WO2024178117A1 - Method of sequence-independent quantification of proteolytically unstable plasma lambda free light chain protein for al amyloidosis diagnosis - Google Patents
Method of sequence-independent quantification of proteolytically unstable plasma lambda free light chain protein for al amyloidosis diagnosis Download PDFInfo
- Publication number
- WO2024178117A1 WO2024178117A1 PCT/US2024/016721 US2024016721W WO2024178117A1 WO 2024178117 A1 WO2024178117 A1 WO 2024178117A1 US 2024016721 W US2024016721 W US 2024016721W WO 2024178117 A1 WO2024178117 A1 WO 2024178117A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- lccd
- fragment
- λflc
- proteins
- level
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/42—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57407—Specifically defined cancers
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/32—Immunoglobulins specific features characterized by aspects of specificity or valency specific for a neo-epitope on a complex, e.g. antibody-antigen or ligand-receptor
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/34—Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/22—Haematology
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/70—Mechanisms involved in disease identification
- G01N2800/7047—Fibrils-Filaments-Plaque formation
Definitions
- This application relates generally to the field of biomarkers and use in health care.
- AL Light chain amyloidosis
- FLC amyloidogenic monoclonal free light chain
- Immunoglobulin light chain and heavy chain proteins are produced by plasma cells, a ty pe of white blood cell found in the bone marrow, in the process of making various ty pes of immunoglobulins, such as IgG, IgA, IgM, etc.
- oncogenically transformed clonal plasma cells proliferate and secret excessive amounts of amyloidogenic free light chain (FLC) into the blood circulation.
- FLC amyloidogenic free light chain
- Amyloidogenic monoclonal FLC differs from the normal polyclonal FLC generated as part of the adaptive immune response in terms of the amount of circulating FLC and its amyloidogenicity.
- Amyloidogenic FLC are less stable and prone to misfolding, proteolysis, and aggregation. Misfolded conformations (soluble aggregates and fibrils) as well as fragments of FLC are believed to be toxic and can eventually deposit onto various organs as amyloids. Organ toxicity manifests as cardiomyopathy, nephrotic syndrome and/or end-stage renal failure, collectively categorized as Light Chain Amyloidosis (AL). Both amyloidogenic and K FLC may cause AL, although ⁇ FLC is frequently identified as the responsible, or involved, FLC in AL. Estimated AL incidence is 10 in every million population worldwide.
- ⁇ FLC monomer is around 227 amino acids and is made up of an N-terminal variable domain (VL), a joint linker region (J), and a C-terminal constant domain (CL).
- VL N-terminal variable domain
- J joint linker region
- CL C-terminal constant domain
- ⁇ FLC usually presents as a homodimer linked by an inter-monomer disulfide bond at the C-terminal constant domain of the protein.
- Variations in FLC sequences are the result of the germline genes and the incorporation of somatic hypermutations. While each AL patient usually possesses a single clonal plasma cell population and therefore one unique monoclonal amyloidogenic FLC sequence, it exists on the background of a highly diverse pool of polyclonal FLC that are present in any human being. Such sequence diversity and the lack of means to differentiate FLC amyloidogenicity pose unique challenges for the diagnosis and targeted treatment of AL.
- AL is often diagnosed late or goes undiagnosed until later stage because the signs and symptoms of AL mimic those of other common diseases.
- Clinical symptom of specific organ involvement is the most common trigger of suspicion of the disease.
- Confirmation of AL diagnosis ty pically takes 6-12 months with visits to 3-4 physicians, while the mortality rate at six months after AL diagnosis approaches 25% with mean survival around four years.
- Laboratory tests of blood and urine M-protein and FLC such as immunofixation and the FreeLite turbidimetry assays (BindingSite. UK), are commonly used to assess FLC levels, dFLC levels (difference in the levels of ⁇ FLC and KFLC) and ⁇ / ⁇ ratio; followed by tissue biopsy where amyloid presence and occasionally protein identity are examined.
- Imaging tests such as echocardiogram and magnetic resonance imaging are also prescribed in cases of organ involvement. These tests are useful tools but nonetheless are beset by low sensitivity, lack of specificity, and the inability to differentiate amyloidogenic from non-amyloidogenic FLC.
- mass spectrometr -based clonal LC detection has significantly improved sensitivity yet is only available in limited numbers of clinics and is not poised for quantification nor indication of amyloidogenicity.
- An aspect of the application is a method of generating a novel biomarker and using a monoclonal antibody for detection of a neo-epitope on the biomarker formed from amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins, comprising the steps of: limited proteolysis of ⁇ free light chain ( ⁇ FLC) proteins by proteinase K, wherein the limited proteolysis results in a neo-epitope exposed on a fragment of the amyloidogenic ⁇ FLC proteins, and wherein the resulting fragment is the intact light chain constant domain dimer and monomer (“LCCD,” or “dLCCD,” or “dLCCD biomarkef ’, or “LCCD biomarker”, or “LCCD fragment,” or “LCCD domain”), wherein the dLCCD biomarker has a representative sequence as in (SEQ ID NO: 1) and can exist as a either a monomer or a dimer formed via disulfide bonding through the C-term Cysteine, and where
- ⁇ FLC may be in the form of recombinant human FLC in buffer, or recombinant and/or endogenous ⁇ FLC in an ex vivo biological matrix such as plasma, serum, urine, etc.
- Other X light chain-containing proteins include any immunoglobulins (IgA, IgG. IgM, IgE, etc.) and non-amyloidogenic ⁇ FLC.
- While optimization includes testing one or more of the following conditions: vary ing the source of the Proteinase K used, vary ing the concentration of the Proteinase K used, varying time of incubation, varying the composition of the matrix containing the test subjects, and varying the temperature of incubation. Specificity is achieved yvhen LCCD fragment is generated from a representative amyloidogenic ⁇ FLC such as WIL (SEQ.ID. No.: 2) at a level at least two times more than that from a representative non-amyloidogenic ⁇ FLC such as JTO (SEQ.ID. No.: 3) under a specific condition.
- WIL SEQ.ID. No.: 2
- JTO SEQ.ID. No.: 3
- the neo-epitope is N-terminal GQP, GQPK, or GQPKA. In certain embodiments, the neo-epitope is N-terminal RQP, RQPK, or RQPKA. In certain embodiments, the neo-epitope is N-terminal SQP, SQPK, or SQPKA. In certain embodiments, the generated antibody (LCCD-G) binds to the neo-epitope of GQP, GQPK, or GQPKA. In certain embodiments, the generated antibody (LCCD-R) binds to the neo-epitope of RQP, RQPK, or RQPKA.
- the generated antibody binds to the neo-epitope of SQP, SQPK, or SQPKA.
- the LCCD fragment of the ⁇ FLC is a ⁇ 23kDa fragment under non-denaturing condition.
- the LCCD fragment of the ⁇ FLC is a ⁇ 11 ,5kDa fragment under denaturing condition.
- the limited proteolysis by proteinase K comprises the steps of: recombinant amyloidogenic ⁇ FLC in a matrix treated yvith an optimized proteolysis condition to produce the LCCD fragment.
