Analytical and therapeutic methods and compositions and uses thereof

文档序号:1525282 发布日期:2020-02-11 浏览:9次 中文

阅读说明:本技术 分析和治疗方法和组合物及其用途 (Analytical and therapeutic methods and compositions and uses thereof ) 是由 N·J·莫兰 P·G·扬 T·普罗夫特 于 2018-04-26 设计创作,主要内容包括:本发明一般涉及用于在有需要的受试者中治疗、检测和辅助诊断化脓性链球菌感染、风湿热或链球菌感染后肾小球肾炎(PSGN)的组合物和方法,以及用于评估发展为风湿热或PSGN的倾向的组合物和方法。本发明还提供重组多肽,包括重组化脓性链球菌SpnA多肽,和包含该多肽的组合物用于本发明方法中的用途。(The present invention relates generally to compositions and methods for treating, detecting, and aiding in the diagnosis of pyogenic streptococcal infection, rheumatic fever, or post-streptococcal glomerulonephritis (PSGN) in a subject in need thereof, as well as compositions and methods for assessing a predisposition to develop rheumatic fever or PSGN. The invention also provides recombinant polypeptides, including recombinant streptococcus pyogenes SpnA polypeptides, and the use of compositions comprising the polypeptides in the methods of the invention.)

1. A method of treating a patient suffering from rheumatic fever or PSGN, the method comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes; and

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iv) wherein the presence of, or a detected amount of, streptococcus pyogenes SpnA antigen-specific complex is above a threshold value, indicating a recent exposure of the subject to streptococcus pyogenes;

v) one or more other diagnostic indicators wherein rheumatic fever or PSGN is present, in combination with the absence of, or a detection of less than a threshold amount of, streptococcus pyogenes SpnA antigen-specific complex, indicating prior exposure of the subject to streptococcus pyogenes; and

vi) administering a treatment for recent onset of rheumatic fever or acute PSGN if the subject has recently been exposed to streptococcus pyogenes; administering a treatment for an established or subsequent Streptococcus pyogenes infection, or administering a treatment for rheumatic fever or PSGN, if the subject was previously exposed to Streptococcus pyogenes.

2. A method of treating a patient suffering from rheumatic fever or PSGN with an antibiotic effective against streptococcus pyogenes, the method comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA and one or more groups of antigens from streptococcus pyogenes dnase b and/or one or more groups of antigens from streptococcus pyogenes SLO, wherein if antigen-specific antibodies are present in the biological sample, said one or more groups of streptococcus pyogenes antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigens antigen-specific antibody complexes; and

iii) detecting the complex, wherein

a. The presence of, or a detected amount of, streptococcus pyogenes SpnA-specific complex above a threshold value indicates a recent exposure of the subject to streptococcus pyogenes; and

b. the presence of a Streptococcus pyogenes DNaseB-specific complex and/or the presence of a Streptococcus pyogenes SLO-specific complex, and the absence of a Streptococcus pyogenes SpnA-specific complex or the detection of a Streptococcus pyogenes SpnA-specific complex being below a threshold value, indicating a previous exposure of the subject to Streptococcus pyogenes,

iv) administering an antibiotic effective against acute streptococcus pyogenes infection if the subject has recently been exposed to streptococcus pyogenes; if the subject was previously exposed to Streptococcus pyogenes, an antibiotic effective against an established or subsequent Streptococcus pyogenes infection is administered.

3. The method according to claim 1 or claim 2, wherein the treatment for recent episodes of rheumatic fever or acute PSGN, or for recent streptococcus pyogenes infections, is administering an antibiotic effective against acute streptococcus pyogenes infections according to a treatment regimen comprising administering a loading dose of the antibiotic within an acute treatment period.

4. The method of claim 3, wherein the acute treatment period is from about 5 days to about 20 days, e.g., from about 8 days to about 15 days, or from about 10 days to about 12 days.

5. The method of any one of claims 2-4, wherein the antibiotic is dicillin.

6. A method for detecting recent exposure of a subject to streptococcus pyogenes, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens is capable of binding to antigen-specific antibodies present in the biological sample to form two or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more complexes above the threshold value indicates a recent exposure of the subject to Streptococcus pyogenes.

7. A method for detecting or diagnosing rheumatic fever or post-streptococcal glomerulonephritis (PSGN), including post-acute streptococcal glomerulonephritis (APSGN), or an increased likelihood of developing rheumatic fever or PSGN in a subject, comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens is capable of binding to antigen-specific antibodies present in the biological sample to form two or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting complexes, wherein an increase in detection of one or more complexes above a threshold value is indicative of an increased likelihood of development of rheumatic fever or APSGN, or is indicative of recent exposure of the subject to streptococcus pyogenes as an indicator of the presence of rheumatic fever or PSGN in the subject;

iv) assessing one or more diagnostic indicators of rheumatic fever or PSGN; and

v) wherein an increase in detection of one or more complexes above the threshold, in combination with one or more other diagnostic indicators of rheumatic fever or APSGN, is indicative of rheumatic fever or APSGN in the subject.

8. A method for detecting the presence of a streptococcus pyogenes infection in a subject, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens binds to antigen-specific antibodies in the biological sample to form two or more groups of antigen-specific antibody complexes if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more of the complexes indicates the presence of Streptococcus pyogenes in the subject or a recent exposure of the subject to Streptococcus pyogenes.

9. A method for detecting an antibody specific for a streptococcus pyogenes antigen in a biological sample, wherein the antibody specific for a streptococcus pyogenes antigen specifically binds to a streptococcus pyogenes antigen, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens binds to antigen-specific antibodies in the biological sample to form two or more groups of antigen-specific antibody complexes if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more of the complexes indicates that the biological sample contains antibodies specific for Streptococcus pyogenes antigens.

10. The method of any one of claims 1 to 9, wherein the increase in detection of one or more complexes is an increase relative to the level of a reference antigen established for each test cohort.

11. The method according to any one of claims 1 to 10, wherein the one or more antibodies specific for one or more Streptococcus pyogenes antigens are one or more serum antibodies.

12. The method of claim 11, wherein the one or more serum antibodies are one or more IgG antibodies.

13. The method of claim 11, wherein the one or more serum antibodies are one or more IgA antibodies or one or more IgM antibodies.

14. The method of claim 1 or claim 7, wherein the one or more diagnostic indicators are the presence or absence of one or more clinical symptoms associated with rheumatic fever or PSGN.

15. The method of claim 14, wherein the one or more clinical symptoms are selected from migrating polyarthritis, myocarditis, hematuria, erythema marginale, subcutaneous nodules, sydenham's chorea, or pyoderma.

16. The method according to any one of claims 1 to 15, wherein the one or more Streptococcus pyogenes antigens are antigens from one of the following proteins:

i) streptococcus pyogenes nuclease A (SpnA),

ii) deoxyribonuclease-B (DNaseB), or

iii) streptolysin-O (SLO).

17. The method according to any one of claims 1 to 16, wherein the one or more Streptococcus pyogenes antigens are selected from the group consisting of:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8, or

iv) deoxyribonuclease-B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5, or

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of, or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) an SLO antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

x) any combination of two or more of the above i) to ix).

18. The method of claim 17, wherein the biological sample is contacted with each of the following groups of Streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8,

and

iv) deoxyribonuclease-B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5,

and

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of, or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) an SLO antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

19. The method of claim 17, wherein the biological sample is contacted with each of the following groups of Streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA),

ii) deoxyribonuclease-B (DNaseB), and

iii) streptolysin-O (SLO).

20. The method of claim 17, wherein the biological sample is contacted with each of the following groups of Streptococcus pyogenes antigens:

i) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No. 8;

ii) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No. 5; and

iii) an SLO antigenic fragment comprising, consisting essentially of or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

21. The method according to any one of the preceding claims, wherein the two or more groups of Streptococcus pyogenes antigens are present in a composition.

22. The method of any one of claims 1 to 21, wherein one or more of the Streptococcus pyogenes antigens are labeled with a detectable label and/or are conjugated to a microparticle, bead, or detectable agent.

23. The method of any one of the preceding claims, wherein one or more of the group of Streptococcus pyogenes antigens are covalently bound to a bead or microparticle.

24. The method of claim 23, wherein each group of Streptococcus pyogenes antigens is covalently bound to a bead or microparticle, optionally wherein each distinct group of beads or microparticles is distinguishable from one another.

25. The method of any one of claims 22 to 24, wherein the bead is a polystyrene bead, a magnetic bead, a carboxylated bead, a functionalized bead, or wherein the microparticle is a polystyrene microparticle, a magnetic microparticle, a carboxylated microparticle, or a functionalized microparticle.

26. The method of any one of claims 1 to 25, wherein detecting the antigen-antibody complex comprises exposing the complex to a specific binding partner with a detectable label and detecting a signal from the label if the antigen-specific antibody is present in the biological sample.

27. The method of claim 26, wherein the specific binding partner comprises an antibody or fragment thereof.

28. The method of claim 27, wherein the specific binding partner is an anti-IgG antibody, an anti-IgG-PE, or a fragment thereof.

29. The method of any one of claims 1 to 28, wherein antigen-antibody complexes are detected using a flow instrument, a plate-based immunoassay, an electrophoretic and/or immunoblot, an immunochromatographic strip, an electronic biosensor, a resonant biosensor or a microfluidic device or sensor.

30. The method of any one of claims 1 to 29, wherein the detection is by an ELISA or luminex assay, or the method of claim 24, wherein the immunoassay is an ELISA or luminex assay.

31. The method of claim 30, wherein the presence of one or more complexes or one or more antigen-specific antibodies is detected using a detectably labeled secondary antibody.

32. The method of claim 31, wherein the detectably labeled second antibody is anti-IgG-PE.

33. The method according to any one of claims 1-32, wherein the streptococcus pyogenes antigen is detectably labeled.

34. The method of claim 33, wherein the detectable label is a fluorophore.

35. The method of any one of claims 1 to 34, wherein the biological sample is obtained from a mammalian species.

36. The method of claim 35, wherein the biological sample is a bodily fluid sample.

37. The method of claim 35 or claim 36, wherein the subject is a human subject.

38. An isolated, purified, or recombinant SpnA polypeptide, wherein the SpnA polypeptide has a relative abundance to a wild-type SpnA:

i) n-terminal truncation;

ii) a C-terminal truncation; or

iii) N-terminal and C-terminal truncations.

39. The isolated, purified, or recombinant SpnA polypeptide of claim 38, wherein the SpnA polypeptide

i) Is immunogenic, or

ii) is immunologically cross-reactive with wild type SpnA, or

iii) is detectably labeled, or

iv) increased stability upon storage at room temperature compared to wild type SpnA, or

v) comprises 10 or more consecutive amino acids from SEQ ID No.8, or

vi) any combination of two or more of the above i) to v).

40. The isolated, purified, or recombinant SpnA polypeptide of claim 38 or claim 39, wherein the polypeptide has enhanced thermostability, enhanced immunogenic stability, or both enhanced thermostability and enhanced immunogenic stability.

41. A composition comprising the isolated, purified, or recombinant SpnA polypeptide of any one of claims 38 to 40.

42. A composition comprising detectably labeled SpnA.

43. A kit for detecting or diagnosing stroke damp heat or PSGN in a subject, for detecting the presence of a streptococcus pyogenes infection in a subject, or for detecting the presence of an antibody specific for a streptococcus pyogenes antigen in a biological sample, the kit comprising a composition comprising at least one streptococcus pyogenes antigen selected from:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8, or

iv) deoxyribonuclease-B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5, or

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of, or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) an SLO antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No.2,

optionally at least one composition comprising a reference antibody control, wherein the antibody control comprises antibodies specific for one of the Streptococcus pyogenes antigens present in the kit,

optionally one or more reagents for constituting a medium for facilitating the contacting of the one or more antigens with the biological sample,

optionally one or more reagents enabling the detection of complexes formed between said one or more antigens and one or more Streptococcus pyogenes antigen-specific antibodies present in the biological sample,

and instructions for use.

44. The kit according to claim 43, wherein at least one of the Streptococcus pyogenes antigens is covalently bound to a bead or microparticle.

45. The kit of claim 43 or claim 44, wherein the composition comprises the group of:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No. 8.

46. The kit of claim 45, wherein the composition comprises each of the following groups of Streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8,

and

iv) deoxyribonuclease-B (DNaseB); or

v) an antigenic fragment of DNaseB comprising, consisting essentially of or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5,

and

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) SLO antigenic fragments comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

47. The method according to any one of the preceding claims, further comprising providing a kit according to any one of claims 43 to 46 prior to step ii).

48. The method or kit according to any one of the preceding claims, wherein the one or more Streptococcus pyogenes antigens are selected from the group consisting of anti-streptolysin (ASO), anti-hyaluronidase (AHase), anti-streptokinase (ASKase), anti-nicotinamide adenine dinucleotide enzyme (anti-NAD).

49. A method for detecting recent exposure of a subject to streptococcus pyogenes, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more complexes above the threshold value indicates a recent exposure of the subject to Streptococcus pyogenes.

50. A method for detecting or diagnosing rheumatic fever or post-streptococcal glomerulonephritis (PSGN), including post-acute streptococcal glomerulonephritis (APSGN), or an increased likelihood of developing rheumatic fever or PSGN in a subject, comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting complexes, wherein an increase in detection of one or more complexes above a threshold value is indicative of an increased likelihood of development of rheumatic fever or APSGN, or is indicative of recent exposure of the subject to streptococcus pyogenes as an indicator of the presence of rheumatic fever or PSGN in the subject;

iv) assessing one or more diagnostic indicators of rheumatic fever or PSGN in the subject; and

v) wherein an increase in detection of one or more complexes above the threshold, in combination with one or more other diagnostic indicators of rheumatic fever or APSGN, is indicative of rheumatic fever or APSGN in the subject.

51. The method of claim 50, wherein the one or more diagnostic indicators are the presence or absence of one or more clinical symptoms associated with rheumatic fever or PSGN.

