Composition for isolating monoclonal antibody against indoxyl sulfate and/or antigen-binding fragment thereof, and use thereof

文档序号:1131753 发布日期:2020-10-02 浏览:12次 中文

阅读说明:本技术 单离抗硫酸吲哚酚的单株抗体及/或其抗原结合片段的组成物及其使用 (Composition for isolating monoclonal antibody against indoxyl sulfate and/or antigen-binding fragment thereof, and use thereof ) 是由 庄詠钧 郑又玮 高誌晖 于 2019-04-18 设计创作,主要内容包括:本发明有关于一种专一性辨识蛋白结合型尿毒素─硫酸吲哚酚(IS)的单离单株抗体及/或其抗原结合片段,编码上述单离单株抗体及/或其抗原结合片段的单离聚核苷酸区段,利用上述单离抗IS的单株抗体及/或其抗原结合片段的免疫分析法、元件组成物及其用于诊断及治疗IS相关疾病的药学组成物。(The invention relates to an isolated monoclonal antibody and/or an antigen binding fragment thereof for specifically identifying protein-bound type uremic toxin-Indoxyl Sulfate (IS), an isolated polynucleotide segment for coding the isolated monoclonal antibody and/or the antigen binding fragment thereof, an immunoassay method and a component composition using the isolated anti-IS monoclonal antibody and/or the antigen binding fragment thereof, and a pharmaceutical composition for diagnosing and treating IS-related diseases.)

1. An isolated monoclonal antibody and/or an antigen-binding fragment thereof, comprising:

a heavy chain Variable (VH) domain comprising a plurality of Complementarity Determining Regions (CDR) CDR-H1, CDR-H2 and CDR-H3; and

a light chain Variable (VL) domain comprising a plurality of complementarity determining regions CDR-L1, CDR-L2 and CDR-L3, and

wherein the complementarity determining regions comprise one or more of the following amino acid sequences:

CDR-H1 is selected from the sequence identification numbers (SEQ ID NO): 1 or 11;

CDR-H2 is selected from SEQ ID NO:2 or 12;

CDR-H3 is selected from SEQ ID NO:3 or 13;

CDR-L1 is selected from SEQ ID NO:6 or 16;

CDR-L2 is selected from SEQ ID NO:7 or 17; and

CDR-L3 is selected from SEQ ID NO:8 or 18.

2. The isolated monoclonal antibody and/or antigen-binding fragment thereof of claim 1, wherein the isolated monoclonal antibody and/or antigen-binding fragment thereof is a full-length immunoglobulin molecule of an isolated human antibody, a chimeric antibody, a humanized antibody, a synthetic antibody, a recombinant antibody and/or antigen-binding fragment thereof, a single-chain variable region fragment, a Fab' fragment, a single domain antibody, a multispecific binding molecule, or any combination thereof.

3. The isolated monoclonal antibody and/or antigen-binding fragment thereof of claim 1, wherein the isolated monoclonal antibody and/or fragment thereof is selected from an integral outer surface presentation system.

4. The isolated monoclonal antibody and/or antigen binding fragment thereof of claim 1, wherein the isolated monoclonal antibody and/or fragment thereof is chemically modified or admixed with a pharmaceutically acceptable carrier.

5. An immunoassay for detecting indoxyl sulfate, comprising the isolated monoclonal antibody and/or antigen-binding fragment thereof according to claim 1, whereby a concentration of indoxyl sulfate in a biological sample collected from a subject is determined.

6. The immunoassay method for detecting indoxyl sulfate according to claim 5, wherein the biological sample comprises blood, serum, plasma, peritoneal fluid, urine and biopsy.

7. The immunoassay of claim 5, wherein the biological sample comprises albumin.

8. A kit for removing indoxyl sulfate, comprising the isolated monoclonal antibody and/or an antigen-binding fragment thereof according to claim 1.

9. The composition as claimed in claim 8, wherein the isolated monoclonal antibody and/or antigen binding fragment thereof is bound to a solid phase.

10. A pharmaceutical composition for removing or reducing indoxyl sulfate in a subject, wherein the pharmaceutical composition comprises the isolated monoclonal antibody and/or antigen-binding fragment thereof of claim 1.

11. The pharmaceutical composition for removing or reducing indoxyl sulfate according to claim 10, further comprising a pharmaceutical adsorbent bound to the isolated monoclonal antibody and/or antigen-binding fragment thereof.

12. The pharmaceutical composition for removing or reducing indoxyl sulfate according to claim 11, wherein the pharmaceutical adsorbent comprises an activated carbon absorbent.

13. An isolated polynucleotide segment encoding an isolated monoclonal antibody and/or antigen-binding fragment thereof directed against indoxyl sulfate, wherein the isolated monoclonal antibody and/or antigen-binding fragment thereof comprises one or more of the following polypeptide sequences:

as shown in SEQ ID NO:4 or 14, a VH domain; and

as shown in SEQ ID NO:9 or 19, and a VL domain.

14. An isolated polynucleotide segment encoding an isolated monoclonal antibody and/or antigen-binding fragment thereof directed against indoxyl sulfate, wherein the isolated monoclonal antibody and/or antigen-binding fragment thereof comprises one or more of the following polypeptide sequences:

CDR-H1 is selected from SEQ ID NO:1 or 11;

CDR-H2 is selected from SEQ ID NO:2 or 12;

CDR-H3 is selected from SEQ ID NO:3 or 13;

CDR-L1 is selected from SEQ ID NO:6 or 16;

CDR-L2 is selected from SEQ ID NO:7 or 17; and

CDR-L3 is selected from SEQ ID NO:8 or 18.

15. An isolated monoclonal antibody and/or antigen binding fragment thereof, wherein the isolated monoclonal antibody and/or antigen binding fragment thereof has an epitope and a scatchard binding affinity for indoxyl sulfate excluding L-tryptophan, indole, and 3-indoleacetic acid.

