COMPOSITIONS AND METHODS FOR IL13 BIOMARKERS

Abstract
Compositions and methods are provided for measuring and determining biomarkers for IL-13.
Description
BACKGROUND OF THE INVENTION

1. Field of the Invention


The invention relates to an assay method for monitoring the biological response to IL-13 or IL-13 therapy using discovered biomarkers for IL-13.


2. Background and Related Art


Asthma is a complex, chronic disorder, with a genetic and an environmental component. It is characterized by reversible airway obstruction, airway hyper-responsiveness, airway inflammation and remodeling. Asthma affects an estimated 15 million Americans and the morbidity and mortality associated with it is on the rise in industrialized countries. Recent reports using murine models of allergic asthma have shown that the Th2 type cytokine IL-13 may play a critical role in the pathogenesis of asthma, either by regulating airway inflammation, mucus hyper-secretion or airway hyper-responsiveness thus making it an attractive target for therapeutic intervention.


The role of biomarkers is becoming increasingly important in the clinical development of therapeutics. A biomarker can be an indicator of normal biological processes, disease processes, or pharmacological responses to therapeutic intervention. Their role ranges from, stratifying the patient population in helping to identify responders versus non-responders, to determining the efficacy of the therapeutic. Biomarkers can be a valuable tool in making better decisions that will reduce the cost for drug development and enable therapies to reach the right patient population faster.


The asthmatic population is very heterogeneous in terms of symptoms and response to therapy. Biomarkers can prove very useful in this by identifying responders versus non-responders. Thus leading to the development of more effective therapeutics.


SUMMARY OF THE INVENTION

This invention discloses the discovery of a panel of genes of which one or more can serve as biomarkers in an anti-interleukin(IL)-13 clinical trial.


The observation originated from data analysis of microarray gene expression profiling of peripheral blood mononuclear cells (PBMCs) from healthy donors. When comparing untreated PBMCs to PBMCs cultured with IL-13 or IL-4 this panel of genes was up regulated in all donors. The observation was confirmed by an independent method—Real Time Polymerase Chain Reaction (PCR) at messenger RNA level and the expression of all these 10 genes was up regulated by IL-13 and IL-4. A subsequent study looking at level of thymus and activation regulated chemokine (TARC) and macrophage derived chemokine (MDC) in the culture media supernatant of PBMCs showed that the neutralization of IL-13 by a fully human anti-interleukin 13 therapeutic antibody, as disclosed in US provisional patent application 60/679,925, filed May 11, 2005, published as U.S. Ser. No. ______, entirely incorporated herein by reference, inhibited the IL-13 induced TARC and MDC proteins as detected by ELISA. This disclosure presents evidence that this panel of genes can be used as biomarkers in an anti-IL-13 clinical study for inflammatory disorders such as asthma, emphysema and chronic obstructive pulmonary disease (COPD), and may also play a role in other lung disorders with an inflammatory component.


The panel of genes responsive to IL-13 disclosed provides methods for diagnosis or prognosis and tracking the efficacy of an anti-IL-13 therapy in IL-13 related disorders, including but not limited to, emphysema, asthma and chronic obstructive pulmonary disorder (COPD). Diagnostic and prognostic methods based on detecting these IL-13 responsive genes, or protein, in a sample is also disclosed.


Thus, in accordance with the invention, there is provided a method of monitoring the response to anti-IL13 therapy in a patient undergoing such therapy by using one or more IL-13 biomarkers, which comprises:

    • (a) Determining the level of expression of at least one of WNT5A or MPIF-1(CCL23) in a tissue sample of a patient;
    • (b) Administering the anti-IL-13 therapy to the patient;
    • (c) Measuring the level of at least one of WNT5A or MPIF-1 (CCL23) expression in said tissue of the patient and determining whether the anti-IL-13 therapy is effective in reducing the level of IL-13 or IL-13 related physiological effects.


      The method can also include at least one of the biomarkers CCL17 (TARC), CCL22 (MDC), CCL23 (MPIF-1) and CCL26 (Eotaxin 3), CD1A, CD1B, CD1C, IgE CD23A, IL-17Rbeta.




FIGURE DESCRIPTIONS


FIG. 1. Microarray data showing the average normalized intensity levels of CCL17, CCL22, CCL23 and CCL26 upon treatment of PBMCs from ten donors with IL-13, IL-4, anti-IL-13 mAb or isotype control mAb for 24 hrs.