- the limited proteolysis by proteinase K comprises the steps of: ex vivo AL plasma sample treated yvith an optimized proteolysis condition to produce the LCCD fragment.
- the optimized proteolysis condition is 0.02-30pM Proteinase K at 15°C - 70°C for 30 seconds to 20 hrs. In particular embodiments, condition is for 1 minute to 2 hours.
- Another aspect of the application is an antibody for detection of a neo-epitope formed from amyloidogenic Z free light chain ( ⁇ FLC) proteins, wherein the antibody is encoded by a nucleotide sequence encoding heavy and light chains of LCCD-G as described herein.
- ⁇ FLC amyloidogenic Z free light chain
- Another aspect of the application is an antibody for detection of a neo-epitope formed from amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins, wherein the antibody is encoded by a nucleotide sequence encoding heavy and light chains of LCCD-R as described herein.
- ⁇ FLC amyloidogenic ⁇ free light chain
- Another aspect of the application is an antibody for detection of a neo-epitope formed from amyloidogenic Z free light chain ( ⁇ FLC) proteins, wherein the antibody is encoded by a nucleotide sequence encoding heavy and light chains of LCCD-S as described herein.
- ⁇ FLC amyloidogenic Z free light chain
- nucleotide sequence that are alterations in the genetic triplet code of that sequence, but do not alter the amino acid being encoded, are also encompassed within this application for any described nucleotide sequences of LCCD-G, LCCD-R, and LCCD-S.
- Another aspect of the application is a method of assessing target engagement or compound potency in the discovery of a small molecule FLC stabilizer, comprising the steps of: limited proteolysis of proteins from a sample containing ⁇ FLC proteins, wherein the sample contains recombinant ⁇ FLC proteins or is an ex vivo biological sample such as serum, plasma, or urine, and wherein the limited proteolysis results in a neo-epitope exposed on an N-terminal of an LCCD fragment of the ⁇ FLC proteins; detecting the presence of the neoepitope on the LCCD fragment of the ⁇ FLC proteins; and quantitating the amount of the LCCD fragment of the ⁇ FLC proteins resulting from the limited proteolysis based on the neoepitope presence detected, wherein target engagement is assessed by comparing a first level of LCCD fragment before the small molecule has been introduced to the test sample, to a second level of LCCD fragment after the small molecule has been introduced to
- Another aspect of the application is a method of assessing target engagement of a FLC stabilizer for treatment of light chain amyloidosis, comprising the steps of: limited proteolysis of proteins from a biological sample, wherein the biological sample is from a subject receiving treatment by a targeted therapeutic molecule for light chain amyloidosis, and wherein the limited proteolysis results in a neo-epitope exposed on an N-terminal of an LCCD fragment of the ⁇ FLC proteins; detecting the presence of the neo-epitope on the LCCD fragment of the ⁇ FLC proteins; and quantitating the amount of the LCCD fragment of the ⁇ FLC proteins resulting from the limited proteolysis based on the neo-epitope presence detected, wherein target engagement is assessed by comparing a first level of LCCD fragment before the targeted therapeutic molecule has been introduced to the subj ect, or to a sample from a subject ex vivo, to a second level of
- Another aspect of the application is a method of assessing target engagement of a FLC stabilizer or pharmacological chaperone for treatment of light chain amyloidosis, comprising the steps of: limited proteolysis of proteins from a biological sample, wherein the biological sample is from a subject receiving treatment by a targeted therapeutic molecule for light chain amyloidosis, and wherein the limited proteolysis results in a neo-epitope exposed on a LCCD fragment of the ⁇ FLC proteins, and wherein the neo-epitope is N-terminal xQP, where x is one of G, R or S; detecting the presence of the LCCD fragment of the ⁇ FLC proteins by binding of a monoclonal antibody to the neo-epitope, wherein the monoclonal antibody binds to the neo-epitope of xQP; and quantitating the level of LCCD fragment resulting from the limited proteolysis based on the signal
- the first level of the LCCD fragment present before the targeted therapeutic molecule has been introduced to the subject is greater than the second level of the LCCD fragment present after the targeted therapeutic molecule has been introduced to the subject, then target engagement has occurred by the targeted therapeutic molecule.
- the biological sample is one of plasma, urine, serum, cerebrospinal fluid (CSF), or tissue biopsy.
- Another aspect of the application is a method of assessing therapeutic response of a therapeutics for treatment of light chain amyloidosis by quantifying level of a LCCD fragment in biological samples subjected to limited Proteinase K proteolysis before and after the therapeutic molecule has been introduced to the subject, or at tw o or more different time points during the treatment, wherein a reduction in the post-treatment or later timepoint levels of LCCD fragment indicates favorable response to the therapeutic molecule, irrespective to the total level of ⁇ FLC.
- Another aspect of the application is a method of assessing if an AL patient would benefit from a light chain stabilizer as a therapy by quantifying level of a LCCD fragment in biological samples from a subject being considered to receive treatment by a light chain stabilizer molecule for light chain amyloidosis, wherein the ex vivo plasma sample is incubated with the said stabilizer or a solvent control, before subjecting the samples to limited Proteinase K proteolysis, and quantification of the resulting LCCD levels, wherein a reduction of LCCD biomarker level in the sample that was incubated with the stabilizer versus the solvent control sample indicates the subject may benefit from treatment by the said light chain stabilizer.
- Another aspect of the application is a method of detection of presence in a subject of amyloidogenic k free light chain ( ⁇ FLC) proteins, comprising the steps of: limited proteolysis of proteins from a biological sample, wherein the limited proteolysis results in a neo-epitope exposed on an N-terminal of the LCCD fragment of the ⁇ FLC proteins; detecting the presence of the neo-epitope on the fragment of the ⁇ FLC proteins; quantitating the level of LCCD fragment of the ⁇ FLC proteins by measuring the neo-epitope by the LCCD mAbs; and comparing LCCD fragment levels from the ⁇ FLC proteins before and after the limited proteolysis, wherein when the LCCD fragment level is greater after limited proteolysis then amyloidogenic ⁇ FLC proteins are detected.
- ⁇ FLC amyloidogenic k free light chain
- Another aspect of the application is a method of detection of presence in a subject of amyloidogenic k free light chain ( ⁇ FLC) proteins, comprising the steps of: limited proteolysis by Proteinase K of proteins from a biological sample, wherein the limited proteolysis results in a neo-epitope exposed on an N-terminal of the LCCD fragment of the ⁇ FLC proteins, and wherein the neo-epitope is xQP, where x is one of G, R or S; detecting the presence of the LCCD fragment of the ⁇ FLC proteins by binding of a monoclonal antibody to the neo-epitope, wherein the monoclonal antibody binds to the neo-epitope of xQP; quantitating the LCCD fragment level by measuring the amount of antibody binding to the neo-epitope; and comparing a first level of quantity of LCCD fragment present after the limited proteolysis in a biological sample
- the LCCD fragment as a 23KDa homodimer, or a 11.5KDa monomer, is detected and quantified using mass spectrometry.
- the level of LCCD fragment from ⁇ FLC is detected after limited proteolysis by Proteinase K and at an elevated level above the reference level, then light chain amyloidosis is present in the subject.