Technical Field

The present invention discloses an antigen binding fragment of uremic toxin-Indoxyl Sulfate (IS), and more particularly relates to an isolated antibody and/or an antigen binding fragment thereof that specifically recognizes IS and the use thereof for diagnosing and treating IS-related diseases.

Background

Uremia (uremia) is a disease caused by the pathological accumulation of various uretoxins in the body, and this debilitating condition can lead to permanent loss of kidney function, gradually entering the stage of Chronic Kidney Disease (CKD), and eventually becoming an end-stage renal disease (ESRD) that is life-threatening. Uremic patients may have various clinical signs and symptoms related to the nervous, digestive, circulatory, skeletal systems (1). Although uremia and its resulting complications are usually controlled by dialysis treatments, including hemodialysis and peritoneal dialysis, when a patient progresses to a state of renal failure, a serious electrolyte imbalance may occur, metabolic wastes have accumulated to a dangerous degree, and a kidney transplant is required for survival. That said, this treatment option is limited by donor kidney shortages, surgical-related mortality risks, and the patient's necessity to suppress immune function for the lifetime after surgery to avoid organ rejection.

Hundreds of potential uremic toxins have been identified. Uremic toxins can be divided into three main groups according to their physicochemical properties or behavior during dialysis: (a) small molecule, water-soluble and non-protein binding compounds (molecular weight <300D, e.g. urea); (b) a "medium-large molecule" compound, mainly peptides, having a molecular weight of 300D to 12,000D, and (c) a small molecule, protein-binding compound (2). Uremic toxins bind to serum proteins, such as albumin, and limit dialysis from removing compounds bound to these proteins. Thus, the average concentration of these toxins in a patient undergoing dialysis may gradually build up to 10-fold to 20-fold the normal concentration.

Indoxyl Sulfate (IS) and p-cresol (PCS) are two typical albumin-bound toxins, both of which are produced by fermentation of tryptophan and tyrosine in food by microorganisms in the gastrointestinal tract. Some groups have demonstrated that the concentration of IS and PCS in CKD patient serum IS pathologically related to total mortality and cardiovascular disease (CVD). Therefore, IS and PCS are not only biomarkers for predicting renal function and disease progression, but also targets for therapeutic procedures (3, 4).

Under normal conditions, uremic toxins can be excreted by urine. Uremic toxins can be divided into three categories, depending on their physiological properties and their reactivity towards dialysis: (1) small molecule, water-soluble and non-protein binding compounds such as urea and creatinine; (2) "mesoscopic molecule" compounds, such as peptides and β 2-microglobulin; (3) small molecule and protein binding compounds. Such as p-cresol, hippuric acid and Indoxyl Sulfate (IS). Binding to serum proteins such as albumin limits dialysis clearance of these protein-binding compounds in these uremic toxins, IS and its precursors (e.g., indoles) and derivatives have been reported to be highly negatively correlated with estimated glomerular filtration rates (egfr rs) in CKD patients, and IS found to be useful in diagnosing CKD and predicting prognostic assessments.

AST-120 [ Kremezin (Kremezin); wu Yu, Inc. (Kureha Corporation), Tokyo, Japan) is an orally administered spherical carbonaceous adsorbent that adsorbs various small-molecule uremic toxins. AST-120 adsorbs indole, a precursor of IS in the intestinal tract, thereby inhibiting IS synthesis and indirectly reducing IS concentrations in serum and urine (5). Although a number of prospective randomized clinical studies have demonstrated the efficacy of AST-120 in slowing CKD progression, the conclusion is that the benefit of adding AST-120 to standard treatment in moderate to severe CKD patients is not supported by these experimental data (6, 7).

IS IS detected and determined by analytical chemistry methods such as liquid chromatography-mass spectrometry (LC-MS) and high-performance liquid chromatography (HPLC), which has high equipment cost, complicated sample preparation, large sample volume and unsuitability for detecting large biological samples (8). On the contrary, Enzyme-linked immunosorbent assay (ELISA) is simple in operation and relatively inexpensive. Omitting complex pre-treatment of the sample, the ELISA protocol can be automated, thereby providing more accurate and repeatable results, faster processing time (turn around time), and lower labor cost. Star (Hoshi) et al developed a competitive ELISA method for quantitative determination of IS, but the monoclonal antibody strain 9A2F6 used in this method was inhibited by bovine or human serum albumin (9, 10). It may be particularly preferred or necessary to perform sample pretreatment such as deproteinization (deproteinization) prior to the performance of the experimental step, although this may reduce the standard for the applicability of the technique in the laboratory or industry.

In summary, despite the continuous advance of technology, there IS a continuing need to provide immunoassays for improved qualitative or quantitative analysis, in which isolation of anti-IS monoclonal antibodies (mabs) IS a necessary functional ingredient. At the same time, the immunoassay method can have the property of being insensitive to plasma proteins, in particular to albumin, in the biological sample to be analyzed. Secondly, there IS also a continuing need to provide methods for effectively removing IS from a subject, thereby treating an IS-associated disease. The methods comprise administering to the subject an effective amount of isolated anti-IS monoclonal antibody, or binding isolated anti-IS monoclonal antibody to a solid support, and contacting IS and isolated anti-IS monoclonal antibody with each other, thereby capturing or removing IS from the biological sample.

Disclosure of Invention

Accordingly, in one aspect, the invention provides an isolated monoclonal antibody and/or antigen-binding fragment thereof that specifically binds indoxyl sulfate (or mAb known as anti-IS).

In another aspect, the present invention provides a multispecific binding molecule comprising a portion or the entirety of the above-described isolated monoclonal antibody and/or antigen-binding fragment thereof.

In another aspect, the present invention provides an immunoassay for detecting indoxyl sulfate, whereby the concentration of indoxyl sulfate in a biological sample collected from a subject is determined.

In another aspect, the present invention provides a device composition for removing indoxyl sulfate.

Furthermore, in still another aspect, the present invention provides a pharmaceutical composition for removing or reducing indoxyl sulfate.

In another aspect, the present invention provides an isolated polynucleotide segment encoding the isolated monoclonal antibody and/or an antigen-binding fragment thereof.

