NZ717890B2 - Methods and Systems for Identifying and Treating Anti-Progestin Sensitive Tumors - Google Patents

Methods and Systems for Identifying and Treating Anti-Progestin Sensitive Tumors Download PDF

Info

Publication number
NZ717890B2
NZ717890B2 NZ717890A NZ71789012A NZ717890B2 NZ 717890 B2 NZ717890 B2 NZ 717890B2 NZ 717890 A NZ717890 A NZ 717890A NZ 71789012 A NZ71789012 A NZ 71789012A NZ 717890 B2 NZ717890 B2 NZ 717890B2
Authority
NZ
New Zealand
Prior art keywords
progesterone receptor
cells
tumor
antibody
progestin
Prior art date
Application number
NZ717890A
Other versions
NZ717890A (en
Inventor
Erard Gilles
Original Assignee
Invivis Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Invivis Pharmaceuticals Inc filed Critical Invivis Pharmaceuticals Inc
Publication of NZ717890A publication Critical patent/NZ717890A/en
Publication of NZ717890B2 publication Critical patent/NZ717890B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41521,2-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. antipyrine, phenylbutazone, sulfinpyrazone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/567Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in position 17 alpha, e.g. mestranol, norethandrolone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00Ā -Ā A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/025Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/72Assays involving receptors, cell surface antigens or cell surface determinants for hormones
    • G01N2333/723Steroid/thyroid hormone superfamily, e.g. GR, EcR, androgen receptor, oestrogen receptor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/743Steroid hormones

Abstract

Discloses the use of an anti-progestin composition in the preparation of a medicament for the treatment of a tumor susceptible to growth inhibition by one or more anti-progestin(s), wherein the tumor is identified as susceptible to growth inhibition by one or more anti-progestin(s) wherein a) a tissue sample suspected of being tumorigenic from a patient is exposed to an anti-progesterone receptor antibody; b) flow cytometry is performed on the tissue sample to identify progesterone receptor positive cells; c) high magnification microscopy is used to identify activated progesterone receptor foci in the progesterone receptor positive cells; and d) the focal binding distribution of the progesterone receptor in nuclei of cells from the tissue is determined, wherein if the focal binding distribution is greater than 5% of the progesterone receptor positive cells in the tissue sample with an A or AD binding pattern, then the tumor is determined to be susceptible to growth inhibition by one or more anti-progestin(s). sue sample suspected of being tumorigenic from a patient is exposed to an anti-progesterone receptor antibody; b) flow cytometry is performed on the tissue sample to identify progesterone receptor positive cells; c) high magnification microscopy is used to identify activated progesterone receptor foci in the progesterone receptor positive cells; and d) the focal binding distribution of the progesterone receptor in nuclei of cells from the tissue is determined, wherein if the focal binding distribution is greater than 5% of the progesterone receptor positive cells in the tissue sample with an A or AD binding pattern, then the tumor is determined to be susceptible to growth inhibition by one or more anti-progestin(s).