FIG. 2. Real Time PCR (Taqman®) analysis showing the level of A) CCL17 B) CCL22 C) CCL23 D) CCL26 upon treatment of PBMCs from ten donors with IL-13, IL-4, anti-IL-13 mAb or isotype control mAb for 24 hrs. The quantity of each gene is normalized to GAPDH.



FIG. 3. Effect of neutralizing IL-13. PBMCs were incubated with IL-13, IL-13+anti-IL-13 mAb, or IL-13+isotype control mAb for 24 hr and 48 hr timepoints. The level of A) CCL17 B) MDC C) CCL26 was assessed in the cell culture supernatants using ELISA.




DETAILED DESCRIPTION

The present invention thus provides a new approach to monitoring anti-IL 13 therapy by detecting one or more biomarkers of IL-13, and IL-13 bioactivity. In summary, we have identified a panel of potential biomarkers, associated with IL-13 biology, which can be useful in an anti-IL-13 clinical trial or for monitoring therapy using methods well known in the art are as disclosed herein. Such a panel of biomarkers can be used as a tool to monitor the efficacy of an anti-IL-13 therapeutic such as an IL-13 antagonist such as a small molecule or a biologic, and provide valuable information in terms of dosing amount and frequency.


The relevance of these genes to disease is very important for them to be effective biomarkers. To that end a number of the genes discussed above have been linked to diseases where IL-13 is believed to play a role. Increased level of TARC has been reported in serum and induced sputum of asthmatics [11], plasma of children during asthma exacerbation [12] and serum of patients with atopic dermatitis [13]. This provides evidence that TARC might be involved in the pathogenesis of such disorders. Eotaxin 3 gene expression has been shown to be upregulated in bronchial biopsies of asthmatics after allergen challenge implying that eotaxin 3 may be important in late-phase cosinophil recruitment to the airways of asthmatics [14]. Also, CCL22 was shown to be up regulated in bronchoalveolar lavage (BAL) after a segmental allergen challenge of asthmatics[15].


Of the panel of potential biomarkers described above, WNT5A and MPIF-1 (CCL23) have not been previously reported to be regulated by IL-13. These are novel IL-13 related biomarkers, which can be useful in any disease where IL-13 plays a major role.


In one embodiment, the improvement is to use of at least one of WNT5A and MPIF-1 (CCL23) as a biomarker for IL-13 antagonist treatment or therapy.


Several methods have been reported for the measurement and any of these methods may be employed in the invention. These include (i) radioimmunoassays and single radial immunodiffusion procedures (Chambers, R. E. and Whicher, J. T. (1983); J. Immunol. Methods 59, 95; Marhaug, G. (1983) supra; Taktak, Y. S. and Lee, M. A. (1991); J. Immunol. Methods 136, 11); (ii) ELISA based assays (Zuckerman, S. H. and Suprenant, Y. M.(1986); J. Immunol. Methods 92, 3743; Dubois, D. Y. and Malmendier, C. L. (1988); J. Immunol. Methods 112, 71-75; Sipe, J. D. et al. (1989); J. Immunol. Methods 125, 125-135; Yamada, T. et al. (1989); Clin. Chim. Acta 179, 169-176; Tino-Casl, M. and Grubb, A. (1993); Arm. Clin. Biochem 30, 278-286); (iii) nephelometric methods (Vermeer, H. et al. (1990); Clin. Chem 36, 1192; Yamada, T. et al. (1993); Ann. Clin. Biochem. 30, 72-76); (iv) an electrophoretic procedure (Godenir, N. L. et al. (1985); J. Immunol. Methods 83, 217); (v) an immunochemiluminescence procedure (Hachem, H. et al. (1991); Clin. Biochem 24, 143-147); (vi) an automated method based on a monoclonal-polyclonal antibody solid phase enzymeimmunoassay (Wilkins, J. W. et al. (1994); Clin. Chem 40(7), 1284-1290); and (vii) time-resolved fluorometric immunoassay (Malle, E., et al. (1995); J. Immunol. Methods 182, 131). See U.S. Pat. No. 6,194,163 which discloses a method for the quantitative measurement of human acute phase serum amyloid A protein.


The following experiments demonstrate that one or more of the biomarkers described herein can be used as accurate biomarker to monitor the response to anti-IL-13 therapies such as anti-IL-13 antibody therapy.


EXAMPLE 1

Microarray Experiment Description:


Peripheral blood mononuclear cells (PBMCs) were isolated from 8 healthy donors. These were cultured in RPMI+10% FBS in the presence of IL-13 (10 ng/ml) or IL-4 (10 ng/ml) or CNTO 607 (10 μg/ml) or human IgG1 isotype control (10 μg/ml) for 24 hrs. Total RNA was isolated from the cells using the RNeasy kit (Qiagen). The quality and quantity of RNA was assessed using the bioanalyzer (Agilent) and microarray analysis was performed using the Affymatrix platform.