- elevated LCCD level in a subject lead to a clinical decision, wherein the clinical decision is treatment and management for light chain amyloidosis (AL).
- AL light chain amyloidosis
- the subject has one of more of clinical suspicion of amyloidosis, Multiple myeloma (MM), Light Chain Amyloidosis (AL), Smoldering Multiple Myeloma (SMM), IgM light chain amyloidosis, Monoclonal Gammopathy of Undetermined Significance (MGUS), Minimal Residual Disease (MRD) of AL, and Relapse of AL.
- the subject received plasma cell therapy and elevated LCCD levels is used to detect residual level of amyloidogenic FLC, indicating minimal residual disease (MRD).
- elevated LCCD levels in a subject who had stable disease indicates disease relapse.
- the biological sample is one of plasma, urine, serum. CSF. or tissue biopsy.
- kits for detection of presence in a subject of amyloidogenic k free light chain ( ⁇ FLC) proteins comprising: one or more of the antibodies of LCCD-G, LCCD-R and LCCD-S; instructions for using the kit in accordance with the methods described herein.
- the kit comprises a protein with an N-terminal neoepitope as the reference standard.
- this protein is dLCCD.
- the neo-epitope is xQP, where x is one of G, R or S.
- the kit comprises one or more of the antibodies to the neo-epitopes described herein.
- the antibody is LCCD-G, LCCD-R or LCCD-S.
- the kit comprises an instruction to generate the LCCD fragment from ⁇ FLC by limited proteolysis by Proteinase K.
- the fragment LCCD and dLCCD names are used interchangeably in this application unless otherwise noted.
- the level of dLCCD of a particular sample is a parameter that is calculated based on one or more readings of the dLCCD signal or signals of the said sample generated under limited proteolysis conditions.
- Figure 1A illustrates an exemplary SDS PAGE gel of Proteinase K (PK) digestion of recombinant lambda light chains, including a random selection of 293F cell produced recombinant amyloidogenic (sequences based on AL patients) and non- amyloidogenic (sequences based on normal healthy person) ⁇ FLC proteins, comparing with and without PK treatment.
- PK digestion resulted in the generation of a proteolytic fragment corresponding to the disulfide linked LC constant domain (LC-C domain, or dLCCD) homodimer.
- LC-C domain disulfide linked LC constant domain
- Figure IB shows LC structure highlighting the hinge region.
- PK cleaves in the hinge region between LC-V and LC-C, producing the dLCCD fragment.
- PK treatment exposes neo-epitopes highly conserved for all LC sequences. Exposure depending on the dynamic/ amyloidogenic nature of the ⁇ FLC.
- Figure 1C shows lambda LC sequence around the cleavage site, indicating the highly conserved PK cleavage site exposing the xQP neo-epitope. Based on analysis of public databases, frequency of sequence occurrence at the N-term of the dLCCD fragment after PK cleavage in AL ⁇ FLC are approximately: 84% GQP, 11% SQP. and 5% RQP.
- Figure 2A and 2B show LCCD-G mAb binding to LCCD vs (a) full length WIL or (b) IgG + PK in buffer limited PK proteolysis in buffer, demonstrating 1) the LCCD mAb specifically binds to the cleaved and exposed xQP epitope, not xQP in a protein sequence; 2) the specific generation of the LCCD-G epitope specifically in amyloidogenic FLC, but not in full length immunoglobulin.
- FIG. 2C show LCCD mAbs highly specific for proteolysis exposed novel epitopes.
- LCCD-G, LCCD-R, and LCCD-S mAbs (green signal around 25 KDa) specifically detect LCCD fragments recombinant FLC proteins containing GQP, RQP, and SQP at the cleavage site, respectively, post-proteolysis by Proteinase K.
- Anti- ⁇ FLC polyclonal antibody red signal around 50KDa is used to indicate all full-length and LCCD fragments from the samples.
- Figure 3 A shows LCCD-G mAb detection of LC proteolysis in 10 treatment naive AL plasma in WB.
- Figure 3B shows quantification of dLCCD fragment level determined in Western blot in pre-treatment AL patients (post-PK treatment).
- Statistical analysis method one-way ANOVA.
- Figure 5 shows an MSD dLCCD immunoassay.
- Figure 5A Using LCCD-X mixture as the capture antibody, dLCCD level generated under limited proteolysis determined on MesoScale Discovery (MSD) platform.
- LCCD-X is an equal molar mixture of the three LCCD mAbs, LCCD-G, LCCD-S. and LCCD-R.
- Recombinant dLCCD was used as the reference standard.
- Figure 6 shows baseline levels of dLCCD correlates to ⁇ AL patient overall survival.
- dLCCD levels were quantified on treatment naive plasma samples from 50 confirmed ⁇ AL patients using the MSD LCCD immunoassay.
- Figure 7 shows concordance of dFLC and dLCCD predicts overall survival.
- Log-rank survival test of patients with low baseline levels of both biomarkers solid line; left- to-right sloping down background shading in lower graph
- those with high levels of both biomarkers dotted line; dotted background shading in lower graph
- those with discordant dFLC and dLCCD dotted-and-dashed line; left-to-right upward sloping up shading in background graph.
- FIG. 8A shows Western blot of the limited proteolysis samples. Using an anti- ⁇ FLC monoclonal antibody (band at 50KDa) and the dLCCD mAb (solid band at 23 KDa).
- Figure 8B Quantification of the antibody-detected bands on the LiCor system and fitted to a variable slope (four parameters) response curve using Prism. EC 50S are similar using the two antibodies).
- Figure 8C Protection against limited proteolysis by PTG-1412 in 13 ex vivo ⁇ AL plasma samples as quantified by dLCCD levels post-proteolysis, with and without PTG-1412.
- Ranges may be expressed herein as from “about” one particular value, and/or to "about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10" is also disclosed.
- this application describes a method of generating neo-epitopes at the N-terminal of the ⁇ FLC cleavage product and the monoclonal antibodies (mAbs) that specifically recognize such epitopes.
- mAbs monoclonal antibodies
- the generation of these neo-epi topes directly correlates to the lack of kinetic stability of the amyloidogenic FLC. which may be corrected with a small molecule stabilizer binding to the dimeric ⁇ FLC. Levels of amyloidogenic FLC may also be reduced by other therapeutic interventions.
- neo-epitopes described herein can be recognized by other epitope-recognizing agents, such as. but not limited to, nanobodies, aptamers, affimers, etc.
- methods of detecting a neo-epitope or novel biomarker as described herein as not limiting on the invention may include, but are not limited to, immunoassay (e.g., ELISA, Simoa, Olink). lateral flow assays, mass spectroscopy, etc.
- antibody refers to a polypeptide or a polypeptide complex that specifically recognizes and binds to antigen through one or more immunoglobulin variable regions. Antibody can be a whole antibody , antigen binding fragment or a single chain thereof.
- antibody encompasses various broad classes of polypeptides that can be distinguished biochemically. Those skilled in the art will appreciate that heavy chains are classified as alpha, delta, epsilon, gamma, and mu, or a, 5, E, y and p) with some subclasses among them (e.g., yl-y 4).