52. The method of claim 51, wherein the one or more clinical symptoms are selected from migrating polyarthritis, myocarditis, hematuria, erythema marginalis, subcutaneous nodules, West-Thomson-Harm's chorea, or pyoderma.

53. A method for detecting the presence of a streptococcus pyogenes infection in a subject, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more of the complexes indicates the presence of Streptococcus pyogenes in the subject or a recent exposure of the subject to Streptococcus pyogenes.

54. A method for detecting an antibody specific for a streptococcus pyogenes antigen in a biological sample, wherein said antibody specific for a streptococcus pyogenes antigen specifically binds streptococcus pyogenes SpnA, said method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with one or more groups of streptococcus pyogenes SpnA antigens, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more of the complexes indicates that the biological sample contains antibodies specific for Streptococcus pyogenes antigens.

Technical Field

The present invention relates generally to compositions and methods for treating, detecting, and aiding in the diagnosis of streptococcus pyogenes (streptococcus pyogenes) infection, rheumatic fever, or post-streptococcal glomerulonephritis (PSGN), as well as compositions and methods for assessing a propensity to develop rheumatic fever or PSGN.

Background

Group a streptococci (GAS, streptococcus pyogenes) cause or are associated with a variety of diseases of varying severity ranging from mild skin infections and pharyngitis to severe invasive diseases and post-infection immune sequelae such as rheumatic fever or poststreptococcal glomerulonephritis (PSGN). Acute rheumatic fever and related rheumatic heart disease are the leading causes of acquired heart disease in developing countries.

Streptococcal serology is crucial for diagnosing immune sequelae after infection, since these sequelae occur weeks after GAS infection, when diagnostic culture of pathogenic bacteria is often not possible. Generally, current clinical practice involves measuring antibody titers for two antigens: streptolysin-O (SLO) and DNAseB. Serological examinations were called antistreptolysin-O (ASO) and antisterneoxyribonuclease-B (ADB), respectively. ASO titers are typically measured using turbidity or turbidimetry, and values are typically reported in international units per milliliter (iu/mL). The degree of standardization of the ADB test was low because, unlike ASO, there was no reference serum for DNaseB. ADB titers are typically measured using an enzyme inhibition assay, in which sera are tested for inhibition of dnase b activity using a colored dye.

At best, significant variability in the immune kinetics of ASO and ADB antibody responses has been reported, with most children with GAS pharyngitis exhibiting elevated ASO and ADB titers (compared to pre-infection levels) for more than one year post-infection [1 ]. Thus, there is a substantial risk of false positive diagnosis.

There remains a need for more effective methods for identifying subjects suffering or recently suffering from GAS infection, as well as identifying subjects having a predisposition to develop rheumatic fever or PSGN, and for more effective detection and diagnosis of rheumatic fever or PSGN.

It is an object of the present invention to address the above problems or to provide methods and compositions for detecting and aiding diagnosis of rheumatic fever or post-streptococcal glomerulonephritis, or for assessing the propensity for development of rheumatic fever or post-streptococcal glomerulonephritis, and/or at least to provide the public with a useful choice.

All references cited in this specification, including any patents or patent applications, are hereby incorporated by reference. No admission is made that any reference constitutes prior art. The discussion of the references states what their authors assert, and the applicants reserve the right to challenge the accuracy and pertinency of the cited documents. It will be clearly understood that, although a number of prior art publications are referred to herein, this reference does not constitute an admission that any of these documents forms part of the common general knowledge in the art, in new zealand or in any other country.

Disclosure of Invention

These and other objects are achieved according to one or more aspects of the present invention.

Thus, in one aspect, the invention relates to a method for detecting recent exposure of a subject to streptococcus pyogenes, the method comprising:

providing a biological sample from a subject, said biological sample being capable of or suspected of containing antibodies specific for one or more streptococcus pyogenes antigens;

contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each group of the two or more groups of Streptococcus pyogenes antigens is capable of binding to antigen-specific antibodies present in the biological sample to form two or more groups of antigens, antigen-specific antibody complexes, if present in the biological sample; and

detecting complexes, wherein an increase in the detection of one or more complexes above the threshold value indicates a recent exposure of the subject to streptococcus pyogenes.

In another aspect, the invention relates to a method for detecting or diagnosing rheumatic fever or post-streptococcal glomerulonephritis (PSGN), including post-acute streptococcal glomerulonephritis (APSGN), in a subject, or an increased likelihood of developing rheumatic fever or PSGN in a subject, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens is capable of binding to antigen-specific antibodies present in the biological sample to form two or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting complexes, wherein an increase in the detection of one or more of the complexes above a threshold value is indicative of an increased likelihood of development of rheumatic fever or APSGN, or is indicative of recent exposure of the subject to streptococcus pyogenes as an indicator of the presence of rheumatic fever or PSGN in the subject;

iv) assessing one or more diagnostic indicators of rheumatic fever or PSGN; and

v) wherein an increase in the detection value of one or more of the compounds above the threshold value, in combination with one or more other diagnostic indicators of rheumatic fever or APSGN, is indicative of rheumatic fever or PSGN in the subject.

In another aspect, the invention relates to a method for detecting the presence of a streptococcus pyogenes infection in a subject, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens binds to the antigen-specific antibodies in the biological sample to form two or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in the detection of one or more of the complexes indicates the presence of Streptococcus pyogenes in the subject or a recent exposure of the subject to Streptococcus pyogenes.

In another aspect, the invention relates to a method for detecting streptococcus pyogenes antigen-specific antibodies in a biological sample, wherein the streptococcus pyogenes antigen-specific antibodies specifically bind to streptococcus pyogenes antigens, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens binds to antigen-specific antibodies in the biological sample to form two or more groups of antigen-specific antibody complexes if present in the biological sample; and

iii) detecting the complex, wherein an increase in the detection of one or more of the complexes indicates that the biological sample contains antibodies specific for Streptococcus pyogenes antigens.

In various embodiments, an increase in the detection of one or more complexes is a detection of the presence of one or more complexes.

Thus, in one embodiment, a method for detecting or diagnosing damp heat or increased likelihood of post-streptococcal glomerulonephritis (PSGN), including post-acute streptococcal glomerulonephritis (APSGN), or the development of rheumatic heat or PSGN in a subject, comprises:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens is capable of binding to antigen-specific antibodies present in the biological sample to form two or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complex, wherein the presence of the one or more antigens, antigen-specific antibody complex, is indicative of an increased likelihood of developing rheumatic fever or APSGN, or of a recent exposure of the subject to streptococcus pyogenes as an indicator of the presence of rheumatic fever or PSGN in the subject;

iv) assessing one or more diagnostic indicators of rheumatic fever or PSGN;

v) an antigen-specific antibody complex, which binds to rheumatic fever or one or more other diagnostic indicators of APSGN, indicative of rheumatic fever or PSGN in the subject.

In another embodiment, a method for detecting the presence of a streptococcus pyogenes infection in a subject comprises:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens binds to antigen-specific antibodies in the biological sample to form two or more groups of antigen-specific antibody complexes if present in the biological sample; and

iii) detecting the complex, wherein the presence of the one or more antigens, the antigen-specific antibody complex indicates the presence of Streptococcus pyogenes in the subject or recent exposure of the subject to Streptococcus pyogenes.

In another embodiment, a method for detecting an antibody specific for a streptococcus pyogenes antigen in a biological sample, wherein said antibody specific for a streptococcus pyogenes antigen specifically binds to a streptococcus pyogenes antigen, said method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for one or more streptococcus pyogenes antigens;

ii) contacting the biological sample with two or more groups of Streptococcus pyogenes antigens, wherein each of the two or more groups of Streptococcus pyogenes antigens binds to antigen-specific antibodies in the biological sample to form two or more groups of antigen-specific antibody complexes if present in the biological sample; and

iii) detecting the complex, wherein the presence of the one or more antigens, the antigen-specific antibody complex indicates that the biological sample contains antibodies specific for Streptococcus pyogenes antigens.

In one aspect, the invention relates to a method of treating a patient suffering from rheumatic fever or PSGN, comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes; and

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iv) wherein the presence of, or a detection value for the amount of, streptococcus pyogenes SpnA antigen-specific complex is above a threshold value, indicating a recent exposure of the subject to streptococcus pyogenes;

v) one or more other diagnostic indicators wherein rheumatic fever or PSGN is present, in combination with the absence of, or the amount of, streptococcus pyogenes SpnA antigen-specific complex being below a threshold value, indicating that the subject has been previously exposed to streptococcus pyogenes; and

vi) administering a therapy for recent onset of rheumatic fever or acute PSGN if the subject has recently been exposed to streptococcus pyogenes; administering a therapy for an established (established) or subsequent infection by s.pyogenes, or administering a therapy for rheumatic fever or PSGN, if the subject was previously exposed to s.pyogenes.

In one aspect, the invention relates to a method of treating a patient suffering from rheumatic fever or PSGN with an antibiotic effective against streptococcus pyogenes, the method comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes; and

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iv) wherein the presence of, or a detection value for the amount of, streptococcus pyogenes SpnA antigen-specific complex is above a threshold value, indicating a recent exposure of the subject to streptococcus pyogenes;

v) one or more other diagnostic indicators wherein rheumatic fever or PSGN is present, in combination with the absence of, or the amount of, streptococcus pyogenes SpnA antigen-specific complex being below a threshold value, indicating prior exposure of the subject to streptococcus pyogenes; and

vi) administering an antibiotic effective against acute or current (currentinfection) streptococcus pyogenes infection if the subject has recently been exposed to streptococcus pyogenes; if the subject is exposed earlier (previously) to s.pyogenes, an antibiotic effective against an established or subsequent s.pyogenes infection is administered.

In one aspect, the invention relates to a method of treating a patient having rheumatic fever or PSGN with an antibiotic effective against streptococcus pyogenes, the method comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA and one or more groups of antigens from streptococcus pyogenes dnase b and/or one or more groups of antigens from streptococcus pyogenes SLO, wherein if antigen-specific antibodies are present in the biological sample, the one or more groups of streptococcus pyogenes antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes; and

iii) detecting the complex, wherein

a. The presence of, or an amount of, streptococcus pyogenes SpnA-specific complex detected above a threshold value indicates a recent exposure of the subject to streptococcus pyogenes; and

b. (ii) the presence of a Streptococcus pyogenes DNaseB-specific complex and/or the presence of a Streptococcus pyogenes SLO-specific complex, and the absence of a Streptococcus pyogenes SpnA-specific complex or the presence of a Streptococcus pyogenes SpnA-specific complex being below a threshold value, indicating prior exposure of the subject to Streptococcus pyogenes,

iv) administering an antibiotic effective against acute streptococcus pyogenes infection if the subject has recently been exposed to streptococcus pyogenes; if the subject was previously exposed to S.pyogenes, an antibiotic effective against an established or subsequent S.pyogenes infection is administered.

In one aspect, the invention relates to a method of treating a patient suffering from rheumatic fever or PSGN, comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA and one or more groups of antigens from streptococcus pyogenes dnase b and/or one or more groups of antigens from streptococcus pyogenes SLO, wherein if antigen-specific antibodies are present in the biological sample, said one or more groups of streptococcus pyogenes antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigens antigen-specific antibody complexes; and

iii) detecting the complex, wherein

a. The presence of, or a detected amount of, streptococcus pyogenes SpnA-specific complex above a threshold value indicates a recent exposure of the subject to streptococcus pyogenes; and

b. (ii) the presence of a Streptococcus pyogenes DNaseB-specific complex and/or the presence of a Streptococcus pyogenes SLO-specific complex, and the absence of a Streptococcus pyogenes SpnA-specific complex or the presence of a Streptococcus pyogenes SpnA-specific complex being below a threshold value, indicating prior exposure of the subject to Streptococcus pyogenes,

iv) administering a therapy for recent onset of rheumatic fever or acute PSGN if the subject has recently been exposed to streptococcus pyogenes; if the subject was previously exposed to Streptococcus pyogenes, a therapy for rheumatic fever or PSGN is administered.

In another aspect, the present invention relates to a method of treating rheumatic fever or PSGN in a subject in need thereof, comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes; and

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample;

iii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iv) wherein the presence of, or a detected amount of, streptococcus pyogenes SpnA antigen-specific complex is above a threshold value, indicating a recent exposure of the subject to streptococcus pyogenes;

v) one or more other diagnostic indicators wherein rheumatic fever or PSGN is present, in combination with the absence of, or the detected amount of, streptococcus pyogenes SpnA antigen-specific complex being below a threshold value, indicating prior exposure of the subject to streptococcus pyogenes; and

vi) administering a therapy for recent onset of rheumatic fever or acute PSGN if the subject has recently been exposed to streptococcus pyogenes; if the subject was previously exposed to Streptococcus pyogenes, a therapy for rheumatic fever or PSGN is administered.

In another aspect, the invention relates to a method of treating rheumatic fever or PSGN in a subject in need thereof, comprising the steps of:

i) determining the presence, absence or amount of one or more streptococcus pyogenes SpnA specific antibodies in a biological sample from the subject; and

ii) optionally, assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iii) predicting that the subject has a recent onset of rheumatic fever or acute PSGN if the sample comprises an amount of one or more streptococcus pyogenes SpnA-specific antibodies above a threshold; and

iv) administering to the subject a therapeutically effective amount of an antibiotic effective against recent Streptococcus pyogenes infection.

In one embodiment, a method of treating rheumatic fever or PSGN in a subject in need thereof comprises the steps of:

i) determining the presence, absence or amount of one or more streptococcus pyogenes SpnA specific antibodies in a biological sample from the subject; and

ii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iii) if the sample comprises one or more streptococcus pyogenes SpnA specific antibodies in an amount above a threshold and the subject has one or more other diagnostic indicators of rheumatic fever or PSGN, predicting that the subject has a recent onset of rheumatic fever or acute PSGN; and

iv) administering to the subject a therapeutically effective amount of an antibiotic effective against acute S.pyogenes infection.