Number of samples: 40


Chip type: Affymetrix GeneChip Human Genome U133 Plus 2.0 arrays were used in this study. Each array is comprised of more than 54K probe sets that analyze the expression level of over 47K transcripts and variants, including 38.5K well-characterized human genes.


Number of chips: 40


Data Processing and Analysis:


Raw intensity data was downloaded from DNA Chip III database. Values below 0.01 were set to 0.01. Using GeneSpring (Redwood City, Calif.; version 7.2), chip-to-chip normalization was performed by dividing the averaged intensity of each probe set by the median intensity of a chip. The intensity of each probe set was then normalized to the median intensity of that probe set in the control group. The control groups in this study were the 8 untreated samples.


A probe set was regarded as reliably detected if it was called Present or Marginally present at least once among the 40 samples. Among 54675 probe sets on a chip, only 36357 probe sets passed the filtering and were analyzed further. Replicate samples were grouped according to their experimental conditions. The average of normalized intensities was used to represent each condition.


Using log 2 transformed normalized intensities, standard ANOVA was conducted in Partek Pro 6.1 (St. Charles, Mo.) to test treatment (untreated, IL13, IL4, CNTO 607, and isotype), and donor was also considered in the model as a random effect. Post-hoc tests were set up to identify genes showing significant differential expression between each treatment condition and untreated. False discovery rate cutoff was set at 0.05, meaning that 5% of identified genes would be false positives. Genes identified by statistical analysis were then filtered by fold change comparison between each treatment and untreated. The fold change cutoff was set at 1.5.


Microarray Resullts:


From the above analysis of the microarray data the following list of genes was identified as potential biomarkers for IL-13. These include:

    • Thymus and activation regulated chemokine (TARC) also known as CCL17
    • Eotaxin 3 (CCL 26)
    • Myeloid progenitor inhibitory factor 1 (MPIF-1), also known as CCL23
    • Macrophage derived chemokine (MDC), also known as CCL22
    • CD1A, CD1B, CD1C antigen
    • WNT5A (wingless-type MMTV integration site family, member 5A)
    • IgE CD23A
    • hIL-17R beta


All of these genes were up regulated with IL-4 and IL-13 when compared to untreated but CNTO 607 or isotype control, as shown in Table 1, did not modulate their expression. For some of the genes more than one probe set was detected by microarray further confirming the results.

TABLE 1The raw and normalized intensities of each of the genes frommicroarray analysis and their Gen Bank accession numbersGeneAverageAverageGenBank(Probe setRawNormalizedAccessionID)TreatmentIntensityIntensityNo.CCL17Untreated138.81.086NM_002987(207900_at)IL-132,26817.62IL-42,87022.42CNTO 607130.81.005Isotype177.11.41controlCCL26Untreated411.64AF096296(223710_at)IL-13188372.26IL-4188875.18CNTO 60741.021.51Isotype42.111.69controlCCL23Untreated35.961.76U58913(210549_s_at)IL-13538.825.8IL-4674.833.8CNTO 60741.051.9Isotype120.16.2controlCCL23Untreated17.81.4U58913(210548_at)IL-13268.121.2IL-4395.732.6CNTO 607252.0Isotype65.15.4controlCCL22Untreated15501.2NM_002990(207861_at)IL-1345073.4IL-443263.3CNTO 60714321Isotype26362controlCD1AUntreated102.61M28825(210325_at)IL-13433.34.3IL-4586.66CNTO 607116.41.1Isotype93.40.9controlCD1BUntreated104.71NM_001764(206749_at)IL-13171517.6IL-4214222.5CNTO 60792.90.9Isotype84.620.9controlCD1CUntreated347.11NM_001765(205987_at)IL-13425112.79IL-4486415.01CNTO 6073531Isotype289.80.9controlWNT5AUntreated33.70.8NM_003392(205990_s_at)IL-13958.123.6IL-4109127.1CNTO 607400.9Isotype111.52.7controlWNT5AUntreated32.11.4NM_003392(213425_at)IL-13390.617.6IL-4463.221CNTO 60731.61.4Isotype64.22.8controlWNT5AUntreated35.81.7NM_003392(231227_at)IL-13248.811.7IL-4256.412.2CNTO 60720.91Isotype381.8controlIgE CD23AUntreated106.81NM_002002(206759_at)IL-13281128.4IL-4339935.3CNTO 60799.51Isotype210.42.2controlIgE CD23AUntreated58.41.4NM_002002(206760_s_at)IL-13209149.4IL-4232156.2CNTO 60727.20.6Isotype52.71.2controlhIL-17 R betaUntreated83.22.3NM_018725(219255_x_at)IL-13379.710.3IL-4372.510.3CNTO 607122.73.3Isotype149.34.2controlhIL-17 R betaUntreated123.91.1NM_018725(224156_x_at)IL-134474IL-4405.63.7CNTO 607153.21.3Isotype180.21.6controlhIL-17 R betaUntreated100.91.2NM_018725(224361_s_at)IL-13362.94.5IL-4340.24.3CNTO 607128.31.6Isotype145.51.8control