- immunoglobulin subclasses e.g., IgGl, IgG2. IgG3, IgG4, etc. are well characterized and are known to confer functional specialization. Modified versions of each of these classes and isotypes are readily discernable to the skilled artisan in view of the instant disclosure and, accordingly , are within the scope of the instant disclosure. All immunoglobulin classes are within the scope of the present disclosure, the following discussion will generally be directed to the IgG class of immunoglobulin molecules.
- Antibodies or antibody antagonists of the present application may include, but are not limited to polyclonal, monoclonal, multispecific, bispecific, trispecific, human, humanized, primatized, chimeric and, single chain antibodies.
- Antibodies disclosed herein may be from any animal origin, including birds and mammals.
- the antibodies are human, murine, rat, donkey, rabbit, goat, guinea pig, camel, llama, horse, or chicken antibodies.
- the variable region may be condricthoid in origin (e.g., from sharks).
- antibody fragment or “antigen-binding fragment” are used with reference to a portion of antibody, such as F(ab')2, F(ab)2, Fab', Fab, Fv, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv), fragments comprising either a VL or VH domain, fragments produced by a Fab expression library and anti-idiotypic (anti- id) antibodies. Regardless of structure, antibody fragment binds with the same antigen that is recognized by the intact antibody.
- antibody fragment includes DARTs and diabodies.
- antibody fragment also includes any synthetic or genetically engineered proteins comprising immunoglobulin variable regions that act like antibody by binding to a specific antigen to form a complex.
- a “single-chain fragment variable” or “scFv” refers to a fusion protein of the variable regions of the heavy (VH) and light chains (VL) of immunoglobulins. In some aspects, the regions are connected with a short linker peptide of ten to about 25 amino acids.
- the linker can be rich in glycine for flexibility, as well as serine or threonine for solubility, and can either connect the N-terminus of the VH with the C-terminus of the VL, or vice versa.
- a standard immunoglobulin molecule comprises two identical light chain polypeptides of molecular weight approximately 23,000 Daltons, and two identical heavy chain polypeptides of molecular weight 53,000-70,000.
- the four chains are typically joined by disulfide bonds in a “Y” configuration where the light chains bracket the heavy chains starting at the mouth of the “Y” and continuing through the variable region.
- both the light and heavy chains are divided into regions of structural and functional homology.
- the terms “constant” and “variable” are used functionally.
- the variable domains of both the light (VL) and heavy (VH) chain portions determine antigen recognition and specificity.
- the constant domains of the light chain (CL) and the heavy chain (CHI, CH2 or CH3) confer important biological properties such as secretion, transplacental mobility, Fc receptor binding, complement binding, and the like.
- the numbering of the constant region domains in conventional antibodies increases as they become more distal from the antigenbinding site or amino-terminus of the antibody.
- the N-terminal portion is a variable region and at the C-terminal portion is a constant region; the CH3 and CL domains actually comprise the carboxy -terminus of the heavy and light chain, respectively.
- a “variant” of antibody, antibody fragment or antibody domain refers to antibody, antibody fragment or antibody domain that (1) shares a sequence identity of at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% with the original antibody, antibody fragment or antibody domain, and (2) binds specifically to the same target that the original antibody, antibody fragment or antibody domain binds specifically. It should be understood that where a measure of sequence identity is presented in the form of the phrase “at least x % identical” or “at least x % identity ”, such an embodiment includes any and all whole number percentages equal to or above the lower limit.
- ammo acid sequence is presented in the present application, it should be construed as additionally disclosing or embracing “variant” amino acid sequences having a sequence identity of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to that amino acid sequence.
- humanized antibody refers to antibody derived from a non-human antibody, typically a mouse monoclonal antibody.
- a humanized antibody may be derived from a chimeric antibody that retains or substantially retains the antigen binding properties of the parental, non-human. antibody but which exhibits diminished immunogenicity as compared to the parental antibody when administered to humans.
- chimeric antibody refers to antibody where the immunoreactive region or site is obtained or derived from a first species and the constant region (which may be intact, partial or modified in accordance with the instant disclosure) is obtained from a second species.
- the target binding region or site will be from a non-human source (e.g., mouse or primate) and the constant region is human.
- compositions and methodologies including asymmetric IgG-like antibodies (e.g., triomab/quadroma. Trion Pharma/Fresemus Biotech); knobs-into-holes antibodies (Genentech); Cross MAbs (Roche); electrostatically matched antibodies (AMGEN); LUZ-Y (Genentech); strand exchange engineered domain (SEED) body (EMD Serono; BioIonic, Merus); Fab-exchanged antibodies (Genmab), symmetric IgG-like antibodies (e.g., triomab/quadroma. Trion Pharma/Fresemus Biotech); knobs-into-holes antibodies (Genentech); Cross MAbs (Roche); electrostatically matched antibodies (AMGEN); LUZ-Y (Genentech); strand exchange engineered domain (SEED) body (EMD Serono; BioIonic, Merus); Fab-exchanged antibodies (Genmab), symmetric IgG-like antibodies (e.g.
- antibody is said to '‘specifically bind” to an epitope when it binds to that epitope via its antigen-binding domain more readily than it would bind to a random, unrelated epitope.
- the term ‘'specificity” is used herein to qualify the relative affinity' by which a certain antibody binds to a certain epitope.
- antibody “A” may be deemed to have a higher specificity for a given epitope than antibody “B,” or antibody “A” may be said to bind to epitope “C” with a higher specificity than it has for related epitope “D.”
- antibody or antibody fragment “has specificity to” antigen if the antibody or antibody fragment forms a complex with the antigen with a dissociation constant (Kd) of 10' 6 M or less, 10' 7 M or less, 10 -8 M or less, 10' 9 M or less, or 10' 10 M or less.
- Kd dissociation constant
- the neo-epitope is GQP. In certain embodiments, the neo-epitope is RQP. In certain embodiments, the neo-epitope is SQP.
- the generated antibody (LCCD or dLCCD-G) binds to the neo-epitope of GQP. In certain embodiments, the generated antibody (LCCD-R or dLCCD-R) binds to the neo-epitope of RQP. In certain embodiments, the generated antibody (LCCD-S or dLCCD-S) binds to the neo-epitope of SQP.
- the fragment of the ⁇ FLC is a homodimeric fragment, which is a ⁇ 23kDa fragment under non-denaturing condition. In certain embodiments, the fragment of the ⁇ FLC is a monomeric fragment, which is a ⁇ 11.5kDa fragment under denaturing condition.
- the limited proteolysis by proteinase K comprises the steps of: a liquid sample treated with an optimized proteolysis condition to produce the LCCD domain.
- the optimized proteolysis condition is 0.02-20.
- OpM Proteinase K at 37°C for 30 seconds to 20 hrs depending on the specific activity of the protease.
- the specific activity of the protease may be independently determined and is not limiting.
- An aspect of the application is an antibody for detection of a neo-epitope formed from amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins, wherein the antibody comprises heavy chain encoded by a nucleotide sequence of LCCD-G SEQ.ID. No.: 4 (HC3) and light chain encoded by a nucleotide sequence of LCCD-G SEQ. ID. No.:5 (LC7).
