tumor diagnosis, comprising 89 Zr-labeled anti-CD25 antibody as an active ingredient.
  • the composition can detect tumors or tumor-infiltrating regulatory T cells, but is not limited thereto.
  • the tumor may be lymphoma, but is not limited thereto.
  • the composition may be for intravenous injection, but is not limited thereto.
  • the present invention provides a method of making 89 Zr labeled anti-CD25 antibody comprising the following steps:
  • step (b) reacting the anti-CD25 antibody reduced in step (a) with deferoxamine-maleimide (DFO-Mal) to produce a DFO-Mal conjugated anti-CD25 antibody; and
  • DFO-Mal deferoxamine-maleimide
  • step (c) reacting the DFO-Mal conjugated anti-CD25 antibody produced in step (b) with 89 Zr-oxalate to obtain an 89 Zr labeled anti-CD25 antibody.
  • the antibody may be IgG, but is not limited thereto.
  • the antibody may have the following characteristics, but is not limited thereto:
  • 89 Zr is specifically bound to the disulfide bond site of the anti-CD25 antibody hinge region
  • the present invention provides a positron emission tomography method for tumor diagnosis, comprising the following steps:
  • the method may use a technology selected from the group consisting of PET (Positron Emission Tomography), PET/CT, PET/MR, and immuno-PET, but is not limited thereto. .
  • the present invention provides a kit for positron emission tomography imaging for tumor diagnosis, including the composition and instructions.
  • the composition is a composition of the invention, and the instructions describe the method as It may be described, but is not limited thereto.
  • the present invention provides a composition comprising an 89 Zr-labeled anti-CD25 antibody as an active ingredient for use in the diagnosis of tumors; or detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.
  • the present invention relates to diagnosis of tumors; Alternatively, it provides the use of a composition comprising 89 Zr-labeled anti-CD25 antibody as an active ingredient for preparing an imaging agent or detection of tumor or intratumoral infiltrating regulatory T cells.
  • the present invention relates to the 89 Zr labeled anti-CD25, for diagnosis of tumors; or detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.
  • the present invention relates to diagnosis of tumors; or for preparing an agent for detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.
  • a medical imaging composition for tumor diagnosis comprising a Zr-labeled anti-CD25 antibody as an active ingredient, and tumor diagnosis using the same; It relates to a method for imaging a tumor or tumor-infiltrating regulatory T cells.
  • the unique 89 Zr-labeled anti-CD25 antibody of the present invention is used to detect lymphoma or regulatory T cells among tissues in which the CD25 antigen is present. Since it shows a very high uptake rate specifically in the area where it exists, it can be usefully used as an effective non-invasive PET-related imaging agent that can distinguish, diagnose, image, and monitor lymphoma and/or regulatory T cells.
  • Figure 1 is an image schematically showing the concept of the development of the 89 Zr-CD25 antibody of the present invention targeting regulatory T cells (Treg) in refractory lymphoma and tumor tissue and the development of immuno-PET imaging technology using the same.
  • Reg regulatory T cells
  • Figure 2a schematically shows the process of changes in chemical bonds within the antibody that occur during the labeling process when labeling an antibody with a conventional labeling technology.
  • Figure 2b schematically shows the binding structure of a labeling substance and an antibody when labeling an antibody using a conventional labeling technology as shown in Figure 2a.
  • Figure 3a shows the results of confirming the size of the molecule using SDS PAGE after TCEP reduction and DFO-conjugation during the process of specifically labeling the hinge-region cysteine residue of the human anti-CD25 antibody with 89 Zr in the present invention. indicates.
  • Figure 3b shows the results of confirming the size of the molecule using SDS PAGE after TCEP reduction and DFO-conjugation during the process of specifically labeling the hinge-region cysteine residue of the mouse anti-CD25 antibody with 89 Zr in the present invention. indicates.
  • Figure 4 schematically shows the binding structure of the 89 Zr-labeled anti-CD25 antibody unique to the present invention.
  • Figure 5a shows the results of analysis by autoradiography on the human 89 Zr-CD25 antibody of the present invention.
  • Figure 5b shows the results of analysis by autoradiography for the mouse 89 Zr-CD25 antibody of the present invention.
  • Figure 6a shows the results of measuring radioactivity for each fraction separated by size-exclusion chromatography when obtaining the human 89 Zr-CD25 antibody of the present invention.
  • Figure 6b shows the results of measuring radioactivity for each fraction separated by size-exclusion chromatography when obtaining the mouse 89 Zr-CD25 antibody of the present invention.
  • Figure 7a shows the results of evaluating the serum stability of the human 89 Zr-CD25 antibody of the present invention after incubation in PBS or FBS for several days and analysis by radio-iTLC.
  • Figure 7b shows the results of evaluating the serum stability of the mouse 89 Zr-CD25 antibody of the present invention after incubation in PBS or FBS for several days and analysis by radio-iTLC.
  • Figure 8a shows the results of western blotting confirming the expression level of CD25 antigen on the cell surface for human lymphoma cell lines H9, Jurkat, and SUDHL1 and mouse lymphoma cell line EL4.
  • Figure 8b shows the results of comparing the binding degree of human 89 Zr-CD25 IgG of the present invention in SUDHL1 human T cell lymphoma cells with high CD25 expression and human H9 and Jurkat cancer cells with low CD25 expression.
  • Figure 8c shows the results of confirming the binding specificity of the human 89 Zr-CD25 antibody of the present invention using SUDHL1 lymphoma cells with high CD25 expression (the blocking group shows high binding specificity inhibited by 0.5 ⁇ M of cold unlabeled antibody). confirmed).
  • Figure 8d shows the results confirming that in EL4 lymphoma cells with low CD25 expression, the low binding of mouse 89 Zr-CD25 IgG was suppressed only to a small extent by a large amount of cold anti-CD25 IgG (0.5 ⁇ M of cold unlabeled antibody in the blocking group). High binding specificity inhibited by was confirmed).
  • Figure 9a shows the results of FACS analysis performed during the process of separating and purifying CD25(-) T cells and CD25(+) Treg cells from the thymus of a mouse using FACS.
  • Figure 9b shows the results of hematoxylin & eosin staining of CD25(+) Treg cells and observation with an optical microscope.
  • Figure 10a shows the results of comparative analysis of the binding rate and specificity of 89 Zr-CD25 IgG of the present invention for CD25(-) T cells and CD25(+) Treg cells isolated from the thymus of mice (blocking group includes High binding specificity inhibited by 0.5 ⁇ M cold unlabeled antibody was confirmed).
  • Figure 10b shows the results of confirming the binding rate and specificity of 89 Zr-CD25 IgG of the present invention for CD25(+) Treg cells isolated from the spleen of mice (in the blocking group, there was a high level of antibody inhibited by 0.5 ⁇ M of cold unlabeled antibody). Binding specificity was confirmed).
  • Figure 11 shows the results of confirming the body distribution and uptake rate in lymphoma 5 days after administration of 89 Zr-CD25 IgG of the present invention in an in vivo mouse lymphoma model (blocking group was treated with 0.8 mg of cold unlabeled antibody) High binding specificity of inhibition was confirmed).
  • Figure 12 is an image showing the results of PET imaging taken 5 days after administration of 89 Zr-CD25 IgG of the present invention in an in vivo mouse lymphoma model, showing high uptake of 89 Zr-CD25 IgG by T cell lymphoma.
  • Figure 13 is an image showing the results of PET imaging on the 5th day of administration of 89 Zr-CD25 IgG of the present invention, 1 hour after administration of 0.8 mg of cold unlabeled Ab, in an in vivo mouse lymphoma model (blocking group). .
  • the contrast was found to be reduced by cold unlabeled antibody (blocking) treatment, confirming the high binding specificity of the 89 Zr-CD25 IgG of the present invention.
  • the present invention developed 89 Zr-labeled CD25 IgG, which selectively targets the CD25 antigen, which is expressed in large quantities on the surface of refractory lymphoma and is expected to be a target of therapeutic antibody drugs, and succeeded in developing a new immuno-PET technology using it. This is disclosed for the first time in the present invention.
  • the present invention provides a composition for positron emission tomography imaging for tumor diagnosis, comprising 89 Zr labeled anti-CD25 antibody and 9 Zr labeled anti-CD25 antibody as active ingredients.
  • CD25 corresponds to the alpha subunit of the heterodimeric interleukin 2 receptor (IL2 receptor) (i.e., IL2RA (Interleukin-2 receptor alpha chain)) and plays an important role in the survival and proliferation of lymphoma cells. Therefore, immuno-PET technology targeting CD25 can be used to detect lymphoma foci in the body and non-invasively and quantitatively evaluate CD25 expression. In addition, it provides pharmacokinetic information to evaluate the delivery of anti-CD25 antibody-based medicines to lymphoma, which can be used to select treatment targets and predict treatment effects.
  • IL2 receptor heterodimeric interleukin 2 receptor
  • IL2RA Interleukin-2 receptor alpha chain
  • CD25 is rarely or only expressed in small amounts in normal cells, but is expressed abundantly only in activated normal regulatory T cells (Treg cells). These immune cells infiltrate the tumor and suppress the immune response of normal cytotoxic T cells. Contributes to treatment resistance through immune evasion. Therefore, immuno-PET technology targeting CD25 can be used not only for imaging lymphomas that express CD25, but also for imaging Treg cells infiltrated within tumors that interfere with treatment. In this case, the 89 Zr radioisotope was optimized for immuno-PET, which evaluates the in vivo pharmacokinetics of antibodies, due to its advantages such as high in vivo stability and physical half-life of 3.3 days.
  • the 89 Zr-labeled anti-CD25 antibody and the immuno-PET imaging technology using it are used to diagnose cancer and predict the effectiveness of anti-CD25 antibody treatment through in-vivo imaging of refractory lymphoma and Treg cells, and to use the 177 Lu-labeled anti-CD25 antibody. It is important for the success of radioimmunotherapy strategies.
  • immuno-PET using an antibody labeled with gamma ray emitting 89 Zr is optimal to evaluate the efficiency of delivery to lymphoma.
  • the 'anti-CD25 antibody' or 'antibody against CD25' refers to a specific protein molecule directed against the antigenic site of CD25.
  • the antibody refers to an antibody that specifically binds to the CD25 protein (cell surface antigen), and includes polyclonal antibodies, monoclonal antibodies, and recombinant antibodies.
  • the antibody it may be preferable for the antibody to be a monoclonal antibody, which is a group of antibodies in which the amino acid sequences of the heavy and light chains of the antibody are substantially identical.
  • Generating antibodies against CD25 as described above can be easily produced using techniques well known in the art.
  • Polyclonal antibodies can be produced by a method well known in the art, which involves injecting the CD25 protein antigen into an animal and collecting blood from the animal to obtain serum containing the antibody.
  • These polyclonal antibodies can be prepared from any animal species host, such as goats, rabbits, sheep, monkeys, horses, pigs, cows and dogs.
  • Monoclonal antibodies can be produced using the hybridoma method or phage antibody library technology well known in the art.
  • Antibodies in the present invention also include functional fragments of the antibody molecule as well as intact forms having two full-length light chains and two full-length heavy chains.
  • a functional fragment of an antibody molecule refers to a fragment that retains at least an antigen-binding function, and preferably, the fragment has at least 50% or 60% of the LRRC15 binding affinity of the parent antibody. Hold 70%, 80%, 90%, 95% or 100% or more.
  • the functional fragment of the antibody may preferably include a hinge region, but is not limited thereto, and may be in the form of F(ab')2, F(ab'), etc., for example.
  • F(ab')2 is a fragment produced by hydrolyzing an antibody with pepsin, and consists of two Fabs connected by a disulfide bond at the heavy chain hinge.
  • F(ab') is a monomeric antibody fragment in which a heavy chain hinge is added to Fab obtained by reducing the disulfur bond of the F(ab')2 fragment.
  • the antibody applied in the present invention is not limited thereto, but may be, for example, selected from the group consisting of IgG, IgA, IgM, IgE, and IgD, and is preferably an IgG antibody.
  • the antibody of the present invention may be selected from the group consisting of IgG1, IgG2, IgG3, and IgG4, and preferably may be IgG1 or IgG2.
  • the antibody of the present invention may most preferably be IgG2a.
  • the 89 Zr (zirconium) is known as a medical radioisotope capable of emitting positrons. Its physical half-life is 78.41 hours, and upon decay, electron capture (76.6%) and positron emission (22.3%) occur spontaneously.
  • the composition can detect tumors or tumor-infiltrating regulatory T cells, but is not limited thereto.
  • Treg cells can move within tissues and have the characteristic of influencing resistance to existing tumor treatments through immune evasion when infiltrating into a tumor.
  • it has been reported to suppress the immune response following the treatment of tumors, for example, lymphoma, and is characterized by abundant expression of CD25.
  • the regulatory T cells may preferably be those that abundantly express CD25 on the cell surface (CD25 positive (+) Treg cells). Since the antibody of the present invention has significantly excellent binding affinity and binding specificity for Treg cells, 89 Zr-CD25 antibody PET imaging can also be used to identify the role of Treg cells in treatment resistance.
  • the tumor may be lymphoma, but is not limited thereto.
  • the tumor includes cancer in the sense commonly known in the art and includes all tumor tissue or tumor cells.
  • the tumor if the tumor abundantly expresses CD25 as a cell surface antigen, The type is not particularly limited.
  • the lymphoma includes non-Hodgkin's lymphoma (NHL), primary, secondary, recurrent lymphoma, indolent lymphoma, aggressive non-Hodgkin's lymphoma (NHL), follicular lymphoma (FL), and chronic lymphocytic lymphoma.
  • NDL leukemia
  • MZL marginal zone lymphoma
  • DLBCL diffuse large B-cell lymphoma
  • MCL mantle cell lymphoma
  • TL transforming lymphoma
  • PTCL peripheral T-cell lymphoma
  • It may be lymphoma or T-cell lymphoma, but is not limited thereto.
  • the 89 Zr-CD25 antibody of the present invention and the medical imaging method using the same are 177 Lu-CD25 antibody-based radiation for the treatment of refractory lymphoma and inhibition of recurrence due to the binding affinity and binding specificity for the above-mentioned tumor and intra-tumor infiltrating Treg cells. It plays a role in promoting the development of immunotherapy technology.
  • the establishment of 89 Zr labeling technology for anti-CD25 antibodies can also be used in 177 Lu labeling technology, contributing to the synthesis of therapeutic 177 Lu-CD25 antibodies.
  • the composition of the present invention may contain the function of a contrast agent used in medical imaging.
  • contrast enhancement may mean that a special drug called a contrast agent is injected into the human body to make lesions or blood vessels more visible in many tests performed in radiology departments.
  • contrast agent is required to view blood vessels, but since the 2000s, MRI has been able to view blood vessels without contrast agent.
  • a contrast enhanced method is used. Radioactive substances or vasodilators are administered during PET or SPECT examinations, but these are not called contrast agents.
  • the composition according to the present invention may include a contrast agent.
  • the composition may be for intravenous injection, but is not limited thereto.
  • the specific administration route is not particularly limited.
  • the composition of the present invention may be used for imaging or monitoring tumors or intra-tumor infiltrating regulatory T cells, but is not limited thereto. Therefore, the present invention provides a method for diagnosing tumors, comprising the 89 Zr labeled anti-CD25 antibody as an active ingredient; A composition for detecting or imaging a tumor or infiltrating regulatory T cells within a tumor is provided.
  • the present invention provides a method of preparing the 89 Zr labeled anti-CD25 antibody comprising the following steps:
  • step (b) reacting the anti-CD25 antibody reduced in step (a) with deferoxamine-maleimide (DFO-Mal) to produce a DFO-Mal conjugated anti-CD25 antibody; and
  • DFO-Mal deferoxamine-maleimide
  • step (c) reacting the DFO-Mal conjugated anti-CD25 antibody produced in step (b) with 89 Zr-oxalate to obtain an 89 Zr labeled anti-CD25 antibody.
  • the reaction in step (a) is a reduction reaction, whereby the disulfide bond chain of the antibody hinge region is selectively reduced and the bond is broken.
  • a pair of thiols reduced disulfide
  • the anti-CD25 antibody and TCEP tris-carboxyethylphosphine
  • the anti-CD25 antibody and TCEP tris-carboxyethylphosphine
  • the anti-CD25 antibody and TCEP tris-carboxyethylphosphine
  • the anti-CD25 antibody and TCEP tris-carboxyethylphosphine
  • the anti-CD25 antibody and TCEP tris-carboxyethylphosphine
  • the reaction in step (a) may be performed at a temperature of 15 to 25° C., but is not limited thereto. In one embodiment of the present invention, the reaction in step (a) may be preferably performed at a temperature of 21 to 23°C.
  • the time for the reaction in step (a) is not particularly limited, and a person skilled in the art can appropriately select and apply the optimal time depending on the yield of the desired reaction product, but a preferred example in the present invention is 5 to 30 minutes. It may be characterized by reacting, and more preferably, reacting for 10 to 20 minutes. In step (a), delicate reaction conditions and control that do not break other double sulfide bonds in the antibody other than the hinge region are very important.
  • the reaction in step (b) is preferably a chelation reaction, thereby producing a DFO-Mal conjugated anti-CD25 antibody.
  • the reaction in step (b) may be preferably carried out in the presence of EDTA.
  • the reaction in step (b) may be performed at a temperature of 15 to 25° C., but is not limited thereto.
  • the reaction in step (b) may be preferably performed at a temperature of 21 to 23°C.
  • the mixing ratio of each material (reduced anti-CD25 antibody and deferoxamine-maleimide (DFO-Mal) obtained in step (a)) is determined by those skilled in the art to obtain the desired reaction product.
  • a person skilled in the art can appropriately select and apply optimal conditions depending on the yield and speed of acquisition.
  • a person skilled in the art can appropriately select and apply the optimal time for the reaction in step (b) according to the yield of the desired reaction product. What can be done is not particularly limited, but a preferred example may be reacting for 30 to 120 minutes.
  • Step (c) is a step for labeling 89 Zr as a tracer that can be used in PET (positron emission tomography) and/or medical imaging techniques that can be used in conjunction with PET, and the reaction in step (b) is preferred. It may be a chelation reaction. Through the above reaction, the 89 Zr labeled anti-CD25 antibody of the present invention is finally obtained.
  • the reaction in step (c) may be performed at a temperature of 15 to 25° C., but is not limited thereto. In one embodiment of the present invention, the reaction in step (c) may be preferably performed at a temperature of 21 to 23°C.
  • step (c) the mixing ratio of each material ( 89 Zr-oxalate and DFO-Mal conjugated anti-CD25 antibody produced in step (b)) is adjusted according to the desired reaction product yield and yield rate by those skilled in the art. A person skilled in the art can appropriately select and apply optimal conditions.
  • the time for the reaction in step (c) is not particularly limited, and a person skilled in the art can appropriately select the optimal time depending on the yield of the desired reaction product, but a preferred example is reaction for 30 to 120 minutes. It could be.
  • 89 Zr-oxalate may be neutralized before being mixed with the DFO-Mal conjugated anti-CD25 antibody produced in step (b).
  • the neutralization (neutralization reaction) is not particularly limited in the type of material (particularly basic material) used as long as it uses an acid and base neutralization reaction known in the art, but for example, it can be mixed with sodium carbonate (Na 2 CO 3 ). It may be neutralized.
  • a person skilled in the art can appropriately select and apply optimal conditions for reaction conditions such as the mixing ratio of each substance, reaction temperature, and reaction time, depending on the desired reaction product yield and acquisition rate.
  • DFO-Mal conjugated anti-CD25 antibody is mixed to perform a chelation reaction with 89 Zr.
  • the antibody may be IgG, but is not limited thereto.
  • the antibody may have the following characteristics, but is not limited thereto:
  • 89 Zr is specifically bound to the disulfide bond site of the anti-CD25 antibody hinge region
  • conjugates As used herein, the term 'conjugate' includes both conjugates and complexes, and the above terms may be used interchangeably in this specification.
  • the antibody of the present invention may preferably be characterized in that the disulfide bond in the hinge region is broken and two 89 Zr atoms are bonded. Most preferably, the antibody of the present invention may be characterized in that one disulfide bond in the hinge region is broken and two 89 Zr atoms are bonded to it (see Figure 4), and it has a structure unique to the present invention.
  • the labeled antibody of the present invention is characterized by high labeling efficiency for target tissues or cells while maintaining high structural stability in vivo.
  • the present invention provides a positron emission tomography method for tumor diagnosis, comprising the following steps:
  • PET positron emission tomography
  • CT Computed Tomography
  • MRI Magnetic Resonance Imaging
  • the imaging may use a technology selected from the group consisting of PET (Positron Emission Tomography), PET/CT, PET/MR, and immuno-PET, but is not limited thereto. .
  • the present invention provides a kit for positron emission tomography imaging for tumor diagnosis, including the composition and instructions.
  • the composition is a composition of the invention, and the instructions describe the method as It may be described, but is not limited thereto.
  • the “kit” of the present invention may include other components, devices, materials, etc. commonly required for positron emission tomography and/or methods for imaging such imaging.
  • all components included in the kit can be used one or more times without limitation, there is no restriction on the order or subsequent use of each substance, and the application of each substance may be carried out simultaneously or in small steps.
  • the kit of the present invention may include a container in addition to the composition and the instructions.
  • the container may serve to package the component, and may also serve to store and secure the component.
  • the material of the container may take the form of, for example, a bottle, a tub, a sachet, an envelope, a tube, an ampoule, etc., which may be partially or entirely made of plastic, glass, paper, or foil. , wax, etc.
  • the container may be equipped with a completely or partially removable closure that may initially be part of the container or may be attached to the container by mechanical, adhesive, or other means, and may also provide access to the contents by needle. A stopper can be installed.
  • the kit may include an external package, and the external package may include, but is not limited to, instructions for use of the components.
  • the present invention provides detection of tumor or/and tumor infiltrating regulatory T cells (regulatory T cells, Tregs);
  • a method of acquiring a PET image is provided using the 89 Zr-labeled anti-CD25 antibody of the present invention as a contrast agent.
  • the present invention provides a composition comprising an 89 Zr-labeled anti-CD25 antibody as an active ingredient for use in the diagnosis of tumors; or detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.
  • the present invention relates to diagnosis of tumors; Alternatively, it provides the use of a composition comprising a water 89 Zr labeled anti-CD25 antibody as an active ingredient for preparing an imaging agent or detection of tumor or intratumoral infiltrating regulatory T cells.
  • 'individual' refers to an object that requires diagnosis or treatment of a disease, and more specifically, human or non-human mammals (primates), mice, rats, dogs, cats, It may be a mammal such as a horse or a cow, but is not limited thereto.
  • FIG. 2a The process of changing the chemical bond within the antibody that occurs during the conventional labeling technology process is schematically shown in Figure 2a, and accordingly, the antibody is labeled with the structure shown in Figure 2b.
  • two 89 Zr atoms are specifically bound to the disulfide bond site of the hinge region of the anti-CD25 antibody in a unique manner, which is different from the conventional techniques of Figures 2a and 2b, and when imaged by immuno-PET, Very excellent images or videos can be obtained.
  • the method for producing the 89 Zr labeled anti-CD25 antibody according to the present invention is described in detail.
  • anti-CD25 antibodies derived from humans or mice InVivoMAb anti-human CD25 (IL-2R ⁇ ; Catalog# BE0014) or InVivoMAb anti-mouse CD25 (IL-2R ⁇ ; Catalog# BE0012) from BioXCell (West Riverside, NH); Mouse monoclonal IgG2a anti- CD25 Ab (clone, 7G7/B6), rat monoclonal IgG1 Ab (clone, PC-61.5.3)) position-specifically ( Site-specific deferoxamine-maleimide (deferoxamine[DFO]-maleimide) was conjugated.
  • the deferoxamine maleimide conjugated anti-CD25 antibody is referred to as 'DFO-conjugated CD25 IgG', or 'DFO-Mal conjugated anti-CD25 antibody'.
  • the anti-CD25 antibody was reduced with TCEP (tris-carboxyethylphosphine) at a molar concentration of 70 to 130 times higher than the molar concentration of the above antibody, and then reacted with deferoxamine-maleimide chelator to form a total of two 89 Zr at the disulfide bond site in the antibody hinge region. It was possible to specifically bind the deferoxamine-maleimide atom, and the specific method is as follows.
  • 2 mg anti-CD25 antibody was reduced by reacting with 100 mM TCEP (tris(2-carboxyethyl)phosphine, 1:100 molar ratio) at room temperature for 20 minutes. Subsequently, the reduced anti-CD25 antibody was diluted in 0.1 M sodium phosphate solution containing 150mM NaCl and 1mM EDTA (ethylene diamine tetraacetic acid). 56.4 ⁇ L of 2mM deferoxamine-maleimide (DFO-Mal) chelator was added and reacted at room temperature for 1 hour to attach to the sulfhydryl group of the cysteine residue in the hinge region of the anti-CD25 antibody.
  • DFO-Mal deferoxamine-maleimide
  • the reaction product was eluted with a PD-10 column to concentrate the DFO-conjugated CD25 antibody fraction, and DFO-conjugated CD25 IgG was obtained from the fraction.
  • the molar ratio of deferoxamine-maleimide:antibody was 60:1.
  • SDS-PAGE Non-reducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis was performed to confirm whether the DFO-conjugated anti-CD25 antibody (human or mouse antibody) prepared by the above method was in a reduced state. Specifically, 2 ⁇ g of DFO-conjugated anti-CD25 antibody (human or mouse antibody) was diluted in water and then mixed with 5x non-reducing sample buffer without dithiothreitol. Afterwards, it was boiled at 95°C for 10 minutes, electrophoresis was performed on an 8% SDS PAGE gel, and stained with 0.5% coomassie blue.
  • CD25 Ab-deferoxamine maleimide also referred to as DFO-conjugated CD25 IgG
  • 89 Zr can be successfully conjugated to CD25 IgG according to Example 1, and the conjugation efficiency is about 70% and the purity is over 98%, making it possible to produce a remarkably excellent 89 Zr-labeled CD25 IgG. This has been confirmed.
  • radio-instant thin layer chromatography (radio-iTLC) was performed. Specifically, iTLC-SG glass microfiber chromatography paper coated with silica gel was used. In addition, 89 Zr-labeled CD25 IgG (human or mouse antibody), respectively, was added to PBS or FBS to serve as an experimental group. Each experimental group was placed in PBS or FBS and reacted at 37°C for 1 to 7 days, and then chromatography was performed using 50 mM EDTA (ethylene diamine tetraacetic acid, pH 5.5) as a solvent. Intact 89 Zr-CD25 IgG remains in the basal position, whereas free 89 Zr 4+ ions and 89 Zr-EDTA migrate along the solvent front and are separated.
  • mM EDTA ethylene diamine tetraacetic acid
  • lymphoma cells with the highest CD25 expression level were selected.
  • the expression levels of the CD25 marker were examined in human lymphoma cell lines H9, Jurkat, and SUDHL1 and mouse lymphoma cell line EL4.
  • the cancer cells were lysed, the proteins were separated and quantified, and 15 ⁇ g of each protein was electrophoresed using a 10% gel and transferred to a membrane, followed by rabbit-anti-CD25 primary antibody (abcam #ab231441; 1:1000 dilution). Reacted overnight at 4°C. After washing with TBST buffer three times for 10 minutes each, it was reacted with HRP-conjugated anti-rabbit secondary antibody (Cell Signaling #7074S; 1:2000 dilution) for 1 hour at room temperature. After washing with TBST buffer three times for 10 minutes each, the membrane was reacted with an enhanced chemiluminescence substrate and the film was exposed to detect and quantify the band intensities of CD25 protein.
  • cancer cells were stained by attaching a FITC or phycoerythrin (PE) fluorescence signal to anti-CD25 IgG, and then FACS analysis (fluorescence-activated cell sorting analysis) was performed.
  • FACS analysis fluorescence-activated cell sorting analysis
  • the cells were harvested with a nonenzymatic cell-dissociation solution, washed, and incubated with phycoerythrin for 30 minutes at room temperature (RT) in a phosphate-buffered saline solution containing 5% fetal bovine serum and 0.2% bovine serum albumin. Incubation was performed with antibody.
  • FACS buffer 500 mL of FACS buffer was added again, and flow cytometry was performed with FACSCalibur (BE Biosciences) using CellQuest software.
  • the cells were cultured and reacted with 89 Zr-CD25 IgG (human), the cells were washed, and the uptake rate was measured in %ID by a conventional method using a gamma counter. At this time, H9 and Jurkat cell lines were set as comparison groups.
  • binding was inhibited with a large amount of cold anti-CD25 antibody (binding inhibition experiment) and binding specificity was quantified as % blocking.
  • the EL4 cell line was used as a comparison group.
  • CD25(+) Treg cells were purified and isolated from the thymus or spleen of normal mice using the Easy prep kit (Stem Cell Tech) according to the manufacturer's protocol. Specifically, CD25(-) T cells and CD25(+) Treg cells were isolated and purified from the thymus of mice using FACS (see Figure 9a), and the Treg cells were stained with hematoxylin and eosin (see Figure 9b). Additionally, the number of CD25(+) Treg cells was expanded for 2 weeks using anti-CD3 antibody and IL2. Afterwards, the degree of binding of 89 Zr-CD25 IgG to CD25(-) T cells and CD25(+) Treg cells was analyzed in the same manner as in Example 3-2.
  • the binding of 89 Zr-CD25 IgG to CD25(+) Treg cells was confirmed to be significantly higher at 391.2 ⁇ 135.2% compared to CD25(-) T cells isolated from the mouse thymus.
  • the high 89 Zr-CD25 IgG binding to CD25(+) Treg cells is inhibited by 72.1% in the presence of 0.5 ⁇ M of cold unlabeled antibody, so the 89 Zr-CD25 IgG according to the present invention has excellent binding ability.
  • Example 5 In vivo mouse lymphoma model 89 Analysis of body distribution of Zr-CD25 IgG
  • the uptake of 89 Zr-CD25 IgG by SUDHL1 lymphoma tumor in the mouse body was confirmed to be 9.4 ⁇ 2.3 %ID/g. This is 10 times more than that in muscle, and the uptake rate in the heart, lungs, liver, spleen, and kidneys was also found to be only about 30 to 40% of that in tumors.
  • the 89 Zr-CD25 IgG antibody of the present invention has excellent tumor-specific targeting and binding effects even in vivo.
  • 89 PET image analysis was performed to confirm the imaging (imaging, contrast) effect of Zr-CD25 IgG.
  • SUDHL1 human T cell lymphoma cells were cultured and 1 x 10 7 SUDHL1 cells were subcutaneously injected into the shoulder area of immunodeficient balb/C nude mice.
  • mice were pre-injected with cold antibodies at a 5:1 molar ratio 1 hr before to analyze the tumor binding (tumor uptake) specificity of 89 Zr-CD25 IgG.
  • PET-based tissue radioactivity was analyzed by applying regions-of-interest (ROIs) in non-attenuation corrected images, including blood pool, major organs, and tumor.
  • ROIs regions-of-interest
  • micro-PET/CT imaging results showed high 89 Zr-CD25 IgG binding (uptake) with excellent contrast in the tumor area.
  • 0.8 mg of cold unlabeled antibody was first injected into the same mouse model and then 89 Zr-CD25 IgG was injected, it was confirmed that the tumor's 89 Zr-CD25 IgG uptake was significantly reduced and the contrast decreased (see Figure 13). .
  • 89 Zr-CD25 IgG prepared according to the method of the present invention has stability in serum and has excellent binding ability to tumors and Tregs infiltrated within the tumor, as well as specificity for this binding ability. It has been done.
  • excellent quality tumor-specific PET images can be obtained through these characteristics, and it can be used in a variety of ways as a composition and method for diagnosis, imaging, and confirmation of treatment effects for lymphomas, including intractable lymphomas. This has been confirmed.
  • the present invention relates to an imaging agent for a PET-based imaging method comprising an 89 Zr-labeled anti-CD25 antibody, and a method for imaging a tumor or a method for imaging tumor-infiltrating regulatory T cells using the same.
  • the 89 Zr-labeled anti-CD25 antibody prepared by the unique method of the present invention is stably maintained in serum, and has a specific high concentration in tissues where CD25 antigen is present, tumors, especially lymphomas, or areas where a large number of regulatory T cells are present. It exhibits an uptake rate and is characterized by significantly excellent binding affinity and binding specificity.
  • lymphoma particularly refractory and/or T-cell lymphoma
  • diagnosis of lymphoma can be used for detecting, imaging, and monitoring lymphoma or infiltrating regulatory T cells within lymphoma, and is expected to be useful as a diagnostic, detection, monitoring, and imaging agent for effective non-invasive medical imaging.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Optics & Photonics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to: an imaging agent of a PET-based imaging method, the agent comprising 89Zr-labeled anti-CD25 antibodies; and a tumor imaging method or tumor-infiltrating regulatory T cell imaging method using same. The unique 89Zr-labeled anti-CD25 antibodies according to the present invention exhibit a very high uptake rate, specifically in tissue in which CD25 antigens are present, and especially in portions in which lymphomas or regulatory T cells are present, and therefore can be effectively used as an effective non-invasive imaging agent capable of identifying, diagnosing, imaging, and monitoring lymphomas and/or regulatory T cells.