In another aspect, the present invention provides an isolated antibody and/or antigen-binding fragment thereof having an epitope (epitope) and Scatchard binding affinity (Scatchard binding affinity) for indoxyl sulfate, excluding L-tryptophan, indole, and 3-indoleacetic acid.

According to the above aspect of the present invention, an isolated monoclonal antibody and/or an antigen-binding fragment thereof is provided. The isolated monoclonal antibody and/or antigen-binding fragment thereof may comprise a heavy chain Variable (VH) domain and a light chain Variable (VL) domain. The VH domain comprises a plurality of Complementarity Determining Regions (CDRs) CDR-H1, CDR-H2 and CDR-H3, and the VL domain comprises a plurality of complementarity determining regions CDR-L1, CDR-L2 and CDR-L3. In one embodiment, the complementarity determining region comprises one or more of the following amino acid sequences: CDR-H1 is selected from the sequence identification numbers (SEQ ID NO): 1 or 11; CDR-H2 is selected from SEQ ID NO:2 or 12; CDR-H3 is selected from SEQ ID NO:3 or 13; CDR-L1 is selected from SEQ ID NO:6 or 16; CDR-L2 is selected from SEQ ID NO:7 or 17; and CDR-L3 is selected from SEQ ID NO:8 or 18.

In one embodiment, the isolated antibody and/or the antibody binding fragment thereof can be a full-length immunoglobulin molecule, a single-chain variable fragment (scFv), a variable fragment (Fv), a Fab fragment, a Fab' fragment, a single domain antibody (dAb), a multispecific binding molecule, or any combination thereof of an isolated human antibody, a chimeric antibody, a humanized antibody, a synthetic antibody, a recombinant antibody, and/or the antibody binding fragment thereof.

In one embodiment, the isolated monoclonal antibodies and/or fragments thereof can be screened by an in vitro surface presentation system.

In one embodiment, the isolated monoclonal antibody and/or fragment thereof can be chemically modified or mixed with a pharmaceutically acceptable carrier.

According to another aspect of the present invention, there is provided a multispecific binding molecule comprising a portion or the entirety of an isolated monoclonal antibody and/or an antigen-binding fragment thereof as described above.

In one embodiment, the multispecific binding molecule specifically binds indoxyl sulfate.

According to another aspect of the present invention, an immunoassay for detecting indoxyl sulfate comprises the isolated monoclonal antibody and/or an antigen-binding fragment thereof.

According to another aspect of the present invention, an immunoassay for detecting indoxyl sulfate-associated diseases is provided, which comprises the above isolated monoclonal antibody and/or antigen-binding fragment thereof, thereby determining the concentration of indoxyl sulfate in a biological sample collected from a subject.

In one embodiment, the biological sample includes blood, serum, plasma, peritoneal fluid, urine, and biopsy (biopsy specmen).

In one embodiment, the biological sample includes albumin.

According to another aspect of the present invention, a composition of matter for removing indoxyl sulfate comprises the isolated monoclonal antibody and/or an antigen-binding fragment thereof.

In one embodiment, the isolated monoclonal antibody and/or antigen binding fragment thereof is bound to a solid phase.

According to another aspect of the present invention, a pharmaceutical composition for removing or reducing indoxyl sulfate in a subject is provided, which comprises the isolated monoclonal antibody and/or the antigen-binding fragment thereof.

In one embodiment, the pharmaceutical composition further comprises a pharmaceutical adsorbent bound to the isolated monoclonal antibody and/or antigen binding fragment thereof.

In one embodiment, the medical adsorbent includes an activated carbon absorbent.

According to other aspects of the present invention, an isolated polynucleotide segment (segment) is provided, wherein the isolated polynucleotide segment encodes an isolated antibody and/or antigen-binding fragment thereof against indoxyl sulfate, and the isolated antibody and/or antigen-binding fragment thereof comprises one or more polypeptide sequences of a VH domain and a VL domain. In one embodiment, the VH domain can be as set forth in SEQ id no:4 or 14, and the VL domain may be as shown in SEQ ID NO:9 or 19.

According to another aspect of the present invention, an isolated polynucleotide segment is provided, wherein the isolated polynucleotide segment encodes an isolated monoclonal antibody against indoxyl sulfate and/or an antigen-binding fragment thereof. In one embodiment, the isolated monoclonal antibody and/or antigen binding fragment thereof comprises one or more of the following polypeptide sequences: CDR-H1 is selected from SEQ ID NO:1 or 11; CDR-H2 is selected from SEQ ID NO:2 or 12; CDR-H3 is selected from SEQ ID NO:3 or 13; CDR-L1 is selected from SEQ ID NO:6 or 16; CDR-L2 is selected from SEQ ID NO:7 or 17; and CDR-L3 is selected from SEQ ID NO:8 or 18.

According to another aspect of the present invention, an isolated monoclonal antibody and/or an antigen-binding fragment thereof is provided. In one embodiment, the isolated monoclonal antibody and/or antigen binding fragment thereof has an antigenic epitope (epitope) and a Scatchard binding affinity (Scatchard binding affinity) for indoxyl sulfate but excludes L-tryptophan, indole, and 3-indoleacetic acid.

The antibody and/or fragment thereof specifically recognizing indoxyl sulfate according to the present invention can be used in various applications, such as immunoassay for indoxyl sulfate detection, a composition for removing indoxyl sulfate, a pharmaceutical composition for removing or reducing indoxyl sulfate from a subject, an isolated polynucleotide segment, and an antibody and/or an antigen-binding fragment thereof having an epitope and a scatchard binding affinity for indoxyl sulfate, but excluding L-tryptophan, indole, and 3-indoleacetic acid.

Drawings

The foregoing and other objects, features, and advantages of the invention will be apparent from the following more particular description of the embodiments of the invention, as illustrated in the accompanying drawings in which:

FIGS. 1A and 1B are graphs showing titration curves of several isolated indoxyl sulfate-resistant monoclonal antibodies LG303 (FIG. 1A) and LG332 (FIG. 1B) against indoxyl sulfate-bovine serum albumin (IS-BSA) according to one embodiment of the present invention.