- ⁇ FLC amyloidogenic ⁇ free light chain
- An aspect of the application is an antibody for detection of a neo-epitope formed from amyloidogenic ⁇ free light chain ( ⁇ FLC ) proteins, wherein the antibody comprises heavy chain encoded by a nucleotide sequence of LCCD-R SEQ.ID. No.: 6 (HC2) and light chain encoded by a nucleotide sequence of LCCD-R SEQ ID No.: 7 (LC9).
- ⁇ FLC amyloidogenic ⁇ free light chain
- An aspect of the application is an antibody for detection of a neo-epitope formed from amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins, wherein the antibody comprises heavy chain needed by a nucleotide sequence of LCCD-S SEQ.ID. No.: 8 (HC2) and light chain encoded by a nucleotide sequence of LCCD-S SEQ ID No: 9 (LC5).
- ⁇ FLC amyloidogenic ⁇ free light chain
- An aspect of the application is a neo-epitope on ⁇ FLC prepared by limited proteolysis of amyloidogenic k free light chain ( ⁇ FLC) proteins by proteinase K.
- the neo-epitope is xQP, where x is one of G, R or S.
- an antibody is generated to the neo-epitope described herein.
- the antibody is LCCD-G, LCCD-R or LCCD-S.
- An aspect of the application is a method of optimizing limited proteolysis condition using Proteinase K, comprising the steps of optimizing one or more of the following enumerated factors: (1) source of the Proteinase K; (2) composition of the testing sample; (3) concentration of Proteinase K: (4) time of proteolysis reaction; (5) temperature of proteolysis reaction; wherein a proteolysis product of the LCCD fragment is differentially obtained from amyloidogenic ⁇ FLC vs. non-amyloidogenic ⁇ FLC.
- biomarker correlates with root cause of AL amyloidosis (i.e. unstable variant LC protein links to amyloidogenicity); (2) applicable to majonty if not all AL variants ( ⁇ FLC AL as the current focus); (3) easy to access immunoassay that uses biofluids (serum, plasma, and urine); (4) sensitive, specific, and quantitative assay; (5) enables early diagnosis or screening based on amyloidogenicity of each ⁇ FLC variant irrespective of the FLC primary sequence or exact ⁇ FLC level (personalized medicine); (6) assesses treatment therapeutic response; (7) follows MRD and predicts relapse.
- amyloidogenic FLC proteins are thermodynamically and/or kinetically less stable, causing them to be more prone to protein misfolding, proteolysis, and aggregation.
- amyloidogenic FLC readily generates the LCCD fragment and reveals novel cryptic epitopes that can be detected, including with antibodies.
- this application discloses the specific stress (i.e. limited proteolysis by Proteinase K) to reveal such neo-epitopes on the LCCD fragment in AL patient plasma samples and mAbs specific for such novel epitopes.
- An aspect of the application is a method of detection of presence in a subject of amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins, comprising the steps of: limited proteolysis of proteins from a biological sample, wherein the limited proteolysis results in a neo-epitope exposed on a LCCD fragment of the ⁇ FLC proteins; detecting the presence of the neo-epitope on the N-termini of the LCCD fragment of the ⁇ FLC proteins; quantitating the presence of the LCCD fragment from the ⁇ FLC proteins based on measuring the level of neoepitope on the fragment using the LCCD mAbs; and comparing levels of the LCCD fragment before and after the limited proteolysis, wherein when the level of the LCCD fragment from ⁇ FLC proteins is greater after limited proteolysis, then amyloidogenic ⁇ FLC proteins are detected.
- the method determining the level (e.g. quantity) of LCCD fragment
- the method further comprises the steps of: measuring a first level of the LCCD fragment of the ⁇ FLC proteins present after the limited proteolysis in a biological sample taken from a healthy subject not having light chain amyloidosis (AL), wherein the first level is a reference level; measuring a second level of the fragment of the ⁇ FLC proteins present after the limited proteolysis of a biological sample from a person suspected of having AL; and wherein when the second level of the LCCD fragment of the ⁇ FLC proteins after the limited proteolysis of the biological sample is greater than the reference level, then amyloidogenic ⁇ FLC proteins are detected.
- AL light chain amyloidosis
- an elevated LCCD fragment level in a subject leads to a clinical decision, wherein the clinical decision is treatment and management for light chain amyloidosis (AL).
- AL light chain amyloidosis
- the LCCD fragment is a 23KDa homodimer and is detected and quantified using mass spectrometry.
- the LCCD fragment is a 11.5KDa monomer and is detected and quantified using mass spectrometry.
- the subject has one of more of clinical suspicion of amyloidosis, Monoclonal Gammopathy of Undetermined Significance (MGUS), Multiple myeloma (MM). Light Chain Amyloidosis (AL), Smoldering Multiple Myeloma (SMM), IgM light chain amyloidosis. Minimal Residual Disease (MRD) of AL, and Relapse of AL.
- MGUS Monoclonal Gammopathy of Undetermined Significance
- MM Multiple myeloma
- AL Light Chain Amyloidosis
- SMM Smoldering Multiple Myeloma
- IgM light chain amyloidosis IgM light chain amyloidosis.
- MRD Minimal Residual Disease
- the biological sample is one of plasma, urine, serum, CSF, or tissue biopsy.
- kits for detection of presence in a subject of amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins comprising: the antibodies of LCCD-G, LCCD-R and LCCD-S; instructions for using the kit in accordance with the methods described herein.
- the method of detection of presence in a subject of amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins as described herein further comprises: obtaining one or more readouts from dLCCD signals obtained by measuring the level of neoepitope on the fragment using the LCCD mAbs.
- method of detection of presence in a subject of amyloidogenic ⁇ free light chain ( ⁇ FLC) proteins as described herein further comprising: obtaining the readout of the dLCCD signal by use of an algorithm where the dLCCD level is a component of.
- Methods of detecting the LCCD fragment resulting from limited PK proteolysis may include mass spectrometry, which will need to account for the limited number of sequence variation in the constant domain of the ⁇ FLC.
- Other methods may include immunoassays using antibodies that can recognize the LCCD fragment. For example, gel electrophoresis coupled with Western blot using an anti- ⁇ FLC antibody. All commercial anti- ⁇ FLC antibodies this study has tested (>20) can recognize not only ⁇ FLC, but also ⁇ light chains associated with immunoglobulins. As reported herein, to solve this problem, the study generated rabbit mAbs that specifically recognize one of the three unique xQP neo-epitopes at the N-termini of the LCCD fragment exposed through limited PK proteolysis.
- immunoassays based on the LCCD-G mAh can sensitively and specifically detect and quantify the LCCD domain post proteolysis and serve as an indicator of the kinetic instability of the ⁇ FLC protein.
- LCCD rabbit mAbs described here may be used alone or together in a mixture of two or three mAbs for the assays.
- an equal-molar mixture of the three mAbs are prepared and used and is referred to LCCD-X.
- LCCD ⁇ 23KDa LCCD or dLCCD fragment is a disulfide-linked homodimer.