Description

METHODS AND SYSTEMS FOR IDENTIFYING AND TREATING ANTI- PROGESTTN IVE TUMORS BACKGROUND This application claims priority to US. Provisional Patent Number 61/542,931, filed on October 4, 2011, the disclosure of which is incorporated by reference herein in its entirety.
The progesterone receptor (PR) is present in cells in two major ms, PRā€”A and PR-B. In the presence of a bound progestin ligand, such as terone, the PR is phosphorylated at speciļ¬c sites, dimerizes, forms a complex with a number of different cellular elements (e.g., p300 and the steroid receptor coactivator), and binds to speciļ¬c DNA sequences known as progesterone responsive elements (PRES) to initiate DNA transcription into RNA. The PR-ligand complex also attracts numerous other co~ activators and coā€”repressors, which form the cellular elements which in turn ribe particular genes. These PR complexes (also referred to as foci) can be visualized in the nuclei of cells which contain the progesterone receptor as ļ¬‚uorescent aggregates using immunohistoļ¬‚uorescence ques and as dense and dark stained nuclear aggregates using the histochemistry techniques described in this patent.
In premenopausal women, during the proliferative phase (the first part of the menstrual cycle) when en is the dominant hormone and progesterone is minimally secreted, staining of normal endometrial cells for PRā€”A and PRā€”B (e.g., using immunofluorescent techniques and confocal microscopy) reveals a e progesterone receptor nuclear staining pattern. In the secretory phase (the second part of the menstrual cycle) when progesterone is the dominant hormone, using the same immunofluorescent techniques and confocal microscopy, staining for PRā€”A and PRā€”B appears as readily detectable ļ¬‚uorescent nuclear foci.
RNA ription inhibitors have been shown to prevent formation of PR foci, and 26S proteasome inhibitors have been shown to t the PR nuclear foci. It is 2012/058732 therefore believed that the presence of PR foci in cells corresponds to active transcriptional complexes, and indicates the tion of the PR and subsequent gene sion. sely, diffuse r staining or the absence of PR foci indicates the presence of PR which is transcriptionally inactive. Upon exposure of normal breast and endometrium tissues (which are physiologically responsive to progesterone) to progestin ligands, a change from a diffuse nuclear staining pattern to focal subnuclear structures can be observed, indicating the activation of the progesterone receptor.
Whereas estrogens are mitogenic (e.g., cause cellular proliferation) for normal breast epithelial and endometrial cells, the effects of progestins are more complex. In the endometrium, progestins inhibit estrogenā€”induced cell cycle progression early in the G1 phase, s in the breast progestins may both stimulate and inhibit proliferation. In normal breast tissue biopsies it has been shown that proliferative activity is stimulated by terone (Am J Obstet Gynecol, 1997). This complexity has led to confounding experimental observations in breast cancer. For example, togens redā€” appear to have a direct proliferative effect on breast cancer cell in vitro when phenol free media is used. H. J. Kloosterboer,J. Steroid Biochem. Molec. Biol. Vol. 49, No. 46, pp. 311ā€”318, 1994. However, when the same contraceptive progestogens that induced proliferation in breast cancer cell lines were studied in an estrogen-dependent DMBA rat breast cancer model, these progestogens inhibited tumor ssion. Id. It has been shown recently that many such in vitro experimental models are inadequate. See, _e_.g,, Lange C. et al. Progesterone Receptor Action: Translating s in Breast Cancer Models to Clinical Insights. Chapter 7 in Innovative Endocrinology of Cancer; 94ā€”111 (2010). While progesterone-induced proliferation has been shown in these experimental models, the majority of proliferating cells were not expressing the PR. Thus, these models do not necessarily predict the efficacy of treatment with antiprogestins.
Malignant cells also exhibit nuclear PR foci, but they are different in size and composition from the foci of normal cells. PR foci observed in cancer indicate a speciļ¬c role for the PR which is pertinent to the malignant nature of the cells. For e, the genes activated by the PR in malignant (cancer) breast cells are different than the genes activated by the PR in normal breast cells; in endometrial cancers PR foci, but not PR levels, are associated with malignant teristics; foci in cancer cells are larger, which may be due to alterations in the chromatin remodeling which are common in cancer, and; PR foci in breast cancer are observed regardless of hormonal status (e.g., in the presence and absence of circulating terone in premenopausal and post- menopausal women respectively). PR foci have been observed (e.g., using immunofluorescent techniques and confocal microscopy) in the tumor cells of imately 50% of PRā€”receptor positive human breast cancer es. Other patientā€™s tumor samples exhibited a diffuse PR nuclear staining pattern in the tumor cells using immunoļ¬‚uorescent techniques and confocal microscopy, indicative of a non- activated or nctional form of the PR.
The majority of breast cancers can be treated with hormonal treatments (i.e., strogens or aromatase inhibitors), which are currently some of the most effective medications used in breast cancer y. Hormonal ent is y indicated based on the identification of e receptors within the cancer cells.