In another aspect, the invention relates to a method of treating rheumatic fever or PSGN in a subject in need thereof, comprising the steps of:

i) determining the presence, absence or amount of one or more streptococcus pyogenes SpnA specific antibodies in a biological sample from the subject; and

ii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iii) predicting that the subject has rheumatic fever or PSGN if the sample comprises one or more streptococcus pyogenes SpnA specific antibodies in an amount below a threshold value and the subject has one or more other diagnostic indicators of rheumatic fever or PSGN; and

iv) administering to the subject a therapeutically effective amount of an antibiotic effective against established or subsequent Streptococcus pyogenes infection.

In various embodiments, the presence, absence, or amount of one or more streptococcus pyogenes SpnA specific antibodies is determined by contacting the biological sample with one or more populations of antigens from streptococcus pyogenes SpnA, wherein if antigen-specific antibodies are present in the biological sample, the one or more populations of streptococcus pyogenes SpnA antigens are capable of binding to the antigen-specific antibodies present in the biological sample to form one or more populations of antigen-specific antibody complexes; and

in various embodiments, the amount of the one or more streptococcus pyogenes SpnA-specific antibodies above the threshold is an antibody titer that correlates with, or is indicative of, a recent exposure of the subject to streptococcus pyogenes.

In various embodiments, the amount of the one or more streptococcus pyogenes SpnA-specific antibodies below the threshold is an antibody titer that correlates with, or is indicative of, a prior exposure of the subject to streptococcus pyogenes.

In various embodiments, the threshold is an amount of SpnA-specific antibody that separates the range of antibody titers or the range of mean antibody titers observed in a population of patients with rheumatic fever or PSGN within 20 days of hospitalization from the range of antibody titers or the range of mean antibody titers observed in a population of patients with rheumatic fever or PSGN after 20 days of hospitalization.

In one embodiment, the reference level, reference threshold, or threshold is the Upper Limit of Normal (ULN), which is the 80 th percentile of matched healthy populations.

In one aspect, the invention relates to a method of treating a patient suffering from or having been exposed to a streptococcus pyogenes infection with an antibiotic effective against streptococcus pyogenes, comprising the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes; and

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) assessing the presence of Streptococcus pyogenes in the subject or one or more other diagnostic indicators of stroke fever or PSGN in the subject;

iv) wherein the presence of, or a detected amount of, streptococcus pyogenes SpnA antigen-specific complex is above a threshold value, indicating a recent exposure of the subject to streptococcus pyogenes;

v) other diagnostic indicators wherein one or more rheumatic fever or PSGN is present, combined with the absence of, or a detection of less than a threshold amount of, streptococcus pyogenes SpnA antigen-specific complex, indicating prior exposure of the subject to streptococcus pyogenes; and

vi) administering an antibiotic effective against acute S.pyogenes infection if the subject has recently been exposed to S.pyogenes; if the subject was previously exposed to S.pyogenes, an antibiotic effective against an established or subsequent S.pyogenes infection is administered.

In one embodiment, a method of treating a patient with an antibiotic effective against streptococcus pyogenes comprises the steps of:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes; and

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA and one or more groups of antigens from streptococcus pyogenes dnase b and/or one or more groups of antigens from streptococcus pyogenes SLO, wherein if antigen-specific antibodies are present in the biological sample, the one or more groups of streptococcus pyogenes antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigens antigen-specific antibody complexes; and

iii) detecting the complex, wherein

a. The presence of, or the detected amount of, a streptococcus pyogenes SpnA-specific complex being above a threshold value indicates a recent exposure of the subject to streptococcus pyogenes, and

b. the presence of a Streptococcus pyogenes DNaseB-specific complex and/or a Streptococcus pyogenes SLO-specific complex, and the absence of a Streptococcus pyogenes SpnA-specific complex or the detected amount of a Streptococcus pyogenes SpnA-specific complex being below a threshold value, indicates that the subject has been previously exposed to Streptococcus pyogenes,

iv) administering an antibiotic effective against acute streptococcus pyogenes infection if the subject has recently been exposed to streptococcus pyogenes; if the subject was previously exposed to S.pyogenes, an antibiotic effective against an established or subsequent S.pyogenes infection is administered.

In various embodiments, the treatment for recent episodes of rheumatic fever or acute PSGN, or for acute or current streptococcus pyogenes infection, is administration of an antibiotic effective against the acute streptococcus pyogenes infection, e.g., according to a treatment regimen. For example, a dosage regimen effective for acute S.pyogenes infection, or for treating recent episodes of rheumatic fever or acute PSGN, includes a 10-day course of treatment with one or more antibiotics, such as a 10-day course of treatment with dicillin (bicillin). Other suitable treatment regimens will be known to those of skill in the art upon reading this disclosure, including certain representative examples disclosed herein.

In various embodiments, the treatment for rheumatic fever or PSGN, or for an established or subsequent streptococcus pyogenes infection, is the administration of an antibiotic effective against an established or subsequent streptococcus pyogenes infection, e.g., administration according to a treatment regimen, such as a prophylactic treatment regimen. For example, a dosage regimen effective for an established or subsequent Streptococcus pyogenes infection, or a dosage regimen effective for the treatment of rheumatic fever or PSGN, comprises monthly administration of one or more antibiotics, such as monthly administration of bifillin. Other suitable treatment regimens will be known to those of skill in the art upon reading this disclosure, including certain representative examples disclosed herein.

In one example, for the treatment of rheumatic fever or PSGN, or for the treatment of established or subsequent streptococcus pyogenes infections, including bed rest and/or hospitalization.

In one example, administration of a treatment for recent episodes of rheumatic fever or acute PSGN comprises administering to the subject an antibiotic effective against acute streptococcus pyogenes infection.

In one example, administration of a treatment for rheumatic fever or PSGN comprises administering to the subject an antibiotic effective against an established or subsequent streptococcus pyogenes infection.

In one example, administering treatment for rheumatic fever or PSGN comprises hospitalizing and/or prescribing the subject or bed rest of the subject.

In one aspect, the invention relates to a method of predicting the responsiveness of a patient with rheumatic fever or PSGN to antibiotic treatment, the method comprising the steps of:

i) determining the presence, absence or amount of one or more streptococcus pyogenes SpnA specific antibodies in a biological sample from the subject; and

ii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iii) predicting that the subject is likely to be responsive to antibiotic treatment if the sample comprises an amount of one or more streptococcus pyogenes SpnA specific antibodies below a threshold value, and the subject has rheumatic fever or one or more other diagnostic indicators of PSGN; and

iv) administering to the subject a therapeutically effective amount of an antibiotic effective against an established or subsequent Streptococcus pyogenes infection.

In one aspect, the invention relates to a method of predicting the responsiveness of a patient with rheumatic fever or PSGN to antibiotic treatment, the method comprising the steps of:

i) determining the presence, absence or amount of one or more streptococcus pyogenes SpnA specific antibodies in a biological sample from the subject; and

ii) optionally assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iii) predicting that the subject is likely to be responsive to antibiotic treatment effective to treat recently onset rheumatic fever or acute PSGN if the sample comprises one or more streptococcus pyogenes SpnA specific antibodies in an amount above a threshold, and optionally if the subject has one or more other diagnostic indicators of rheumatic fever or PSGN; and

iv) administering to the subject a therapeutically effective amount of an antibiotic effective against acute S.pyogenes infection.

In another aspect, the invention relates to a method of determining a treatment regimen for a patient suffering from rheumatic fever or PSGN, the method comprising the steps of:

i) determining the presence, absence or amount of one or more streptococcus pyogenes SpnA specific antibodies in a biological sample from the subject; and

ii) assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iii) if the sample comprises an amount of one or more streptococcus pyogenes SpnA specific antibodies below a threshold value, and the subject has one or more other diagnostic indicators of rheumatic fever or PSGN, determining that the subject should undergo a treatment regimen suitable for treating rheumatic fever or PSGN; and

iv) treating the subject according to a treatment regimen suitable for treating rheumatic fever or PSGN.

In one embodiment, the treatment of rheumatic fever or PSGN is the treatment of chronic rheumatic heart disease.

In one embodiment, the treatment of rheumatic fever or PSGN comprises administering to the subject a therapeutically effective amount of an antibiotic effective against established or subsequent streptococcus pyogenes infection, e.g., a prophylactically effective amount of such antibiotic.

In one embodiment, a suitable treatment regimen for rheumatic fever or PSGN is monthly antibiotic administration, such as monthly penicillin (Benzathine).

In another aspect, the invention relates to a method of determining a treatment regimen for a patient suffering from rheumatic fever or PSGN, the method comprising the steps of:

i) determining the presence, absence or amount of one or more streptococcus pyogenes SpnA specific antibodies in a biological sample from the subject; and

ii) optionally assessing one or more other diagnostic indicators of stroke damp heat or PSGN in the subject;

iii) if the sample comprises one or more streptococcus pyogenes SpnA specific antibodies in an amount above a threshold, and optionally the subject has one or more other diagnostic indicators of rheumatic fever or PSGN, determining that the subject should undergo a treatment regimen suitable for treating a recent episode of rheumatic fever or acute PSGN; and

iv) optionally administering to the subject a therapeutically effective amount of an antibiotic effective against acute streptococcus pyogenes infection according to a treatment regimen.

In one embodiment, if the subject has recently been exposed to streptococcus pyogenes, an antibiotic effective against the acute streptococcus pyogenes infection is administered at a dosage rate effective against the acute streptococcus pyogenes infection.

In one embodiment, the antibiotic effective for acute streptococcus pyogenes infection is administered if the subject has recently been exposed to streptococcus pyogenes, wherein the dosage rate administered is greater than the dosage rate administered to a subject with an established or subsequent streptococcus pyogenes infection or rheumatic fever or PSGN.

In one embodiment, if the subject has recently been exposed to streptococcus pyogenes, an antibiotic effective against acute streptococcus pyogenes infection is administered in combination with one or more other therapeutic agents, such as anti-inflammatory agents, e.g., aspirin (aspirin), glucocorticoids, e.g., prednisone (prednisone), neuroleptics, e.g., haloperidol (haloperidol), inotropic agents, e.g., digoxin (digoxin), or in combination with one or more other therapies, e.g., long term hospitalization, bed rest, etc.

In one embodiment, the risk of adverse antibiotic reactions or sequelae from administration of an antibiotic after administration of an antibiotic effective against chronic pyogenic streptococcal infection in a patient exposed to, but not recently exposed to, streptococcus pyogenes is lower than if the antibiotic administered was an antibiotic administered to a subject having an acute pyogenic streptococcal infection or a recent episode of rheumatic fever or acute PSGN.

In one embodiment, after administration of an antibiotic effective against Streptococcus pyogenes infection in an amount effective against established or chronic Streptococcus pyogenes infection in a patient exposed but not recently exposed to Streptococcus pyogenes, the risk of adverse antibiotic reactions or sequelae from administration of the antibiotic is lower than if the antibiotic were administered in an amount effective to treat acute Streptococcus pyogenes infection or recent episodes of rheumatic fever or acute PSGN.

In one embodiment, if the subject has recently been exposed to streptococcus pyogenes, administering an antibiotic effective against acute streptococcus pyogenes infection comprises parenterally administering the antibiotic.

In various embodiments, the antibiotic effective against streptococcus pyogenes is selected from the group consisting of penicillin, Amoxicillin (Amoxicillin), Oxacillin (oxacilin), Erythromycin (Erythromycin), Azithromycin (Azithromycin), Clarithromycin (Clarithromycin), cephalosporin (Cephalothin), Cefoxitin (Cefoxitin), Cefixime (Cefixime), Cefuroxime (Cefuroxime), Cefotaxime (Cefotaxime), Ceftriaxone (Ceftriaxone), Vancomycin (Vancomycin), Clindamycin (Clindamycin), rifampin (rifapecin), Ciprofloxacin (Ciprofloxacin), Tetracycline (tetratetracycline), sulfamethoxazole (Cotrimoxazole), and Chloramphenicol (chloreninol).

In various embodiments, the antibiotic effective against Streptococcus pyogenes is selected from the group consisting of β -lactams such as penicillin, Amoxicillin (Amoxicillin), Cefixime (Cefixim), cefpodoxime (Cefpodoxine), Cefotaxime (Cefotam), Ceftriaxone (Ceftriaxone), Oxacillin (Oxalilin), macrolides such as Erythromycin (Erythromycin), spiramycin (Spiramycin), Azithromycin (Azithromycin), lincomamines (Lincosamides) such as Clindamycin (Clramycin), streptogramins (streptamycins) such as Pristinamycin (Primatinmycin), ketolides (Ketolide) such as Telithromycin (Telithromycin), enPhocinols such as Chloramphenicol (Chlorophyceins), Glycopeptides (glucopeptide) such as Teicoplanin, and tetracycline (Vasinoclines), Tetracyclines such as Fluoromycin (Vasinoclines), and Tetracyclines such as Levofloxacin (Fluoromycin).

In one embodiment, the antibiotic effective for acute S.pyogenes infection is a penicillin, e.g., penicillin G, including penicillin G procaine (e.g., Crytillin) and benzathine penicillin G (e.g., Bicillin L-A), penicillin VK (e.g., Beepen-VK, Betapen-VK, Robicillin VK, Veetids), erythromycin, e.g., E-Mycin, Ery-Tab, Erythrocin, or Sulfadiazine (Sulfadiazine), e.g., Microsulforin.

In one embodiment, if the patient has recently been exposed to streptococcus pyogenes, administering an antibiotic effective for acute streptococcus pyogenes infection includes parenteral administration of penicillin G, erythromycin, or sulfadiazine, e.g., intravenous or intramuscular administration of penicillin G, erythromycin, or sulfadiazine. In another embodiment, oral administration is substituted for parenteral administration.

In various embodiments, an antibiotic effective against acute streptococcus pyogenes infection is administered according to a dosage regimen. In various examples, the dosage regimen comprises administration of a loading dose (loading dose) of the antibiotic, e.g., during an acute treatment period.