Real Time PCR (TaqMan) Confirmation:


In order to confirm the microarray finding by an independent means, Real Time PCR technology was employed. Total RNA from PBMCs cultured in the presence of IL-13 (10 ng/ml) or IL-4 (10 ng/ml) or CNTO 607 (10 μg/ml) or human IgG1 isotype control (10 μg/ml) for 24 hrs was reverse transcribed and used in Real Time PCR analysis for each of the 10 genes using the Applied Biosystems Gene Expression Assays on Demand on the ABI PRISM® 7900HT Sequence Detection System. The results are summarized in Table 2.

TABLE 2The mean RNA quantity for each of the genes at various treatmentconditions using TaqManMean RNA quantity(Normalized toGeneTreatmentGAPDH)Assay ID #CCL17Untreated0.96Hs00171074_mlIL-13289.6IL-4489CNTO 6070.96Isotype2.4controlCCL26UntreatedUndetectedHs00171146_mlIL-131.1IL-41.3CNTO 607UndetectedIsotypeUndetectedcontrolCCL23Untreated0.57Hs00270756_mlIL-135.6IL-46.3CNTO 6070.5Isotype1.24controlCCL22Untreated1.4Hs00171080_mlIL-135.1IL-45.1CNTO 6071.3Isotype2.4controlCD1AUntreated1.1Hs00233332_mlIL-1332.4IL-442.5CNTO 6070.79Isotype1controlCD1BUntreated1.3Hs00233507_mlIL-1330.5IL-438.4CNTO 6071.1Isotype1.4controlCD1CUntreated0.9Hs00233509_mlIL-1318.3IL-419.8CNTO 6070.77Isotype0.5controlWNT5AUntreated1.2Hs00180103_mlIL-13147IL-4190.4CNTO 6072.1Isotype7.2controlIgE CD23AUntreated0.79Hs00233627_mlIL-1333.3IL-435.8CNTO 6070.7Isotype1.4controlhIL-17 RUntreated0.7Hs00218889_mlbetaIL-136IL-45.8CNTO 6070.7Isotype1.1control


The Real Time PCR data seems to confirm the microarray data for each of the 10 genes.


Protein Analysis via ELISA:


To further confirm these data at the protein level a follow up study in PBMCs from 5 healthy donors was performed. PBMCs were cultured with IL-13 (10 ng/ml), IL-13 (10 ng/ml) plus anti-IL-13 therapeutic antibody, CNTO 607 (2 pg/ml) or IL-13 (10 ng/ml) plus isotype control mAb, human IgG1 (2 μg/ml) for 24 and 48 hr time points. The PBMC culture supernatant was assayed for TARC protein using a human TARC ELISA assay (R&D Systems) and for MDC protein using a human MDC ELISA assay (R&D Systems). The data showed that the neutralization of IL-13 by CNTO 607 resulted in a down regulation of IL-13 induced TARC and MDC at the protein level (Table 3). These data suggest that TARC and MDC could serve as potential biomarkers in an anti-IL-13 clinical study.

TABLE 3Mean protein level in PBMC culture supernatants from n = 5donors following various treatment conditionsMDCTreatmentTime pointTARC (pg/ml)(pg/ml)Untreated24 hr0.670.6IL-13 (10 ng/ml)24 hr89519.4IL-13 (10 ng/ml) + human24 hr86.2473.6IgG1(2 μg/ml)IL-13 (10 ng/ml) + CNTO24 hr0.893.2607(2 μg/ml)Untreated48 hr2110IL-13 (10 ng/ml)48 hr287851.2IL-13 (10 ng/ml) + human48 hr361.61398.2IgG1(2 μg/ml)IL-13 (10 ng/ml) + CNTO48 hr2.4240.2607(2 μg/ml)


The role of biomarkers is becoming increasingly important in the clinical development of therapeutics. Their role ranges from, stratifying the patient population in helping to identify responders versus non-responders, to determining the efficacy of the therapeutic. Biomarkers can be a valuable tool in making better decisions that will reduce the cost for drug development and enable therapies to reach the right patient population faster.