Description

89ZR 표지된 항-CD25 항체를 이용한 IMMUNO-PETIMMUNO-PET using 89ZR labeled anti-CD25 antibody

본 발명은 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 의료 영상 촬영용 조성물에 관한 것이다. The present invention relates to a composition for medical imaging comprising 89 Zr labeled anti-CD25 antibody as an active ingredient.

본 출원은 2022년 08월 11일에 출원된 한국특허출원 제10-2022-0100554호에 기초한 우선권을 주장하며, 해당 출원의 명세서 및 도면에 개시된 모든 내용은 본 출원에 원용된다. This application claims priority based on Korean Patent Application No. 10-2022-0100554 filed on August 11, 2022, and all contents disclosed in the specification and drawings of the application are incorporated in this application.

양전자 방출 단층 촬영(Positron emission tomography, PET)은 양전자를 방출하는 방사성 의약품을 이용하여 질병으로 인한 인체의 생물학적 변화를 영상화 하여 질병의 조기진단 및 질환의 치료방법 결정에 정확한 정보를 제공할 수 있다.Positron emission tomography (PET) uses radioactive pharmaceuticals that emit positrons to image biological changes in the human body caused by disease, and can provide accurate information for early diagnosis of disease and decision-making on treatment methods.

임상에서 가장 많이 이용되고 있는 암 진단용 방사성 의약품은 [18F]FDG 이다. 18F-FDG는 방사성 동위원소를 지닌 포도당 유사체로서, 포도당 수송체(glucose transporter: Glut)를 통하여 세포 내로 유입되어 인산화(phosphorylation)된 후 더 이상 대사가 되지 않고 세포 내에 머무르며 양전자를 방출한다. 암세포는 세포 표면에 포도당 수송체의 수가 정상 세포보다 증가되어 있어 더 많은 18F-FDG가 암세포내로 유입된다. 따라서 기존의 영상의학적 검사는 인체 내 구조를 영상화하여 해부학적으로 병소부위를 찾아 진단하는 반면 18F-FDG PET/CT는 해부학 변화 이전의 생화학적 변화를 영상화할 수 있다. The radiopharmaceutical for cancer diagnosis most commonly used in clinical practice is [ 18 F]FDG. 18 F-FDG is a glucose analogue with a radioactive isotope that enters the cell through the glucose transporter (Glut) and is phosphorylated. It is no longer metabolized and stays in the cell, emitting positrons. Cancer cells have an increased number of glucose transporters on the cell surface compared to normal cells, so more 18 F-FDG flows into cancer cells. Therefore, while existing radiological tests image structures within the human body to find and diagnose anatomical lesions, 18 F-FDG PET/CT can image biochemical changes before anatomical changes.

림프종(lymphoma)은 림프계 조직에서 발생하는 가장 흔한 악성 혈액암이다. 크게 비호지킨성 림프종 및 호지킨성 림프종으로 분류되며, 그 중 대다수의 종류를 차지하는 비호지킨성 림프종은 B세포 림프종과 T세포 림프종으로 분류될 수 있다. 림프종의 치료에 있어서 항체기반 의약품이 면역반응을 통한 암세포 제거효과가 현저히 크게 나타나는 것으로 보고되고 있고, 일례로 B 세포 림프종에는 리툭시맵이라는 단일클론 항체가 림프종 치료 경과를 크게 개선하는 것으로 알려져 있다. Lymphoma is the most common malignant blood cancer that occurs in lymphoid tissue. It is broadly classified into non-Hodgkin's lymphoma and Hodgkin's lymphoma, of which non-Hodgkin's lymphoma, which accounts for the majority, can be classified into B-cell lymphoma and T-cell lymphoma. In the treatment of lymphoma, antibody-based medicines are reported to have a significantly greater effect in eliminating cancer cells through immune responses. For example, in B-cell lymphoma, a monoclonal antibody called rituximab is known to significantly improve the course of lymphoma treatment.

그러나 T 세포 림프종을 포함한 난치성 림프종 환자가 현저히 증가하고 있고, 표준 항암치료에 반응이 불량하며 재발이 흔할 뿐만 아니라, 다른 암종 대비 효과적인 항체기반 의약품이 아직 개발되지 못한 실정으로 인하여 상기 질환을 극복할 수 있는 새로운 치료전략이 절실하다.However, the number of patients with refractory lymphoma, including T-cell lymphoma, is significantly increasing, the response to standard anti-cancer treatment is poor, recurrence is common, and antibody-based medicines that are more effective than other cancer types have not yet been developed, making it difficult to overcome the disease. New treatment strategies are urgently needed.