In certain embodiments, the acute treatment period is from about 5 days to about 20 days, e.g., from about 8 days to about 15 days, or from about 10 days to about 12 days, including about 10 days.

In one example, a dosage regimen effective for acute streptococcus pyogenes infection or for treating recent episodes of rheumatic fever or acute PSGN includes a 10-day course of treatment with one or more antibiotics, such as a 10-day course of treatment with penicillin. For example, certain exemplary dosage regimens effective for acute S.pyogenes infection, or for recent episodes of rheumatic fever or acute PSGN, include

i. Benzathine penicillin (pivampicillin): for adults and children above 30kg, about 900mg as a single dose, usually administered by deep IM; for 30kChildren below g, about 450- 6IU as single dose IM; all usually over a period of 10 days.

Phenoxymethylpenicillin: from 125-250mg twice daily, if IM is not possible, orally, typically for 10 days; or 10mg/kg up to 500mg twice a day for 10 days;

erythromycin: 10mg/kg to a maximum of 500mg twice a day for 10 days;

erythromycin ethyl succinate: 40 mg/kg/day, administered in 2-4 times, with a maximum dose of 1 g/day for children.

In various embodiments, an antibiotic effective against an established or chronic streptococcus pyogenes infection is administered according to a dosage regimen. For example, certain exemplary dosage regimens effective for the already established or subsequent infection by Streptococcus pyogenes, or for the treatment of rheumatic fever or PSGN (including chronic rheumatic heart disease), include

i. Benzathine penicillin: about 900mg per 4 weeks for adults and children above 30kg, usually by deep IM administration, about 450 and 675mg per 4 weeks for children below 30kg, 600,000IU per 4 weeks in muscle for patients below 20kg, 1.2X 10 per 4 weeks for patients above 20kg 6IU administered intramuscularly;

phenoxymethylpenicillin: 125-250mg twice daily, if IM is not possible, orally, typically for a period of 10 days;

erythromycin: 250mg, twice daily;

erythromycin ethyl succinate: 400mg twice daily.

In another aspect, the invention relates to a method for detecting recent exposure of a subject to streptococcus pyogenes, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein if SpnA antigen-specific antibodies are present in the biological sample, the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to SpnA antigen-specific antibodies present in the biological sample to form one or more groups of SpnA antigen: SpnA antigen-specific antibody complexes; and

iii) detecting the presence or absence of the complex,

iv) optionally assessing the presence of Streptococcus pyogenes infection or rheumatic fever or one or more other diagnostic indicators of PSGN in the subject;

v) wherein the streptococcus pyogenes SpnA antigen-specific complex is present, or the amount of streptococcus pyogenes SpnA antigen-specific complex is above a threshold, indicating a recent exposure of the subject to streptococcus pyogenes;

vi) one or more other diagnostic indicators wherein rheumatic fever or PSGN is present, in combination with the absence or amount of streptococcus pyogenes SpnA antigen-specific complex being below a threshold, is indicative of prior exposure of the subject to streptococcus pyogenes.

In another aspect, the invention relates to a method for detecting or diagnosing rheumatic fever or post-streptococcal glomerulonephritis (PSGN), including post-acute streptococcal glomerulonephritis (APSGN), in a subject, or an increased likelihood of developing rheumatic fever or PSGN in a subject, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to SpnA antigen-specific antibodies present in the biological sample to form one or more groups of SpnA antigen: SpnA antigen-specific antibody complexes, if antigen-specific antibodies are present in the biological sample; and

iii) detecting the presence or absence of the complex;

iv) assessing one or more other diagnostic indicators of rheumatic fever or PSGN in the subject;

v) one or more other diagnostic indicators wherein rheumatic fever or PSGN is present, in combination with the presence of streptococcus pyogenes SpnA specific complex, or a detected amount of streptococcus pyogenes SpnA specific complex above a threshold value, is indicative of an increased likelihood of development of rheumatic fever or APSGN, or of a recent exposure of the subject to streptococcus pyogenes as an indicator of the presence of rheumatic fever or PSGN in the subject;

vi) one or more other diagnostic indicators wherein rheumatic fever or PSGN is present, in combination with the absence of, or the amount of, streptococcus pyogenes SpnA-specific complex being below a threshold value, indicates rheumatic fever or PSGN in the subject, or indicates prior exposure of the subject to streptococcus pyogenes as an indicator that rheumatic fever or PSGN is present in the subject, such as an increased risk of or from a subsequent streptococcus pyogenes infection.

In one embodiment, the one or more other diagnostic indicators are the presence or absence of antibodies specific for one or more streptococcus pyogenes antigens other than SpnA. For example, one or more other diagnostic indicators are the presence or absence of antibodies specific for streptococcus pyogenes dnase b, or antibodies specific for streptococcus pyogenes SLO.

Any embodiment disclosed herein may be directed to any aspect set forth herein. To avoid any confusion, it will be apparent from the disclosure herein that any aspect disclosed herein, e.g., any method described herein, will in certain embodiments employ one or more streptococcus pyogenes nuclease a (SpnA) polypeptides, such as one or more truncated SpnA polypeptides, or one or more SpnA fragments, as described herein. For example, in various embodiments, one or more antigens or one or more antigenic groups from streptococcus pyogenes SpnA are present as one or more SpnA polypeptides, such as one or more truncated SpnA polypeptides or one or more SpnA fragments, as described herein.

Similarly, when used in any aspect disclosed herein, e.g., in any of the methods described herein, the one or more streptococcus pyogenes dnase b antigens or polypeptides will in certain embodiments be one or more dnase b polypeptides, such as one or more dnase b antigen fragments, as disclosed herein. For example, in various embodiments, the one or more antigens or one or more groups of antigens from streptococcus pyogenes DNaseB are present as one or more DNaseB polypeptides, e.g., as one or more DNaseB fragments, as described herein.

Similarly, when used in any aspect disclosed herein, e.g., in any of the methods described herein, the one or more streptococcus pyogenes SLO antigens or SLO polypeptides will in certain embodiments be one or more dnase b polypeptides, such as one or more SLO antigen fragments, as disclosed herein. For example, in various embodiments, one or more antigens or one or more antigenic groups from a streptococcus pyogenes SLO are present as one or more SLO polypeptides, such as one or more SLO fragments, as described herein.

In one embodiment, the presence of two or more groups of antigens, the antigen-specific antibody complex indicates the presence of Streptococcus pyogenes in the subject, or indicates recent exposure of the subject to Streptococcus pyogenes, or indicates that the biological sample contains antibodies specific for two or more Streptococcus pyogenes antigens.

In one embodiment, the increase in detection of one or more complexes is an increase relative to a reference level of antigen established for each test population.

In one embodiment, the one or more antibodies specific for one or more streptococcus pyogenes antigens are one or more serum antibodies.

In one embodiment, the one or more serum antibodies are one or more IgG antibodies.

In one embodiment, the one or more serum antibodies are one or more IgA antibodies or one or more IgM antibodies.

In one embodiment, the one or more other diagnostic indicators are the presence or absence of one or more clinical symptoms associated with rheumatic fever or PSGN.

In one embodiment, the one or more clinical symptoms are selected from migratory polyarthritis (miglityorthritis), myocarditis (cardiis), hematuria (hematura), erythema marginalis (erythromatriatium), subcutaneous nodules (subeutaneous nodules), sydenham's chorea, or pyoderma (pyoderma).

In one embodiment, the one or more streptococcus pyogenes antigens are antigens from one of the following proteins:

i) streptococcus pyogenes nuclease A (SpnA),

ii) deoxyribonuclease-B (DNaseB), or

iii) streptolysin-O (SLO).

In one embodiment, the one or more streptococcus pyogenes antigens are selected from the group consisting of:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8, or

iv) deoxyribonuclease-B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5, or

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of, or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) an SLO antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

x) any combination of two or more of the above i) to ix).

In one embodiment, the biological sample is contacted with each of the following groups of streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8,

and

iv) deoxyribonuclease-B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5,

and

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of, or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) an SLO antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

In one embodiment, the biological sample is contacted with each of the following groups of streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA),

ii) deoxyribonuclease-B (DNaseB), and

iii) streptolysin-O (SLO).

In one embodiment, the biological sample is contacted with each of the following groups of streptococcus pyogenes antigens:

i) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No. 8;

ii) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No. 5; and

iii) an SLO antigenic fragment comprising, consisting essentially of or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

In one embodiment, two or more groups of streptococcus pyogenes antigens are present in the composition.

In one embodiment, one or more Streptococcus pyogenes antigens are labeled with a detectable label and/or are conjugated to a microparticle, bead or detectable reagent.

In one embodiment, one or more groups of Streptococcus pyogenes antigens are covalently bound to the bead or microparticle.

In one embodiment, the streptococcus pyogenes antigens of each group are covalently bound to the beads or microparticles, optionally wherein the beads or microparticles of different groups are distinguishable from each other.

In one embodiment, the beads are polystyrene beads, magnetic beads, carboxylated beads, functionalized beads, or wherein the microparticles are polystyrene microparticles, magnetic microparticles, carboxylated microparticles, or functionalized microparticles. In various examples, the beads are suitable for use in multiplex assays, such as those comprising two or more populations of beads or microparticles, wherein each population is conjugated to a different antigen. In various examples, the beads or microparticles are suitable for use in immunoassays, such as CBA, luminex assays, and the like.

In one embodiment, detecting the antigen-antibody complex comprises exposing the complex to a specific binding partner bearing a detectable label and detecting a signal from the label if the antigen-specific antibody is present in the biological sample.

In one embodiment, the specific binding partner comprises an antibody or fragment thereof.

In one embodiment, the specific binding partner is an anti-IgG antibody, an anti-IgG-PE, or a fragment thereof.

In one embodiment, the antigen-antibody complex is detected using a flow instrument, immunoassay, such as a plate-based immunoassay, electrophoresis and/or immunoblotting, immunochromatographic strip, electronic biosensor, resonance biosensor, or microfluidic device or sensor.

In one embodiment, the immunoassay, such as a plate-based immunoassay, is an ELISA or luminex assay.

In one embodiment, the antigen-antibody complex is detected in a luminex assay, such as the luminex assay exemplified herein.

In one embodiment, the presence of one or more complexes or one or more antigen-specific antibodies is detected using a detectably labeled second antibody.

In one embodiment, the detectably labeled second antibody is anti-IgG-PE.

In one embodiment, the Streptococcus pyogenes antigen is detectably labeled.

In one embodiment, the detectable label is a fluorophore.

In one embodiment, the biological sample is obtained from a mammalian species.

In one embodiment, the biological sample is a bodily fluid sample.

In one embodiment, the subject is a human subject.

In another aspect, the invention relates to an isolated, purified, or recombinant SpnA polypeptide, wherein the SpnA polypeptide has a relative abundance to a wild-type SpnA:

i) n-terminal truncation;

ii) a C-terminal truncation; or

iii) N-terminal and C-terminal truncations.

In one embodiment, the SpnA polypeptide

i) Is immunogenic, or

ii) is immunologically cross-reactive with wild type SpnA, or

iii) is detectably labeled, or

iv) increased stability upon storage at room temperature compared to wild type SpnA, or

v) comprises 10 or more consecutive amino acids from SEQ ID No.8, or

vi) any combination of two or more of the above i) to v).

In one embodiment, the SpnA polypeptide has an increased average Tagg compared to a wild-type SpnA, wherein Tagg is the temperature at which 50% of the protein aggregates, as determined, for example, by SDS-PAGE analysis. For example, the SpnA polypeptide has an average Tagg of at least about 50 ℃, as determined by SDS-PAGE analysis of at least about 50 ℃.

In one embodiment, the SpnA polypeptide has a greater degree of thermostability at a temperature of about 35 ℃ to about 60 ℃ as compared to a wild-type SpnA polypeptide.

In one embodiment, the polypeptide has enhanced thermostability, enhanced immunogenic stability, or both enhanced thermostability and enhanced immunogenic stability.

Again, for the avoidance of doubt, the skilled person will understand that any aspect disclosed herein, for example any of the methods described herein, will in certain embodiments employ one or more truncated streptococcus pyogenes nuclease a (SpnA) polypeptides, such as one or more truncated SpnA polypeptides described above, including one or more SpnA fragments described herein. For example, in various embodiments, one or more antigens or one or more antigenic groups from streptococcus pyogenes SpnA are present as one or more truncated SpnA polypeptides, such as one or more N-terminally truncated SpnA polypeptides, or one or more C-terminally truncated SpnA polypeptides, or one or more SpnA fragments, as described herein.

In another aspect, the invention relates to a composition comprising an isolated, purified, or recombinant SpnA polypeptide as described herein.

In another aspect, the invention relates to a composition comprising a detectably labeled SpnA polypeptide, such as a recombinant SpnA polypeptide described herein.

In one embodiment, the detectably labeled SpnA is a truncated SpnA polypeptide, one or more N-terminally truncated SpnA polypeptides, or one or more C-terminally truncated SpnA polypeptides, or one or more SpnA fragments, as described herein.

In another aspect, the invention relates to beads or microparticles comprising or having bound thereto one or more streptococcus pyogenes antigens or one or more groups of streptococcus pyogenes antigens, e.g., one or more streptococcus pyogenes nuclease a (spna) polypeptides as described herein.

In another aspect, the present invention relates to a composition comprising one or more beads or microparticles described herein.

In a further aspect, the present invention relates to a kit for detecting or diagnosing moist heat of stroke or PSGN in a subject, for detecting the presence of a streptococcus pyogenes infection in a subject, or for detecting the presence of antibodies specific for a streptococcus pyogenes antigen in a biological sample, the kit comprising a composition comprising at least one streptococcus pyogenes antigen selected from the group consisting of:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8, or

iv) deoxyribonuclease-B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5, or

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of, or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) an SLO antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No.2,

optionally at least one composition comprising a reference antibody control, wherein the antibody control comprises antibodies specific for one of the Streptococcus pyogenes antigens present in the kit,

optionally one or more reagents for constituting a medium for facilitating the contacting of the one or more antigens with the biological sample,

optionally one or more reagents capable of detecting complexes formed between the one or more antigens and one or more Streptococcus pyogenes antigen-specific antibodies present in the biological sample,

and instructions for use.