Here we describe the use of the Microarray technology platform to identify biomarkers that are associated with the biology of IL-13, which could prove useful in an anti-IL-13 clinical trial. Peripheral blood mononuclear cells (PBMCs) from 10 healthy donors were cultured in the presence of IL-13, IL-4, anti-IL-13 mAb or an isotype control mAb and RNA from the treated cells was subjected to microarray. This revealed a number of genes that showed increased expression in the IL-13 and IL-4 treatment groups e.g., CCL17 (TARC), CCL22 (MDC), CCL23 (MPIF-1) and CCL26 (Eotaxin 3). These results were confirmed by Real Time PCR. A follow up study in PBMCs from five healthy donors that were cultured with IL-1 3, IL-13 plus anti-IL-13 mAb or IL-13 plus isotype control mAb showed that the neutralization of IL-13 resulted in a down regulation of IL-13 induced TARC, MDC and eotaxin 3 at the protein level. These data suggest that TARC, MDC and eotaxin 3 could serve as a potential efficacy specific biomarkers in an anti-IL-13 clinical study.


EXAMPLE 2

Background


Asthma is a complex, chronic disorder, with a genetic and an environmental component (ST 1999). It is characterized by reversible airway obstruction, airway hyper-responsiveness, airway inflammation and remodeling (1997). Asthma affects an estimated 15 million Americans and the morbidity and mortality associated with it is on the rise in industrialized countries. Recent reports using murine models of allergic asthma have shown that the Th2 type cytokine IL-13 may play a critical role in the pathogenesis of asthma, either by regulating airway inflammation, mucus hyper-secretion or airway hyper-responsiveness (Grunig G 1998; Wills-Karp M 1998; Walter D M 2001; Venkayya R 2002; Akbari O 2003) thus making it an attractive target for therapeutic intervention.


The role of biomarkers is becoming increasingly important in the clinical development of therapeutics. A biomarker can be an indicator of normal biological processes, disease processes, or pharmacological responses to therapeutic intervention (2001).Their role ranges from, stratifying the patient population in helping to identify responders versus non-responders, to determining the efficacy of the therapeutic. Biomarkers can be a valuable tool in making better decisions that will reduce the cost for drug development and enable therapies to reach the right patient population faster.


The asthmatic population is very heterogeneous in terms of symptoms and response to therapy. Biomarkers can prove very useful in this by identifying responders versus non-responders. Thus leading to the development of more effective therapeutics. One approach uses the DNA Microarray platform to identify biomarkers using precilinical models. Here we describe the use of the Microarray technology platform to identify biomarkers that are associated with the biology of IL-13, which could prove useful in an anti-IL-13 clinical trial.


Methods


Peripheral Blood Mononuclear Cell Isolation and Culture


Peripheral blood mononuclear cells (PBMCs) from healthy donors were isolated using Ficoll and density gradient centrifugation. The PBMCs were cultured in RPMI+10% FBS and left untreated or treated with IL-13 (10 ng/ml) or IL-4 (10 ng/ml) or anti-IL-13 mAb (10 μg/ml) or isotype control mAb (10 μg/ml ) for 24 hr.


RNA Isolation and Microarray


Total cellular RNA was isolated from the cells using the RNeasy mini kit (Qiagen, Inc. Valencia, Calif.) as per manufacturer's instructions. The IL-13 and IL-4 was purchased from R&D Systems (Minneapolis, Minn.). The quality and quantity of RNA was assessed using the Agilent 2100 Bioanalyzer (South Plainfield, N.J.). Samples that demonstrated high quality (ratio of 28S rRNA and 18S rRNA is greater than 1.7) were submitted for microarray analysis on the Affymatrix chip.


Microarray Processing


RNA amplification, probe synthesis and labeling, cDNA chip hybridization and washing were performed as described previously [17]. Agilent Image Scanner was used to scan the cDNA chips (Agilent Technologies, Palo Alto, Calif.). Fluorescence intensity for each feature of the array was obtained by using ImaGene software (BioDiscovery, Los Angeles, Calif.).