FIGS. 2A-2D are B/B% bar graphs showing several isolated indoxyl sulfate-resistant monoclonal antibodies LG303 and LG332, respectively, in the presence of different concentrations of indoxyl sulfate (FIG. 2A), L-tryptophan (L-tryptophan; FIG. 2B), indole (FIG. 2C), and 3-indoleacetic acid (3-indoleacetic acid; FIG. 2D), in accordance with one embodiment of the present invention.

FIG. 3 is a standard graph showing the blank test limit (limit of blank, LoB) and the detection limit (limit of detection, LoD) of the monoclonal antibody LG332 isolated against indoxyl sulfate by using a competitive ELISA method at sequential indoxyl sulfate concentration according to another embodiment of the present invention.

FIG. 4 is a flowchart illustrating an experimental strategy for animal models of chronic kidney disease, according to one embodiment of the present invention.

Fig. 5A and 5B are images of tissue sections of a kidney (fig. 5A) and a heart (fig. 5B) of a test animal stained with hematoxylin and eosin (H & E) according to an embodiment of the present invention.

FIG. 6 is a tissue section image showing the collagen detection of kidney and heart sections of a test animal stained with sirius red (Siriusred) according to an embodiment of the present invention.

FIG. 7 is an Immunohistochemical (IHC) image of kidney and heart sections of experimental animals using type IV collagen antibody according to one embodiment of the present invention.

FIG. 8 is an IHC image showing the detection of fibroplasia areas in kidney and heart sections of experimental animals of one embodiment of the present invention by alpha-smooth muscle actin (alpha-SMA) antibodies.

FIG. 9 is an IHC image showing the detection of apoptotic regions by M30 antibody on kidney and heart sections of experimental animals according to one embodiment of the present invention.

Detailed Description

The following detailed description of the embodiments of the invention will be best understood when read in conjunction with the accompanying drawings, where like elements are indicated with like reference numerals.

All documents cited herein are hereby incorporated by reference to the same extent as if each individual document or patent application was specifically and individually indicated to be incorporated by reference. If a definition or use of a term in a reference, which is inconsistent or contrary to the definition of that term provided herein, applies the definition of that term provided herein and does not apply to the definition of that term provided herein.

For the purpose of interpreting the specification, the following definitions will apply, where appropriate, to singular nouns also including plural, and vice versa. Additional definitions are set forth throughout the detailed description.

Unless the context is otherwise appropriate, the terms "a" and "an" and "the" are defined as "one or more" and include the plural.

The invention discloses a method for diagnosing and treating Chronic Kidney Disease (CKD) patients with uremia and removing the same by using an isolated antibody of anti-protein-bound uremic toxin-Indoxyl Sulfate (IS) and/or an antigen-binding fragment thereof. For diagnostic use, the present invention provides an immunoassay for the simultaneous qualitative and quantitative detection of IS in a biological sample. To effectively remove circulating IS from a subject, an effective amount of isolated anti-IS monoclonal antibody can be administered to the subject, or a biological sample can be passed over a solid support that binds isolated anti-IS monoclonal antibody to adsorb IS. The biological sample may be blood, serum, plasma, peritoneal fluid, urine, biological specimen (biopsy specimen), or the like. Since the above biological samples collected from the subjects often contain various plasma proteins in normal or abnormal physiological states, the presentation of the binding activity of the above isolated anti-IS monoclonal antibodies should not be interfered with by any plasma proteins, especially albumin, which IS the most common. Although IS believed to exacerbate CKD and some other renal pathologies, the application of the present invention IS not limited to renal pathologies.

The term "subject" as used herein includes a human or non-human animal. Non-human animals include all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dogs, cattle, chickens, amphibians, and reptiles. Unless otherwise indicated, the "patient" or "subject" as described herein are interchangeable.

The term "nucleic acid" as used herein, interchangeably with "polynucleotide" refers to deoxyribonucleic acid or ribonucleic acid, and polymers thereof, in either single-or double-stranded form. The term nucleic acid includes nucleic acids containing known nucleotide analogs or modified backbone residues or linkers, which may be synthetic, naturally occurring, and non-naturally occurring, which bind in a manner similar to that of a reference nucleic acid, and which are metabolized in a manner similar to that of a reference nucleotide. Specific examples of the aforementioned analogs may include, but are not limited to, phosphorothioates, phosphoramidates, methylphosphonates, optically active-methylphosphonates, 2-O-methylribonucleotides, peptide-nucleic acids (PNAs).

Unless otherwise indicated, a particular nucleic acid sequence also implies conservatively modified variants thereof (e.g., degenerate codon substitutions), complementary sequences and the sequence explicitly indicated. In addition, degenerate codon substitutions may be made by substituting the third position of a selected codon or codons with mixed base and/or deoxyinosine residues.

The term "polypeptide" as used herein, interchangeably with "protein" refers to a polymer of amino acid residues. The term "polypeptide" as applied to amino acid polymers, wherein one or more amino acid residues can be artificial chemical mimetics of corresponding naturally occurring amino acids, naturally occurring amino acid polymers, and non-naturally occurring amino acid polymers. Unless otherwise indicated, conservatively modified variants thereof are also implied by a particular polypeptide sequence.