In one embodiment, at least one of the streptococcus pyogenes antigens is covalently bound to the bead or microparticle.

In one embodiment, the composition comprises the group of:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No. 8.

In one embodiment, the composition comprises each of the following groups of streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8,

and

iv) deoxyribonuclease-B (DNaseB); or

v) an antigenic fragment of DNaseB comprising, consisting essentially of or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5,

and

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) SLO antigenic fragments comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

In various embodiments of the methods provided herein, the method further comprises providing a kit described herein prior to step ii).

In various embodiments of the methods or kits provided herein, the one or more streptococcus pyogenes antigens are selected from the group consisting of anti-streptolysin (ASO), anti-hyaluronidase (AHase), anti-streptokinase (ASKase), anti-nicotinamide adenine dinucleotide enzyme (anti-NAD).

In one embodiment, the method comprises the use of a composition or kit comprising two or more groups of beads or microparticles, wherein each group of beads or microparticles comprises a different streptococcus pyogenes antigen, respectively, and wherein at least one group of beads or microparticles comprises a group of one of the following streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) a SpnA antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8,

and wherein the beads or microparticles are suitable for use in a flow instrument, immunoassay such as a plate-based immunoassay, electrophoresis and/or immunoblotting, immunochromatographic strip, electronic biosensor, resonance biosensor, or microfluidic device or sensor, including for example ELISA, Luminex or CBA assays.

In various embodiments, the use is in any of the methods described herein. For example, the application is an application including, but not limited to, any of: a method of detecting recent exposure to Streptococcus pyogenes in a subject, a method of detecting or diagnosing rheumatic fever or post-streptococcal glomerulonephritis (PSGN) including post-acute streptococcal glomerulonephritis (APSGN) in a subject, a method of detecting or diagnosing an increased likelihood of developing rheumatic fever or PSGN in a subject, a method of detecting the presence of a Streptococcus pyogenes infection in a subject, a method of detecting Streptococcus pyogenes antigen-specific antibodies in a biological sample, a method of treating a patient having rheumatic fever or PSGN with an antibiotic effective against Streptococcus pyogenes, a method of treating a patient having or having been exposed to a Streptococcus pyogenes infection, a method of predicting the responsiveness of a patient having rheumatic fever or PSGN to antibiotic treatment, a method of determining a treatment regimen for a patient having rheumatic fever or PSGN, or detecting recent exposure to Streptococcus pyogenes in the subject.

Optionally, one or more of the two or more bead or microparticle populations comprises a population of one of the following streptococcus pyogenes antigens:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8, or

iv) deoxyribonuclease B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5, or

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) SLO antigenic fragments comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

In another aspect, the invention relates to: an isolated, purified, or recombinant SpnA polypeptide, beads or microparticles comprising or having bound thereto one or more streptococcus pyogenes antigens or one or more groups of streptococcus pyogenes antigens (e.g., one or more streptococcus pyogenes nuclease a (SpnA) polypeptides), a composition comprising an isolated, purified, or recombinant SpnA polypeptide described herein, a composition comprising a detectably labeled SpnA, a composition comprising one or more beads or microparticles described herein, a kit for detecting or diagnosing rheumatic fever or PSGN in a subject, a kit for detecting the presence of a streptococcus pyogenes infection in a subject, or a kit for detecting an antibody specific for a streptococcus pyogenes antigen in a biological sample, a kit for any one of: detecting recent exposure of streptococcus pyogenes in the subject; detecting or diagnosing rheumatic fever or post-streptococcal glomerulonephritis (PSGN) including post-acute streptococcal glomerulonephritis (APSGN) in a subject; detecting or diagnosing an increased likelihood of developing rheumatic fever or PSGN in a subject; detecting the presence of a streptococcus pyogenes infection in a subject; detecting antibodies specific for a streptococcus pyogenes antigen in a biological sample; treating patients with rheumatic fever or PSGN; treating a patient with rheumatic fever or PSGN with an antibiotic effective against streptococcus pyogenes; treating a patient with an antibiotic effective against Streptococcus pyogenes, wherein the patient is suffering from or has been exposed to a Streptococcus pyogenes infection; predicting the responsiveness of a patient with rheumatic fever or PSGN to antibiotic treatment; determining a treatment regimen for a rheumatic fever or PSGN patient; or detecting recent exposure to streptococcus pyogenes in the subject.

In various embodiments, the polypeptide, bead or microparticle, composition or kit is or comprises at least one of the streptococcus pyogenes antigens selected from the group consisting of:

i) streptococcus pyogenes nuclease A (SpnA), or

ii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of the amino acid sequence of SEQ ID No.8, or

iii) an antigenic fragment of SpnA comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.8, or

iv) deoxyribonuclease B (DNaseB), or

v) an antigenic fragment of DNaseB comprising, consisting essentially of or consisting of the amino acid sequence of SEQ ID No.5, or

vi) a DNaseB antigenic fragment comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids of the amino acid sequence of SEQ ID No.5, or

vii) streptolysin-O (SLO), or

viii) an SLO antigenic fragment comprising, essentially consisting of or consisting of the amino acid sequence of SEQ ID No.1 or SEQ ID No.2, or

ix) SLO antigenic fragments comprising, consisting essentially of, or consisting of at least 10 contiguous amino acids in the amino acid sequence of SEQ ID No.1 or SEQ ID No. 2.

In various embodiments, the subject or patient is a subject having an amount or titer of anti-streptococcus pyogenes SpnA antibodies above a threshold, wherein an amount or titer of said antibodies above said threshold is indicative of a recent exposure to streptococcus pyogenes. For example, the threshold is the Upper Limit of Normal (ULN), the 80 th percentile of the matched healthy population.

In other embodiments, the subject or patient is a subject having an amount or titer of anti-streptococcus pyogenes SpnA antibodies below a threshold, wherein an amount or titer of said antibodies below said threshold is indicative of a prior exposure to streptococcus pyogenes. For example, the threshold is the Upper Limit of Normal (ULN), the 80 th percentile of the matched healthy population.

In another aspect, the invention relates to a method for detecting recent exposure of a subject to streptococcus pyogenes, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in the detection of one or more complexes above the threshold value indicates a recent exposure of the subject to Streptococcus pyogenes.

In another aspect, the invention relates to a method for detecting or diagnosing rheumatic fever or post-streptococcal glomerulonephritis (PSGN), including post-acute streptococcal glomerulonephritis (APSGN), or an increased likelihood of developing rheumatic fever or PSGN in a subject, comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in the detection of one or more of the complexes above the threshold value is indicative of an increased likelihood of development of rheumatic fever or APSGN, or of a recent exposure of the subject to streptococcus pyogenes as an indicator of the presence of rheumatic fever or PSGN in the subject;

iv) assessing one or more diagnostic indicators of rheumatic fever or PSGN in the subject;

v) wherein an increase in detection of the one or more complexes above a threshold value, in combination with one or more other diagnostic indicators of rheumatic fever or APSGN, is indicative of rheumatic fever or APSGN in the subject.

In one embodiment, the one or more diagnostic indicators are the presence or absence of one or more clinical symptoms associated with rheumatic fever or PSGN.

In one embodiment, the one or more clinical symptoms are selected from the group consisting of migratory polyarthritis, myocarditis, hematuria, erythema marginale, subcutaneous nodules, sydenham chorea, and pyoderma.

In another aspect, the invention relates to a method for detecting the presence of a streptococcus pyogenes infection in a subject, the method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more of the complexes indicates the presence of Streptococcus pyogenes in the subject or a recent exposure of the subject to Streptococcus pyogenes.

In another aspect, the invention relates to a method for detecting an antibody specific for a streptococcus pyogenes antigen in a biological sample, wherein said antibody specific for a streptococcus pyogenes antigen specifically binds streptococcus pyogenes SpnA, said method comprising:

i) providing a biological sample from a subject, said biological sample being capable of or suspected of containing one or more antibodies specific for streptococcus pyogenes SpnA;

ii) contacting the biological sample with the one or more groups of antigens from streptococcus pyogenes SpnA, wherein the one or more groups of streptococcus pyogenes SpnA antigens are capable of binding to antigen-specific antibodies present in the biological sample to form one or more groups of antigen-specific antibody complexes, if present in the biological sample; and

iii) detecting the complexes, wherein an increase in detection of one or more of the complexes indicates that the biological sample contains antibodies specific for Streptococcus pyogenes antigens.

In one embodiment, the post-streptococcal glomerulonephritis is post-acute streptococcal glomerulonephritis (APSGN).

In various embodiments, the increase in detection of the one or more complexes is an increase in detection of the one or more complexes above a threshold.

In one embodiment, a reference level, reference threshold, or threshold (also referred to herein interchangeably as a cutoff value) is determined for a particular population. In one example, a recent onset of rheumatic fever (ARF) in a given country, e.g., an ARF in NZ, uses a group of healthy, well-matched volunteers to determine the threshold. The cutoff value (or reference level) thus established is then applied to all ARFs in NZ. It will be appreciated that the reference threshold may vary between countries, ethnic groups and populations, and thus in some embodiments, different countries or groups each determine their own reference threshold. In one embodiment, the reference level, reference threshold, or threshold is the Upper Limit of Normal (ULN), i.e., the 80 th percentile of the matched healthy population.

In one embodiment, the reference threshold for detecting a streptococcus pyogenes antigen is the average titer of the antibody observed in samples obtained from patients with rheumatic fever or PSGN within 20 days of hospitalization. For example, the streptococcus pyogenes antigen reference threshold for use in the methods described herein is the mean antigen-specific antibody titer observed in samples obtained using the methods described herein in a population of patients with rheumatic fever or PSGN that is demographically comparable to the subject within 20 days of hospitalization.

In one embodiment, for detecting an anti-SpnA antibody: the reference threshold for SpnA complexes, e.g., the SpnA reference threshold for determining recent exposure to streptococcus pyogenes, is the average anti-SpnA antibody titer observed in samples obtained from patients with rheumatic fever or PSGN patients within 20 days of hospitalization. For example, the reference threshold for SpnA for determining recent exposure to streptococcus pyogenes is the mean anti-SpnA antibody titer observed in samples obtained using the methods described herein in a population of patients with rheumatic fever or PSGN that is demographically comparable to the subject within 20 days of hospitalization.

Other objects, aspects, features, and advantages of the present invention will become apparent from the following description. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.

Drawings

Other aspects of the invention will become apparent from the following description, given by way of example only, with reference to the accompanying drawings, wherein:

FIG. 1 shows a schematic view of aScatter plots are given showing the correlation between the median fluorescence values (MFI) for singleplex and multiplex (multiplex) by cytometric bead array method (cytometric bead array) for SLO (fig. 1A), dnase B (fig. 1B) and SpnA (fig. 1C) in 10 serum samples. Performing linear regression analysis and determining R 2The following were used: SLO ═ 0.999; dnase B ═ 0.998; and SpnA ═ 0.998;

FIG. 2 shows the ELISA results for purified IgG against three group A streptococcal antigens. The antibody was purified from IVIG using affinity chromatography to yield IgG specific for slo (a), dnase b (b), and spna (c). Error bars represent standard deviation;

FIG. 3 gives a scatter plot showing serum antibody concentrations determined by the cytometry bead array method for SLO (Panel A), DNaseB (Panel B) and SpnA (Panel C). The ULN value for each antigen is shown (dashed line). Kruskal-Wallis one-way analysis of variance was performed to determine the p-value;

FIG. 4 shows a scatter plot showing commercial assays and cells for SLO (A), DNaseB (B)Correlation between the bead array methods. Performing linear regression analysis and determining R 2SLO ═ 0.968 and DNaseB ═ 0.934;

FIG. 5 shows standard curves for SLO, DNAse B and SpnA for cytometry bead Array methods fitted with a five parameter logistic formula on FCAP Array software. SLO (500ng/ml), DNAse B (500ng/ml) and SpnA (1500ng/ml) specific purified IgG were diluted two-fold and incubated with antigen-conjugated beads;

FIG. 6 depicts the amino acid sequence of a recombinant SLO fragment comprising amino acids 34-571 (FIG. 6A), while the amino acid sequence of a detoxified SLO analog is shown in FIG. 6B. Substituted amino acids are highlighted and underlined;

FIG. 7 depicts the amino acid sequence of a recombinant DNaseB fragment comprising amino acids 43-271;

FIG. 8 shows the amino acid sequence of a recombinant SpnA fragment comprising amino acids 28-854;

FIG. 9 presents three scatter plots comparing a single luminex assay (each antigen independently) with multiple luminex assays in which three antigen bead aliquots are mixed and incubated with test serum in a single assay. FIG. 9A shows the correlation between MFI of SLO in single and multiple luminex assays; FIG. 9B shows the correlation between MFI of DNaseB in the single luminex assay and the multiple luminex assay; FIG. 9C shows the correlation between MFI for SpnA in the single and multiple luminex assays. As described in example two herein;

FIG. 10 gives two scatter plots showing the correlation between the commercial assay and the luminex assay for SLO (FIG. 10A) and DNaseB (FIG. 10B). Performing linear regression analysis, and subjecting R 2Determined as SLO ═ 0.933 and DNaseB ═ 0.942;

figure 11 gives three graphs showing the level of IgG antibodies present in serum collected from patients as determined by the luminex assay, where the serum is split according to the number of days of hospitalization. No significant difference was observed in anti-SLO antibody concentrations in sera collected at hospitalization <20 days (fig. 11, left panel), nor in anti-dnase b antibody concentrations between these two groups (fig. 11, middle panel), compared to sera collected at hospitalization >20 days. In contrast, as described in example two herein, a significant decrease in anti-SpnA antibody concentration was observed in sera collected at >20 days of hospitalization compared to sera collected at <20 days of hospitalization (fig. 11, right panel).