난치성 림프종의 항체 치료제 개발이 성공하기 위해서는 종양에 표적 발현이 충분한지 평가하고 림프종에 치료제가 잘 도달하는지 모니터링하는 영상기술이 필수적이나, 기존의 18F-FDG PET/CT로는 제공할 수 없는 생물학적 정보이다(비특허문헌 1). 이를 극복하기 위해서 종양 특이적 세포-표면 항원을 표적하는 immuno-PET 영상기술이 관심을 받고 있다. immuno-PET 영상기술은 신약개발 전임상시험 기간을 단축시키고, 림프종 세포가 충분한 양의 표적 항원을 발현하는 환자를 선별하며, 항체 치료의 효과를 예측할 수 있다는 특징이 있으나, 여전히 림프종 특이적인 새로운 항원의 표적이 필요하다. In order to successfully develop antibody treatments for refractory lymphoma, imaging technology is essential to evaluate whether target expression is sufficient in the tumor and monitor whether the treatment reaches the lymphoma. However, biological information cannot be provided by existing 18 F-FDG PET/CT. (Non-patent Document 1). To overcome this, immuno-PET imaging technology targeting tumor-specific cell-surface antigens is attracting attention. Although immuno-PET imaging technology has the characteristics of shortening the preclinical trial period for new drug development, selecting patients whose lymphoma cells express a sufficient amount of target antigen, and predicting the effect of antibody treatment, it still has the potential to produce new lymphoma-specific antigens. A target is needed.

현재 항-CD25 항체를 기반으로 난치성 림프종 암세포만 찾아서 제거시키는 면역치료가 임상시험 진행 중에 있다. 항체 치료제 개발에는 몸속에서 어떻게 이동하고 표적 암에 얼마나 전달되는지 보여주는 immuno-PET가 중요하지만, 항-CD25 항체 치료제 개발을 위한 immuno-PET 영상기술은 아직 개발되지 못한 상황이다. 또한 종양 내 침윤한 Treg cell(tumor-infiltrating regulatory T cell) 역시 CD25를 발현하면서 면역감시 회피(immune evasion)를 통한 치료 내성에 중요한 역할을 하지만 이를 이미징하는 기술은 아직 없다. 따라서 CD25 표적 immuno-PET 기술을 개발하면 몸속 림프종의 CD25 발현을 정량적으로 평가할 수 있을 뿐만 아니라 종양에 침윤한 Treg cell을 이미징하여 림프종 치료의 발전에 기여할 것이다. Currently, clinical trials are underway for immunotherapy that finds and removes only refractory lymphoma cancer cells based on anti-CD25 antibodies. In the development of antibody treatments, immuno-PET is important to show how it moves in the body and how much is delivered to the target cancer, but immuno-PET imaging technology for developing anti-CD25 antibody treatments has not yet been developed. In addition, Treg cells (tumor-infiltrating regulatory T cells) that have infiltrated within the tumor also express CD25 and play an important role in treatment resistance through immune evasion, but there is still no technology to image them. Therefore, the development of CD25-targeted immuno-PET technology will not only allow quantitative evaluation of CD25 expression in lymphoma in the body, but will also contribute to the development of lymphoma treatment by imaging Treg cells infiltrating tumors.

나아가, 항체 의약품 중에 항체-약물 결합체(antibody-drug conjugate)가 부상하고 있는데, 그 중에서 베타방출 동위원소를 탑재한 항체 결합체가 림프종의 방사면역치료제로 기대를 모으고 있으며, 현재 방사면역치료에는 177Lu 방사성동위원소가 선호되고 있다. 난치성 림프종에 대한 방사선 탑재 항-CD25 항체 방사면역치료도 전임상 및 임상 개발 단계에 있지만 이 기술을 뒷받침하기 위한 immuno-PET 영상기술은 아직 없다. Furthermore, among antibody drugs, antibody-drug conjugates are emerging. Among them, antibody conjugates loaded with beta-emitting isotopes are raising expectations as radioimmunotherapy agents for lymphoma. Currently, radioimmunotherapy uses 177 Lu. Radioactive isotopes are preferred. Radiation-loaded anti-CD25 antibody radioimmunotherapy for refractory lymphoma is also in the preclinical and clinical development stages, but there is still no immuno-PET imaging technology to support this technology.

이에 본 발명자들은 본 발명 특유의 89Zr 표지된 항-CD25 항체를 이용하는 immuno-PET 영상기술을 개발하여 CD25를 발현하는 림프종 세포와 Treg 세포에 높은 친화도로 결합함을 확인하였을 뿐만 아니라, 마우스 모델의 몸속 림프종에 높은 섭취와 방사성 표지되지 않은 항-CD25 항체에 의해 섭취가 억제되는 우수한 표적 특이도를 확인하여 본 발명을 완성하였다.Accordingly, the present inventors developed an immuno-PET imaging technology using the 89 Zr-labeled anti-CD25 antibody unique to the present invention and confirmed that it binds with high affinity to lymphoma cells and Treg cells expressing CD25, as well as to The present invention was completed by confirming the high uptake by lymphoma in the body and the excellent target specificity in which uptake was inhibited by non-radioactive anti-CD25 antibodies.

즉, 본 발명의 목적은, 새로운 치료표적으로 난치성 림프종과 Treg 세포를 몸속에서 영상화하여, 항체 치료제의 개발에 기여할 뿐만 아니라 치료 대상의 선정과 치료효과 예측에 활용할 수 있도록 마우스 및 사람 89Zr-CD25 항체를 개발하고 이를 이용한 immuno-PET 영상기술을 개발하여 이를 제공하는 데 있다. In other words, the purpose of the present invention is to image refractory lymphoma and Treg cells in the body as new treatment targets, so that it can be used to not only contribute to the development of antibody treatments, but also to select treatment targets and predict treatment effects, using mouse and human 89 Zr-CD25 The goal is to develop and provide antibodies and immuno-PET imaging technology using them.

이에 구체적으로, 본 발명의 목적은, 89Zr 표지된 항-CD25 항체를 제공하는 것이다.Specifically, the purpose of the present invention is to provide an 89 Zr labeled anti-CD25 antibody.

본 발명의 다른 목적은 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 조성물을 제공하는 것이다.Another object of the present invention is to provide a composition for positron emission tomography imaging for tumor diagnosis, comprising 89 Zr labeled anti-CD25 antibody as an active ingredient.

본 발명의 또 다른 목적은 하기 단계를 포함하는 89Zr 표지된 항-CD25 항체를 제조하는 방법을 제공하는 것이다:Another object of the present invention is to provide a method for preparing 89 Zr labeled anti-CD25 antibody comprising the following steps:

(a) 항-CD25 항체를 TCEP(tris-carboxyethylphosphine)와 반응시켜 환원 시키는 단계; (a) reducing the anti-CD25 antibody by reacting it with TCEP (tris-carboxyethylphosphine);

(b) 상기 (a) 단계에서 환원된 항-CD25 항체를 데페록사민-말레이미드(deferoxamine-maleimide, DFO-Mal)와 반응시켜, DFO-Mal 접합된 항-CD25 항체를 생성하는 단계; 및(b) reacting the anti-CD25 antibody reduced in step (a) with deferoxamine-maleimide (DFO-Mal) to produce a DFO-Mal conjugated anti-CD25 antibody; and

(c) 상기 (b) 단계에서 생성된 DFO-Mal 접합된 항-CD25 항체를 89Zr-옥살레이트(89Zr-oxalate)와 반응시킨 후, 89Zr 표지된 항-CD25 항체를 수득하는 단계.(c) reacting the DFO-Mal conjugated anti-CD25 antibody produced in step (b) with 89 Zr-oxalate to obtain an 89 Zr labeled anti-CD25 antibody.

본 발명의 또 다른 목적은, 종양 또는 종양 내 침윤(tumor infiltrating) 조절 T 세포(regulatory T cell, Treg)의 검출; 또는 종양 진단에 필요한 정보를 제공하기 위하여, 상기 본 발명의 89Zr 표지된 항-CD25 항체를 조영제로 사용하여 PET 영상을 획득하는 방법을 제공하는 것이다. Another object of the present invention is to detect tumor or tumor infiltrating regulatory T cells (regulatory T cells, Tregs); Alternatively, in order to provide information necessary for tumor diagnosis, a method of acquiring a PET image using the 89 Zr-labeled anti-CD25 antibody of the present invention as a contrast agent is provided.

본 발명의 또 다른 목적은 하기 단계를 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 방법을 제공하는 것이다:Another object of the present invention is to provide a positron emission tomography method for tumor diagnosis, comprising the following steps:

(i) 89Zr 표지된 항-CD25 항체를 개체에 투여하는 단계; 및(i) administering 89 Zr labeled anti-CD25 antibody to the subject; and

(ii) 상기 개체의 검출 이미지 또는 영상을 획득하는 단계.(ii) Obtaining a detection image or video of the object.

본 발명의 또 다른 목적은 상기 조성물, 및 설명서를 포함하는 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 키트를 제공하는 것이다.Another object of the present invention is to provide a kit for positron emission tomography imaging for tumor diagnosis, including the composition and instructions.

그러나, 본 발명이 이루고자 하는 기술적 과제는 이상에서 언급한 과제에 제한되지 않으며, 언급되지 않은 또 다른 과제들은 아래의 기재로부터 본 발명이 속하는 기술 분야의 통상의 지식을 가진 자에게 명확하게 이해될 수 있을 것이다.However, the technical problem to be achieved by the present invention is not limited to the problems mentioned above, and other problems not mentioned can be clearly understood by those skilled in the art from the description below. There will be.

상기와 같은 목적을 달성하기 위하여, 본 발명은 89Zr 표지된 항-CD25 항체를 제공한다.In order to achieve the above object, the present invention provides an 89 Zr labeled anti-CD25 antibody.

본 발명은 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 조성물을 제공한다.The present invention provides a composition for positron emission tomography imaging for tumor diagnosis, comprising 89 Zr-labeled anti-CD25 antibody as an active ingredient.

본 발명의 일 실시예에 있어서, 상기 조성물은 종양 또는 종양 내 침윤 조절 T 세포(tumor-infiltrating regulatory T cell)를 검출할 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the composition can detect tumors or tumor-infiltrating regulatory T cells, but is not limited thereto.

본 발명의 일 실시예에 있어서, 상기 종양은 림프종일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the tumor may be lymphoma, but is not limited thereto.

본 발명의 일 실시예에 있어서, 상기 조성물은 정맥주사용일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the composition may be for intravenous injection, but is not limited thereto.

본 발명은 하기 단계를 포함하는 89Zr 표지된 항-CD25 항체를 제조하는 방법을 제공한다:The present invention provides a method of making 89 Zr labeled anti-CD25 antibody comprising the following steps:

(a) 항-CD25 항체를 TCEP(tris-carboxyethylphosphine)와 반응시켜 환원 시키는 단계; (a) reducing the anti-CD25 antibody by reacting it with TCEP (tris-carboxyethylphosphine);

(b) 상기 (a) 단계에서 환원된 항-CD25 항체를 데페록사민-말레이미드(deferoxamine-maleimide, DFO-Mal)와 반응시켜, DFO-Mal 접합된 항-CD25 항체를 생성하는 단계; 및(b) reacting the anti-CD25 antibody reduced in step (a) with deferoxamine-maleimide (DFO-Mal) to produce a DFO-Mal conjugated anti-CD25 antibody; and

(c) 상기 (b) 단계에서 생성된 DFO-Mal 접합된 항-CD25 항체를 89Zr-옥살레이트(89Zr-oxalate)와 반응시킨 후, 89Zr 표지된 항-CD25 항체를 수득하는 단계.(c) reacting the DFO-Mal conjugated anti-CD25 antibody produced in step (b) with 89 Zr-oxalate to obtain an 89 Zr labeled anti-CD25 antibody.

본 발명의 일 실시예에 있어서, 상기 항체는 IgG일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the antibody may be IgG, but is not limited thereto.

본 발명의 일 실시예에 있어서, 상기 항체는 하기를 특징으로 할 수 있으나, 이에 제한되는 것은 아니다:In one embodiment of the present invention, the antibody may have the following characteristics, but is not limited thereto:

항-CD25 항체 힌지 영역(hinge region)의 이황화 결합(disulfide bond) 부위에 특이적으로 89Zr이 결합됨; 및 89 Zr is specifically bound to the disulfide bond site of the anti-CD25 antibody hinge region; and

89Zr 및 항-CD25 항체의 접합체(conjugate) 형태임.It is a conjugate form of 89 Zr and anti-CD25 antibody.

본 발명은 하기 단계를 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 방법을 제공한다:The present invention provides a positron emission tomography method for tumor diagnosis, comprising the following steps:

(i) 89Zr 표지된 항-CD25 항체를 개체에 투여하는 단계; 및(i) administering 89 Zr labeled anti-CD25 antibody to the subject; and

(ii) 상기 개체의 검출 이미지 또는 영상을 획득하는 단계.(ii) Obtaining a detection image or video of the object.

본 발명의 일 실시예에 있어서, 상기 방법은 PET(Positron Emission Tomography), PET/CT, PET/MR, 및 immuno-PET로 이루어진 군으로부터 선택되는 하나의 기술을 이용할 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the method may use a technology selected from the group consisting of PET (Positron Emission Tomography), PET/CT, PET/MR, and immuno-PET, but is not limited thereto. .

본 발명은 상기 조성물, 및 설명서를 포함하는 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 키트를 제공한다.The present invention provides a kit for positron emission tomography imaging for tumor diagnosis, including the composition and instructions.

본 발명의 일 실시예에 있어서, 상기 조성물은 본 발명의 조성물이고, 상기 설명서는 상기 방법이 기재된 것일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the invention, the composition is a composition of the invention, and the instructions describe the method as It may be described, but is not limited thereto.

또한, 본 발명은 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 조성물의, 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화 용도를 제공한다.In addition, the present invention provides a composition comprising an 89 Zr-labeled anti-CD25 antibody as an active ingredient for use in the diagnosis of tumors; or detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.

또한, 본 발명은 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화제를 제조하기 위한, 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 조성물의 용도를 제공한다.Additionally, the present invention relates to diagnosis of tumors; Alternatively, it provides the use of a composition comprising 89 Zr-labeled anti-CD25 antibody as an active ingredient for preparing an imaging agent or detection of tumor or intratumoral infiltrating regulatory T cells.

또한, 본 발명은 89Zr 표지된 항-CD25 항의, 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화 용도를 제공한다.In addition, the present invention relates to the 89 Zr labeled anti-CD25, for diagnosis of tumors; or detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.

또한, 본 발명은 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화제를 제조하기 위한, 89Zr 표지된 항-CD25 항체의 용도를 제공한다.Additionally, the present invention relates to diagnosis of tumors; or for preparing an agent for detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.

89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 종양 진단을 위한 의료 촬영 영상화용 조성물, 이를 이용한 종양 진단; 종양 또는 종양 내 침윤 조절 T 세포(tumor-infiltrating regulatory T cell)의 영상화 방법에 관한 것이다.본 발명 특유의 89Zr 표지된 항-CD25 항체는 CD25 항원이 존재하는 조직 중에서도 특히 림프종 또는 조절 T 세포가 존재하는 부위에서 특이적으로 매우 높은 섭취율을 나타낸다는 점에서, 림프종 및/또는 조절 T 세포를 구별, 진단, 이미징 및 모니터링할 수 있는 효과적인 비침습적 PET 관련 촬영 영상화제로서 유용하게 사용 가능하다. 89 A medical imaging composition for tumor diagnosis comprising a Zr-labeled anti-CD25 antibody as an active ingredient, and tumor diagnosis using the same; It relates to a method for imaging a tumor or tumor-infiltrating regulatory T cells. The unique 89 Zr-labeled anti-CD25 antibody of the present invention is used to detect lymphoma or regulatory T cells among tissues in which the CD25 antigen is present. Since it shows a very high uptake rate specifically in the area where it exists, it can be usefully used as an effective non-invasive PET-related imaging agent that can distinguish, diagnose, image, and monitor lymphoma and/or regulatory T cells.

도 1은 난치성 림프종(lymphoma)과 종양 조직 내 regulatory T 세포(Treg)를 표적하는 본 발명의 89Zr-CD25 항체 개발과 이를 이용한 immuno-PET 영상기술 개발에 대한 개념을 개략적으로 나타낸 이미지이다.Figure 1 is an image schematically showing the concept of the development of the 89 Zr-CD25 antibody of the present invention targeting regulatory T cells (Treg) in refractory lymphoma and tumor tissue and the development of immuno-PET imaging technology using the same.

도 2a는 종래의 표지기술로 항체를 표지할 때, 표지 과정 중에 나타나는 항체 내 화학적 결합(bond)의 변화 과정을 개략적으로 나타낸 것이다. Figure 2a schematically shows the process of changes in chemical bonds within the antibody that occur during the labeling process when labeling an antibody with a conventional labeling technology.

도 2b는 상기 도 2a와 같은 종래의 표지 기술로 항체를 표지할 때, 표지물질 및 항체의 결합 구조를 개략적으로 나타낸 것이다. Figure 2b schematically shows the binding structure of a labeling substance and an antibody when labeling an antibody using a conventional labeling technology as shown in Figure 2a.

도 3a는 본 발명에서 89Zr을 인간 항-CD25 항체의 hinge-region cysteine 잔기에 특이적으로 표지하기 위한 과정 중, TCEP 환원 후 및 DFO-conjugation 후 분자의 크기를 SDS PAGE를 이용하여 확인한 결과를 나타낸다.Figure 3a shows the results of confirming the size of the molecule using SDS PAGE after TCEP reduction and DFO-conjugation during the process of specifically labeling the hinge-region cysteine residue of the human anti-CD25 antibody with 89 Zr in the present invention. indicates.

도 3b는 본 발명에서 89Zr을 마우스 항-CD25 항체의 hinge-region cysteine 잔기에 특이적으로 표지하기 위한 과정 중, TCEP 환원 후 및 DFO-conjugation 후 분자의 크기를 SDS PAGE를 이용하여 확인한 결과를 나타낸다. Figure 3b shows the results of confirming the size of the molecule using SDS PAGE after TCEP reduction and DFO-conjugation during the process of specifically labeling the hinge-region cysteine residue of the mouse anti-CD25 antibody with 89 Zr in the present invention. indicates.

도 4는 본 발명 특유의 89Zr 표지 항-CD25 항체의 결합구조를 개략적으로 도시한다. Figure 4 schematically shows the binding structure of the 89 Zr-labeled anti-CD25 antibody unique to the present invention.

도 5a는 본 발명의 인간 89Zr-CD25 항체에 대하여 자가방사법(autoradiography)으로 분석한 결과를 나타낸다. Figure 5a shows the results of analysis by autoradiography on the human 89 Zr-CD25 antibody of the present invention.

도 5b는 본 발명의 마우스 89Zr-CD25 항체에 대하여 자가방사법(autoradiography)으로 분석한 결과를 나타낸다.Figure 5b shows the results of analysis by autoradiography for the mouse 89 Zr-CD25 antibody of the present invention.

도 6a는 본 발명의 인간 89Zr-CD25 항체 수득 시, size-exclusion chromatography로 분리한 각 분획들에 대하여, 방사성(radioactivity)을 측정한 결과를 나타낸다.Figure 6a shows the results of measuring radioactivity for each fraction separated by size-exclusion chromatography when obtaining the human 89 Zr-CD25 antibody of the present invention.

도 6b는 본 발명의 마우스 89Zr-CD25 항체 수득 시, size-exclusion chromatography로 분리한 각 분획들에 대하여, 방사성(radioactivity)을 측정한 결과를 나타낸다.Figure 6b shows the results of measuring radioactivity for each fraction separated by size-exclusion chromatography when obtaining the mouse 89 Zr-CD25 antibody of the present invention.