The term "antibody" refers to an intact immunoglobulin molecule or a functional fragment thereof. Naturally occurring antibodies generally comprise tetramers (tetramers), typically consisting of at least a double (H) chain and at least a di-light (L) chain. Each heavy (H) chain comprises a heavy chain Variable (VH) domain and a heavy chain Constant (CH) domain, wherein the heavy chain constant domain comprises three constant domains of CH1, CH2, and CH 3. A light chain variable (hereinafter abbreviated as VL) domain. The heavy chain may be of any isotype (isotype), including IgG (IgG1, IgG2, IgG3 and IgG4 subtypes), IgA (IgA1 and IgA2 subtypes), IgM and IgE. Each light chain comprises a light chain Variable (VL) domain and a light chain Constant (CL) domain. Light chains include kappa (. kappa.) chains and lambda (. lamda.) chains. The binding of the VH and VL domains is generally responsible for antigen recognition, while the CH domain mediates immunoglobulin binding to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the 1 st complement component of the complement system of the classical pathway (C1 q). The VH and VL domains can be subdivided into highly variable (hypervariability) regions, also known as Complementarity Determining Regions (CDRs), interspersed with more conserved antibody Framework Regions (FRs). Each VH and VL domain is composed of three CDRs and four FRs, which are arranged in the following order from N-terminus to C-terminus: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR 4. The heavy and light chain variable regions contain binding domains for interaction with an antigen.

The term "antibody fragment" or "specific binding fragment thereof" refers to the complete structure or a portion of an antibody, and can include Fab fragments, Fab ', F (ab') 2 fragments, Fd fragments, Fv fragments, and disulfide-linked Fv fragments; single chain variable fragment (scFv), single chain variable fragment dimer [ (scFv)2, also known as diabodies ], single chain variable fragment trimer [ (scFv)3, also known as triabodies, single chain variable fragment tetramer [ (scFv)4, also known as tetravalent antibodies (tetrabodies); single domain antibodies (dabs), minibodies (minibodies), nanobodies (nanobodies), and multispecific antibodies formed from antibody fragments. Next, the term "antibody fragment" or "specific binding fragment thereof" may also refer to a monoclonal antibody, a polyclonal antibody, a multispecific antibody (e.g., bispecific antibody) formed from at least two antibody fragments, a human antibody, a humanized antibody, a camelized (camelized) antibody, a chimeric antibody, or a murine antibody.

The term "Antibody Drug Complex (ADC)" or "immune complex" as used herein refers to an antibody or antigen-binding fragment thereof linked to another agent, such as a chemotherapeutic agent, a toxin, an immunotherapeutic agent, an imaging probe (imaging probe), and the like. The attachment means may be covalent bonds, or non-covalent interactions, for example by electrostatic forces. Various linking groups known in the art may be used to form antibody drug complexes. In addition, the antibody drug complex may be in the form of a fusion protein expressed from a polynucleotide encoding an immune complex. The term "fusion protein" as used herein refers to a protein produced by linking two or more genes or gene fragments, each of which originally encodes a different protein (including a peptide and a polypeptide). The fusion gene is translated to produce a single protein with multiple functional properties derived from the original protein.

The term "isolated" refers to an immunoglobulin, antibody or polypeptide, optionally present in a physical environment, but distinct from that found in nature.

The term "epitope" or "antigenic determinant" refers to the portion of an antigenic molecule that is responsible for specific interaction with the antigen-binding site of an antibody. The antigen binding site may be provided by one or more antibody variable domains. Functional antigen-binding fragments can be obtained by separating the VH and VL domains of a monoclonal antibody (mAb) and performing multiple recombinations and screenings.

The term "Complementary Determining Regions (CDRs)" is defined as portions of the variable chain of an antibody that bind to its specific antigen. In the present disclosure, the CDRs of the heavy chain are generally referred to as CDR-H1, CDR-H2 and CDR-H3, and the CDRs of the light chain are generally referred to as CDR-L1, CDR-L2 and CDR-L3. The CDRs are numbered in sequence from the N end to the C end.

The monoclonal antibody or antigen binding fragment thereof herein refers to a protein or peptide produced from a cDNA-derived source, a recombinant RNA-derived source, or any synthetic source, or a source that binds to any of the foregoing, and also includes proteins or peptides produced from any one of the following sources or derived sources: (1) unrelated to proteins found in nature; (2) other proteins not containing the same source, e.g., not containing murine proteins; (3) expressed by cells of different species; or (4) one not present in nature.

The term "anti-indoxyl sulfate monoclonal antibody (anti-IS mAb)" as used herein refers to a generic term for the type of antibody and/or fragment thereof that specifically binds indoxyl sulfate.

Furthermore, the present invention also provides methods for producing isolated anti-IS monoclonal antibodies and/or fragments thereof. The isolated anti-IS monoclonal antibodies are exemplified by two monoclonal antibodies LG303 and LG332, which are named according to the names of antibody-producing hybridoma cell lines.

The present invention also provides a polynucleotide sequence encoding the variable domain of the anti-IS monoclonal antibody, which comprises a heavy chain Variable (VH) domain and a light chain Variable (VL) domain. For example, the VH domains of antibodies LG303 and LG332 are respectively the sequence identification numbers (SEQ ID NOs): 1 and 11, and VL domains of antibodies LG303 and LG332 are SEQ ID NOs: 6 and 16.

The invention also provides amino acid sequences translated from the above-described polynucleotide sequences. For example, the VH domains of antibodies LG303 and LG332 are respectively the sequence identification numbers (SEQ ID NOs): 2 and 12, and VL domains of antibodies LG303 and LG332 are SEQ ID NOs: 7 and 17. The complementarity determining regions CDR-H1, CDR-H2 and CDR-H3 in the VH domain and CDR-L1, CDR-L2 and CDR-L3 in the VL domain of the above antibodies were also confirmed.

In one embodiment, the amino acid sequences of the CDRs corresponding to antibodies LG303 and LG332 are as follows: CDR-H1 is selected from the sequence identification numbers (SEQ ID NO): 1 or 11; CDR-H2 is selected from SEQ ID NO:2 or 12; CDR-H3 is selected from SEQ ID NO:3 or 13; CDR-L1 is selected from SEQ ID NO:6 or 16; CDR-L2 is selected from SEQ ID NO:7 or 17; and CDR-L3 is selected from SEQ ID NO:8 or 18.