Microarray Data Analysis


In this study, Affymetrix GeneChip Human Genome U133 Plus 2.0 arrays were used to profile gene expression in human peripheral blood mononuclear cells from 10 donors stimulated with IL-13, or IL-4 at 1 time point (24 hr). Untreated samples from the same group of donors were used as control. Each array is comprised of more than 54K probe sets that analyze the expression level of over 47K transcripts and variants, including 38.5K well-characterized human genes. Raw intensity data was downloaded from DNA Chip III database. Values below 0.01 were set to 0.01. Using GeneSpring (Redwood City, Calif.; version 7.2), chip-to-chip normalization was performed by dividing the averaged intensity of each probe set by the median intensity of a chip.


The intensity of each probe set was then normalized to the median intensity of that probe set in the control group. The control groups in this study were the 8 untreated samples.


A probe set was regarded as reliably detected if it was called Present or Marginally present at least once among the 40 samples. Among 54675 probe sets on a chip, only 36357 probe sets passed the filtering and were analyzed further. Replicate samples were grouped according to their experimental conditions. The average of normalized intensities was used to represent each condition.


Using log 2 transformed normalized intensities, standard ANOVA was conducted in Partek Pro 6. 1 (St. Charles, Mo.) to test treatment (untreated, IL13, IL4, anti-IL-13 mAb, and isotype control mAb), and donor was also considered in the model as a random effect. Post-hoc tests were set up to identify genes showing significant differential expression between each treatment condition and untreated. False discovery rate cutoff was set at 0.05, meaning that 5% of identified genes would be false positives. Genes identified by statistical analysis were then filtered by fold change comparison between each treatment and untreated. The fold change cutoff was set at 1.5.


Reverse Transcription and Real Time PCR


1 μg of total RNA from each of the treated and untreated groups of PBMCs were used for the reverse transcription (RT) reaction. The RT reaction was performed as per protocol using TaqMan® RT reagents (Applied Biosystems) at 37° C. for 120 min followed by 25° C. for 10 min. Forty ng of cDNA per reaction were used in the Real Time PCR using the ABI Prism® 7900 sequence detection system (Foster City, Calif.). In the presence of AmpliTaq Gold DNA plolymerase (ABI biosystem, Foster City, Calif.), the reaction was incubated for 2 min at 50° C. followed by 10 min at 95° C. Then the reaction was run for 40 cycles at 15 sec, 95° C. and 1 min, 60° C. per cycle. Assays-on-Demand™ primers and probes (Applied Biosystems) were used in the PCR. The Real Time PCR data was analyzed using the standard curve method.


PBMC Culture and ELISA


PBMCs were cultured in the presence of IL-13 (10 ng/ml) or IL-13 (10 ng/ml)+anti-IL-13 mAb (10 μg/ml) or IL-13 (10 ng/ml)+isotype control mAb (10 μg/ml). Culture media supernatants were collected and tested for levels of TARC, MDC and eotaxin 3 protein at 24 hr and 48 hr time points using human TARC specific, MDC specific and human eotaxin 3 specific ELISA as per the manufacturer's instructions (R&D Systems)


Results


IL-13 Upregulates Genes in PBMCs


In order to identify genes downstream of IL-13, which could serve as potential efficacy specific biomarkers cDNA microarray technology platform was used. IL-4 was also included in the treatment groups to try and differentiate between IL-13 and IL-4 and identify any markers that could be uniquely regulated by IL-13 alone.


Ten donors were used and after analyzing the microarray data it became apparent that both IL-13 and IL-4 generated a very similar expression profile i.e., genes upregulated by IL-13 were the same as those upregulated by IL-4 at the 1.5 fold cutoff. From this a list of potential biomarkers was identified (Table 4). FIG. 2 shows a graphical representation of the microarray data for four of them.


Real Time PCR Validation


TaqMan™ Real Time PCR was used to validate the potential biomrkers. As shown in FIG. 2, CCL17, CCL22, CCL23 and CCL26 were all upregulated by IL-13 or IL-4 while their expression was not modulated by anti-IL-13 mAb or the isotype control mAb. This is very comparable to the microarray data.


Validation of Expression at Protein Level and Inhibition of IL-13 Induced Protein by an Anti-IL-13 mAb


In order to validate the modulation of genes by IL-13 at the protein level, ELISA was used to detect potein in cell culture supernatants and confirm upregulation of CCL17, CCL22 and CCL26 by IL-13. As shown in FIG. 3, IL-13 (10 ng/ml, 24 hr and 48 hr) stimulates the protein expression of CCL17, CCL22 and CCL26. Also neutralization of IL-13 by an anti-IL-13 mAb results in a down regulation of IL-13 induced CCL17, CCL22 and CCL26.