Figure 12 gives three graphs showing the levels of IgG antibodies present in serum collected from patients as determined by the luminex assay, where the serum is split according to the number of days of hospitalization. No significant difference in anti-SLO antibody concentration was observed in sera collected at <20 days of hospitalization (fig. 12, left panel), nor was there any significant difference in anti-dnase b antibody concentration between these two groups (fig. 12, middle panel) compared to sera collected at >20 days of hospitalization. In contrast, as described in example three herein, a significant decrease in anti-SpnA antibody concentration was observed in sera collected at >20 days of hospitalization compared to sera collected at <20 days of hospitalization (fig. 12, right panel).

Fig. 13 presents two graphs showing analysis of the thermostability of native SpnA and the truncated SpnA polypeptides disclosed herein, as described in example four. FIG. 13A is a chromatogram of an SDS-PAGE analysis of full-length SpnA polypeptide stored under optimal conditions (day 0) and 5 days at room temperature (day 5). FIG. 13B is a chromatogram of an SDS-PAGE analysis of truncated SpnA polypeptides stored under optimal conditions (day 0) and 5 days at room temperature (day 5).

Fig. 14 presents a chromatogram and two graphs showing an analysis of the thermostability of native SpnA and the truncated SpnA polypeptides disclosed herein, as described in example five. FIG. 14A is an SDS-PAGE analysis chromatogram of the percentage of folded protein stored at various temperatures. Fig. 14B shows Tagg (temperature at which 50% of the protein aggregates) of each protein at each temperature. FIG. 14C is a graph of the mean Tagg values for each polypeptide, depicting a higher mean Tagg at 51.0+/-0.6 ℃ as determined for truncated constructs, significantly higher than the mean Tagg (47.5+/-0.9 ℃) as determined for native SpnA polypeptides.

Detailed Description

The present invention relates generally to methods and compositions for detecting the presence of GAS-specific antibodies in a biological sample. The detection of such antibodies can be used to identify subjects at increased risk of immune sequelae following infection, as well as to identify subjects who may benefit from a particular treatment, as well as other uses that will become apparent to those skilled in the art upon review of the following disclosure.

Throughout the specification and claims, the words "comprise", "comprising", and the like, are to be construed in an inclusive sense as opposed to an exclusive or exhaustive sense, that is, in a sense "including but not limited to", unless the context clearly requires otherwise.

In certain embodiments, the invention relates to a method for aiding in the diagnosis of rheumatic fever or post-streptococcal glomerulonephritis (PSGN) or assessing the propensity to develop rheumatic fever or PSGN in a subject, the method comprising determining the presence or amount of one or more antibodies specific for one or more streptococcus pyogenes antigens in a biological sample from the subject, wherein an elevated level of said antibodies in the biological sample relative to the level of said antibodies in a control is indicative of rheumatic fever or PSGN or is indicative of an increased propensity to develop rheumatic fever or PSGN.

In other embodiments, the invention relates to a method of determining the efficacy of a treatment for stroke moist heat or PSGN in a subject, the method comprising determining the presence or amount of one or more antibodies specific for one or more streptococcus pyogenes antigens in one or more biological samples obtained from the subject prior to or during the treatment, wherein a decrease in the level of the one or more antibodies specific for the one or more streptococcus pyogenes antigens in the sample obtained from the subject over time indicates that the treatment is effective.

In another embodiment, the invention relates to a method of selecting a subject for treatment of rheumatic fever or PSGN, the method comprising: (a) determining the presence or amount of one or more antibodies specific for one or more streptococcus pyogenes antigens in a biological sample obtained from the subject; (b) comparing the level of the antibody in the biological sample to the level of the antibody in a control; and (c) selecting the subject for treatment when the level of said antibody in the biological sample is higher than the level of said antibody in the control.

The present invention relies in part on the affinity of antibodies for antigenic components-the ability of antibodies to specifically recognize and bind to an antigen or epitope, and the determination of such specific recognition and binding. In other words, the present invention relies in part on the detection and identification of complexes formed when antibodies recognize and bind to particular epitopes.

The term "affinity" refers to a measure of the binding strength of an individual epitope to the Complementarity Determining Regions (CDRs) of a binding molecule, typically an immunoglobulin molecule, in the context of the present invention. The term "affinity" refers to the overall stability of the complex between a population of immunoglobulins and antigen(s), i.e., the functional binding strength of the immunoglobulin mixture to the antigen. One skilled in the art will appreciate that affinity is related not only to the affinity of each immunoglobulin molecule in the population for a particular epitope, but also to the valency of these immunoglobulins and antigens. For example, the interaction between a bivalent monoclonal antibody and an antigen (e.g., a polymer) with a highly repetitive epitope structure will be a high affinity. The affinity or avidity of an antibody for an antigen may be determined experimentally using any suitable method well known in the art, including certain methods described herein. General techniques for measuring the affinity of an antibody for an antigen include ELISA, RIA and surface plasmon resonance. The measured affinity of a particular antibody-antigen interaction may differ if measured under different conditions (e.g., salt concentration, pH, buffer, temperature). Thus, especially when data comparison is required, affinity and other antigen binding parameters (e.g., KD, IC 50) are typically measured using standardized solutions of antibody and antigen, standardized buffers, and standardized assay conditions.

"specific binding" or "specific recognition" as used interchangeably herein generally refers to binding molecules, such as antibodies, that bind to an epitope through their antigen binding domain, and binding requires some complementarity between the antigen binding domain and the epitope. Thus, an antibody is said to "specifically bind" to an epitope when it more readily binds to that epitope through its antigen binding domain than it does to a random, unrelated epitope. Such antibodies may be referred to herein as "specific" antibodies to the epitope or epitope class. The term "specificity" is used herein to characterize the relative affinity of an antibody for binding to an epitope. For example, antibody "a" can be said to have a higher specificity for a given epitope than antibody "B", or antibody "a" can be said to have a higher binding specificity for epitope "x" than it does for the relevant epitope "y".

As used herein, reference to "determining" includes estimating, quantifying, calculating, or otherwise deducing the amount of a reference substance (e.g., an antibody, antigen, or biomarker) present in a particular sample. This can be accomplished by measuring an endpoint indicator, which can be, for example, the appearance of a detectable product, any detectable change (e.g., a change in the level of substrate), or any change in the rate of appearance of a product or disappearance of a substrate, or measuring the amount of antibody bound to an antigen, biomarker, complex, or other agent, as described herein.

The term "immunological binding characteristics" or other binding characteristics of an antibody to an antigen, when present, refers to the specificity, affinity, cross-reactivity, and other binding characteristics of the antibody, in various grammatical expressions.

As used herein, the terms "immunogenic stability", "immune stability" and grammatical equivalents encompass the maintenance of one or more immunogenic or immunological characteristics, such as the ability to produce a given (including specific) immune response, such as the ability to be bound or recognized by an antibody, or the ability to elicit a cell-mediated immune response. For example, when used in connection with a particular antigen, polypeptide, or other agent, immunogenic stability or immune stability includes, for example, maintenance of specific recognition by the antibody, such as the ability of the antigen, polypeptide, or other agent to be bound by a specific antibody. It is understood that immune stability may also refer to the stability of an antibody, such as maintaining the ability to recognize and bind an epitope.

As used herein, the term "preferentially binds" encompasses binding, e.g., binding of an antibody to one epitope occurs more readily than to another epitope (e.g., a related, similar, homologous, or similar epitope). Thus, an antibody that "preferentially binds" to a given epitope will be more likely to bind to that epitope than to another epitope, even where such an antibody may exhibit some cross-reactivity with another epitope.

For example, if the antibody is smaller than the antibody K to the second epitope DDissociation constant (K) of D) Binding to the first epitope, the antibody can be considered to bind preferentially to the first epitope. In one embodiment, antibody K is a second epitope if the antibody aligns to the second epitope DAn antibody can be considered to bind preferentially to the first antigen if it binds to the first epitope with an affinity that is at least one order of magnitude less. In another embodiment, antibody K is a second epitope if the antibody is aligned with the second epitope DAn antibody can be considered to bind a first epitope preferentially if it binds the first epitope with an affinity that is at least two orders of magnitude less.

In another example, an antibody may be considered to preferentially bind a first epitope if it binds the first epitope with a binding rate constant (onrate, (k (on)) that is less than the binding rate constant (k (on)) of antibody k (on) to the second epitope. In one embodiment, an antibody may be considered to preferentially bind a first epitope if it binds the first epitope with a k (on) that is at least one order of magnitude greater than antibody k (on) of the second epitope. In another embodiment, an antibody can be considered to preferentially bind a first epitope if it binds the first epitope with a k (on) that is at least two orders of magnitude greater than antibody k (on) of the second epitope.

In other examples, an antibody may be considered to preferentially bind a first epitope if it binds the first epitope with an off rate constant (off (k) (off)) that is less than the antibody k (off) for the second epitope.

In certain methods useful herein, competitive binding of two or more antibodies is typically assessed by assessing the ability of one antibody to inhibit the binding of one or more other antibodies to an epitope. In certain embodiments, an antibody is considered to competitively inhibit binding of a reference antibody to a given epitope if it preferentially binds to that epitope such that binding of the reference antibody to that epitope is blocked to some extent. Competitive inhibition can be determined by any method known in the art, such as a competitive ELISA assay. Generally, an antibody can be said to competitively inhibit binding of a reference antibody to a given epitope by at least 90%, at least 80%, at least 70%, at least 60%, or at least 50%.

An antibody or antigen-binding fragment thereof, variant or derivative thereof as described herein may also be described or defined in terms of its cross-reactivity. As used herein, the term "cross-reactivity" refers to the ability of an antibody specific for one antigen to recognize and bind to a second antigen. Typically, the observation of antibody cross-reactivity is taken as a measure of the correlation between two (or more) different antigenic substances that support this cross-reactivity. Thus, an antibody is cross-reactive if it binds to an epitope other than the epitope that induced its formation.

It is to be understood that in certain instances, epitopes may also be referred to or described as cross-reactive, whereby two or more different epitopes may be recognized and/or bound by one particular antibody. A cross-reactive epitope typically comprises many of the same complementary structural features as the inducing or initiating epitope. In some cases, the cross-reactive epitope may be bound with greater affinity than the starting epitope.

It will be appreciated that for diagnostic purposes, antibodies and antigens with low or no cross-reactivity are generally preferred.

As used herein, the term "polypeptide" is intended to encompass a single "polypeptide" as well as a plurality of "polypeptides" and refers to a molecule consisting of monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds). The term "polypeptide" refers to any chain or chains of two or more amino acids and does not refer to a particular length of the product. Thus, included within the definition of "polypeptide" are peptides, dipeptides, tripeptides, oligopeptides, "proteins," "amino acid chains," or any other term used to refer to one or more chains of two or more amino acids, and the term "polypeptide" may be used instead of or interchangeably with any of these terms.

The term "polypeptide" is also intended to refer to the product of post-expression modification of the polypeptide, including, but not limited to, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification of non-naturally occurring amino acids. The polypeptides may be derived from natural biological sources or produced by recombinant techniques, but are not necessarily translated from a specified nucleic acid sequence. It may be produced in any manner, including by chemical synthesis.

The polypeptides of the invention may be about 3 or more, 5 or more, 10 or more, 20 or more, 25 or more, 50 or more, 75 or more, 100 or more, 200 or more, 500 or more, 1,000 or more, or 2,000 or more amino acids in size. Polypeptides may have a defined three-dimensional structure, although they do not necessarily have such a structure. The term glycoprotein refers to a protein coupled to at least one carbohydrate moiety linked to the protein by an oxygen-or nitrogen-containing side chain of an amino acid residue, such as a serine residue or an asparagine residue.

An "isolated" polypeptide or fragment, variant or derivative thereof refers to a polypeptide that is not in its natural environment. No particular level of purification is required. For example, an isolated polypeptide may be removed from its natural or native environment. For the purposes of the present invention, recombinantly produced polypeptides and proteins expressed in host cells are considered isolated, and native or recombinant polypeptides that have been isolated, fractionated or partially or substantially purified by any suitable technique are also considered isolated.

The polypeptides of the present invention also include fragments, derivatives, analogs, or variants of the above polypeptides, and any combination thereof. When referring to an antibody or antibody polypeptide of the invention, the terms "fragment", "variant", "derivative" and "analogue" include any polypeptide that retains at least some of the antigen binding properties of the corresponding native binding molecule, antibody or polypeptide. Fragments of the polypeptides of the invention include proteolytic fragments as well as deletion fragments. Variants of the polypeptides include fragments as described above, as well as polypeptides having altered amino acid sequences due to amino acid substitutions, deletions, or insertions. Variants may occur naturally or non-naturally. Non-naturally occurring variants can be generated using mutagenesis techniques known in the art. Variant polypeptides may comprise conservative or non-conservative amino acid substitutions, deletions or additions. Variant polypeptides may also be referred to herein as "polypeptide analogs". Derivatives of polypeptides are considered herein to be polypeptides that have been altered to exhibit other characteristics not found on the native polypeptide. Examples include fusion proteins or functionally modified polypeptides, such as pegylated polypeptides, polypeptides attached to beads, and polypeptides covalently linked to one or more other agents or compounds. In one embodiment, a "derivative" of a polypeptide refers to the subject polypeptide having one or more residues chemically derivatized by functional side chain group reaction. "derivatives" also include those peptides that contain naturally occurring amino acid derivatives of the twenty standard amino acids. For example, proline may be substituted with 4-hydroxyproline; lysine may be substituted with 5-hydroxylysine; histidine may be replaced by 3-methylhistidine; homoserine may be substituted for serine; ornithine may be substituted for lysine.