The term "percent (%) sequence similarity" or "homology" is defined as the percentage of amino acid residues or nucleotides in a candidate sequence that are identical to a reference sequence, after aligning the candidate sequence with the reference sequence and introducing gaps therebetween, if necessary, to achieve an upper limit on the percent sequence similarity and to exclude nucleic acid conservative substitutions. In addition to manual methods, better sequence alignments can be generated by well-known regional similarity algorithms or computer programs (e.g., BLAST P) using these algorithms.

Furthermore, the above-mentioned nucleic acid sequences and amino acid sequences are listed in the attached sequence listing.

The present invention also provides a method for using the isolated anti-indoxyl sulfate monoclonal antibody as a reagent for diagnosing indoxyl sulfate-related diseases.

In addition, the invention also provides a method for using the isolated monoclonal antibody against indoxyl sulfate as a reagent for treating indoxyl sulfate related diseases.

The present invention also provides a method of using the isolated monoclonal antibody against indoxyl sulfate as an IS-substituting reagent for biological fluids to be cleaned in vitro.

Furthermore, the invention also provides a method for using the isolated monoclonal antibody against indoxyl sulfate in immunoassay and a device composition (device composition).

In addition, the invention also provides methods for developing the above immunoassays, device compositions, and pharmaceutical compositions.

In addition, the invention also provides an immunoassay comprising the isolated anti-indoxyl sulfate monoclonal antibody, so as to qualitatively and quantitatively detect indoxyl sulfate in a biological sample.

Also, the present invention provides a method for diagnosing indoxyl sulfate-related diseases by using the above immunoassay.

Furthermore, the invention also provides a device composition comprising the isolated anti-indoxyl sulfate monoclonal antibody, so that indoxyl sulfate in a body fluid of a subject is removed in vitro or the uremic burden is reduced.

In addition, the invention also provides a method for removing indoxyl sulfate or reducing the uremic burden, which is a pharmaceutical composition containing an effective dose of the isolated anti-indoxyl sulfate monoclonal antibody.

In one embodiment, when the isolated monoclonal antibody against indoxyl sulfate is used in an immunoassay or a device composition, the isolated monoclonal antibody against indoxyl sulfate can be bound to a solid phase as a solid support.

In one embodiment, the pharmaceutical composition described above can be administered to reduce the risk of renal failure or CVD associated with CVD in a subject in need thereof.

In one embodiment, the pharmaceutical composition may be combined with other drugs, such as oral administration spherical carbon absorbents, angiotensin-converting enzyme inhibitors (ACEIs), Angiotensin Receptor Blockers (ARBs), and active vitamin D.

The "element composition" as used herein refers to a composition for implementation in a specific apparatus. In one embodiment, the above-described compositions of elements can be integrated into a hemodialysis (hemidialysis) or plasmapheresis (plasmapheresis) system to reduce the risk of renal failure or CVD in a subject with a need for CVD.

In one embodiment, the above-described component composition may be combined with a device for extracorporeal cleaning of blood, wherein the blood may be divided into a first cleaning portion and a second cleaning portion. After removal of protein-bound toxins and/or toxins dissolved in the plasma, a second, clean fraction is produced.

In one embodiment, the biological sample (or liquid) to be analyzed includes blood, serum, plasma, peritoneal fluid, urine, and biopsy (biopsy specimen).

In one embodiment, the biological sample includes albumin.

While the invention has been described with reference to specific exemplary embodiments, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention.

Compositions specifically binding Indoxyl Sulfate (IS)

This example provides binding molecules, which can be isolated monoclonal antibodies and/or antigen-binding fragments thereof (hereinafter also referred to as anti-IS monoclonal antibodies or anti-IS mabs), that specifically recognize/bind Indoxyl Sulfate (IS). anti-IS monoclonal antibodies comprise a heavy chain Variable (VH) domain comprising a plurality of Complementarity Determining Regions (CDRs) CDR-H1, CDR-H2 and CDR-H3, and a light chain Variable (VL) domain comprising a plurality of complementarity determining regions CDR-L1, CDR-L2 and CDR-L3. The Complementarity Determining Regions (CDRs) comprise one or more of the following amino acid sequences: CDR-H1 is selected from the sequence identification numbers (SEQ ID NOs): 1 or 11; CDR-H2 is selected from SEQ ID NO:2 or 12; CDR-H3 is selected from SEQ ID NO:3 or 13; CDR-L1 is selected from SEQ ID NO:6 or 16; CDR-L2 is selected from SEQ ID NO:7 or 17; and CDR-L3 is selected from SEQ ID NO:8 or 18.

In some embodiments, the amino acid sequence of the VH domain of the anti-IS monoclonal antibody can be SEQ ID NO:4 or 14, or a variant of SEQ ID NO:4 or 14 (identity) represents at least 80%, 90%, 95%, or 99% (or any percentage between the above values). Furthermore, the amino acid sequence of the VL domain of the anti-IS monoclonal antibody may be seq id NO:9 or 19, or a variant of SEQ ID NO:9 or 19 (or any percentage therebetween) exhibits at least 80%, 90%, 95%, or 99% (or any percentage therebetween).

Immunoassay and clinical diagnosis for detecting and quantifying IS

The term "immunoassay" refers to a method of detecting an analyte in a sample that involves contacting the sample with an antibody that specifically binds to the analyte, and detecting the binding of the antibody to the analyte. The immunoassay described herein can be applied to various forms of immunological or biochemical diagnostic techniques using the antibody-antigen reaction caused by the aforementioned monoclonal antibody. The aforementioned analytical methods include, but are not limited to, radioimmunoassay (radioimmunoassay), immunohistochemistry assay (immunohistochemistry assay), in situ hybridization assay (in situ hybridization assay), competitive-binding assay (competitive-binding assay), Western Blot analysis (Western Blot assays), and ELISA assay. Then, the assay technique comprises (a) labeling the monoclonal antibody with a fluorescent antibody or a chemical staining method using a dye (e.g., a fluorescent dye) so that the binding of the monoclonal antibody to the antigen is observable by the naked eye; (b) enzyme is used to replace fluorescent dye, and the enzyme-antibody method is used for marking the monoclonal antibody; (c) performing ELISA method by using protein-labeled secondary antibody to measure the content of antigen; (d) radioimmunoassay of isotope-labeled monoclonal antibodies; and (e) immunoprecipitation using an agglutination reaction caused by an antibody-antigen reaction.