- LCCD presents as a monomer at about 11.5KDa (SEQ ID NO: 2), yet still containing the N-terminal xQP epitope.
- SEQ ID NO: 2 amino acid sequence sequence
- Such a monomer can also be utilized for the uses intended by the current application.
- variants of the constant domain of the ⁇ FLC proteins may include the neo-epitope; the present application also encompasses antibody or antibody fragments that recognize the neo-epitope when it is a part of variants of the constant domain of the ⁇ FLC proteins.
- Solution based LCCD immunoassays such as ELISA, MSD, Simoa and Olink can afford improved sensitivity over Western blot method. Such improved sensitivity is especially valuable when applied to early diagnosis and following minimal residual diseases of AL, where the total ⁇ FLC levels are much lower.
- the LCCD immunoassay can be used to demonstrate target engagement for treatment targeting stabilization of amyloidogenic ⁇ FLC (as described in, but not limited to, WO2022/226166). This method may be used as companion diagnostics to select AL patients who are most likely to respond to an LC stabilizer in the clinic, enabling personalized medicine.
- LCCD signals can also be used in combination with other clinical biomarkers and assessments, such as the ⁇ FLC level, dFLC, Troponin, and NT-proBNP to guide clinical practice.
- Levels of the LCCD biomarker measured using ex vivo samples from the patient can also be used to assess AL disease prognosis. Due to the pathogenic nature of the LCCD biomarker, higher level of LCCD level may indicate worse clinical outcomes.
- the method of combining limited proteolysis on ⁇ FLC and detection of the resulting LCCD fragment can also be used in the course of drug discovery in identifying molecules that can influence ⁇ FLC stability, and for the purpose of finding potent drug candidates.
- reagents that specifically bind to the N-terminal exposed xQP neoepitope such as aptamer, nanobody, etc. may also be generated and used for the detection and quantification of the LCCD biomarker.
- An aspect of the application is assessing target engagement of a FLC stabilizer for treatment of light chain amyloidosis, comprising the steps of: limited proteolysis of proteins from a biological sample, wherein the biological sample is a recombinant protein or from a subject receiving treatment by a targeted therapeutic molecule for light chain amyloidosis, and wherein the limited proteolysis results in a neo-epitope exposed on a LCCD fragment of the ⁇ FLC proteins; detecting the presence of the neo-epitope on an N-termini of the LCCD fragment of the ⁇ FLC proteins; and quantitating the level of the fragment of the ⁇ FLC proteins resulting from the limited proteolysis based on the neo-epitope presence detected, wherein target engagement is assessed by comparing a first level of quantity of the LCCD fragment present before the targeted therapeutic molecule has been introduced to the subject, or to a sample from a subject ex vivo, to a second level
- An aspect of the application is a method of assessing therapeutic response of a therapeutics for treatment of light chain amyloidosis by quantifying a LCCD fragment in plasma samples before and after the therapeutic molecule has been introduced to the subject, or at two different time points during the treatment, wherein a reduction in the post-treatment or second timepoint level of LCCD indicates favorable response to the therapeutic molecule, irrespective to the total level of ⁇ FLC.
- Said therapeutics include but are not limited to those that are Light Chain stabilizers, plasma cell-targeted therapies, gene and cell therapies, stem cell transplant, and chemotherapeutic agents.
- Assessment based on the dLCCD biomarker may be done one time or multiple times following the said therapeutic treatment.
- An aspect of the application is a method of assessing if an AL patient would benefit from a light chain stabilizer as a therapy by quantifying a LCCD fragment in plasma samples from a subject being considered to receive treatment by a light chain stabilizer molecule for light chain amyloidosis, wherein the ex vivo plasma sample is incubated with the said stabilizer or a solvent control, before subjecting the samples to limited proteolysis, and quantification of the resulting LCCD levels, wherein a reduction of LCCD level in the sample that was incubated with the stabilizer versus the solvent control sample indicates the subject may benefit from treatment by the said light chain stabilizer.
- the first level of the LCCD fragment present before the targeted therapeutic molecule has been introduced to the subject is greater than the second level of the LCCD fragment present after the targeted therapeutic molecule has been introduced to the subject, then target engagement or treatment response has occurred by the targeted therapeutic molecule.
- the biological sample is one of plasma, urine, serum, cerebrospinal fluid (CSF), or tissue biopsy.
- xQP mAbs may have other general utilities in biotechnology.
- xQP may be the smallest non-charged N-terminal protein tag known.
- N-terminal or C-terminal tags are widely used in recombinant proteins and cellular systems for the purpose of target protein detection and/or purification. Antibodies binding to these tags are important tools in studying target proteins and protein complexes. Commonly used tags include His tag, Flag tag, HA tag, and myc tag, all containing 6-10 charged amino acids.
- the three new mAbs (LCCD-G, LCCD-S, and LCCD-R) discussed in the application specifically bind to small N-terminal tags of 1-5 amino acids, with GQP and SQP non-charged.
- small tags epitopal tags
- Such small tags can be placed at the N-terminal of a protein of interest through genetic engineering to facilitate such utility.
- the methods and compositions described herein can be used for diagnosis of Light Chain Amyloidosis (AL) amongst plasma cell disorder patients suspected of Multiple myeloma (MM), Light Chain Amyloidosis (AL), Smoldering Multiple Myeloma (SMM), Monoclonal Gammopathy of Undetermined Significance (MGUS), and IgM-AL, also including those who have no clinical symptoms.
- MM Multiple myeloma
- MM Light Chain Amyloidosis
- SMM Smoldering Multiple Myeloma
- MGUS Monoclonal Gammopathy of Undetermined Significance
- IgM-AL also including those who have no clinical symptoms.
- the methods and compositions described herein can be used to demonstrate target engagement by measuring the stabilization effect of a light chain stabilizer clinical candidate (or drug) for its protection against proteolysis in individual AL patient plasma sample post treatment.
- the methods and compositions described herein can be used to monitor response to light chain stabilizer or plasma cell-targeting therapeutic treatment in AL amyloidosis patients.
- the methods and compositions described herein can be used for prognostic biomarkers, such as the level of LCCD biomarker can be used to indicate disease severity and clinical outcomes.
- the methods and compositions described herein can be used for detection of Minimal Residual Disease (MRD) and Relapse: Detection of Light Chain Amyloidosis (AL) MRD and relapse by measuring the amount of LCCD signal in patient plasma samples.
- MRD Minimal Residual Disease
- AL Light Chain Amyloidosis
- the methods and compositions described herein can be used for companion diagnostics, such as to test the ability of a LC stabilizer clinical candidate (or drug) for its protection against proteolysis in individual pre-treatment AL patient plasma sample ex vivo and use the outcome to determine if the patient would benefit from treatment with the LC stabilizer.
- the methods and compositions described herein can be used for surrogate biomarkers or therapeutic response markers, such as to demonstrate the proteolysis protection effect of a LC stabilizer clinical candidate (or drug) and use that as an indication of clinical efficacy.