Discussion


It is becoming increasing evident that there is a great need for biomarkers to become an integral part of drug discovery. Biomarkers can be especially valuable in helping to make early decisions on lead compounds and thereby drive down the cost of drug development. The microarray technology platform provides a unique approach in profiling the entire genome and thereby provide insight into potential markers that can help stratify a patient population or predict respone to therapy.


The asthmatic patient population is highly heterogenous both in terms of sysmptoms and response to therapy. Biomarkers can play a key role in identifying responders versus non-responders and thereby lead to the development of more effective therapies. A number of studies have been described where microarray platform has been used to identify asthma signature genes.


Here we describe the use of microarray technology to identify genes associated with the biology of IL-13.We have identified a panel of potential biomarkers, associated with IL-13 biology, which are down regulated upon neutralizing IL-13 and thus can be useful in an anti-IL-13 clinical trial. Such a panel of biomarkers can be used as a tool to monitor the efficacy of an anti-IL-13 therapeutic antibody.


The relevance of these genes to disease is very important for them to be effective biomarkers. To that end a number of the genes discussed above have been linked to diseases where IL-13 is believed to play a role. Increased level of TARC has been reported in serum and induced sputum of asthmatics (T Sekiya 2002), plasma of children during asthma exacerbation (Leung, Wong et al. 2003) and serum of patients with atopic dermatitis (Hijnen, de Bruin-Weller et al. 2004). This provides evidence that TARC might be involved in the pathogenesis of such disorders. Eotaxin 3 gene expression has been shown to be upregulated in bronchial biopsies of asthmatics after allergen challenge implying that eotaxin 3 may be important in late-phase eosinophil recruitment to the airways of asthmatics (Berkman, Ohnona et al. 2001). Also, CCL22 was shown to be up regulated in bronchoalveolar lavage (BAL) after a segmental allergen challenge of asthmatics(Pilette, Francis et al. 2004). Of the panel of potential biomarkers described above, WNT5A and MPIF-1 (CCL23) have not been previously reported to be regulated by IL-13. These are novel IL-13 related biomarkers, which can be useful in any disease where IL-13 plays a major role.


It is worth pointing out that he ultimate validity of a potential biomarker or panel of biomarkers can only be tested in the clinic. However, the purpose of this study and studies like this is to identify potential candidates which can be put forth for validation in a clinical setting.

TABLE 4Potential biomarkers associated with IL-13 biologyCCL17 (TARC thymus & activation regulatedchemokine)CCL26 (Eotaxin 3)CCL23 (MPIF-1 myeloid progenitor inhibitory factor1)CCL22 (MDC macrophage derived chemokine)CD1 A, B, CWNT5AIgE CD23AhIL-17 R beta


Figure Legends:



FIG. 1. Microarray data showing the average normalized intensity levels of CCL17, CCL22, CCL23 and CCL26 upon treatment of PBMCs from ten donors with IL-13, IL-4, anti-IL-13 mAb or isotype control mAb for 24 hrs.



FIG. 2. Real Time PCR (Taqman®) analysis showing the level of A) CCL17 B) CCL22 C) CCL23 D) CCL26 upon treatment of PBMCs from ten donors with IL-13, IL-4, anti-IL-13 mAb or isotype control mAb for 24 hrs. The quantity of each gene is normalized to GAPDH.



FIG. 3. Effect of neutralizing IL-13. PBMCs were incubated with IL-1 3, IL-13+anti-IL-13 mAb, or IL-13+isotype control mAb for 24 hr and 48 hr timepoints. The level of A) CCL17 B) MDC C) CCL26 was assessed in the cell culture supernatants using ELISA.


REFERENCES



  • (1997). “National Institutes of Health. Guidelines for the diagnosis and management of asthma. Rep. No. 97-4051. Washington, D.C.: U.S. Dep. Health Human Serv., Natl. Heart Lung Blood Inst.”

  • (2001). “Biomarkers and surrogate endpoints: preferred definitions and conceptual framework.” Clin Pharmacol Ther 69(3): 89-95.

  • Akbari O, S. P., Meyer E, Kronenberg M, Sidobre S, Nakayama T, Taniguchi M, Grusby M J, DeKruyff R H, Umetsu D T. (2003). “Essential role of NKT cells producing IL-4 and IL-13 in the development of allergen-induced airway hyperreactivity.” Nat Med 9: 582-588.