도 7a는 본 발명의 인간 89Zr-CD25 항체를 PBS 또는 FBS에서 수일동안 인큐베이션한 후, radio-iTLC로 분석하여 혈청 안정성을 평가한 결과를 나타낸다.Figure 7a shows the results of evaluating the serum stability of the human 89 Zr-CD25 antibody of the present invention after incubation in PBS or FBS for several days and analysis by radio-iTLC.

도 7b는 본 발명의 마우스 89Zr-CD25 항체를 PBS 또는 FBS에서 수일동안 인큐베이션한 후, radio-iTLC로 분석하여 혈청 안정성을 평가한 결과를 나타낸다. Figure 7b shows the results of evaluating the serum stability of the mouse 89 Zr-CD25 antibody of the present invention after incubation in PBS or FBS for several days and analysis by radio-iTLC.

도 8a는 사람 림프종 세포주인 H9, Jurkat 및 SUDHL1과, 마우스 림프종 세포주 EL4에 대하여, 세포 표면에 CD25 항원의 발현 정도를 western blotting으로 확인한 결과를 나타낸다. Figure 8a shows the results of western blotting confirming the expression level of CD25 antigen on the cell surface for human lymphoma cell lines H9, Jurkat, and SUDHL1 and mouse lymphoma cell line EL4.

도 8b는 CD25 발현이 높은 SUDHL1 사람 T cell 림프종 세포와 CD25 발현량이 낮은 사람 H9 및 Jurkat 암세포에서, 본 발명의 인간 89Zr-CD25 IgG의 결합정도를 비교한 결과를 나타낸다.Figure 8b shows the results of comparing the binding degree of human 89 Zr-CD25 IgG of the present invention in SUDHL1 human T cell lymphoma cells with high CD25 expression and human H9 and Jurkat cancer cells with low CD25 expression.

도 8c는 본 발명의 인간 89Zr-CD25 항체의 결합 특이도를, CD25 발현이 높은 SUDHL1 림프종 세포를 이용하여 확인한 결과를 나타낸다(blocking group에는 0.5 μM의 cold unlabeled antibody로 억제되는 높은 결합 특이도를 확인하였다). Figure 8c shows the results of confirming the binding specificity of the human 89 Zr-CD25 antibody of the present invention using SUDHL1 lymphoma cells with high CD25 expression (the blocking group shows high binding specificity inhibited by 0.5 μM of cold unlabeled antibody). confirmed).

도 8d는 CD25 발현이 낮은 EL4 림프종 세포에서, 마우스 89Zr-CD25 IgG의 낮은 결합이, 다량의 cold anti-CD25 IgG에 의해 소량만 억제됨을 확인한 결과를 나타낸다(blocking group에는 0.5 μM의 cold unlabeled antibody로 억제되는 높은 결합 특이도를 확인하였다). Figure 8d shows the results confirming that in EL4 lymphoma cells with low CD25 expression, the low binding of mouse 89 Zr-CD25 IgG was suppressed only to a small extent by a large amount of cold anti-CD25 IgG (0.5 μM of cold unlabeled antibody in the blocking group). High binding specificity inhibited by was confirmed).

도 9a는 FACS를 이용하여 마우스의 흉선으로부터 CD25(-) T 세포와 CD25(+) Treg 세포를 분리정제 하는 과정 중에 수행한 FACS 분석 결과를 나타낸다. Figure 9a shows the results of FACS analysis performed during the process of separating and purifying CD25(-) T cells and CD25(+) Treg cells from the thymus of a mouse using FACS.

도 9b는 CD25(+) Treg 세포를 hematoxylin & eosin 염색하고 광학현미경으로 관찰한 결과를 나타낸다. Figure 9b shows the results of hematoxylin & eosin staining of CD25(+) Treg cells and observation with an optical microscope.

도 10a는 마우스의 흉선으로부터 분리된 CD25(-) T 세포 및 CD25(+) Treg 세포에 대하여, 본 발명의 89Zr-CD25 IgG 결합율 및 특이도를 비교적으로 분석한 결과를 나타낸다(blocking group에는 0.5 μM의 cold unlabeled antibody로 억제되는 높은 결합 특이도를 확인하였다).Figure 10a shows the results of comparative analysis of the binding rate and specificity of 89 Zr-CD25 IgG of the present invention for CD25(-) T cells and CD25(+) Treg cells isolated from the thymus of mice (blocking group includes High binding specificity inhibited by 0.5 μM cold unlabeled antibody was confirmed).

도 10b는 마우스의 비장으로부터 분리한 CD25(+) Treg 세포에 대하여, 본 발명의 89Zr-CD25 IgG 결합율 및 특이도를 확인한 결과를 나타낸다(blocking group에는 0.5 μM의 cold unlabeled antibody로 억제되는 높은 결합 특이도를 확인하였다).Figure 10b shows the results of confirming the binding rate and specificity of 89 Zr-CD25 IgG of the present invention for CD25(+) Treg cells isolated from the spleen of mice (in the blocking group, there was a high level of antibody inhibited by 0.5 μM of cold unlabeled antibody). Binding specificity was confirmed).

도 11은 in vivo 마우스 림프종 모델에서, 본 발명의 89Zr-CD25 IgG의 투여 후 5일 째에, 체내 분포 및 림프종에서의 섭취 비율을 확인한 결과를 나타낸다(blocking group에는 0.8 mg의 cold unlabeled antibody로 억제되는 높은 결합 특이도를 확인하였다).Figure 11 shows the results of confirming the body distribution and uptake rate in lymphoma 5 days after administration of 89 Zr-CD25 IgG of the present invention in an in vivo mouse lymphoma model (blocking group was treated with 0.8 mg of cold unlabeled antibody) High binding specificity of inhibition was confirmed).

도 12는 in vivo 마우스 림프종 모델에서, 본 발명의 89Zr-CD25 IgG의 투여 후 5일 째에 PET 영상을 촬영한 결과를 나타내는 이미지로, T cell lymphoma의 높은 89Zr-CD25 IgG 섭취를 보여준다. Figure 12 is an image showing the results of PET imaging taken 5 days after administration of 89 Zr-CD25 IgG of the present invention in an in vivo mouse lymphoma model, showing high uptake of 89 Zr-CD25 IgG by T cell lymphoma.

도 13은 in vivo 마우스 림프종 모델(blocking group)에서, 0.8 mg의 cold unlabeled Ab를 투여하고 1시간 후에 본 발명의 89Zr-CD25 IgG의 투여 5일 째에 PET 영상을 촬영한 결과를 나타내는 이미지이다. Cold unlabeled antibody(blocking) 처리에 의하여 대조도가 줄어드는 소견을 보여, 이로써 본 발명 89Zr-CD25 IgG의 높은 결합 특이도를 확인한 결과를 나타낸다.Figure 13 is an image showing the results of PET imaging on the 5th day of administration of 89 Zr-CD25 IgG of the present invention, 1 hour after administration of 0.8 mg of cold unlabeled Ab, in an in vivo mouse lymphoma model (blocking group). . The contrast was found to be reduced by cold unlabeled antibody (blocking) treatment, confirming the high binding specificity of the 89 Zr-CD25 IgG of the present invention.

본 발명은 난치성 림프종 표면에 다량 발현하여 치료용 항체 의약품의 표적으로 기대를 모으고 있는 CD25 항원을 선택적으로 표적하는, 89Zr 표지 CD25 IgG를 개발하고 이를 이용한 새로운 immuno-PET 기술 개발에 성공하였으며, 이는 본 발명에서 최초로 공개하는 것이다. The present invention developed 89 Zr-labeled CD25 IgG, which selectively targets the CD25 antigen, which is expressed in large quantities on the surface of refractory lymphoma and is expected to be a target of therapeutic antibody drugs, and succeeded in developing a new immuno-PET technology using it. This is disclosed for the first time in the present invention.

본 발명은 89Zr 표지된 항-CD25 항체 및 9Zr 표지된 항-CD25 항체를 유효성분으로 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 조성물을 제공한다.The present invention provides a composition for positron emission tomography imaging for tumor diagnosis, comprising 89 Zr labeled anti-CD25 antibody and 9 Zr labeled anti-CD25 antibody as active ingredients.

본 발명에서, CD25는 heterodimeric interleukin 2 receptor(IL2 수용체)의 alpha subunit에 해당하며(즉, IL2RA (Interleukin-2 receptor alpha chain)), 림프종 세포의 생존과 증식에 중요한 역할을 한다. 따라서 CD25를 표적하는 immuno-PET 기술은 몸속 림프종 병소를 검출하고 CD25 발현을 비침습적이면서 정량적으로 평가하는데 이용할 수 있다. 뿐만 아니라 항-CD25 항체 기반 의약품의 림프종 전달을 평가하는 약리 동태 정보를 제공하여 치료 대상을 선정하고 치료효과를 예측하는데 이용 가능하다. In the present invention, CD25 corresponds to the alpha subunit of the heterodimeric interleukin 2 receptor (IL2 receptor) (i.e., IL2RA (Interleukin-2 receptor alpha chain)) and plays an important role in the survival and proliferation of lymphoma cells. Therefore, immuno-PET technology targeting CD25 can be used to detect lymphoma foci in the body and non-invasively and quantitatively evaluate CD25 expression. In addition, it provides pharmacokinetic information to evaluate the delivery of anti-CD25 antibody-based medicines to lymphoma, which can be used to select treatment targets and predict treatment effects.

CD25는 정상세포에서는 거의 발현하지 않거나, 소량만 발현하지만 예외적으로 활성화된 정상 regulatory T cell(Treg cell)에만 풍부하게 발현하는데, 이 면역세포는 종양 안으로 침투하여 정상 cytotoxic T cell의 면역반응을 억제하여 immune evasion을 통한 치료 저항에 기여한다. 따라서 CD25를 표적하는 immuno-PET 기술은 이를 발현하는 림프종의 영상뿐 만 아니라 치료에 방해가 되는 종양 내 침윤한 Treg cell의 영상에도 이용할 수 있다. 이 경우 항체의 체내 약리 동태를 평가하는 immuno-PET에는 89Zr 방사선동위원소가 높은 체내 안정성, 3.3일의 물리적 반감기 등의 장점으로 최적화되었다. CD25 is rarely or only expressed in small amounts in normal cells, but is expressed abundantly only in activated normal regulatory T cells (Treg cells). These immune cells infiltrate the tumor and suppress the immune response of normal cytotoxic T cells. Contributes to treatment resistance through immune evasion. Therefore, immuno-PET technology targeting CD25 can be used not only for imaging lymphomas that express CD25, but also for imaging Treg cells infiltrated within tumors that interfere with treatment. In this case, the 89 Zr radioisotope was optimized for immuno-PET, which evaluates the in vivo pharmacokinetics of antibodies, due to its advantages such as high in vivo stability and physical half-life of 3.3 days.

상기와 같은 이유로 89Zr 표지 항-CD25 항체와 이를 이용한 immuno-PET 영상기술은 난치성 림프종 및 Treg cell의 체내 이미징을 통해 암진단과 항-CD25 항체 치료제의 효과 예측, 그리고 177Lu 표지 항-CD25 항체 방사면역치료 전략의 성공을 위해 중요하다. 특히 항체-약물 결합체(antibody-drug conjugate)로 치료용 베타선 방출 177Lu를 탑재한 항체 의약품의 경우 림프종에 전달되는 효율 평가하기 위해서는 감마선 방출 89Zr로 표지한 항체를 이용한 immuno-PET이 최적이다. For the above reasons, the 89 Zr-labeled anti-CD25 antibody and the immuno-PET imaging technology using it are used to diagnose cancer and predict the effectiveness of anti-CD25 antibody treatment through in-vivo imaging of refractory lymphoma and Treg cells, and to use the 177 Lu-labeled anti-CD25 antibody. It is important for the success of radioimmunotherapy strategies. In particular, in the case of antibody drugs loaded with therapeutic beta ray emitting 177 Lu as an antibody-drug conjugate, immuno-PET using an antibody labeled with gamma ray emitting 89 Zr is optimal to evaluate the efficiency of delivery to lymphoma.

본 발명에서 상기 '항-CD25 항체' 또는 'CD25에 대한 항체'는 CD25의 항원성 부위에 대해서 지시되는 특이적인 단백질 분자를 의미한다. 본 발명의 목적상, 상기 항체는 CD25 단백질(세포 표면 항원)에 대해 특이적으로 결합하는 항체를 의미하며, 다클론 항체, 단일클론 항체 및 재조합 항체를 모두 포함한다. 본 발명의 목적상 항체의 중쇄와 경쇄의 아미노산 서열이 실질적으로 동일한 항체의 집단인 단일클론 항체인 것이 바람직할 수 있다.In the present invention, the 'anti-CD25 antibody' or 'antibody against CD25' refers to a specific protein molecule directed against the antigenic site of CD25. For the purposes of the present invention, the antibody refers to an antibody that specifically binds to the CD25 protein (cell surface antigen), and includes polyclonal antibodies, monoclonal antibodies, and recombinant antibodies. For the purposes of the present invention, it may be preferable for the antibody to be a monoclonal antibody, which is a group of antibodies in which the amino acid sequences of the heavy and light chains of the antibody are substantially identical.

상기한 바와 같은 CD25에 대한 항체를 생성하는 것은 당업계에 널리 공지된 기술을 이용하여 용이하게 제조할 수 있다. 다클론 항체는 상기 CD25 단백질 항원을 동물에 주사하고 동물로부터 채혈하여 항체를 포함하는 혈청을 수득하는 당업계에 널리 공지된 방법에 의해 생산할 수 있다. 이러한 다클론 항체는 염소, 토끼, 양, 원숭이, 말, 돼지, 소 개 등의 임의의 동물 종 숙주로부터 제조 가능하다.Generating antibodies against CD25 as described above can be easily produced using techniques well known in the art. Polyclonal antibodies can be produced by a method well known in the art, which involves injecting the CD25 protein antigen into an animal and collecting blood from the animal to obtain serum containing the antibody. These polyclonal antibodies can be prepared from any animal species host, such as goats, rabbits, sheep, monkeys, horses, pigs, cows and dogs.

단일클론 항체는 당업계에 널리 공지된 하이브리도마 방법(hybridoma method), 또는 파지 항체 라이브러리 기술을 이용하여 제조될 수 있다.Monoclonal antibodies can be produced using the hybridoma method or phage antibody library technology well known in the art.

또한 본 발명에서 항체는 2개의 전체 길이의 경쇄 및 2개의 전체 길이의 중쇄를 가지는 완전한 형태뿐 만 아니라, 항체 분자의 기능적인 단편을 포함한다. 항체 분자의 기능적인 단편이란 적어도 항원 결합 기능을 보유하고 있는 단편을 뜻하며, 바람직하게 상기 단편은 모(母)항체의 LRRC15 결합 친화도의 적어도 50%, 60%. 70%, 80%, 90%, 95% 또는 100% 또는 그 이상을 보유한다. 구체적으로 본 발명에서 상기 항체의 기능적 단편은 힌지 영역을 포함하는 것이 바람직할 수 있으며, 이에 제한되지 않으나 일례로 F(ab')2, F(ab') 등의 형태일 수 있다. F(ab')2는 항체를 펩신으로 가수분해시켜서 생성되는 단편으로, 두 개의 Fab가 중쇄 경첩(hinge)에서 이황결합(disulfide bond)으로 연결된 형태를 하고 있다. F(ab')는 F(ab')2 단편의 이황결합을 환원하여 분리시킨 Fab에 중쇄 경첩이 부가된 형태의 단량체 항체 단편이다. Antibodies in the present invention also include functional fragments of the antibody molecule as well as intact forms having two full-length light chains and two full-length heavy chains. A functional fragment of an antibody molecule refers to a fragment that retains at least an antigen-binding function, and preferably, the fragment has at least 50% or 60% of the LRRC15 binding affinity of the parent antibody. Hold 70%, 80%, 90%, 95% or 100% or more. Specifically, in the present invention, the functional fragment of the antibody may preferably include a hinge region, but is not limited thereto, and may be in the form of F(ab')2, F(ab'), etc., for example. F(ab')2 is a fragment produced by hydrolyzing an antibody with pepsin, and consists of two Fabs connected by a disulfide bond at the heavy chain hinge. F(ab') is a monomeric antibody fragment in which a heavy chain hinge is added to Fab obtained by reducing the disulfur bond of the F(ab')2 fragment.

본 발명에 적용되는 항체는 이에 제한되지 않으나 일례로 IgG, IgA, IgM, IgE 및 IgD로 이루어진 군에서 선택되는 것일 수 있으며, 바람직하게는 IgG 항체일 수 있다. 본 발명의 일 실시 양태에서, 본 발명의 항체는 IgG1, IgG2, IgG3 및 IgG4로 이루어진 군에서 선택되는 것일 수 있으며, 바람직하게는 IgG1 또는 IgG2 일 수 있다. 본 발명의 구체적 실시 양태에서, 상기 본발명의 항체는 가장 바람직하게는 IgG2a일 수 있다. The antibody applied in the present invention is not limited thereto, but may be, for example, selected from the group consisting of IgG, IgA, IgM, IgE, and IgD, and is preferably an IgG antibody. In one embodiment of the present invention, the antibody of the present invention may be selected from the group consisting of IgG1, IgG2, IgG3, and IgG4, and preferably may be IgG1 or IgG2. In a specific embodiment of the present invention, the antibody of the present invention may most preferably be IgG2a.

상기 89Zr(지르코늄)은 양전자 방출이 가능한 의료용 방사성동위원소로서 알려져 있으며, 물리적 반감기는 78.41 시간이고, 붕괴 시 전자포획(76.6%) 및 양전자 방출(22.3%)이 자발적으로 일어난다.The 89 Zr (zirconium) is known as a medical radioisotope capable of emitting positrons. Its physical half-life is 78.41 hours, and upon decay, electron capture (76.6%) and positron emission (22.3%) occur spontaneously.

본 발명의 일 실시예에 있어서, 상기 조성물은 종양 또는 종양 내 침윤 조절 T 세포(tumor-infiltrating regulatory T cell)를 검출할 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the composition can detect tumors or tumor-infiltrating regulatory T cells, but is not limited thereto.

본 발명에서, Treg 세포는 조직 내에서 이동할 수 있고, 종양 내에 침윤되는(infiltration) 경우에 면역감시 회피(immune evasion)를 통해 기존 종양치료제들에 대한 내성에 영향을 주는 특성을 가진다. 또한, 종양, 예를 들어, 림프종 치료에 따른 면역반응을 억제하는 것으로 보고되어 있고, 이 때, CD25가 풍부하게 발현되는 것을 특징으로 한다. 본 발명에서 상기 조절 T 세포 (regulatory T cell, Treg)는 바람직하게 CD25를 세포 표면에 풍부하게 발현하고 있는 것(CD25 양성 (+) Treg 세포)일 수 있다. 본 발명의 항체는 Treg 세포에 대한 결합 친화도 및 결합 특이도가 현저히 우수하므로 89Zr-CD25 항체 PET 영상 촬영은 치료 내성에 대한 Treg 세포의 역할 규명에도 활용될 수 있다. In the present invention, Treg cells can move within tissues and have the characteristic of influencing resistance to existing tumor treatments through immune evasion when infiltrating into a tumor. In addition, it has been reported to suppress the immune response following the treatment of tumors, for example, lymphoma, and is characterized by abundant expression of CD25. In the present invention, the regulatory T cells (Treg) may preferably be those that abundantly express CD25 on the cell surface (CD25 positive (+) Treg cells). Since the antibody of the present invention has significantly excellent binding affinity and binding specificity for Treg cells, 89 Zr-CD25 antibody PET imaging can also be used to identify the role of Treg cells in treatment resistance.