In one embodiment, an analog of a specifically identified polypeptide can have about 70% sequence identity to the specified polypeptide sequence (e.g., the amino acid sequence shown in the figure or a fragment thereof), for example, over a sequence of 10 or more contiguous amino acids. That is, 70% of the residues of each polypeptide are identical. In another embodiment, analogs of the specifically identified polypeptides will have greater than 75% identity. In another embodiment, analogs of the specifically identified polypeptides will have greater than 80% identity. In another embodiment, analogs of the specifically identified polypeptides will have greater than 85% identity. In another embodiment, analogs of the specifically identified polypeptides will have greater than 90% identity. In another embodiment, analogs of the specifically identified polypeptides will have greater than 95% identity. In another embodiment, analogs of the specifically identified polypeptides will have greater than 99% identity. In another embodiment, analogs of a polypeptide of the invention will have fewer than about 20 amino acid residue substitutions, modifications, or deletions, relative to the sequence of a given polypeptide, e.g., fewer than 10.

In another embodiment, the polypeptide will have greater than 70% homology. In another embodiment, the polypeptide will have greater than 75% homology. In another embodiment, the polypeptide will have greater than 80% homology. In another embodiment, the polypeptide will have greater than 85% homology. In another embodiment, the polypeptide will have greater than 90% homology. In another embodiment, the polypeptide will have greater than 95% homology. In another embodiment, the polypeptide will have greater than 99% homology. In another embodiment, derivatives and analogs of the polypeptides of the invention will have fewer than about 20 amino acid residue substitutions, modifications or deletions, more preferably fewer than 10. Preferred substitutions are conservative substitutions known in the art, i.e., the substituted residues share physical or chemical properties, such as hydrophobicity, size, charge, or functional groups.

Analogs of the polypeptides having a specified degree of identity over a specified number of consecutive amino acid residues are also contemplated. For example, in one embodiment, an analog of a given polypeptide has greater than 90% amino acid sequence identity over 10 or more contiguous amino acids of the reference/given sequence. In another embodiment, an analog of a given polypeptide has greater than 90% amino acid sequence identity over 20 or more contiguous amino acids of the reference/given sequence.

The term "polynucleotide" is intended to encompass both a single nucleic acid and a plurality of nucleic acids, and refers to an isolated nucleic acid molecule or construct, including, for example, messenger rna (mrna) or plasmid dna (pdna). Polynucleotides may comprise conventional phosphodiester bonds or unconventional bonds (e.g., amide bonds, as found in Peptide Nucleic Acids (PNAs)). The term "nucleic acid" refers to any one or more nucleic acid fragments, such as DNA or RNA fragments, present in a polynucleotide. An "isolated" nucleic acid or polynucleotide refers to a nucleic acid molecule, DNA or RNA, that has been removed from its natural environment. For example, for the purposes of this disclosure, a recombinant polynucleotide encoding an antibody or antigen contained in a vector is considered to be isolated. Other examples of isolated polynucleotides include recombinant polynucleotides maintained in heterologous host cells or (partially or substantially) purified polynucleotides in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the polynucleotides described herein. Isolated polynucleotides or nucleic acids described herein also include synthetically produced such molecules. In addition, the polynucleotide or nucleic acid may be or may include regulatory elements such as a promoter, ribosome binding site or transcription terminator.

As used herein, the term "sample" refers to any biological material obtained from a subject or patient, but it is to be understood that in most embodiments (e.g., methods) described herein, a sample in which one or more antibodies can be present will be preferred. In one embodiment, the sample may comprise blood, plasma, serum, cerebrospinal fluid ("CSF"), or urine. For example, the sample may comprise whole blood, plasma, B cells enriched from a blood sample, or cultured cells (e.g., B cells from a subject). Samples may also include biopsy or tissue samples, including mucosal or neural tissue. In other embodiments, the sample may comprise whole cells and/or lysates of cells. Methods of collecting and/or preparing samples are well known in the art.

By "subject" or "individual" or "animal" or "patient" or "mammal" is meant any subject, particularly a mammalian subject, e.g., a human patient, in need of diagnosis, prognosis, prevention or treatment.

As used herein, the term "treatment" refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as rheumatic fever or the occurrence or progression of APSGN. Beneficial or desired clinical results include, but are not limited to, remission, decreased disease extent, stable (i.e., not worsening) disease state, delayed or slowed disease progression, improved or reduced disease state, pathogen clearance, and remission (whether partial or total), whether detectable or undetectable. "treatment" may also mean an extended survival period as compared to the expected survival period without treatment. Those in need of treatment include those already with the disorder or condition as well as those prone to the disorder or condition or those in whom the manifestation of the disorder or condition is to be prevented.

In certain embodiments, the antigens described herein are used to quantitatively or qualitatively detect streptococcus pyogenes-specific antibodies in a sample. This can be achieved by techniques that provide a visually detectable signal, which can be any of fluorescence (immunofluorescence), chromogenic product of an enzymatic reaction, production of a precipitate, chemiluminescence, or bioluminescence. Certain embodiments use fluorescent or colored labeled antibodies in conjunction with light microscopy, flow cytometry, or fluorescence detection. Other techniques and labels that can be used to detect antibodies include, but are not limited to, colloidal gold, radiolabels, GFP (green fluorescent protein), etc., avidin/streptavidin-biotin, magnetic beads, and physical systems, such as nanotechnology systems, that are sensitive to actual binding.

The antigen, antibody or fragment thereof may be used for histological staining, such as immunohistochemistry, immunofluorescence or immunoelectron microscopy, and for in situ detection of the antibody or protein. One of ordinary skill will readily recognize that any of a variety of histological methods, such as staining procedures, may be modified to achieve such in situ detection.

One of the methods that can be used to label and directly detect the antibodies or antigens described herein is to attach the antibody or antigen to an enzyme and use in an Enzyme Immunoassay (EIA). upon subsequent exposure to a suitable substrate, the enzyme will react with the substrate to produce a chemical moiety that can be detected, for example, by spectrophotometry, fluorimetry, or by visual means.

In some embodiments, detection of the reaction of an antibody with an antigen may be further aided, where appropriate, by the use of a second antibody or other binding partner that binds the antigen, an antibody complex, which may be reactive with the antigen or more generally with the antibody. Typically, the second antibody or ligand is detectably labeled, wherein detection of the second antibody allows the presence of an antigen-antibody complex to be inferred.

Specific binding partners (e.g., secondary antibodies) are often reactive with conserved regions of immunoglobulins of the species from which the sample is derived. In the examples specifically contemplated herein, the second antibody or specific binding partner has affinity for a human immunoglobulin such as IgG. The choice of the second antibody will depend, at least in part, on the source of the sample and, therefore, on the nature of the antibody expected to be present therein.

Many well known detection methods are suitable for practicing the methods described herein. For example, enzyme immunoassays, such as immunofluorescence assays (IFA), photometric assays, enzyme-linked immunosorbent assays (ELISA), ELISPOT assays, and immunoblots, can be readily adapted for accomplishing the detection of specific antibodies.

Other detection systems that may also be used include those based on the use of protein a derived from staphylococcus aureus Cowan strain I, protein G derived from group C streptococcus (e.g., strain 26RP66), or systems that employ a biotin-avidin binding reaction.

Other immunoenzymatic detection methods in which the antigens and antibodies described herein may be used are western blotting and dot blotting, in which the reagents are separated by electrophoresis and transferred to nitrocellulose membrane or other suitable support. The sample to be tested (e.g. plasma) is then brought into contact with the membrane and the presence of the immunocomplexes formed is detected by the methods already described. In one variation of this method, the purified antigen is applied to the membrane in a line or spot and allowed to bind to any antibodies present in the sample, and the immune complexes formed are detected using the techniques described herein.

The presence of antibody-antigen complexes can also be detected by aggregation. In a representative example of such a method, the antigens described herein are used to coat, for example, latex particles to form a uniform suspension. When mixed with a sample (e.g., serum containing specific antibodies capable of recognizing an antigen), latex particles are caused to aggregate, and the presence of large aggregates can be visually detected.

The reaction of the detection antibody with the antigen may be facilitated using an antibody or ligand labeled with a detectable moiety by methods known in the art. Such detectable moieties allow visual detection of precipitates or color changes, visual detection by microscopy, or automatic detection by spectroscopy or radiometry, etc. Examples of detectable moieties include fluorescein and rhodamine (for fluorescence microscopy), horseradish peroxidase and alkaline phosphatase (for light or electron microscopy and biochemical detection, and for biochemical detection by color change), and biotin-streptavidin (for light or electron microscopy). The detection method and moieties used may be selected from the above list or other suitable examples according to standard conditions known in the art to apply to such selection.

Although methods that rely on radioisotopes are now generally less appreciated than before, radioactive detection methods can be used, which are accomplished by radiolabeling an antigen, antibody or antibody fragment and using Radioimmunoassay (RIA).

More commonly, the antibody or antigen to be detected is labeled with a non-radioactive detectable label, such as a fluorescent compound. When a fluorescently labeled antibody or antigen is exposed to light of the appropriate wavelength, the presence of the antibody or antigen can be detected due to fluorescence. The most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine.

Metals that emit fluorescence such as 152E or other lanthanide metals detectably label the antibody or antigen. These metals can be attached to antibodies or antigens using metal chelating groups such as diethylenetriaminepentaacetic acid (ETPA).

The antibody or antigen may also be detectably labeled by coupling to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody or antigen is then determined by detecting the presence of luminescence generated during the chemical reaction. Examples of particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.

Also, bioluminescent compounds can be used to label the antibody or antigen. Bioluminescence typically occurs in biological systems through the activity of catalytic proteins, which increases the efficiency of the chemiluminescent reaction. The presence of a given bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for labeling purposes are luciferin, luciferase and aequorin.

Specific examples of methods for detecting antibodies in biological samples are disclosed, for example, in the following patents, including methods employing dipsticks or other immobilized assay devices: U.S. Pat. No.5,965,356 (herpes simplex virus-like specific serological assay); U.S. Pat. No.6,114,179 (methods and test kits for detecting antigens and/or antibodies); U.S. Pat. No.6,077,681 (diagnosis of motor neuropathy by detection of antibodies); us patent No.6,057,097 (markers for pathology including autoimmune reactions and/or for inflammatory diseases); and U.S. patent No.5,552,285 (immunoassay methods, compositions, and kits for antibodies to oxidized DNA bases).

For example, microsphere assays (also known as flow bead assays) may also be used to detect one or more antibodies specific for one or more streptococcus pyogenes antigens in a biological fluid, such as a blood or serum sample from a subject. This technique, represented by the system developed by Luminex Corporation and other systems developed by Becton Dickinson, enables the processing of very small volumes of sample (typically 20. mu.L) for the detection of one or several analytes. The principle of the assay is based on coupling a capture antibody to a microsphere containing specific amounts of red dye and infrared dye. After incubation of these microspheres with the sample, Phycoerythrin (PE) -conjugated second detection antibody, the beads were analyzed by flow cytometry. One laser detects the beads and the other laser detects the intensity of the PE bound to these beads. This technique has been used in multiplex assays to detect a number of biologically important molecules or agents, including cytokines, serotyping of Streptococcus pneumoniae (Streptococcus pneumoniae), simultaneous measurement of human chorionic gonadotropin (hCG) and alpha-fetoprotein (AFP), simultaneous detection of serum IgG for Toxoplasma (Toxoplasma gondii), rubella virus, cytomegalovirus, and herpes simplex virus types 1 and 2 (see the technical description provided by luminexcomp. e.g., on its website or by its catalog of products).

In certain embodiments, one or more antibodies specific for one or more streptococcus pyogenes antigens present in the biological sample bind to antigenic proteins coupled to the microspheres. In some embodiments, the second detection antibody (an example of which is also referred to herein as a specific binding partner) is a monoclonal antibody, e.g., a monoclonal antibody against human IgG. In other embodiments, the second detection antibody is a polyclonal antibody, such as a polyclonal antibody directed against human IgG. The second antibody used in such methods may be conjugated to, for example, biotin, or otherwise detectably labeled.

Immunological techniques suitable for use herein include immunological methods that rely on the formation of antibody-antigen complexes, for example, using labeled antibodies or antibody fragments, including labeled secondary antibodies. Exemplary methods include ELISA methods, such as indirect ELISA, competitive ELISA, sandwich ELISA, and other immunological-based methods, reagents or devices, such as immunochromatographic strips, fluorescent immunomicroparticles, western blots, biosensors based on electrochemical reactions catalyzed by enzymes linked to antibodies, magnetic particles coated with antibodies, surface plasmon resonance, and other techniques for detecting analytes bound to antibodies. Various methods and techniques exist for implementing these methods and will be well known to those skilled in the art and may include microfluidic techniques, automation, and/or high throughput techniques.

Suitable assays may be quantitative techniques such as ELISA, or qualitative, such as rapid immunochromatographic assays, immunoblots, dipsticks, and the like. It will be appreciated that in some cases assays commonly used as quantitative assays may be employed qualitatively.

Qualitative, alternative rapid assays, such as immunochromatographic assays, particularly those employing immunochromatographic strips, are particularly useful in areas (e.g., developing countries) where more complex assay techniques are not feasible or practical, or as a first-line diagnosis to identify subjects who may benefit from further evaluation.

For example, in one embodiment of a qualitative assay that detects whether the presence or amount of one or more antibodies specific for one or more streptococcus pyogenes antigens is greater than or less than a certain amount, a qualitative ELISA assay is used in which two or more antigens described herein are used. The procedure may be performed with a kit as described herein, e.g. the kit may contain a composition, such as a buffer or sample preparation solution (which comprises one or more streptococcus pyogenes antigens), optionally a negative control (in which no antibodies specific for the streptococcus pyogenes antigens are present), optionally a positive control (in which one or more antibodies specific for the streptococcus pyogenes antigens are present), and components of an ELISA (such as a reaction vessel, e.g. a multi-well plate), and one or more second detection antibodies for detecting complexes formed between one or more antibodies specific for the streptococcus pyogenes antigens and the one or more streptococcus pyogenes antigens, and optionally one or more components for immobilizing the complexes and/or for determining color development. In one embodiment, first, a composition comprising one or more streptococcus pyogenes antigens is introduced into a reaction vessel, such as an ELISA plate, under conditions that immobilize the one or more streptococcus pyogenes antigens on the vessel. In another embodiment, a reaction vessel, such as an ELISA plate, is provided having one or more streptococcus pyogenes antigens already immobilized thereon. In another particularly contemplated example, one or more second detection antibodies may already be immobilized on or in the reaction vessel, or may be provided in a composition and introduced into the reaction vessel under conditions that allow the detection antibodies to be immobilized on the reaction vessel. Those skilled in the art will appreciate that there are various alternative methods in which the formation, capture and labeling of the complex dependent on the analyte may provide a detectable signal, and these methods typically rely on the immobilization of an antibody-antigen complex for detection.