The invention relates to an immunoassay method for qualitatively and quantitatively detecting IS of a biological sample, which comprises the steps of judging normal and abnormal levels. In some embodiments, the biological sample may be blood, serum, plasma, peritoneal fluid, urine, a biological specimen sample, or the like. Secondly, the biological sample to be analyzed can still be analyzed in the presence of plasma proteins, i.e. albumin. In yet another embodiment, the immunoassay provided herein comprises the isolated anti-IS monoclonal antibody described above, which can be used to diagnose an abnormality or disease, as assessed by the concentration of a biological sample collected from the subject. One embodiment of the aforementioned pathophysiological condition or clinical symptom can be uremia or CKD.

IS-removing element composition

According to an embodiment of the present disclosure, a device composition for removing or reducing IS provided. In some embodiments, the isolated anti-IS monoclonal antibody IS attached to a solid phase, such as agarose, glass, plastic or magnetic resin beads, dialysis membranes and hollow fiber tubes, as a solid support. Second, the dialysis membrane or hollow fiber tube described above linked to the isolated monoclonal antibody to anti-IS can be integrated into a system for hemodialysis or plasmapheresis (plasmapheresis).

Methods for removing albumin-bound uremic toxins from a patient's blood are described in U.S. patent publication No. (u.s.pat. No.) 8,206,591B 2. The method comprises introducing a replacement substance (displastissubstance) into the blood, such that the replacement substance replaces the albumin-bound uremic toxin. Subsequently, unbound uremic toxins are removed using extracorporeal renal replacement therapy (extracorporal renal replacement) before the blood is returned to the patient. The substitution substance may be, for example, the isolated anti-IS monoclonal antibody described above. The method suggests that other drugs, i.e., oral spherical carbon absorbents, angiotensin-converting enzyme inhibitors (ACEIs), Angiotensin Receptor Blockers (ARBs) and active vitamin D, may be used in combination.

In yet another embodiment, isolated anti-IS monoclonal antibodies can be subjected to a reductive amination reaction (reductive amination reaction) to attach to agarose beads, and the monoclonal antibody-attached beads are packed into an empty column to form an IS removal column. The biological fluid was run through the IS removal column twice using a peristaltic pump (peristatic pump). Untreated and treated plasma samples are analyzed using a quantitative mass spectrometer, such as a liquid chromatography tandem mass spectrometer (LC-MS/MS), to assess IS removal. The percentage of IS removal (%) was calculated using the formula ([ IS of the untreated group ] - [ IS of the treated group ])/[ IS of the untreated group ] × 100%.

Pharmaceutical composition for removing or reducing IS in subject

According to one embodiment of the present disclosure, a pharmaceutical composition for removing or reducing IS in a subject IS provided. Dosages and therapies for administering anti-IS monoclonal antibodies can be readily determined by those of ordinary skill in the clinical art of treating IS-related disorders. Administration of a predetermined dose in one or more applications may achieve a predetermined result. According to embodiments of the present invention, pharmaceutical compositions in unit dosage form may be provided. The pharmaceutical composition can be administered in any suitable pharmaceutical carrier. The pharmaceutical composition can be administered in any form to prevent, alleviate, avoid or treat uremia in a human or animal patient. As applied to this example, the isolated anti-IS monoclonal antibody can be provided in the form of a pharmaceutical composition comprising a pharmaceutically acceptable carrier or diluent and the active ingredient IS the isolated anti-IS monoclonal antibody and/or binding fragment thereof. The compositions comprise isolated anti-IS monoclonal antibodies in an amount sufficient to fully or partially antagonize IS cytotoxicity, or to antagonize IS's natural binding to uremic cellular biological receptors, such as anion transporters (OATs) (10) or aromatic hydrocarbon receptors (AhRs) (11), in patients in need of such antagonism. The pharmaceutical composition comprises at least one of the isolated anti-IS monoclonal antibodies described above. Secondly, the pharmaceutical composition can be used in combination with other drugs, i.e., oral spherical carbon absorbent, angiotensin-converting enzyme inhibitors (ACEIs), Angiotensin Receptor Blockers (ARBs) and active vitamin D. In some embodiments, the drug may be, for example, an activated carbon absorbent, such as AST-120.

Polynucleotide and polypeptide sequences for isolating anti-IS monoclonal antibody variable regions

According to one embodiment of the present disclosure, a polynucleotide sequence or segment (segment) encoding the isolated anti-IS monoclonal antibody and/or antigen-binding fragment described above IS provided. In some embodiments, the nucleic acid sequence encoding SEQ ID NO:4 may be SEQ ID NO:5, or a sequence identical to SEQ ID NO:5 exhibits at least 80%, 90%, 95%, or 99% (or any percentage between the above values) of any sequence. Meanwhile, the above-mentioned code SEQ ID NO:9 can be SEQ ID NO:10, or a sequence identical to SEQ ID NO:10 exhibits at least 80%, 90%, 95%, or 99% (or any percentage between the above values) of any sequence.

Alternatively, the nucleic acid sequence described above encoding SEQ ID NO: the polynucleotide sequence of the VH domain of 14 may be SEQ ID NO:15, or a sequence identical to SEQ ID NO:15 exhibits at least 80%, 90%, 95%, or 99% (or any percentage between the above values) of any sequence. Meanwhile, the above-mentioned code SEQ ID NO:19 the polynucleotide sequence of the VL domain of SEQ ID NO:20, or a sequence identical to SEQ ID NO:20 exhibits at least 80%, 90%, 95%, or 99% (or any percentage between the above values) of any sequence.