본 발명의 일 실시예에 있어서, 상기 종양은 림프종일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the tumor may be lymphoma, but is not limited thereto.

본 발명에서 상기 종양은 당업계에 통상적으로 알려진 의미의 암(cancer)을 포함하는 의미로 종양 조직 또는 종양 세포를 모두 아우르는 의미이며, 본 발명에서 CD25를 세포 표면 항원으로서 풍부하게 발현하고 있는 종양이라면 그 종류가 특별히 제한되지 않는다. In the present invention, the tumor includes cancer in the sense commonly known in the art and includes all tumor tissue or tumor cells. In the present invention, if the tumor abundantly expresses CD25 as a cell surface antigen, The type is not particularly limited.

본 발명에서, 상기 림프종은 비-호지킨 림프종(NHL), 1차, 2차, 재발성 림프종, 지연성 림프종, 공격성 비-호지킨 림프종(NHL), 여포성 림프종(FL), 만성 림프구성 백혈병(CLL), 변연부 림프종(MZL), 미만성 거대 B-세포 림프종(DLBCL), 외투 세포 림프종(MCL), 변형성 림프종(TL), 말초 T-세포 림프종 (PTCL)일 수 있고, 바람직하게는 난치성 림프종, T세포 림프종일 수 있으나, 이에 제한되는 것은 아니다. In the present invention, the lymphoma includes non-Hodgkin's lymphoma (NHL), primary, secondary, recurrent lymphoma, indolent lymphoma, aggressive non-Hodgkin's lymphoma (NHL), follicular lymphoma (FL), and chronic lymphocytic lymphoma. It may be leukemia (CLL), marginal zone lymphoma (MZL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), transforming lymphoma (TL), peripheral T-cell lymphoma (PTCL), and is preferably refractory. It may be lymphoma or T-cell lymphoma, but is not limited thereto.

본 발명의 89Zr-CD25 항체 및 이를 이용한 의료 촬영 방법은 상술된 종양 및 종양 내 침윤 Treg 세포에 대한 결합 친화도 및 결합 특이성으로 인하여 난치성 림프종의 치료와 재발 억제를 위한 177Lu-CD25 항체 기반 방사면역치료 기술의 개발을 촉진시키는 역할을 한다. 또한, 항-CD25 항체의 89Zr 표지 기술 정립은 177Lu 표기 기술에도 활용되어 치료용 177Lu-CD25 항체 합성에 기여할 수 있다. The 89 Zr-CD25 antibody of the present invention and the medical imaging method using the same are 177 Lu-CD25 antibody-based radiation for the treatment of refractory lymphoma and inhibition of recurrence due to the binding affinity and binding specificity for the above-mentioned tumor and intra-tumor infiltrating Treg cells. It plays a role in promoting the development of immunotherapy technology. In addition, the establishment of 89 Zr labeling technology for anti-CD25 antibodies can also be used in 177 Lu labeling technology, contributing to the synthesis of therapeutic 177 Lu-CD25 antibodies.

본 발명의 상기 조성물은 의료 영상 촬영에서 사용되는 조영제의 기능을 포함할 수 있다. 일반적으로 조영제(contrast enhancement)란 영상의학과에서 실시하는 상당수의 검사에서 조영제라고 불리는 특수한 약물을 인체에 주입하여 병소나 혈관을 더 잘보이게 하는 것을 의미할 수 있다. CT와 뇌혈관조영술, 관상동맥조영술 등 X선 기반 검사에서는 혈관을 보려면 조영제가 필수지만, 2000년대 이후 MRI에서는 조영제 없이도 혈관을 볼 수 있다. 조영제 없이 MRI로 혈관을 보는 경우는 time of flight라는 방식이고 조영제 투여시에는 contrast enhanced 방식이 될 수 있다. PET나 SPECT 검사시에 방사성 물질이나 혈관확장제를 투여하지만 이들을 조영제라고 하지는 않으나, 당업계에서 일반적으로 조영제의 의미에 따라 본 발명에 따른 조성물은 조영제를 포함하는 의미일 수 있다.The composition of the present invention may contain the function of a contrast agent used in medical imaging. In general, contrast enhancement may mean that a special drug called a contrast agent is injected into the human body to make lesions or blood vessels more visible in many tests performed in radiology departments. In X-ray-based tests such as CT, cerebral angiography, and coronary angiography, contrast agent is required to view blood vessels, but since the 2000s, MRI has been able to view blood vessels without contrast agent. When viewing blood vessels with MRI without a contrast agent, a time of flight method is used, and when contrast agent is administered, a contrast enhanced method is used. Radioactive substances or vasodilators are administered during PET or SPECT examinations, but these are not called contrast agents. However, depending on the general meaning of contrast agents in the art, the composition according to the present invention may include a contrast agent.

본 발명의 일 실시예에 있어서, 상기 조성물은 정맥주사용일 수 있으나, 이에 제한되지 않는다. 당업계에 알려진 조영제 투여 경로라면 그 구체적 투여 경로가 특별히 제한되지 않는다. In one embodiment of the present invention, the composition may be for intravenous injection, but is not limited thereto. As long as it is a contrast agent administration route known in the art, the specific administration route is not particularly limited.

본 발명의 조성물은 종양 또는 종양 내 침윤 조절 T 세포의 이미징(imaging)용 또는 모니터링(monitoring)용인 것을 특징으로 하는 것일 수 있으나, 이에 제한되지 않는다. 따라서, 본 발명은 상기 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는, 종양의 진단; 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화(imaging)용 조성물을 제공한다. The composition of the present invention may be used for imaging or monitoring tumors or intra-tumor infiltrating regulatory T cells, but is not limited thereto. Therefore, the present invention provides a method for diagnosing tumors, comprising the 89 Zr labeled anti-CD25 antibody as an active ingredient; A composition for detecting or imaging a tumor or infiltrating regulatory T cells within a tumor is provided.

본 발명은 하기 단계를 포함하는 상기 89Zr 표지된 항-CD25 항체를 제조하는 방법을 제공한다:The present invention provides a method of preparing the 89 Zr labeled anti-CD25 antibody comprising the following steps:

(a) 항-CD25 항체를 TCEP(tris-carboxyethylphosphine)와 반응시켜 환원 시키는 단계; (a) reducing the anti-CD25 antibody by reacting it with TCEP (tris-carboxyethylphosphine);

(b) 상기 (a) 단계에서 환원된 항-CD25 항체를 데페록사민-말레이미드(deferoxamine-maleimide, DFO-Mal)와 반응시켜, DFO-Mal 접합된 항-CD25 항체를 생성하는 단계; 및(b) reacting the anti-CD25 antibody reduced in step (a) with deferoxamine-maleimide (DFO-Mal) to produce a DFO-Mal conjugated anti-CD25 antibody; and

(c) 상기 (b) 단계에서 생성된 DFO-Mal 접합된 항-CD25 항체를 89Zr-옥살레이트(89Zr-oxalate)와 반응시킨 후, 89Zr 표지된 항-CD25 항체를 수득하는 단계.(c) reacting the DFO-Mal conjugated anti-CD25 antibody produced in step (b) with 89 Zr-oxalate to obtain an 89 Zr labeled anti-CD25 antibody.

상기 (a) 단계의 반응은 환원반응으로, 상기 반응에 의하여 항체 힌지 영역의 이황화 결합 쇄(chain)가 선택적으로 환원되어 상기 결합이 끊어진다. 그 결과, 이에 제한되지 않으나, 바람직하게 한쌍의 티올기(pair of thiols (reduced disulfide)가 생성된다. 상기 (a) 단계에서, 항-CD25 항체와 TCEP(tris-carboxyethylphosphine)는 1: 70 내지 130의 몰농도 비율로 혼합되는 것이 특징이다. 본 발명에서 목적하는 항체 표지 구조를 위하여, 상기 (a) 단계에서는 힌지 영역 이외에 항체 내 다른 이중황화결합을 끊지 않는 섬세한 반응 조건 및 이의 조절이 매우 중요하다. 본 발명의 바람직한 일 실시 양태에서, 상기 (a) 단계에서, 항-CD25 항체와 TCEP(tris-carboxyethylphosphine)는 1: 100의 몰농도 비율로 혼합될 수 있다. The reaction in step (a) is a reduction reaction, whereby the disulfide bond chain of the antibody hinge region is selectively reduced and the bond is broken. As a result, but not limited thereto, a pair of thiols (reduced disulfide) is preferably generated. In step (a), the anti-CD25 antibody and TCEP (tris-carboxyethylphosphine) are 1: 70 to 130. It is characterized by mixing at a molar concentration ratio of For the purpose of the antibody label structure of the present invention, in step (a), delicate reaction conditions and control that do not break other double sulfide bonds in the antibody other than the hinge region are very important. In a preferred embodiment of the present invention, in step (a), the anti-CD25 antibody and TCEP (tris-carboxyethylphosphine) may be mixed at a molar concentration ratio of 1:100.

본 발명의 일 실시예에 있어서, 상기 (a) 단계의 반응은 15 내지 25℃ 온도로 수행되는 것일 수 있으나, 이에 제한되지 않는다. 본 발명의 일 실시예에 있어서, 바람직하게 상기 (a)단계의 반응은 21 내지 23℃ 온도로 수행되는 것일 수 있다. 상기 (a) 단계 반응의 시간은 당업자가 목적하는 반응 산물의 수율에 따라 당업자가 적절하게 최적의 시간을 선택 적용할 수 있는 것으로, 특별히 제한되지 않으나, 본 발명에서 바람직한 일례로 5 내지 30분 동안 반응시키는 것을 특징으로 할 수 있으며, 더욱 바람직하게는 10 내지 20분 동안 반응시키는 것일 수 있다. 상기 (a) 단계에서는 힌지 영역 이외에 항체 내 다른 이중황화결합을 끊지 않는 섬세한 반응 조건 및 이의 조절이 매우 중요하다.In one embodiment of the present invention, the reaction in step (a) may be performed at a temperature of 15 to 25° C., but is not limited thereto. In one embodiment of the present invention, the reaction in step (a) may be preferably performed at a temperature of 21 to 23°C. The time for the reaction in step (a) is not particularly limited, and a person skilled in the art can appropriately select and apply the optimal time depending on the yield of the desired reaction product, but a preferred example in the present invention is 5 to 30 minutes. It may be characterized by reacting, and more preferably, reacting for 10 to 20 minutes. In step (a), delicate reaction conditions and control that do not break other double sulfide bonds in the antibody other than the hinge region are very important.

상기 (b) 단계의 반응은 바람직하게 킬레이트화 반응으로, 상기 반응에 의하여 DFO-Mal 접합된(DFO-conjugated) 항-CD25 항체가 생성된다. 이에 제한되지 않으나, 상기 (b) 단계의 반응은 바람직하게 EDTA 존재 하에 수행되는 것일 수 있다. 본 발명의 일 실시예에 있어서, 상기 (b) 단계의 반응은 15 내지 25℃ 온도로 수행되는 것일 수 있으나, 이에 제한되지 않는다. 본 발명의 일 실시예에 있어서, 바람직하게 상기 (b)단계의 반응은 21 내지 23℃ 온도로 수행되는 것일 수 있다. 상기 (b) 단계에서, 각 물질(상기 (a) 단계에서 수득된 환원된 항-CD25 항체 및 데페록사민- 말레이미드(deferoxamine-maleimide, DFO-Mal)의 혼합 비율은 당업자가 목적하는 반응 산물 수율 및 수득 속도에 따라 당업자가 적절하게 최적의 조건을 선택 적용할 수 있다. 또한 상기 (b) 단계 반응의 시간은 당업자가 목적하는 반응 산물의 수율에 따라 당업자가 적절하게 최적의 시간을 선택 적용할 수 있는 것으로, 특별히 제한되지 않으나, 바람직한 일례로 30 내지 120분 동안 반응시키는 것일 수 있다. The reaction in step (b) is preferably a chelation reaction, thereby producing a DFO-Mal conjugated anti-CD25 antibody. Although not limited thereto, the reaction in step (b) may be preferably carried out in the presence of EDTA. In one embodiment of the present invention, the reaction in step (b) may be performed at a temperature of 15 to 25° C., but is not limited thereto. In one embodiment of the present invention, the reaction in step (b) may be preferably performed at a temperature of 21 to 23°C. In step (b), the mixing ratio of each material (reduced anti-CD25 antibody and deferoxamine-maleimide (DFO-Mal) obtained in step (a)) is determined by those skilled in the art to obtain the desired reaction product. A person skilled in the art can appropriately select and apply optimal conditions depending on the yield and speed of acquisition.In addition, a person skilled in the art can appropriately select and apply the optimal time for the reaction in step (b) according to the yield of the desired reaction product. What can be done is not particularly limited, but a preferred example may be reacting for 30 to 120 minutes.

상기 (c) 단계는 PET(양전자방출단층촬영) 및/또는 PET와 함께 사용될 수 있는 의료 촬영 기법에 사용될 수 있는 추적물질로서 89Zr을 표지하기 위한 단계로, 상기 (b) 단계의 반응은 바람직하게 킬레이트화 반응일 수 있다. 상기 반응에 의하여, 최종적으로 본 발명의 89Zr 표지된 항-CD25 항체가 수득된다. Step (c) is a step for labeling 89 Zr as a tracer that can be used in PET (positron emission tomography) and/or medical imaging techniques that can be used in conjunction with PET, and the reaction in step (b) is preferred. It may be a chelation reaction. Through the above reaction, the 89 Zr labeled anti-CD25 antibody of the present invention is finally obtained.

본 발명의 일 실시예에 있어서, 상기 (c) 단계의 반응은 15 내지 25℃ 온도로 수행되는 것일 수 있으나, 이에 제한되지 않는다. 본 발명의 일 실시예에 있어서, 바람직하게 상기 (c)단계의 반응은 21 내지 23℃ 온도로 수행되는 것일 수 있다. 상기 (c) 단계에서, 각 물질(89Zr-옥살레이트 및 상기 (b) 단계에서 생성된 DFO-Mal 접합된 항-CD25 항체)의 혼합 비율은 당업자가 목적하는 반응 산물 수율 및 수득 속도에 따라 당업자가 적절하게 최적의 조건을 선택 적용할 수 있다. 또한 상기 (c) 단계 반응의 시간은 당업자가 목적하는 반응 산물의 수율에 따라 당업자가 적절하게 최적의 시간을 선택 적용할 수 있는 것으로, 특별히 제한되지 않으나, 바람직한 일례로 30 내지 120분 동안 반응시키는 것일 수 있다. In one embodiment of the present invention, the reaction in step (c) may be performed at a temperature of 15 to 25° C., but is not limited thereto. In one embodiment of the present invention, the reaction in step (c) may be preferably performed at a temperature of 21 to 23°C. In step (c), the mixing ratio of each material ( 89 Zr-oxalate and DFO-Mal conjugated anti-CD25 antibody produced in step (b)) is adjusted according to the desired reaction product yield and yield rate by those skilled in the art. A person skilled in the art can appropriately select and apply optimal conditions. In addition, the time for the reaction in step (c) is not particularly limited, and a person skilled in the art can appropriately select the optimal time depending on the yield of the desired reaction product, but a preferred example is reaction for 30 to 120 minutes. It could be.

상기 (c) 단계에서, 89Zr-옥살레이트(89Zr-oxalate)는, 상기 (b) 단계에서 생성된 DFO-Mal 접합된 항-CD25 항체와 혼합되기 전에, 중화(neutralization) 처리될 수 있다. 상기 중화(중화 반응)은, 당업계에 공지된 산 및 염기 중화 반응을 이용하는 것이라면 이에 사용되는 물질(특히, 염기성 물질)의 종류가 특별히 제한되지 않으나, 일례로 탄산나트륨(Na2CO3)과 혼합되어 중화되는 것일 수 있다. 상기 중화반응에서, 각 물질의 혼합 비율, 반응 온도, 반응 시간 등의 반응 조건은 당업자가 목적하는 반응 산물 수율 및 수득 속도에 따라 당업자가 적절하게 최적의 조건을 선택 적용할 수 있다. 89Zr-옥살레이트 중화 후, DFO-Mal 접합된 항-CD25 항체를 혼합하여, 89Zr과의 킬레이트화 반응을 수행한다.In step (c), 89 Zr-oxalate may be neutralized before being mixed with the DFO-Mal conjugated anti-CD25 antibody produced in step (b). . The neutralization (neutralization reaction) is not particularly limited in the type of material (particularly basic material) used as long as it uses an acid and base neutralization reaction known in the art, but for example, it can be mixed with sodium carbonate (Na 2 CO 3 ). It may be neutralized. In the neutralization reaction, a person skilled in the art can appropriately select and apply optimal conditions for reaction conditions such as the mixing ratio of each substance, reaction temperature, and reaction time, depending on the desired reaction product yield and acquisition rate. After neutralizing 89 Zr-oxalate, DFO-Mal conjugated anti-CD25 antibody is mixed to perform a chelation reaction with 89 Zr.

본 발명의 일 실시예에 있어서, 상기 항체는 IgG일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the antibody may be IgG, but is not limited thereto.

본 발명의 일 실시예에 있어서, 상기 항체는 하기를 특징으로 할 수 있으나, 이에 제한되는 것은 아니다:In one embodiment of the present invention, the antibody may have the following characteristics, but is not limited thereto:

항-CD25 항체 힌지 영역(hinge region)의 이황화 결합(disulfide bond) 부위에 특이적으로 89Zr이 결합됨; 및 89 Zr is specifically bound to the disulfide bond site of the anti-CD25 antibody hinge region; and

89Zr 및 항-CD25 항체의 접합체(conjugate) 형태임.It is a conjugate form of 89 Zr and anti-CD25 antibody.

본 명세서에서 용어 '접합체'는, 결합체 또는 복합체의 의미를 모두 포함하는 것으로, 본 명세서에서 상기 용어들을 혼용하여 지칭될 수 있다. As used herein, the term 'conjugate' includes both conjugates and complexes, and the above terms may be used interchangeably in this specification.

본 발명의 상기 항체는, 바람직하게, 힌지 영역의 이황화 결합을 끊고 89Zr 2개 원자가 결합하는 것을 특징으로 하는 것일 수 있다. 가장 바람직하게, 본 발명의 상기 항체는, 힌지 영역의 이황화 결합 1개를 끊고 89Zr 2개 원자가 결합하는 것을 특징으로 하는 것일 수 있으며(도 4 참조), 이러한 본 발명 특유의 구조를 보유함을 통해 본 발명의 표지 항체는 생체 내에서 높은 구조 안정성을 유지하면서도, 타겟(target)하는 조직 또는 세포에 대해 표지 효율이 높은 것이 특징이다. The antibody of the present invention may preferably be characterized in that the disulfide bond in the hinge region is broken and two 89 Zr atoms are bonded. Most preferably, the antibody of the present invention may be characterized in that one disulfide bond in the hinge region is broken and two 89 Zr atoms are bonded to it (see Figure 4), and it has a structure unique to the present invention. The labeled antibody of the present invention is characterized by high labeling efficiency for target tissues or cells while maintaining high structural stability in vivo.