In another embodiment of the qualitative assay, an immunochromatographic strip is used, in which the presence of one or more antibodies specific for one or more Streptococcus pyogenes antigens is determined, or in which it is determined whether the amount of said antibodies is greater than or less than a certain threshold. In one example, the test strip comprises two or more antigens described herein, and is typically provided with one or more compositions, such as a buffer solution or sample preparation solution, optionally a negative control (in which no antibodies specific for streptococcus pyogenes antigens are present), optionally a positive control (in which one or more antibodies specific for one or more antigens are present), and a specific binding partner for detecting complexes formed between one or more antibodies specific for one or more streptococcus pyogenes antigens and one or more streptococcus pyogenes antigens, and optionally one or more components for determining color development. In one example, a separate immunochromatographic strip in which streptococcus pyogenes antigen is absent is provided to serve as a negative control. In other embodiments, the one or more immunochromatographic strips have at least two distinct zones, wherein one zone comprises one or more streptococcus pyogenes antigens, optionally wherein another zone does not comprise streptococcus pyogenes antigens, and optionally another zone comprises one or more immobilized antibodies or other reagents capable of being bound by a second detection antibody or specific binding partner as a positive control zone.

In a particular embodiment, the detection is performed by capture ELISA. A capture ELISA (also known as a "sandwich" ELISA) is a sensitive assay for quantifying very small amounts (from picograms to micrograms) of substances (such as hormones, cell signaling chemicals, infectious disease antigens and cytokines, and in the context of this disclosure, antibodies). This type of ELISA is generally considered when the substance to be analyzed may be too dilute to bind to a support material such as polystyrene microtiter plates (e.g., proteins in cell culture supernatants), or may bind poorly to plastics (e.g., small organic molecules). The optimal dilutions of capture reagents (e.g., capture antigen), sample, control and detection antibodies, and incubation times are often determined empirically, and may require extensive titration. Ideally, an enzyme-labeled detection antibody or detection antigen may be used. However, if the detection antibody or antigen is unlabeled, the second antibody should not cross-react with the coating antibody or antigen.

As used herein in this specification, the terms "detectable moiety," "detectable label," and grammatical equivalents refer to any atom, molecule or portion thereof, agent or substance whose presence, absence or level can be monitored directly or indirectly. One example includes a radioisotope. Other examples include (i) enzymes that can catalyze a color or luminescence (luminescence) reaction and (ii) fluorophores. The detection of the detectable moiety may be direct, provided that the detectable moiety itself is detectable, for example in the case of a fluorophore. Alternatively, detection of the detectable moiety may be indirect. In the latter case, a second moiety is typically used which reacts with the detectable moiety, the second moiety itself being directly detectable. The detectable moiety may be inherent to the antibody or labelled antigen, for example by covalent linkage. For example, the constant region of an antibody can be used as an indirectly detectable moiety that can specifically bind to a second antibody having a directly detectable moiety.

Thus, a second antibody is a particularly suitable means for detecting the antibody in the methods described herein, which may itself be conjugated to a detectable moiety one method that may be used to detectably label an antibody according to the invention is to link it to an enzyme which, upon subsequent exposure to a suitable substrate, will then react with the substrate to produce a chemical moiety which may be detected by, for example, spectrophotometric, fluorescent or visual means.

Detection can be accomplished by colorimetric methods that use chromogenic substrates for enzymes. Detection can also be accomplished by visual comparison of the extent of enzymatic reaction of the substrate with similarly prepared standards.

In embodiments (e.g., embodiments of methods, assays, and kits) that employ a solid support bound to a reagent (e.g., an antigen or an antibody, as applicable), the solid support can be any water-insoluble, non-suspended solid support. Examples of suitable solid supports include, for example, beads of polystyrene, filter paper, test tubes, dipsticks, and microtiter plates. The bound agent may be bound to the solid support by covalent bonds or by adsorption. An advantage of using a solid support is that no centrifugation step is required for separation of the solid and liquid phases, although centrifugation, such as bead-based assays, may be used in certain embodiments to facilitate processing.

Such solid supports may comprise polymers such as polystyrene, agarose, sepharose, cellulose, glass beads, and magnetizable particles of cellulose or other polymers. The solid support may be in the form of large or small beads or particles, tubes, plates, strips, or other forms.

As solid support, typically test tubes or microtiter plates are used, the inner wall of which is coated with a first antibody or antigen, e.g., an antigen specific for the streptococcus pyogenes antibody to be detected or any fragment or derivative thereof, such as those specifically disclosed herein.

It is also contemplated to provide a kit comprising one or more components necessary to perform one or more of the methods described herein. The kit will typically comprise a composition comprising at least one streptococcus pyogenes antigen as described herein, e.g. a composition comprising two or more of said antigens. Other components useful for performing the methods according to the present disclosure may be usefully included in the kit. For example, a kit may comprise one or more reagents for constituting a medium for facilitating contacting the one or more antigens with a biological sample. The kit may also comprise a device for sample collection, such as a swab, pipette or similar collection means, and a device for performing one or more reaction steps of contacting the reagents, such as an incubation device (including a liquid or semi-solid medium placed on a plate, test tube, glass or plastic surface, well, or on an absorbent paper strip), or similar device. Other kit components may include one or more reagents that enable detection of a complex formed between one or more antigens and one or more streptococcus pyogenes antigen-specific antibodies present in a biological sample.

In certain embodiments, the methods, assays, and kits contemplated herein may include or utilize contacting one or more samples with other assay reagents (e.g., one or more streptococcus pyogenes antigens), wherein the latter are arranged in an array to allow, for example, rapid processing of multiple samples, including high throughput performance.

As used herein, the term "array" refers to a "addressed" spatial arrangement of one or more identifiers (e.g., a combination of two or more streptococcus pyogenes antigens). Each "address" of the array is a predetermined specific spatial region containing one or more identifiers. For example, the array may be a plurality of wells (test tubes), plates, microwells in a microplate, each containing a set of antigens. Various array embodiments are contemplated. In one embodiment, each address contains a similar or identical identifier and multiple samples are assayed, one sample at each address. In another embodiment, each address contains a different identifier or a different combination of identifiers, and an aliquot of a single sample is introduced to each address. In other embodiments, a combination of these methods is employed. The array may also be any solid support that holds known identifiers in different areas (spots, lines, columns), e.g., where different identifiers are placed in different combinations at one or more different locations, or contact different samples at different locations. In certain embodiments, the array includes built-in appropriate controls, e.g., a region without the sample, a region without the antigen, a region without either (e.g., with only solvent and reagents), and a region containing synthetic or isolated antibodies (as positive controls) that bind the one or more antigens.

Solid supports useful herein, e.g., solid supports for arrays or kits, are typically substantially insoluble in the liquid phase. The solid support of the present invention is not limited to a specific type of support. In fact, a large number of supports are available and known to those of ordinary skill in the art. Thus, useful solid supports include solid and semi-solid substrates such as aerogels and hydrogels, resins, beads, biochips (including biochips coated with thin films), microfluidic chips, silicon chips, multiwell plates (also known as microtiter plates or microplates), membranes, filters, conductive and non-conductive metals, glass (including microscope slides), and magnetic supports. More specific examples of useful solid supports include silica gel, polymer membranes, particles, derivatized plastic films, glass beads, cotton, plastic beads, alumina gels, polysaccharides such as sepharose, nylon, latex beads, magnetic beads, paramagnetic beads, superparamagnetic beads, starch, and the like.

Antigens

It is to be understood that, in principle, any streptococcus pyogenes antigen against which antibodies produced in a subject are directed may be used in the methods described herein, in particular the multiplex methods described herein. As shown in the examples herein, the clinically established antigens dnase b and SLO can be cross-linked to beads for use in multiplex CBA assays. In certain instances, and without wishing to be bound by any theory, applicants have established that certain antigens, such as SpnA, can be effectively stabilized to facilitate their inclusion in the multiplex assays described herein. In the case of SpnA, truncation of the full-length polypeptide improves immunological stability, particularly when cross-linked or otherwise bound to a solid support (in this case a bead) used in embodiments of the methods described herein.

In one embodiment, two or more streptococcus pyogenes antigens are utilized, and in particular, it is contemplated that three or more streptococcus pyogenes antigens are utilized, e.g., in a single multiplex assay. In one example, each antigen is bound to a bead or other substrate, where each antigen-binding bead population is different from each other. An example of such a multiplex assay is provided in the examples herein, where SLO, dnase b and SpnA are each covalently linked to a different bead population such that each antigen-labelled bead population can be identified separately, whereby the amount of antibody specific for each antigen can be determined in a single assay.

In certain embodiments, e.g., when contacted with a sample, two or more streptococcus pyogenes antigens are provided in a single composition (e.g., a single solution comprising any desired buffers or carriers, stabilizers, etc.), and thus are particularly suitable for multiple embodiments. In one example, the streptococcus pyogenes SpnA antigen is provided together with at least one other streptococcus pyogenes antigen, e.g., SpnA and at least one other streptococcus pyogenes antigen are present in a single composition, e.g., when contacted with a sample. In a particularly contemplated example, the streptococcus pyogenes SpnA antigen and SLO are provided, for example, in a single composition, e.g., when contacted with a sample. In another specifically contemplated example, the streptococcus pyogenes SpnA antigen and dnase b are provided, for example, in a single composition, e.g., when contacted with a sample. In another particularly contemplated example, the streptococcus pyogenes SpnA antigen, dnase b and SLO are provided, for example, in a single composition, e.g., when contacted with a sample.

SLO (AAK33267.1) is a secreted, pore-forming lysin. In one embodiment, the SLO is derived from Streptococcus pyogenes M1 GAS (strain: SF370, serotype: M1, locus tag: SPy-0167 (NCBI: NP-268546.1.) in one example, a recombinant fragment of the SLO is used, e.g., a recombinant polypeptide comprising amino acids 34-571 (see FIG. 6A, SEQ ID NO. 1.) in one example, the recombinant polypeptide is tagged, e.g., to aid purification, and specifically to account for the N-terminal or C-terminal His tag. in another embodiment, a "detoxified" SLO analog is used, wherein one or more amino acids determined to contribute to toxicity are substituted with amino acids resulting in a polypeptide that immunologically cross-reacts with wild-type SLO but exhibits reduced toxicity (e.g., to reduce production, handling or containment limitations or requirements.) the amino acid sequence of a representative example of a detoxified SLO analog is given in FIGS. 6B and SEQ ID NO. 2. in this example of a detoxified SLO, two point mutations in the encoding nucleotide sequence resulted in P427L and W535F substitutions: the substituted amino acids are underlined in FIGS. 6A and 6B.

DNase-B (AAK34710.1) is a secretase that degrades DNA. In one embodiment, DNaseB is derived from Streptococcus pyogenes M1 GAS (strain: SF370, serotype: M1, locus tag: SPy _2043 (NCBI: NP. RTM. 269989.1)). In one example, a recombinant fragment of dnase b, e.g., a recombinant polypeptide comprising amino acids 43-271 (see fig. 7, SEQ ID No.5) is used. In one example, the recombinant polypeptide is tagged, for example to aid in purification, and N-terminal or C-terminal His tags are specifically contemplated.

SpnA (AAK33693.1) is a cell wall anchored enzyme that also degrades DNA. In one embodiment, SpnA is derived from Streptococcus pyogenes M1 GAS (strain: SF370, serotype: M1, locus tag: SPy _0747 (NCBI: NP. RTM. 268972.1)). In one example, a recombinant fragment of SpnA, e.g., a recombinant polypeptide comprising amino acids 28-854 (see fig. 8, SEQ ID No.8), is used. In one example, the recombinant polypeptide is tagged, for example to aid in purification, and N-terminal or C-terminal His tags are specifically contemplated.

One skilled in the art will recognize that in certain embodiments of the invention disclosed herein, including the methods, polypeptides, beads, compositions and kits described herein, particularly embodiments related to the multiplex diagnostic methods and compositions described herein, provide accurate and rapid diagnosis and/or identification of: patients in need of treatment for recent episodes of rheumatic fever or acute PSGN, or in need of treatment for acute streptococcus pyogenes infection, but in need of long-term treatment (including long-term prophylactic treatment) of rheumatic fever, PSGN, or persistent streptococcus pyogenes infection, or in need of continuous monitoring (e.g., to rapidly identify subsequent streptococcus pyogenes infection).

In various embodiments, for example, when a patient is identified as having or has had a recent streptococcus pyogenes exposure or a recent streptococcus pyogenes infection using the methods described herein, the patient will receive a treatment suitable for treating the recent streptococcus pyogenes infection, and an ongoing treatment, such as a prophylactic treatment to prevent subsequent streptococcus pyogenes infection, or to alleviate one or more symptoms of rheumatic fever, PSGN, chronic rheumatic heart disease, or to monitor the disease state. In other embodiments, when a patient is identified as having or having had a prior exposure to streptococcus pyogenes or a prior infection with streptococcus pyogenes using the methods described herein, the patient will receive ongoing treatment, such as prophylactic treatment to prevent subsequent infection with streptococcus pyogenes, or to alleviate one or more symptoms of rheumatic fever, PSGN, chronic rheumatic heart disease, or to monitor the disease state. Representative treatments include those described herein.

The following examples, sequence listing and figures are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims. It will be appreciated that modifications may be made to the proposed solution without departing from the spirit of the invention.

Examples

80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:单步ATPS增强型LFA诊断设计

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!