In this embodiment, the polynucleotide sequence comprises a polypeptide substantially identical to the polypeptide encoding the amino acid sequence. Substantially equivalent sequences include mutated sequences, including sequences containing silent mutations (silent mutations). The mutation may comprise a variation of one or more nucleotide residues, a deletion of one or more nucleotide residues, or an insertion of one or more additional nucleotide residues. Substantially equivalent sequences are also included in the above-disclosed amino acid sequences, based on the degeneracy of the nucleic acid code (degeneration), as are various nucleotide sequences that encode the same amino acid at a predetermined amino acid position. The polynucleotide sequences encoding isolated anti-IS monoclonal antibodies and/or antigen-binding fragments thereof can be obtained by any conventional method. For example, if the nucleotide sequence of an antibody is known, the polynucleotide sequence encoding the antibody may be composed of a combination of chemically synthesized oligonucleotide sequences. The oligonucleotide sequences may involve, for example, synthesizing overlapping oligonucleotide sequences comprising portions encoding the antibody sequences, adhesively ligating the oligonucleotide sequences, and then amplifying the ligated oligonucleotide sequences using PCR (12).

In summary, the polynucleotide sequences encoding the isolated anti-IS monoclonal antibodies and/or antigen binding fragments thereof described above can be recombined into expression vectors as fragments of an open reading frame (open reading frame). In some applications, the expression vector can be introduced into a subject or host cell (e.g., E.coli BL21 from prokaryotic cells, CHO cells from eukaryotic cells) to produce a protein product that specifically recognizes IS, as provided in the preceding examples. In particular, the polynucleotide sequence segments of this embodiment are recombinant DNA or RNA, and in some cases, these segments may be separated by non-coding regulatory elements (introns) or introns. Preferably, the isolated nucleic acid may be a cDNA molecule. To generate the scFv genes, the DNA fragments encoding VH and VL may be operably linked to another fragment, which may encode a flexible linker, for example encoding the amino acid sequence (Gly4-Ser)3 (SEQ ID NO: 21), rendering the VH and VL sequence listings as a contiguous single-chain protein in which the VH and VL regions are linked by a flexible linker. For antibodies obtained using phage display technologies, phage strains carrying isolated nucleic acids isolated anti-IS monoclonal antibodies and/or antigen binding fragments thereof as provided in the preceding examples can be recovered by screening phage display libraries (library) (13). The polynucleotides disclosed above may also be produced from any other suitable source of nucleic acid, such as an antibody cDNA library, or a cDNA library isolated from any tissue or cell expressing the antibody (e.g., from a hybridoma cell selected to express an antibody).

In some embodiments, any of the above disclosed polynucleotide sequences may be inserted into an expression vector and expressed under the control of a predetermined promoter. Well known vectors suitable for expression in human and animal cell types are well known in the art. Suitable host cells may include, but are not limited to, HEK293, CHO, yeast (Pichia), SF9, e.coli BL21, and other human or non-human cell lines. In some embodiments, the nucleic acid of the expression vector IS expressed transiently or stably upon culturing of the cells, and methods of producing monoclonal antibodies and/or antigen-binding fragments against IS are disclosed herein.

5/6 model of Chronic Kidney Disease (CKD) in rats following nephrectomy (5/6Nx) and treatment

Typical animal models of CKD are Unilateral Ureteral Obstruction (UUO), an erythromycin (Adriamycin, ADR) nephropathy, and 5/6 nephrectomy. The pattern of UUO rapidly and completely occludes the tubules and presents features of different pathological stages and fibrosis, including inflammatory cell infiltration, tubular apoptosis, myofibroblast (fibroblast) activation, fibronectin and type I collagen deposition. The pattern of ADR nephropathy is currently commonly used to mimic human glomerular disease, characterized by massive proteinuria (proteinuria), tubular columns (renal tubular cassettes), tubular collapse (renal tubular collagen), focal segmental glomerulosclerosis (FSGS; also known as progressive local segmental sclerosis (progressive focal segmental sclerosis) and glomerulosclerosis, severe interstitial and inflammatory formation of the kidney. 5/6 the pattern of nephrectomy is characterized by arterial hypertension, proteinuria and glomerulosclerosis. Among the possible experimental models described above, 5/6 nephrectomized rats are most commonly used to study progressive renal disease because of the features of this experimental approach in common with the observed CKD in humans. 5/6 nephrectomy has also been established to test new treatment modalities and has been shown to be clinically relevant (14, 15).

Tubulointerstitial fibrosis (tubulointerstitial fibrosis) is often pathologically associated with CKD and presents common features including persistent inflammatory cell infiltration; the number of mesenchymal cells increases with the appearance of the myofibroblast-expressed cytoskeletal protein alpha-smooth muscle actin (alpha-SMA); tubular atrophy and epithelial apoptosis; the microvasculature (peritubular capillaries) surrounding the renal tubules is completely damaged; and accumulation of extracellular matrix (ECM) (16). In normal kidney, different collagen molecules are produced in different regions, Tubular Epithelial Cells (TECs) as the cells that produce type IV collagen primarily, while resident fibroblasts produce type I and type III collagen. In tubulointerstitial fibrosis, more significantly, interstitial collagen accumulates and resident fibroblasts transdifferentiate into α -SMA positive myofibroblasts, causing ECM expansion. In addition, type IV collagen, which is normally expressed in the glomerular and tubular basement membranes, is no longer restricted and is also observed in the interstitium. 5/6 rats after nephrectomy induce apoptosis due to kidney injury, and then the proximal tubular and podocytes release the cytokeratin (caspase-cleaved) 18(M30), so M30 in different regions can be considered as a pharmacodynamic biomarker of the treatment scheme. Since type I and type IV collagen are associated with excessive ECM deposition, α -SMA can assess glomerulosclerosis, and M30 can be used as an index of glomerulocyte apoptosis, and the effects of anti-IS monoclonal antibodies on IS-associated tissue atrophy should be demonstrated using tissue staining and specific Immunohistochemical (IHC) assays.

Several examples are provided below to illustrate the utility of the isolated monoclonal antibody against indoxyl sulfate and/or antigen-binding fragments thereof of the present invention.

38页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:抗血管生成素-2抗体及其用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!