본 발명은 하기 단계를 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 방법을 제공한다:The present invention provides a positron emission tomography method for tumor diagnosis, comprising the following steps:

(i) 89Zr 표지된 항-CD25 항체를 개체에 투여하는 단계; 및(i) administering 89 Zr labeled anti-CD25 antibody to the subject; and

(ii) 상기 개체의 검출 이미지 또는 영상을 획득하는 단계.(ii) Obtaining a detection image or video of the object.

본 발명에서, 양전자 방출 단층 촬영(Positron Emission Tomography)은 PET로 약칭되는 핵의학과에서 사용하는 기능적 영상진단 기술 중 하나로, 생체 내 특정 대사 작용이 활발한 정도의 3차원 분포를 영상화한다. 당업계에서는 PET를 단독으로 사용할 경우에는 연구목적인 경우가 대부분이며, 일반적으로 해부학적 진단 방법인 CT(Computed Tomography, 컴퓨터 단층 촬영) 또는 MRI(Magnetic Resonance Imaging, 자기 공명 영상)와 동시에 수행되어 PET/CT, PET/MR(PET/MRI)로 사용될 수 있으며, 면역-PET(immuno-PET)의 방법도 사용될 수 있는 것으로 알려져 있다. In the present invention, positron emission tomography (Positron Emission Tomography), abbreviated as PET, is one of the functional imaging techniques used in nuclear medicine, and images the three-dimensional distribution of the degree to which specific metabolic activity is active in the living body. In the industry, when PET is used alone, it is mostly for research purposes, and is generally performed simultaneously with CT (Computed Tomography) or MRI (Magnetic Resonance Imaging), which are anatomical diagnostic methods. It can be used as CT, PET/MRI (PET/MRI), and it is known that immuno-PET (immuno-PET) method can also be used.

본 발명의 일 실시예에 있어서, 상기 촬영은 PET(Positron Emission Tomography), PET/CT, PET/MR, 및 immuno-PET로 이루어진 군으로부터 선택되는 하나의 기술을 이용할 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the present invention, the imaging may use a technology selected from the group consisting of PET (Positron Emission Tomography), PET/CT, PET/MR, and immuno-PET, but is not limited thereto. .

본 발명은 상기 조성물, 및 설명서를 포함하는 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 키트를 제공한다.The present invention provides a kit for positron emission tomography imaging for tumor diagnosis, including the composition and instructions.

본 발명의 일 실시예에 있어서, 상기 조성물은 본 발명의 조성물이고, 상기 설명서는 상기 방법이 기재된 것일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment of the invention, the composition is a composition of the invention, and the instructions describe the method as It may be described, but is not limited thereto.

본 발명의 “키트”는 상기 구성 이외에도 양전자 방출 단층 촬영 및/또는 상기 촬영의 영상화를 위한 방법에 통상적으로 필요한 다른 구성 성분, 장치, 물질 등이 포함될 수 있다. 또한, 키트에 포함된 모든 구성은 1회 이상 횟수에 제한 없이 사용할 수 있으며, 각 물질을 사용하는 선후에는 제한이 없고, 각 물질의 적용은 동시에 진행될 수도 있고 미시에 진행될 수도 있다.In addition to the above components, the “kit” of the present invention may include other components, devices, materials, etc. commonly required for positron emission tomography and/or methods for imaging such imaging. In addition, all components included in the kit can be used one or more times without limitation, there is no restriction on the order or subsequent use of each substance, and the application of each substance may be carried out simultaneously or in small steps.

본 발명의 키트는 상기 조성물, 및 상기 설명서 이외에도 컨테이너를 포함할 수 있다. 상기 컨테이너는 상기 구성을 포장하는 역할을 할 수 있고, 보관 및 고정하는 역할을 할 수도 있다. 상기 컨테이너의 재질은 예컨대, 병, 통(tub), 작은 봉지 (sachet), 봉투(envelope), 튜브, 앰플(ampoule) 등과 같은 형태를 취할 수 있고, 이들은 부분적 또는 전체적으로 플라스틱, 유리, 종이, 호일, 왁스 등으로부터 형성될 수 있다. 상기 용기는 처음에는 용기의 일부이거나 또는 기계적, 접착성, 또는 기타 수단에 의해 용기에 부착될 수 있는, 완전히 또는 부분적으로 분리가 가능한 마개를 장착할 수 있으며, 또한 주사바늘에 의해 내용물에 접근할 수 있는 스토퍼가 장착될 수 있다. 상기 키트는 외부 패키지를 포함할 수 있으며, 외부 패키지는 구성 요소들의 사용에 관한 지시서를 포함할 수 있으나, 이에 제한되는 것은 아니다.The kit of the present invention may include a container in addition to the composition and the instructions. The container may serve to package the component, and may also serve to store and secure the component. The material of the container may take the form of, for example, a bottle, a tub, a sachet, an envelope, a tube, an ampoule, etc., which may be partially or entirely made of plastic, glass, paper, or foil. , wax, etc. The container may be equipped with a completely or partially removable closure that may initially be part of the container or may be attached to the container by mechanical, adhesive, or other means, and may also provide access to the contents by needle. A stopper can be installed. The kit may include an external package, and the external package may include, but is not limited to, instructions for use of the components.

또한, 본 발명은 종양 또는/및 종양 내 침윤(tumor infiltrating) 조절 T 세포(regulatory T cell, Treg)의 검출; 또는 종양 진단에 필요한 정보를 제공하기 위하여, 상기 본 발명의 89Zr 표지된 항-CD25 항체를 조영제로 사용하여 PET 영상을 획득하는 방법을 제공한다. In addition, the present invention provides detection of tumor or/and tumor infiltrating regulatory T cells (regulatory T cells, Tregs); Alternatively, in order to provide information necessary for tumor diagnosis, a method of acquiring a PET image is provided using the 89 Zr-labeled anti-CD25 antibody of the present invention as a contrast agent.

또한, 본 발명은 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 조성물의, 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화 용도를 제공한다.In addition, the present invention provides a composition comprising an 89 Zr-labeled anti-CD25 antibody as an active ingredient for use in the diagnosis of tumors; or detection or imaging of a tumor or infiltrating regulatory T cells within a tumor.

또한, 본 발명은 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화제를 제조하기 위한, 물89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 조성물의 용도를 제공한다. Additionally, the present invention relates to diagnosis of tumors; Alternatively, it provides the use of a composition comprising a water 89 Zr labeled anti-CD25 antibody as an active ingredient for preparing an imaging agent or detection of tumor or intratumoral infiltrating regulatory T cells.

본 발명에서 '개체'란 질병의 진단 또는 치료를 필요로 하는 대상을 의미하고, 보다 구체적으로는 인간 또는 비-인간인 포유류(영장류), 생쥐 (mouse), 쥐 (rat), 개, 고양이, 말, 및 소 등의 포유류일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, 'individual' refers to an object that requires diagnosis or treatment of a disease, and more specifically, human or non-human mammals (primates), mice, rats, dogs, cats, It may be a mammal such as a horse or a cow, but is not limited thereto.

이하, 본 발명의 이해를 돕기 위하여 바람직한 실시예 및 실험예를 제시한다. 그러나 하기의 실시예 및 실험예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 하기 실시예 및 실험예에 의해 본 발명의 내용이 한정되는 것은 아니다.Below, preferred examples and experimental examples are presented to aid understanding of the present invention. However, the following examples and experimental examples are provided only to make the present invention easier to understand, and the content of the present invention is not limited by the following examples and experimental examples.

실시예 1. Example 1. 8989 Zr 표지 항-CD25 항체 제조Preparation of Zr-labeled anti-CD25 antibody

종래의 표지 기술 과정 중에 나타나는 항체 내 화학적 결합(bond)의 변화 과정을 도 2a에 개략적으로 나타내었으며, 이에 따라 항체는 도 2b와 같은 구조로 표지된다. 본 발명은 도 2a 및 도 2b의 종래의 기술과는 다른 본 발명 특유의 방식으로 항-CD25 항체의 힌지 영역 이황화 결합부위에 특이적으로 89Zr 원자 2개를 결합시켜 immuno-PET로 촬영하였을 때 매우 우수한 이미지 또는 영상을 수득할 수 있다. 이하에서, 본 발명에 따른 89Zr 표지 항-CD25 항체를 제작하는 방법을 상세히 기술하였다.The process of changing the chemical bond within the antibody that occurs during the conventional labeling technology process is schematically shown in Figure 2a, and accordingly, the antibody is labeled with the structure shown in Figure 2b. In the present invention, two 89 Zr atoms are specifically bound to the disulfide bond site of the hinge region of the anti-CD25 antibody in a unique manner, which is different from the conventional techniques of Figures 2a and 2b, and when imaged by immuno-PET, Very excellent images or videos can be obtained. Below, the method for producing the 89 Zr labeled anti-CD25 antibody according to the present invention is described in detail.

1-1. 항-CD25 항체 환원 및 시스테인-특이적 데페록사민-말레이미드 접합 단계1-1. Anti-CD25 antibody reduction and cysteine-specific deferoxamine-maleimide conjugation steps

먼저, 사람 또는 마우스 유래의 항-CD25 항체(BioXCell 사(West Lebanon, NH) InVivoMAb anti-human CD25(IL-2Rα; Catalog# BE0014) 또는 InVivoMAb anti-mouse CD25(IL-2Rα; Catalog# BE0012), Mouse monoclonal IgG2a anti- CD25 Ab (clone, 7G7/B6), rat monoclonal IgG1 Ab (clone, PC-61.5.3))의 시스테인(cysteine) 잔기 내 설프히드릴기(sulfohydryl residues)에 위치 특이적으로(site-specific) 데페록사민-말레이미드(deferoxamine[DFO]-maleimide)를 접합하였다. 본 명세서에서 데페록사민 말레이미드 접합된 항-CD25 항체를 'DFO-conjugated CD25 IgG', 또는 'DFO-Mal 접합된 항-CD25 항체'등으로 표기한다. First, anti-CD25 antibodies derived from humans or mice (InVivoMAb anti-human CD25 (IL-2Rα; Catalog# BE0014) or InVivoMAb anti-mouse CD25 (IL-2Rα; Catalog# BE0012) from BioXCell (West Lebanon, NH); Mouse monoclonal IgG2a anti- CD25 Ab (clone,  7G7/B6), rat monoclonal IgG1 Ab (clone,  PC-61.5.3)) position-specifically ( Site-specific deferoxamine-maleimide (deferoxamine[DFO]-maleimide) was conjugated. In this specification, the deferoxamine maleimide conjugated anti-CD25 antibody is referred to as 'DFO-conjugated CD25 IgG', or 'DFO-Mal conjugated anti-CD25 antibody'.

항-CD25 항체를 상기 항체 몰농도 대비 70 ~ 130배 이상의 몰농도에 해당하는 TCEP(tris-carboxyethylphosphine)로 환원시킨 후 deferoxamine-maleimide chelator와 반응시켜 항체 힌지 영역의 이황화 결합부위에 총 2개 89Zr 원자의 데페록사민-말레이미드를 특이적으로 결합시킬 수 있었으며, 구체적인 방법은 다음과 같다. The anti-CD25 antibody was reduced with TCEP (tris-carboxyethylphosphine) at a molar concentration of 70 to 130 times higher than the molar concentration of the above antibody, and then reacted with deferoxamine-maleimide chelator to form a total of two 89 Zr at the disulfide bond site in the antibody hinge region. It was possible to specifically bind the deferoxamine-maleimide atom, and the specific method is as follows.

2 mg 항-CD25 항체를 100 mM TCEP(tris(2-carboxyethyl)phosphine, 1:100 molar ratio)와 실온에서 20분 동안 반응시켜 환원시켰다(reducing). 이어서, 150 mM NaCl 및 1 mM EDTA(ethylene diamine tetraacetic acid)를 포함하는 0.1 M 인산나트륨(sodium phosphate) 용액에 상기 환원시킨 항-CD25 항체를 희석하였다. 여기에 2 mM deferoxamine-maleimide(DFO-Mal) chelator 56.4 μL를 첨가하여 실온에서 1시간 반응시킴에 따라 항-CD25 항체의 힌지영역(hinge region) 시스테인 잔기의 설프히드릴기에 부착시켰다. 반응 결과물을 PD-10 column으로 elution하여 DFO-conjugated CD25 antibody 분획(fraction)을 농축시켰으며, 분획으로부터 DFO-conjugated CD25 IgG를 수득하였다. 데페록사민-말레이미드:항체의 몰비는 60:1이었다.2 mg anti-CD25 antibody was reduced by reacting with 100 mM TCEP (tris(2-carboxyethyl)phosphine, 1:100 molar ratio) at room temperature for 20 minutes. Subsequently, the reduced anti-CD25 antibody was diluted in 0.1 M sodium phosphate solution containing 150mM NaCl and 1mM EDTA (ethylene diamine tetraacetic acid). 56.4 μL of 2mM deferoxamine-maleimide (DFO-Mal) chelator was added and reacted at room temperature for 1 hour to attach to the sulfhydryl group of the cysteine residue in the hinge region of the anti-CD25 antibody. The reaction product was eluted with a PD-10 column to concentrate the DFO-conjugated CD25 antibody fraction, and DFO-conjugated CD25 IgG was obtained from the fraction. The molar ratio of deferoxamine-maleimide:antibody was 60:1.

상기 방법으로 항-CD25 항체에 2개 89Zr 원자가 결합하였는지 확인하였다. 구체적으로, 유사한 항체에 동일한 기법으로 표지를 수행했을 때에 Mass spectrometry로 측정한 MALDI peak에서 unmodified antibody의 질량은 147786 Da으로 조사되었다. 반면, 본 발명에 따른 방법으로 제조된 DFO-conjugated IgG의 질량은 75443 Da으로 나타났고, 이는 한 개의 항체당 2.18개의 deferoxamine-maleimide conjugations에 해당한다는 점에서 항체 힌지 영역의 이황화 결합부위에 총 2개 89Zr 원자의 결합이 형성되었음이 검증되었다.Using the above method, it was confirmed that two 89 Zr atoms were bound to the anti-CD25 antibody. Specifically, when similar antibodies were labeled using the same technique, the mass of the unmodified antibody in the MALDI peak measured by mass spectrometry was found to be 147786 Da. On the other hand, the mass of DFO-conjugated IgG prepared by the method according to the present invention was found to be 75443 Da, which corresponds to 2.18 deferoxamine-maleimide conjugations per antibody, so there are a total of two at the disulfide bond site in the antibody hinge region. It was verified that bonds of 89 Zr atoms were formed.

또한, 상기 방법으로 제조된 DFO-conjugated 항-CD25 항체(사람 또는 마우스 항체)가 환원 상태인지 여부를 확인하기 위하여 SDS-PAGE(Non-reducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis)분석을 수행하였다. 구체적으로, DFO-conjugated 항-CD25 항체(사람 또는 마우스 항체) 2 μg을 물에 희석시킨 다음 dithiothreitol이 빠진 5x non-reducing 샘플 버퍼와 혼합하였다. 그 후, 95℃에서 10분 동안 끓였으며 8% SDS PAGE gel상에서 전기영동(electrophoresis)을 수행하고, 0.5% coomassie blue로 염색하였다.In addition, SDS-PAGE (Non-reducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis) analysis was performed to confirm whether the DFO-conjugated anti-CD25 antibody (human or mouse antibody) prepared by the above method was in a reduced state. Specifically, 2 μg of DFO-conjugated anti-CD25 antibody (human or mouse antibody) was diluted in water and then mixed with 5x non-reducing sample buffer without dithiothreitol. Afterwards, it was boiled at 95°C for 10 minutes, electrophoresis was performed on an 8% SDS PAGE gel, and stained with 0.5% coomassie blue.

실험결과 도 3a 및 도 3b에 나타낸 바와 같이, 사람 또는 마우스 CD25 IgG 항체의 disulfide bond는 TCEP에 의해 완전히 환원되었으며 DFO conjugation과 농축 후에도 환원상태가 유지되는 것이 확인되었다.As shown in Figures 3a and 3b, the disulfide bond of human or mouse CD25 IgG antibody was completely reduced by TCEP and it was confirmed that the reduced state was maintained even after DFO conjugation and concentration.

1-2. 데페록사민-말레이미드 접합된 항-CD25 항체(CD25 Ab-deferoxamine maleimide, 또는 DFO-conjugated CD25 IgG로도 표기)에 1-2. Deferoxamine-maleimide conjugated anti-CD25 antibody (CD25 Ab-deferoxamine maleimide, also referred to as DFO-conjugated CD25 IgG) 8989 Zr 표지 단계Zr labeling steps

상기 실시예 1-1에서 제작된 DFO-conjugated CD25 IgG를 89Zr-oxalate와 실온에서 반응시켜 도 4에서 나타낸 구조의 89Zr-CD25 IgG를 합성하였다. DFO-conjugated CD25 IgG prepared in Example 1-1 was reacted with 89 Zr-oxalate at room temperature to synthesize 89 Zr-CD25 IgG with the structure shown in FIG. 4.

구체적으로, 89Zr-oxalate(50 μL; Korea Atomic Energy Research Institute)를 2 M Na2CO3 25 μL로 중화(neutralized)시켰다. 75 μL의 0.5 M 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid buffer(pH 7.5)에 DFO-Mal 접합된 CD25 IgG(사람 또는 마우스 항체)를 희석한 뒤, 89Zr-oxalate와 실온에서 1시간 인큐베이션하여 반응시켜 표지하였다. 0.5% gentisic acid를 포함하는 0.25 M sodium acetate 용액을 용출 버퍼로 이용하였으며, 반응 결과물을 PD-10 column으로 size-exclusion chromatography하여 정제하였고, 최종적으로 89Zr 표지 CD25 IgG가 용출(elute)되는 분획(fraction)을 분리 정제하여 89Zr 표지 CD25 IgG를 수득하였다.Specifically, 89 Zr-oxalate (50 μL; Korea Atomic Energy Research Institute) was neutralized with 25 μL of 2 M Na 2 CO 3 . DFO-Mal conjugated CD25 IgG (human or mouse antibody) was diluted in 75 μL of 0.5 M 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid buffer (pH 7.5), then incubated with 89 Zr-oxalate for 1 hour at room temperature. The reaction was incubated and labeled. A 0.25 M sodium acetate solution containing 0.5% gentisic acid was used as an elution buffer, and the reaction product was purified by size-exclusion chromatography with a PD-10 column, and finally, the fraction in which 89 Zr-labeled CD25 IgG elutes ( fraction) was separated and purified to obtain 89 Zr-labeled CD25 IgG.