  • Berkman, N., S. Ohnona, et al. (2001). “Eotaxin-3 but Not Eotaxin Gene Expression Is Upregulated in Asthmatics 24 Hours after Allergen Challenge.” Am. J. Respir. Cell Mol. Biol. 24(6): 682-687.

  • Elias J, Z. A., Chupp G, Homer R. (1999). “Airway Remodeling in asthma.” J Clin Invest 104(8): 1001-6.

  • Grunig G, W. M., Wakil A E, Venkayya R, Brombacher F, Rennick D M, Sheppard D, Mohrs M, Donaldson D D, Locksley R M, Corry D B. (1998). “Requirement for IL-13 independently of IL-4 in experimental asthma.” Science 282: 2261-2263.

  • Hijnen, D., M. de Bruin-Weller, et al. (2004). “Serum thymus and activation-regulated chemokine (TARC) and cutaneous T cell-attracting chemokine (CTACK) levels in allergic diseases: TARC and CTACK are disease-specific markers for atopic dermatitis.” Journal of Allergy and Clinical Immunology 113(2): 334-340.

  • Leung, T. F., C. K. Wong, et al. (2003). “Plasma TARC concentration may be a useful marker for asthmatic exacerbation in children.” Eur Respir J 21(4): 616-620.

  • Pilette, C., J. N. Francis, et al. (2004). “CCR4 ligands are up-regulated in the airways of atopic asthmatics after segmental allergen challenge.” Eur Respir J 23(6): 876-884.

  • Society, A. T. (2000). “Proceedings of the ATS workshop on refractory asthma: current understanding, recommendations, and unanswered questions.” Am J Respir Crit Care Med 162(6): 2341-51.

  • ST, H. (1999). “The epidemic of allergy and asthma.” Nature 402: B2-B4.

  • T Sekiya, H. Y., M Yamaguchi, K Yamamoto, A Ishii, O Yoshie, Y Sano, A Morita, K Matsushima, K Hirai (2002). “Increased levels of a TH2-type CC chemokine thymus and activation-regulated chemokine (TARC) in serum and induced sputum of asthmatics.” Allergy 57(2): 173-177.

  • Venkayya R, L. M., Willkom M, Grunig G, Corry D B, Erle D J. (2002). “The Th2 lymphocyte products IL-4 and IL-13 rapidly induce airway hyperresponsiveness through direct effects on resident airway cells.” Am J Respir Cell Mol Biol 26: 202-208.

  • Walter D M, M. J., Berry G, McKenzie A N, Donaldson D D, DeKruyff R H, Umetsu D T. (2001). “Critical role for IL-13 in the development of allergen-induced airway hyperreactivity.” J Immunol 167: 4668-4675.

  • Wills-Karp M, L. J., Xu X, Schofield B, Neben T Y, Karp C L, Donaldson D D (1998). “Interleukin-13: central mediator of allergic asthma.” Science 282: 2258-2261.


Claims
  • 1. A method of monitoring the response to anti-IL13 therapy in a patient undergoing such therapy by using one or more IL-13 biomarkers, which comprises: (d) Determining the level of expression of at least one of WNT5A or MPIF-1(CCL23) in a tissue sample of a patient; (e) Administering the anti-IL-13 therapy to the patient; (a) Measuring the level of at least one of WNT5A or MPIF-1 (CCL23) expression in said tissue of the patient and determining whether the anti-IL-13 therapy is effective in reducing the level of IL-13 or IL-13 related physiological effects.
  • 2. The method of claim 1 wherein the therapy is for the treatment of asthma, COPD or emphysema and said expression of at least one of WNT5A or MPIF-1 is employed as an IL-13 therapy responsive biomarker.
  • 3. The method of claim 1 wherein the anti-IL-13 therapy is selected from agents that neutralize or inhibit IL-13.
  • 4. The method of claim 3 wherein the anti-IL-13 therapy is an anti-IL-13 antibody or fusion protein.
  • 5. The method of claim 4 wherein the anti-IL-13 therapy is the administration of an anti-IL-13 neutralizing antibody.
  • 6. The method of claim 1, wherein said biomarkers optionally further include at least one of the biomarkers CCL17 (TARC), CCL22 (MDC), CCL23 (MPIF-1) and CCL26 (Eotaxin 3), CD1A, CD1B, CD1C, IgE CD23A, IL-17Rbeta.
PRIOR APPLICATION

This application claims priority to U.S. application No. 60/722,296, filed Sep. 30, 2005, which is incorporated herein by reference.

Provisional Applications (1)
Number Date Country
60722296 Sep 2005 US