- LCCD mAbs were tested for their binding to WIL and IgG with and without treatment of Proteinase K (PK) in a Sandwich ELISA binding assay. Briefly, goat anti-IgG (H+L) antibody from Bethyl on a high-binding ELISA plate at 1 pg/mL concentration in a pH 9.6 carbonate buffer and incubated at 4 °C for overnight. After blocking with Superblock, serial dilutions of WIL LC and purified human IgG with and without PK (0.2 pM) digestion were added to the plate and incubated at room temperature for 1 hour. After washing away the unbound protein.
- PK Proteinase K
- LCCD mAb at 1 pg/mL was added to the wells and the plate was incubated at room temperature for 1 hour. Anti-rabbit HRP was then added to the washed plate for 30 minutes, the plate was washed thoroughly, then the enzyme substrate tetramethylbenzidine (TMS) was added to each well. The reaction was stopped by adding 0.5 M of sulfuric acid and absorbance at 450 nm was read on a plate reader.
- TMS enzyme substrate tetramethylbenzidine
- plasma samples were incubated with IpM of Proteinase K at 37°C for 2 hrs before the reaction was stopped by adding phenylmethylsulfonyl fluoride protease inhibitor.
- Plasma samples treated with Proteinase K using the limited proteolysis condition were run on non-reducing SDS-PAGE gels, followed by standard Western blot procedure.
- the gel was transferred onto a nitrocellulose membrane.
- the membrane was then blocked with blocking reagent and incubated with 0.5 pg/mL LCCD-G mAb at 4 °C overnight.
- Anti-rabbit conjugated with 800 dye was used and the washed blot was imaged on an LiCor Odyssey system. The fluorescence intensity of the dLCCD band at around 23 KDa was recorded and the fluorescence signal quantified.
- Example 1 Identification of a neo-epitope after limited proteolysis of
- Amyloidogenic /.FLC tends to be kinetically less stable compared to the non- amyloidogenic counterparts, rendering it susceptible to denaturing conditions.
- Limited proteolysis has been widely used to probe protein structural dynamicity.
- kinetic stability of ⁇ FLC is assessed using limited proteolysis where the FLC protein is incubated with a protease. Quantification of the intact full-length FLC protein remaining or the proteolysis product(s) is used as an indication of the FLC protein stability.
- Proteinase K (PK) has previously been used to study the stability of ⁇ FLC. although it was assumed ⁇ FLC would yield small peptides after PK treatment, and only the full-length intact ⁇ FLC was followed and analyzed.
- PK is considered a broad-spectrum protease.
- the predominant site of PK cleavage is the peptide bond adjacent to the carboxyl group of aliphatic and aromatic amino acids with blocked alpha amino groups. Proteins can be completely digested by PK if the incubation time is long and PK concentration high enough. 5pM of the LC recombinant protein was treated with 200nM Proteinase K at 37°C for 2 hrs. The gel was run under nonreducing condition, stained and visualized with Coomassie dye.
- the cleavage site constituted by the GQPKA sequence is a neo-epitope exposed only through limited proteolysis by PK.
- Monoclonal antibodies were generated through immunizing rabbits using peptide-KLH conjugates as the antigens.
- Anti-serum titer was monitored using ELISA against the antigen. Upon detection of high titer, antigen-specific B cells were isolated for specific recognition of the antigen. Cells were expanded and antibody genes amplified and cloned. The amplified antibody heavy and light chains were expressed in HEK293 cell, and the supernatant assayed by ELISA to confirm the antibody specificity.
- mAbs from the immunizations were selected, sequenced, and the mAbs were named LCCD-G, LCCD-S, and LCCD-R, respectively, based on the identity of x in the xQP neo-epitopes.
- LCCD mAb was used to detect and quantify the proteolysis product in AL patient plasma samples using standard Western blot (WB) and imaged on a LiCor system ( Figure 3A).
- LCCD mAb is highly specific to the LCCD or dLCCD fragment generated after the limited proteolysis and does not detect any other signal in the AL plasma samples ( Figure 3A).
- WB Western blot
- Figure 3A LiCor system
- the mAb detects signal in 32 randomly selected ⁇ AL plasma samples in this application employing standard Western blot analysis using a few microliters of samples.
- the mean signal in AL samples is 10x-200x above that of normal or most MM plasma samples and the result is statistically significant. It may be that the single MM patient with high signal had co-existing AL. This result establishes the assay’s utility as AL diagnostics.
- FIG. 5A shows the steps of an ELISA-based dLCCD immunoassay that differentiates AL from MM, Normal, and ATTR post limited proteolysis using ex vivo blood samples.
- the Meso Scale Discovery (MSD) platform is used in the example.
- the steps of the plasma/serum assay led to a combination of a tagged detection antibody (anti-LC pAb), which binds to the analy te (dLCCD), which is in turn bound to a capture antibody (dLCCD mAbs). which is supported on a working electrode.
- the MSD electrochemiluminescence has high sensitivity, and a large dynamic range (>400x); typically, a 96- or 384-well format is used with as little as approx.
- FIG. 5B shows the outcome of a dLCCD assay with plasma sample post limited proteolysis.
- Recombinant dLCCD homodimer of SEQ ID NO: 2 linked by disulfide bond was used as the reference standard.
- Figure 6 shows baseline levels of dLCCD correlates to ⁇ AL patient overall survival. dLCCD in the experiment was detected at the 2 hour time point.
- Figure 7 shows concordance of dFLC and dLCCD predicts overall survival based on the data from Figure 6.
- High and low levels of dFLC and dLCCD are defined by the median value of 18 mg/dL for baseline dFLC and 300 nM for dLCCD. The results are based on treatment naive plasma samples from 50 confirmed ⁇ AL patients using an MSD LCCD immunoassay as described herein.
- dLCCD low /dLCCD low 162 months; dFLC high /dLCCD high 36 months; discordant 16 months.
- dLCCD is a pathogenic-indicating biomarker in most ⁇ AL patients.
- Baseline dLCCD is a better OS predictor than dFLC. Accordingly, reducing dLCCD levels by therapeutics is important to achieve clinical benefits; for example, by eradicating amyloidogenic FLC producing clonal plasma cells, or by stabilizing ⁇ FLC to minimize its amyloidogenicity, or a combination of the above two.
- Example 8 dLCCD biomarker level as an indicator of LC stabilizer target engagement in biofluids or as response to therapy
- LC stabilizer PTG-1412 w as incubated with the ex vivo AL plasma, and the assay showed dose-dependent protection of the ⁇ FLC in the sample.
- One skilled in the art may conduct the same assay on biofluid from an AL patient before and after therapeutic treatment and use the difference in the dLCCD levels as an indicator for response to therapy or the lack thereof.
- the N-terminal G can be S or R in the xQP epitope.