상기 방법으로 수득된 89Zr 표지 CD25 IgG(사람 또는 마우스 항체)의 89Zr 표지가 성공적으로 이루어졌는지를 확인하기 위하여 자가방사기록(autoradiography) 실험을 수행하였다. 구체적으로, 이를 위해 89Zr-CD25 IgG를 8% native PAGE로 분리하여 감광판에 노출시켰다.An autoradiography experiment was performed to confirm whether 89 Zr labeling of the 89 Zr labeled CD25 IgG (human or mouse antibody) obtained by the above method was successful. Specifically, for this purpose, 89 Zr-CD25 IgG was separated using 8% native PAGE and exposed to a photosensitive plate.

실험결과 도 5a 및 도 5b에 나타낸 바와 같이, 사람 또는 마우스 89Zr-CD25 IgG에 대하여 예상한 170kD 위치에 radioactive IgG band가 확인되었다.As shown in Figures 5a and 5b, a radioactive IgG band was confirmed at the 170kD position expected for human or mouse 89 Zr-CD25 IgG.

또한, PD-10 size-exclusion chromatography로 분리 정제한 후 용출된 분획(eluted fraction)들을 감마 계수한 결과, 첫 번째 방사능 절정(peak)에 나타난 사람 또는 마우스 89Zr-CD25 IgG 표지 효율은 각각 68.7%와 67.2%인 것으로 조사되었고, 최종적으로 수득한 89Zr-CD25 IgG의 방사화학적 순도는 >98%, 비방사능은 >0.8 mCi/mg으로 확인되었다 (도 6a 및 도 6b 참조).In addition, as a result of gamma counting the eluted fractions after separation and purification using PD-10 size-exclusion chromatography, the labeling efficiency of human or mouse 89 Zr-CD25 IgG at the first radioactivity peak was 68.7%, respectively. and 67.2%, and the radiochemical purity of the finally obtained 89 Zr-CD25 IgG was confirmed to be >98% and the specific radioactivity was confirmed to be >0.8 mCi/mg (see Figures 6a and 6b).

이와 같은 결과에 따라, 실시예 1에 따라 CD25 IgG에 89Zr가 성공적으로 접합될 수 있고, 이 접합 효율은 약 70%, 순도는 98% 이상으로 현저히 우수한 89Zr 표지 CD25 IgG를 제조할 수 있음이 확인되었다.According to these results, 89 Zr can be successfully conjugated to CD25 IgG according to Example 1, and the conjugation efficiency is about 70% and the purity is over 98%, making it possible to produce a remarkably excellent 89 Zr-labeled CD25 IgG. This has been confirmed.

실시예 2. Example 2. 8989 Zr-CD25 IgG의 안정성 및 혈청 내 표지 안정성 확인Confirmation of stability of Zr-CD25 IgG and labeling in serum

상기 실시예 1-2에서 최종적으로 수득된 89Zr 표지 CD25 IgG(사람 또는 마우스 항체)의 혈청 안정성을 확인하기 위하여 radio-instant thin layer chromatography(radio-iTLC)을 수행하였다. 구체적으로, silica gel을 입힌 iTLC-SG glass microfiber chromatography paper를 이용하였다. 또한, 89Zr 표지 CD25 IgG(사람 또는 마우스 항체) 각각을 PBS 또는 FBS에 넣어 실험군으로 하였다. 각 실험군을 PBS 또는 FBS에 넣고 37℃에서 1일 내지 7일 동안 반응시킨 후, 50 mM EDTA(ethylene diamine tetraacetic acid, pH 5.5)를 용매로 이용하여 크로마토그래피를 수행하였다. 온전한(Intact) 89Zr-CD25 IgG는 기저 위치에 남아 있는데 반하여, 유리된 89Zr4+ 이온과 89Zr-EDTA는 용매 전선을 따라 이동하게 되어 분리된다.To confirm the serum stability of the 89 Zr-labeled CD25 IgG (human or mouse antibody) finally obtained in Example 1-2, radio-instant thin layer chromatography (radio-iTLC) was performed. Specifically, iTLC-SG glass microfiber chromatography paper coated with silica gel was used. In addition, 89 Zr-labeled CD25 IgG (human or mouse antibody), respectively, was added to PBS or FBS to serve as an experimental group. Each experimental group was placed in PBS or FBS and reacted at 37°C for 1 to 7 days, and then chromatography was performed using 50 mM EDTA (ethylene diamine tetraacetic acid, pH 5.5) as a solvent. Intact 89 Zr-CD25 IgG remains in the basal position, whereas free 89 Zr 4+ ions and 89 Zr-EDTA migrate along the solvent front and are separated.

실험결과 도 7a 및 도 7b에 나타낸 바와 같이, 각각의 실험군인 사람 또는 마우스 89Zr-CD25 IgG의 안정성은 PBS에서는 >90%(90% 초과)으로, FBS에서는 >80%(80% 초과)(n = 2)으로 나타나, 89Zr 표지 CD25 IgG는 사람 또는 마우스 항체 여부에 무관하게 방사성 표지가 안정한 것으로 확인되었다. As shown in Figures 7a and 7b, the stability of human or mouse 89 Zr-CD25 IgG in each experimental group was >90% (>90%) in PBS and >80% (>80%) in FBS ( n = 2), 89 Zr-labeled CD25 IgG was confirmed to be radioactively stable regardless of whether it was a human or mouse antibody.

실시예 3. In vitro 림프종 세포 모델에서의 Example 3. In vitro lymphoma cell model 8989 Zr-CD25 IgG의 우수한 결합 친화도 및 특이도 확인Confirmed excellent binding affinity and specificity of Zr-CD25 IgG

3-1. 림프종 세포 종류별 CD25 발현정도 확인과 림프종 세포모델 준비3-1. Confirmation of CD25 expression level by lymphoma cell type and preparation of lymphoma cell model

89Zr-CD25 IgG의 결합 친화도 및 특이도를 분석하기 위하여 CD25 발현 수준이 가장 우수한 림프종 세포를 선별하였다. 89 To analyze the binding affinity and specificity of Zr-CD25 IgG, lymphoma cells with the highest CD25 expression level were selected.

구체적으로, 웨스턴 블롯(western blotting) 분석을 통해 사람 림프종 세포주인 H9, Jurkat 및 SUDHL1과, 마우스 림프종 세포주 EL4의 CD25 마커 발현 수준을 조사하였다. 먼저, 상기 암세포들을 용해하여 단백질을 분리 및 정량하여 각 15 μg의 단백질을 10% gel로 전기영동하고 membrane에 옮긴 후, rabbit-anti-CD25 1차 항체(abcam #ab231441; 1:1000 희석)를 4℃에서 밤새 반응시켰다. 이어 TBST 버퍼로 10분씩 3회 세척한 다음, HRP-conjugated anti-rabbit 2차 항체(Cell Signaling #7074S; 1:2000 희석)와 실온에서 1시간 반응시켰다. 다시 TBST 버퍼로 10분씩 3회 세척한 다음 membrane을 enhanced chemiluminescence substrate와 반응시키고 필름을 노출하여 CD25 단백질의 band intensities를 검출 및 정량하였다.Specifically, Through western blotting analysis The expression levels of the CD25 marker were examined in human lymphoma cell lines H9, Jurkat, and SUDHL1 and mouse lymphoma cell line EL4. First, the cancer cells were lysed, the proteins were separated and quantified, and 15 μg of each protein was electrophoresed using a 10% gel and transferred to a membrane, followed by rabbit-anti-CD25 primary antibody (abcam #ab231441; 1:1000 dilution). Reacted overnight at 4°C. After washing with TBST buffer three times for 10 minutes each, it was reacted with HRP-conjugated anti-rabbit secondary antibody (Cell Signaling #7074S; 1:2000 dilution) for 1 hour at room temperature. After washing with TBST buffer three times for 10 minutes each, the membrane was reacted with an enhanced chemiluminescence substrate and the film was exposed to detect and quantify the band intensities of CD25 protein.

또한 세포 표면의 CD25 마커 발현 수준에 대한 정량 분석을 위해 항-CD25 IgG에 FITC 또는 phycoerythrin(PE) 형광신호를 부착하여 암세포를 염색시킨 다음 FACS 분석(fluorescence-activated cell sorting analysis)을 수행하였다. 먼저, nonenzymatic cell-dissociation solution으로 세포들을 회수(harvested)한 뒤 세척하고, 5% fetal bovine serum 및 0.2% bovine serum albumin을 포함하는 phosphate-buffered saline 용액 내에서 실온(RT) 상태로 30분 동안 phycoerythrin antibody와 인큐베이션 하였다. FACS buffer로 세척한 후 다시 500 mL의 FACS buffer를 가하여 CellQuest software를 이용하는 FACSCalibur(BE Biosciences)로 flow cytometry를 수행하였다. Additionally, for quantitative analysis of the expression level of the CD25 marker on the cell surface, cancer cells were stained by attaching a FITC or phycoerythrin (PE) fluorescence signal to anti-CD25 IgG, and then FACS analysis (fluorescence-activated cell sorting analysis) was performed. First, the cells were harvested with a nonenzymatic cell-dissociation solution, washed, and incubated with phycoerythrin for 30 minutes at room temperature (RT) in a phosphate-buffered saline solution containing 5% fetal bovine serum and 0.2% bovine serum albumin. Incubation was performed with antibody. After washing with FACS buffer, 500 mL of FACS buffer was added again, and flow cytometry was performed with FACSCalibur (BE Biosciences) using CellQuest software.

실험결과 도 8a와 같이, SUDHL1 사람 T 림프종 암세포주에서 CD25 발현 수준이 현저히 높은 것으로 확인되어 상기 SUDHL1 세포주를 이하의 실험에서 대표적인 CD25 과발현 림프종 세포로서 이용하였다. As a result of the experiment, as shown in Figure 8a, it was confirmed that the expression level of CD25 was significantly high in the SUDHL1 human T lymphoma cancer cell line, and the SUDHL1 cell line was used as a representative CD25-overexpressing lymphoma cell in the following experiments.

3-2. In vitro SUDHL1 림프종 세포 모델에서 3-2. In an in vitro SUDHL1 lymphoma cell model 8989 Zr-CD25 IgG의 우수한 결합 친화도 및 특이도 확인Confirmed excellent binding affinity and specificity of Zr-CD25 IgG

실시예 1에 따라 제조된 89Zr-CD25 IgG의 결합 친화도 및 특이도(binding specificity)를 In vitro SUDHL1 림프종 세포 모델에서 확인하였다. The binding affinity and specificity of 89 Zr-CD25 IgG prepared according to Example 1 were confirmed in an in vitro SUDHL1 lymphoma cell model.

구체적으로, 상기 세포를 배양하여 89Zr-CD25 IgG(사람)와 반응시킨 후, 세포를 세척하고 감마계수기를 이용하여 통상적인 방법으로 섭취율을 %ID 단위로 측정하였다. 이때, H9 및 Jurkat 세포주를 비교군으로 설정하였다.Specifically, the cells were cultured and reacted with 89 Zr-CD25 IgG (human), the cells were washed, and the uptake rate was measured in %ID by a conventional method using a gamma counter. At this time, H9 and Jurkat cell lines were set as comparison groups.

실험결과 도 8b에 나타낸 바와 같이, CD25 발현이 높은 SUDHL1 사람 T cell 림프종 세포와 CD25 발현량이 낮은 사람 H9 및 Jurkat 암세포에서 사람 89Zr-CD25 IgG와의 결합정도를 비교한 결과, SUDHL1 림포종 세포에서 수십 배 높은 결합 수준이 확인되었다(n = 6, both P <0.001).As shown in Figure 8b, as a result of comparing the degree of binding to human 89 Zr-CD25 IgG in SUDHL1 human T cell lymphoma cells with high CD25 expression and human H9 and Jurkat cancer cells with low CD25 expression, dozens of binding was found in SUDHL1 lymphoma cells. A fold higher binding level was confirmed (n = 6, both P <0.001).

또한 다량의 cold anti-CD25 antibody로 결합을 억제하여(binding inhibition 실험) 결합 특이도를 % blocking으로 정량하였으며, 이 때, EL4 세포주를 비교군으로 하여 실험하였다. In addition, binding was inhibited with a large amount of cold anti-CD25 antibody (binding inhibition experiment) and binding specificity was quantified as % blocking. At this time, the EL4 cell line was used as a comparison group.

실험결과 도 8c에 나타낸 바와 같이, CD25 발현이 높은 SUDHL1 림프종 세포에서 결합 특이도를 측정하기 위해 다량의 cold anti-CD25 antibody를 이용하여 binding inhibition 실험을 한 결과, 89Zr-CD25 IgG는 97% 이상의 결합 특이도를 가지는 것이 나타나(n = 6, P <0.001), 본 발명에 따른 사람 89Zr-CD25 IgG의 결합이 거의 완벽하게 억제되는 것으로 확인되었다. CD25 발현이 낮은 EL4 림프종 세포에서는 마우스 89Zr-CD25 IgG의 낮은 결합이 다량의 cold anti-CD25 antibody에 의해 소량만 억제되었다(도 8d).As shown in Figure 8c, as a result of a binding inhibition experiment using a large amount of cold anti-CD25 antibody to measure binding specificity in SUDHL1 lymphoma cells with high CD25 expression, 89 Zr-CD25 IgG showed an inhibition rate of more than 97%. It was shown to have binding specificity (n = 6, P <0.001), and it was confirmed that the binding of human 89 Zr-CD25 IgG according to the present invention was almost completely inhibited. In EL4 lymphoma cells with low CD25 expression, the low binding of mouse 89 Zr-CD25 IgG was only slightly inhibited by a large amount of cold anti-CD25 antibody (Figure 8d).

실시예 4. Treg 면역세포에서의 Example 4. In Treg immune cells 8989 Zr-CD25 IgG의 우수한 결합 친화도 및 특이도 확인Confirmed excellent binding affinity and specificity of Zr-CD25 IgG

실시예 1에 따라 제조된 89Zr-CD25 IgG의 Treg에 대한 결합 친화도 및 특이도(binding specificity)를 분석하였다. The binding affinity and specificity of 89 Zr-CD25 IgG prepared according to Example 1 to Tregs were analyzed.

먼저, Easy prep kit(Stem Cell Tech 社)를 제조사의 프로토콜에 따라 정상 마우스의 흉선 또는 비장으로부터 CD25(+) Treg 세포를 정제 및 분리하였다. 구체적으로, FACS를 이용하여 마우스의 흉선으로부터 CD25(-) T 세포와 CD25(+) Treg 세포를 분리정제 하였고(도 9a 참조), Treg 세포를 hematoxylin & eosin 염색하였다(도 9b 참조). 또한, anti-CD3 antibody와 IL2를 이용하여 CD25(+) Treg 세포의 수를 2주 동안 expansion 시켰다. 그 후, 실시예 3-2와 동일한 방법으로 CD25(-) T 세포와 CD25(+) Treg 세포에 대한 89Zr-CD25 IgG의 결합정도를 분석하였다. First, CD25(+) Treg cells were purified and isolated from the thymus or spleen of normal mice using the Easy prep kit (Stem Cell Tech) according to the manufacturer's protocol. Specifically, CD25(-) T cells and CD25(+) Treg cells were isolated and purified from the thymus of mice using FACS (see Figure 9a), and the Treg cells were stained with hematoxylin and eosin (see Figure 9b). Additionally, the number of CD25(+) Treg cells was expanded for 2 weeks using anti-CD3 antibody and IL2. Afterwards, the degree of binding of 89 Zr-CD25 IgG to CD25(-) T cells and CD25(+) Treg cells was analyzed in the same manner as in Example 3-2.

실험 결과 도 10a에 나타낸 바와 같이, 마우스의 흉선으로부터 분리된 CD25(-) T 세포 대비 CD25(+) Treg 세포에 대한 89Zr-CD25 IgG 결합이 391.2 ± 135.2%로 현저히 높은 것을 확인하였다. 특히, CD25(+) Treg 세포에서의 높은 89Zr-CD25 IgG 결합은 0.5 μM의 cold unlabeled antibody가 있는 조건에서는 72.1%이나 억제된다는 점에서, 본 발명에 따른 89Zr-CD25 IgG는 결합력이 우수할 뿐만 아니라 표적 특이도도 현저히 우수한 것으로 확인되었다(n =6, P <0.001, 도 9a 참조). As shown in FIG. 10A, the binding of 89 Zr-CD25 IgG to CD25(+) Treg cells was confirmed to be significantly higher at 391.2 ± 135.2% compared to CD25(-) T cells isolated from the mouse thymus. In particular, the high 89 Zr-CD25 IgG binding to CD25(+) Treg cells is inhibited by 72.1% in the presence of 0.5 μM of cold unlabeled antibody, so the 89 Zr-CD25 IgG according to the present invention has excellent binding ability. In addition, target specificity was confirmed to be significantly excellent (n =6, P <0.001, see Figure 9a).

또한, 도 10b에 나타낸 바와 같이, 마우스의 비장으로부터 분리된 CD25(+) Treg 세포의 경우에도 흉성과 동일한 양상의 결합력 및 특이도가 확인되었다. 구체적으로, 89Zr-CD25 IgG는 CD25(+) Treg 세포에 대하여 높은 수준으로 결합을 형성하였으며 상기의 결과는 0.5 μM의 cold unlabeled antibody 조건에서는 87.0 ± 0.7%이나 억제되는 것으로 확인되었다(n = 3, P <0.01, 도 9b 참조).In addition, as shown in Figure 10b, the same binding affinity and specificity as for CD25(+) Treg cells isolated from the spleen of mice were confirmed. Specifically, 89 Zr-CD25 IgG formed a high level of binding to CD25(+) Treg cells, and the above results were confirmed to be inhibited by 87.0 ± 0.7% under the condition of 0.5 μM cold unlabeled antibody (n = 3). , P < 0.01, see Figure 9b).

이와 같은 결과는 Treg 세포의 유래와 관계없이 본 발명에 따른 89Zr-CD25 IgG가 CD25(+) Treg 세포에 대한 높은 결합력과 표적 특이도를 동시에 가지는 것을 시사하는 것이다.These results suggest that 89 Zr-CD25 IgG according to the present invention has both high binding affinity and target specificity for CD25(+) Treg cells, regardless of the origin of the Treg cells.

실시예 5. In vivo 마우스 림프종 모델에서의 Example 5. In vivo mouse lymphoma model 8989 Zr-CD25 IgG의 체내분포 분석Analysis of body distribution of Zr-CD25 IgG

89Zr-CD25 IgG의 체내분포를 분석하기 위하여 우선적으로 In vivo 마우스 림프종 모델을 준비하였다. 구체적으로, SUDHL1 사람 T cell 림프종 세포를 배양하여 면역결핍 balb/C nude 마우스의 어깨 부위에 SUDHL1 세포 1 x 107개를 피하 주사하였다. 이식 후 21일 정도 되어 1 cm의 종양이 형성된 것이 확인된 마우스(n = 6) 꼬리정맥에 89Zr-CD25 IgG(사람)를 주사하였다. 5일 경과 후, 경구탈구 방법으로 희생시켰으며, 종양, 혈액 및 주요장기를 적출하여 무게를 측정한 다음 방사성 계수를 고에너지 감마카운터로 측정하여 섭취율을 % injected-dose per gram(%ID/g) 단위로 산출하였다. 89 To analyze the body distribution of Zr-CD25 IgG, an in vivo mouse lymphoma model was first prepared. Specifically, SUDHL1 human T cell lymphoma cells were cultured and 1 x 10 7 SUDHL1 cells were subcutaneously injected into the shoulder area of immunodeficient balb/C nude mice. About 21 days after transplantation, 89 Zr-CD25 IgG (human) was injected into the tail vein of mice (n = 6) in which 1 cm tumors were confirmed to have formed. After 5 days, they were sacrificed by oral dislocation, and the tumor, blood, and major organs were removed and weighed, and the radioactivity coefficient was measured with a high-energy gamma counter to determine the intake rate as % injected-dose per gram (%ID/g). ) was calculated in units.

실험 결과 도 11에 나타낸 바와 같이, 마우스 체내에서 SUDHL1 림프종 종양의 89Zr-CD25 IgG 섭취가 9.4 ± 2.3 %ID/g으로 확인되었다. 이는 근육 대비 10배를 초과하는 수준이고, 심장, 폐, 간, 비장, 신장에서의 섭취율 역시 종양 대비 30 내지 40% 정도에 불과한 것으로 조사되었다. As shown in FIG. 11, the uptake of 89 Zr-CD25 IgG by SUDHL1 lymphoma tumor in the mouse body was confirmed to be 9.4 ± 2.3 %ID/g. This is 10 times more than that in muscle, and the uptake rate in the heart, lungs, liver, spleen, and kidneys was also found to be only about 30 to 40% of that in tumors.

또한 상기 마우스 모델에 0.8 mg의 cold unlabeled antibody를 먼저 주사한 후 89Zr-CD25 IgG를 주입한 경우에는 SUDHL1 림프종 종양에서 89Zr-CD25 IgG의 섭취가 3.8 ± 0.8 %ID/g로 현저히 감소하는 것으로 나타났다. 이는 앞선 실험 결과 대비 60%나 줄어든 수치로 89Zr-CD25 IgG의 높은 결합 특이도를 시사하는 것이다(n = 6, P <0.001, 도 11 참조). 반면, 혈액 및 기타 장기에는 cold unlabeled antibody에 의해 89Zr-CD25 IgG 섭취에 변화가 없었다. In addition, when 0.8 mg of cold unlabeled antibody was first injected into the mouse model and then 89 Zr-CD25 IgG was injected, the uptake of 89 Zr-CD25 IgG in SUDHL1 lymphoma tumor was significantly reduced to 3.8 ± 0.8 %ID/g. appear. This is a 60% reduction compared to the previous experiment result, suggesting the high binding specificity of 89 Zr-CD25 IgG (n = 6, P < 0.001, see Figure 11). On the other hand, there was no change in the uptake of 89 Zr-CD25 IgG in the blood and other organs by cold unlabeled antibody.

이에 따라 본 발명의 89Zr-CD25 IgG 항체는 체내에서도(in vivo 상에서도) 종양 특이적으로 타겟팅(targeting) 및 결합되는 효과가 우수한 것이 검증되었다.Accordingly, it was verified that the 89 Zr-CD25 IgG antibody of the present invention has excellent tumor-specific targeting and binding effects even in vivo.

실시예 6. In vivo 마우스 림프종 모델에서의 Example 6. In vivo mouse lymphoma model 8989 Zr-CD25 IgG의 우수한 이미징 효과 확인Confirm the excellent imaging effect of Zr-CD25 IgG

89Zr-CD25 IgG의 이미징(영상화, 조영) 효과를 확인하기 위하여 PET 영상 분석을 실시하였다. 먼저, SUDHL1 사람 T cell 림프종 세포를 배양하여 면역결핍 balb/C nude 마우스의 어깨 부위에 SUDHL1 세포 1 x 107개를 피하 주사하였다. 이식 후 21일 정도 되어 1 cm의 종양이 형성된 마우스(n = 6) 꼬리정맥에 0.2 mCi 89Zr-CD25 IgG(사람)를 주사하고 5일 후, 소동물 전용 Inveon micro PET/CT scanner(Siemens, USA)를 이용하여 PET/CT 영상을 획득하였다. 이때 일부 마우스에는 5:1 molar ratio의 cold 항체를 1 hr 전에 pre-injection하여 89Zr-CD25 IgG의 종양 결합(종양 섭취) 특이도를 분석하였다. PET-based 조직 방사능은 non-attenuation corrected 영상에서 regions-of-interest(ROIs)를 적용하여 분석하였으며, 이때 blood pool, 주요장기, 종양을 포함시켜 분석하였다. 89 PET image analysis was performed to confirm the imaging (imaging, contrast) effect of Zr-CD25 IgG. First, SUDHL1 human T cell lymphoma cells were cultured and 1 x 10 7 SUDHL1 cells were subcutaneously injected into the shoulder area of immunodeficient balb/C nude mice. About 21 days after transplantation, 0.2 mCi 89 Zr-CD25 IgG (human) was injected into the tail vein of mice (n = 6) with 1 cm tumors formed, and 5 days later, small animal-specific Inveon micro PET/CT scanner (Siemens, PET/CT images were acquired using USA). At this time, some mice were pre-injected with cold antibodies at a 5:1 molar ratio 1 hr before to analyze the tumor binding (tumor uptake) specificity of 89 Zr-CD25 IgG. PET-based tissue radioactivity was analyzed by applying regions-of-interest (ROIs) in non-attenuation corrected images, including blood pool, major organs, and tumor.

micro-PET/CT 이미징 수행 결과 도 12에 나타낸 바와 같이, 종양 부위에서 우수한 대조도로 높은 89Zr-CD25 IgG 결합(섭취)가 나타났다. 또한 동일한 마우스 모델에 0.8 mg의 cold unlabeled antibody를 먼저 주사한 다음 89Zr-CD25 IgG를 주입한 경우에는 종양의 89Zr-CD25 IgG 섭취가 상당히 감소하며 대조도가 줄어드는 것이 확인되었다(도 13 참조).As shown in Figure 12, micro-PET/CT imaging results showed high 89 Zr-CD25 IgG binding (uptake) with excellent contrast in the tumor area. In addition, when 0.8 mg of cold unlabeled antibody was first injected into the same mouse model and then 89 Zr-CD25 IgG was injected, it was confirmed that the tumor's 89 Zr-CD25 IgG uptake was significantly reduced and the contrast decreased (see Figure 13). .

이와 같은 결과에 따르면, 본 발명의 방법에 따라 제조된 89Zr-CD25 IgG는 혈청 내에서 안정성을 가지면서도, 종양 및 종양 내 침윤한 Treg에 대한 결합력뿐만 아니라 상기 결합력에 대한 특이도 역시 우수한 것으로 확인되었다. 또한, 이와 같은 특징을 통해 우수한 품질의 종양 특이적 PET 영상을 획득할 수 있다는 점이 확인되어, 난치성 림프종 등을 포함한 림프종의 진단용, 영상화용, 치료 효과 확인용 조성물 및 방법으로 다양하게 활용될 수 있음이 확인되었다. According to these results, it was confirmed that 89 Zr-CD25 IgG prepared according to the method of the present invention has stability in serum and has excellent binding ability to tumors and Tregs infiltrated within the tumor, as well as specificity for this binding ability. It has been done. In addition, it has been confirmed that excellent quality tumor-specific PET images can be obtained through these characteristics, and it can be used in a variety of ways as a composition and method for diagnosis, imaging, and confirmation of treatment effects for lymphomas, including intractable lymphomas. This has been confirmed.

전술한 본 발명의 설명은 예시를 위한 것이며, 본 발명이 속하는 기술분야의 통상의 지식을 가진 자는 본 발명의 기술적 사상이나 필수적인 특징을 변경하지 않고서 다른 구체적인 형태로 쉽게 변형이 가능하다는 것을 이해할 수 있을 것이다. 그러므로 이상에서 기술한 실시예들은 모든 면에서 예시적인 것이며 한정적이 아닌 것으로 이해해야만 한다.The description of the present invention described above is for illustrative purposes, and those skilled in the art will understand that the present invention can be easily modified into other specific forms without changing the technical idea or essential features of the present invention. will be. Therefore, the embodiments described above should be understood in all respects as illustrative and not restrictive.

본 발명은 89Zr 표지된 항-CD25 항체를 포함하는 PET 기반 촬영 방법의 영상화제, 및 이를 이용한 종양의 영상화 방법 또는 종양 내 침윤 조절 T 세포(tumor-infiltrating regulatory T cell)의 영상화 방법에 관한 것이다. 본 발명의 고유한 방법으로 제조된 89Zr 표지된 항-CD25 항체는 혈청 내 안정적으로 유지되면서도, CD25 항원이 존재하는 조직, 종양, 특히 림프종 또는 조절 T 세포가 다량 존재하는 부위에서 특이적으로 높은 섭취율을 나타내 현저히 우수한 결합 친화도 및 결합 특이도를 특징으로 한다. 따라서, 림프종, 특히, 난치성 및/또는 T세포 림프종의 진단; 또는 림프종 또는 림프종 내 침윤 조절 T 세포를 검출, 이미징, 및 모니터링에 사용할 수 있는 바, 효과적인 비침습적 의료 영상 촬영용 진단, 검출, 모니터링, 영상화제 등으로 유용하게 활용될 수 있을 것으로 기대된다.The present invention relates to an imaging agent for a PET-based imaging method comprising an 89 Zr-labeled anti-CD25 antibody, and a method for imaging a tumor or a method for imaging tumor-infiltrating regulatory T cells using the same. . The 89 Zr-labeled anti-CD25 antibody prepared by the unique method of the present invention is stably maintained in serum, and has a specific high concentration in tissues where CD25 antigen is present, tumors, especially lymphomas, or areas where a large number of regulatory T cells are present. It exhibits an uptake rate and is characterized by significantly excellent binding affinity and binding specificity. Accordingly, diagnosis of lymphoma, particularly refractory and/or T-cell lymphoma; Alternatively, it can be used for detecting, imaging, and monitoring lymphoma or infiltrating regulatory T cells within lymphoma, and is expected to be useful as a diagnostic, detection, monitoring, and imaging agent for effective non-invasive medical imaging.

Claims (13)

89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 조성물. 89 A composition for positron emission tomography imaging for tumor diagnosis, comprising a Zr-labeled anti-CD25 antibody as an active ingredient. 제1항에 있어서,According to paragraph 1, 상기 조성물은 종양 또는 종양 내 침윤 조절 T 세포(tumor-infiltrating regulatory T cell)를 검출하는 것을 특징으로 하는, 조성물. The composition is characterized in that it detects a tumor or tumor-infiltrating regulatory T cells. 제1항에 있어서, According to paragraph 1, 상기 종양은 림프종인 것을 특징으로 하는, 조성물.Composition, wherein the tumor is lymphoma. 제1항에 있어서,According to paragraph 1, 상기 조성물은 정맥주사용인 것을 특징으로 하는, 조성물.The composition is characterized in that it is for intravenous injection. 하기 단계를 포함하는 제1항의 89Zr 표지된 항-CD25 항체를 제조하는 방법:A method of producing the 89 Zr labeled anti-CD25 antibody of claim 1 comprising the following steps: (a) 항-CD25 항체를 TCEP(tris-carboxyethylphosphine)와 반응시켜 환원 시키는 단계; (a) reducing the anti-CD25 antibody by reacting it with TCEP (tris-carboxyethylphosphine); (b) 상기 (a) 단계에서 환원된 항-CD25 항체를 데페록사민-말레이미드(deferoxamine-maleimide, DFO-Mal)와 반응시켜, DFO-Mal 접합된 항-CD25 항체를 생성하는 단계; 및(b) reacting the anti-CD25 antibody reduced in step (a) with deferoxamine-maleimide (DFO-Mal) to produce a DFO-Mal conjugated anti-CD25 antibody; and (c) 상기 (b) 단계에서 생성된 DFO-Mal 접합된 항-CD25 항체를 89Zr-옥살레이트(89Zr-oxalate)와 반응시킨 후, 89Zr 표지된 항-CD25 항체를 수득하는 단계.(c) reacting the DFO-Mal conjugated anti-CD25 antibody produced in step (b) with 89 Zr-oxalate to obtain an 89 Zr labeled anti-CD25 antibody. 제5항에 있어서, According to clause 5, 상기 항체는 IgG인 것을 특징으로 하는, 방법.A method, characterized in that the antibody is IgG. 제5항에 있어서, According to clause 5, 상기 항체는 하기를 특징으로 하는 것인, 방법:A method wherein the antibody has the following characteristics: 항-CD25 항체 힌지 영역(hinge region)의 이황화 결합(disulfide bond) 부위에 특이적으로 89Zr이 결합됨; 및 89 Zr is specifically bound to the disulfide bond site of the anti-CD25 antibody hinge region; and 89Zr 및 항-CD25 항체의 접합체(conjugate) 형태임.It is a conjugate form of 89 Zr and anti-CD25 antibody. 하기 단계를 포함하는, 종양 진단을 위한 양전자 방출 단층 촬영 방법:A positron emission tomography method for tumor diagnosis comprising the following steps: (i) 89Zr 표지된 항-CD25 항체를 개체에 투여하는 단계; 및(i) administering 89 Zr labeled anti-CD25 antibody to the subject; and (ii) 상기 개체의 검출 이미지 또는 영상을 획득하는 단계.(ii) Obtaining a detection image or video of the object. 제8항에 있어서,According to clause 8, 상기 촬영은 PET(Positron Emission Tomography), PET/CT, PET/MR, 및 immuno-PET로 이루어진 군으로부터 선택되는 하나의 기술을 이용하는 것을 특징으로 하는, 방법.The method, wherein the imaging uses a technology selected from the group consisting of PET (Positron Emission Tomography), PET/CT, PET/MR, and immuno-PET. 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 조성물, 및 설명서를 포함하는 종양 진단을 위한 양전자 방출 단층 촬영 영상화용 키트. 89 A kit for positron emission tomography imaging for tumor diagnosis, comprising a composition comprising a Zr-labeled anti-CD25 antibody as an active ingredient, and instructions. 제10항에 있어서,According to clause 10, 상기 조성물은 제1항의 조성물이고, 상기 설명서는 제5항 및 제8항의 방법이 기재된 것을 특징으로 하는, 키트.The composition is the composition of claim 1, and the instructions are the methods of claims 5 and 8. A kit, characterized as described. 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 조성물의, 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화 용도. 89 Diagnosis of a tumor of a composition comprising a Zr-labeled anti-CD25 antibody as an active ingredient; or for the detection or imaging of tumors or infiltrating regulatory T cells within tumors. 종양의 진단; 또는 종양 또는 종양 내 침윤 조절 T 세포의 검출 또는 영상화제를 제조하기 위한, 89Zr 표지된 항-CD25 항체를 유효성분으로 포함하는 조성물의 용도. Diagnosis of tumors; or use of a composition comprising 89 Zr-labeled anti-CD25 antibody as an active ingredient for preparing an imaging agent or detection of tumor or intratumoral infiltrating regulatory T cells.
PCT/KR2023/002337 2022-08-11 2023-02-17 Immuno-pet using 89zr-labeled anti-cd25 antibodies Ceased WO2024034759A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0100554 2022-08-11
KR1020220100554A KR102759386B1 (en) 2022-08-11 2022-08-11 Immuno-PET using 89Zr labelled anti-CD25 antibody

Publications (1)

Publication Number Publication Date
WO2024034759A1 true WO2024034759A1 (en) 2024-02-15

Family

ID=89851893

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/002337 Ceased WO2024034759A1 (en) 2022-08-11 2023-02-17 Immuno-pet using 89zr-labeled anti-cd25 antibodies

Country Status (2)

Country Link
KR (1) KR102759386B1 (en)
WO (1) WO2024034759A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004110390A2 (en) * 2003-06-17 2004-12-23 Immunomedics, Inc. Anti-cd74 immunoconjugates and methods
WO2015153772A2 (en) * 2014-04-01 2015-10-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Zirconium-89 oxine complex as a cell labeling agent for positron emission tomography
US10905784B2 (en) * 2017-02-10 2021-02-02 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-LAG3 antibodies for immuno-PET imaging
WO2021113450A2 (en) * 2019-12-05 2021-06-10 Imaginab, Inc. Methods of imaging using multiple imaging agents
KR20220093103A (en) * 2019-09-13 2022-07-05 어드메어 테라퓨틱스 소사이어티 Anti-oncolytic virus antigen antibody and method of use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004110390A2 (en) * 2003-06-17 2004-12-23 Immunomedics, Inc. Anti-cd74 immunoconjugates and methods
WO2015153772A2 (en) * 2014-04-01 2015-10-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Zirconium-89 oxine complex as a cell labeling agent for positron emission tomography
US10905784B2 (en) * 2017-02-10 2021-02-02 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-LAG3 antibodies for immuno-PET imaging
KR20220093103A (en) * 2019-09-13 2022-07-05 어드메어 테라퓨틱스 소사이어티 Anti-oncolytic virus antigen antibody and method of use thereof
WO2021113450A2 (en) * 2019-12-05 2021-06-10 Imaginab, Inc. Methods of imaging using multiple imaging agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LEE, K. H.: "Site-specific 89Zr-CD25 and PD-1 IgG for Immuno-PET of Lymphomas", A3 FORESIGHT PROGRAM SYMPOSIUM 2022, 27 February 2022 (2022-02-27), pages 1 - 4 *

Also Published As

Publication number Publication date
KR20240022186A (en) 2024-02-20
KR102759386B1 (en) 2025-01-24

Similar Documents

Publication Publication Date Title
SK12162001A3 (en) Radiolabeling kit and binding assay
JPWO2017217347A1 (en) Site-specific RI-labeled antibody by IgG binding peptide
KR20230066119A (en) Hydrophilic antibody-drug conjugates
EP3783016A1 (en) Modified antibody and radioactive metal-labelled antibody
JP2014074036A (en) Methods of impairing hematologic cancer progenitor cells and compounds related thereto
US7781212B2 (en) Antitumor antibodies, proteins, and uses thereof
US5344919A (en) Integrin from human epithelial cells
JP2011219477A (en) Antitumor antibody, protein, and use thereof
WO2024034759A1 (en) Immuno-pet using 89zr-labeled anti-cd25 antibodies
US6252087B1 (en) Nitroaromatic compounds for the detection of hypoxia
WO2022244908A1 (en) Anti-bcam antibody or antigen-binding fragment thereof
JP3513173B2 (en) Monoclonal anti-ganglioside antibody, production thereof and therapeutic agent for tumor containing the same
CA1339709C (en) Monoclonal antibodies to human pancreatic cancer
WO2025211768A1 (en) Radioisotope-labeled anti-her2 antibody–chelator conjugate, pharmaceutical composition comprising same, and preparation method therefor
JP4575692B2 (en) LOX-1 diagnostic agent
US6855828B1 (en) Detection of hypoxia
AU2021361746A1 (en) Radioactive complexes of anti-her2 antibody, and radiopharmaceutical
Turhanen et al. A novel strategy for the synthesis of enzymatically stable biotin–DOTA conjugates for in vivo use
WO2019103531A1 (en) Anti-icam4 antibody and composition for diagnosis and treatment of icam4 expression cell-associated disease
JPH06509102A (en) Tissue-specific contrast agent using in-vivo contrast anti-idiotype antibody

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23852657

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 23852657

Country of ref document: EP

Kind code of ref document: A1