- Monomeric LCCD can form dimeric
- LCCD LCCD
- dLCCD dLCCD
- dLCCD biomarker LCCD fragment
- Amyloidogemc FLC protein WIL (SEQ ID No.: 2):
- Non-amyloidogenic FLC protein JTO (SEQ ID No.: 3):
- NFMLNQPHSVSES PGKTVTISCTRSSGNIDSNYVQWYQQRPGSAPITVIYEDNQRPSGVPDR FAGS IDRSSNSASLTISGLKTEDEADYYCQSYDARNWFGGGTRLTVLGQPKAAPSVTLFPP SSEELQANKATLVCLISDFYPGAVTVAWKADSS PVKAGVETTTPSKQSNNKYAAS SYLSLTP EQWKSHKSYSCQVTHEGSTVEKTVAPTECS
- LCCD-S mAb LC5 (SEQ ID NO.: 9) ATGGACACGAGGGCCCCCACTCAGCTGCTGGGGCTCCTGCTGCTCTGGCTCCCAGGTGCCA
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Molecular Biology (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- Medicinal Chemistry (AREA)
- Hematology (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Urology & Nephrology (AREA)
- Food Science & Technology (AREA)
- General Physics & Mathematics (AREA)
- Cell Biology (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biotechnology (AREA)
- Oncology (AREA)
- Microbiology (AREA)
- Pathology (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Hospice & Palliative Care (AREA)
- Investigating Or Analysing Biological Materials (AREA)
- Other Investigation Or Analysis Of Materials By Electrical Means (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
Claims
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| AU2024226949A AU2024226949A1 (en) | 2023-02-23 | 2024-02-21 | Method of sequence-independent quantification of proteolytically unstable plasma lambda free light chain protein for al amyloidosis diagnosis |
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363486587P | 2023-02-23 | 2023-02-23 | |
| US63/486,587 | 2023-02-23 | ||
| US202363594556P | 2023-10-31 | 2023-10-31 | |
| US63/594,556 | 2023-10-31 | ||
| US202363607763P | 2023-12-08 | 2023-12-08 | |
| US63/607,763 | 2023-12-08 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024178117A1 true WO2024178117A1 (en) | 2024-08-29 |
Family
ID=90473296
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/016721 Ceased WO2024178117A1 (en) | 2023-02-23 | 2024-02-21 | Method of sequence-independent quantification of proteolytically unstable plasma lambda free light chain protein for al amyloidosis diagnosis |
Country Status (2)
| Country | Link |
|---|---|
| AU (1) | AU2024226949A1 (en) |
| WO (1) | WO2024178117A1 (en) |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022226166A1 (en) | 2021-04-22 | 2022-10-27 | Protego Biopharma, Inc. | Spirocyclic imidazolidinones and imidazolidinediones for treatment of light chain amyloidosis |
-
2024
- 2024-02-21 WO PCT/US2024/016721 patent/WO2024178117A1/en not_active Ceased
- 2024-02-21 AU AU2024226949A patent/AU2024226949A1/en active Pending
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022226166A1 (en) | 2021-04-22 | 2022-10-27 | Protego Biopharma, Inc. | Spirocyclic imidazolidinones and imidazolidinediones for treatment of light chain amyloidosis |
Non-Patent Citations (3)
| Title |
|---|
| JIANG XIN ET AL: "Blood-Based Diagnostic Assay for [lambda] Light Chain Amyloidosis through Quantification of an Amyloidogenicity-Indicating Neo-Epitope", BLOOD, vol. 142, no. Supplement 1, 2 November 2023 (2023-11-02), US, pages 536 - 536, XP093170928, ISSN: 0006-4971, Retrieved from the Internet <URL:https://ashpublications.org/blood/article-pdf/142/Supplement%201/536/2191691/blood-5140-main.pdf> DOI: 10.1182/blood-2023-175005 * |
| MARK RENZ ET AL: "2A4 binds soluble and insoluble light chain aggregates from AL amyloidosis patients and promotes clearance of amyloid deposits by phagocytosis", AMYLOID, vol. 23, no. 3, 5 August 2016 (2016-08-05), GB, pages 168 - 177, XP055708123, ISSN: 1350-6129, DOI: 10.1080/13506129.2016.1205974 * |
| SANDRA DAVERN ET AL: "Immunodiagnostic Capabilities of Anti?Free Immunoglobulin Light Chain Monoclonal Antibodies", AMERICAN JOURNAL OF CLINICAL PATHOLOGY, AMERICAN SOCIETY FOR CLINICAL PATHOLOGY, US, vol. 130, 1 January 2008 (2008-01-01), pages 702 - 711, XP007913119, ISSN: 0002-9173, DOI: 10.1309/AJCPNS6K1CYJPDBA * |
Also Published As
| Publication number | Publication date |
|---|---|
| AU2024226949A1 (en) | 2025-09-04 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| KR102716144B1 (en) | Analysis to detect neurodegeneration | |
| CN102947707B (en) | Progastrin and liver pathology | |
| JP2023065484A (en) | New tau species | |
| WO2011034128A1 (en) | Collagen neoepitope antibody | |
| WO2004029242A1 (en) | Antibody against enzyme specifically cleaving von villebrand factor and assay system using the same | |
| EP3132268B1 (en) | Immunoassay for the detection of chromogranin a | |
| JP6175058B2 (en) | In vitro evaluation of cardiovascular events by neoepitope assay of titin protein | |
| US20150355199A1 (en) | Agents, kits and methods for complement factor h-related protein 1 detection | |
| WO2024178117A1 (en) | Method of sequence-independent quantification of proteolytically unstable plasma lambda free light chain protein for al amyloidosis diagnosis | |
| JP5749643B2 (en) | Use of IRAP proteins for carrying out diagnostic and prognostic methods | |
| JP2025506203A (en) | Anti-HER2/NEU Antibodies and Methods of Use | |
| US20210388108A1 (en) | Antibodies specific for glycosylated apoj and uses thereof | |
| CN105980857A (en) | Biomarkers for Cardiac Disorders | |
| US20250076313A1 (en) | Anastellin Biomarker | |
| JP5605879B2 (en) | Antibody to sAPPβ | |
| WO2024219421A1 (en) | ANTIBODY AGAINST NT-proBNP, METHOD FOR DETECTING NT-proBNP USING THE ANTIBODY, AND COMPOSITION AND KIT FOR NT-proBNP DETECTION | |
| JP4423426B2 (en) | Method of using increased concentration of adrenomedullin precursor C-terminal peptide as an indicator of cardiovascular disease or inflammatory disease | |
| CA3210996A1 (en) | Anastellin biomarker | |
| JP2025533448A (en) | Immunoassay for detecting N-terminal peptides of proteolytic cleavage products of cartilage intermediate lamina protein-1 | |
| JP5885158B2 (en) | Antibodies against polypeptides containing sulfur-containing amino acid residues | |
| JP2012070648A (en) | Monoclonal antibody to mouse hig1 polypeptide | |
| KR20140015283A (en) | Immunological cofilin-1 protein measurement method | |
| KR20200067193A (en) | Copy protection against antibodies | |
| HK1229884B (en) | Biomarker for cardiac disorders | |
| HK1229425A1 (en) | Immunoassay for the detection of chromogranin a |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24714332 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: AU2024226949 Country of ref document: AU |
|
| ENP | Entry into the national phase |
Ref document number: 2024226949 Country of ref document: AU Date of ref document: 20240221 Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024714332 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2024714332 Country of ref document: EP Effective date: 20250923 |
|
| ENP | Entry into the national phase |
Ref document number: 2024714332 Country of ref document: EP Effective date: 20250923 |