Onapristone (ONA) is an anti-progestin drug which was originally developed for contraceptive use. However, it has demonstrated substantial activity in advanced breast with breast cancer, with a 10% se rate in a study of 101 poor prognosis patients cancer in whom prior hormonal therapy had failed (e.g., breast cancer progressed despite the patient receiving the antiestrogen tamoxifen). In a small breast cancer study using DNA as a ļ¬rst line hormone treatment, ONA produced a 56% objective response rate, an efficacy in the upper range of the best ble treatments in this disease. ONA binds to the PR, does not induce PR phosphorylation and does not allow the PR to dimerize. The PR-ONA complex binds weakly, or not at all, to its target DNA segment and therefore does not activate the chromatin remodeling which is a necessary process for DNA transcription. In in vitro systems, DNA has been shown to reverse the PR nuclear aggregates produced by binding of an artiļ¬cial ligand to the PR. Gene activation studies have consistently shown that, while progestins and other anti~progestins activate progesterone responsive genes, DNA has minimal activation (i.c., 3 genes).
In addition, DNA is a pure PR antagonist at concentrations which can be physiologically achieved. ONA does not interfere with other steroid receptors and does not increase en secretion in human ts, which is an undesirable sideā€”effect breast cancer therapy ted by other anti-progestins such as mifepristone.
While onapristone has previously been igated as a potential therapeutic agent for breast cancer, its development was stopped due to toxicity ns. son et al., Onaprz'stone, a Progesterone Receptor Antagonist, as First-Zine Therapy in Primary Breast Cancer European J. of Cancer 35(2) 214ā€”21 8 (1999). It is important to identify the subset of the patients with tumors most likely to respond and equally as important to identify the subset of the patients with tumors least likely to respond to treatment with DNA and other anti-progestins. Identifying these subsets of patients will allow those patients with APF access to a potentially effective cancer treatment and will avoid exposing patients with those cancers for which DNA or other antiā€”progestins may not provide beneļ¬t to unnecessary toxicity.
Currently, only the presence or absence of the estrogen or progesterone whether to use an endocrine or is considered when making therapeutic decisions on treatment in certain cancers (e.g., breast cancer). Accordingly, conventional assays for PR fy the tumors from patients with cancer into two categories: PRā€”positive or PRā€” negative. One type of assay quantitates the amount of PR per total protein of the cell.
These methods can be automated and are quantitative, but are not satisfactory with respect to accuracy, sensitivity and analysis of cellular subnuclear receptor structures. second type of assay includes immunohistochemical methods using in ļ¬xed tissue specimens and ļ¬‚uorescent or chromophore labeled monoclonal antibodies ing the or (either an antibody for each of PRā€”A and PRā€”B, or a single antibody that izes both). With immunohistochemical methods, any microscopically detectable nuclear staining reaction in more than a certain percentage of cells (typically 2 1%), is reported as being PR positive as per professional society ines. Typically, a clinical cut off of 210% ER or PR positive cells is used to make therapeutic decisions regarding the use of anti-hormone treatments. No consideration is given to the pattern of cellular or nuclear staining. Relative staining intensity (i.e., low, medium, or high) is also use as a qualitative measure of hormone receptor positivity. This second type of assay is more labor intensive and it is not standardized. Typically, low magniļ¬cation microscopic examination is used for the IHC analysis to identify the presence of the hormone receptor (either estrogen receptor (ER) or PR). Using tional methods, no analysis of cellular distribution is done other then an estimate of the percentage of the tumor cells sing the identified hormone receptor. Analysis of the subnuclear distribution pattern of the PR requires high powered microscopy. In contrast, high powered microscopy is not needed for standard IHC determination of hormone receptors in tumor tissue. These conventional methods of hormone receptor determination are thus unable to provide information regarding lear PR distribution. tins have complex actions in the breast and other hormone sensitive tissues by targeting distinct cells and having ct effects on cells not expressing the PR. PR foci complexes are not qualitatively the same in normal tissue and cancerous tissue, and they do not necessarily activate the same progesterone receptor associated genes. Available clinical data does not fully support the position that conventional techniques for identifying hormone or positive cells are predictive of ormone efļ¬cacy, whether it be for antiā€”estrogen or antiā€”progestin directed treatments. Currently, the decision to utilize a hormone ent (e.g., antiestrogens or ase inhibitors) for patients with breast cancer and other hormone sensitive tumors is based on the simple presence of hormone-receptors in tumor samples. The presence of hormone receptors (ER or PR) does not fully predict for response to e treatment, as only 50ā€”60% of hormone-receptor positive tumor cases are expected to benefit from ent.
There is a need for a consistent method for ting the efļ¬cacy of DNA and other antiā€”progestins with respect to heterogeneous ā€œnaturally ingā€ tumors.
Further, there is a need for an assay which is predictive of therapeutic efficacy of DNA and other antiā€”progestins against the cancers in individual patients.
SUMMARY An important question pertinent to anti-progestin treatment is how to fy activated PRs that are relevant clinical therapeutic targets. The present exemplary methods are aimed at characterizing PRs that are present in a functional (activated) state in the human tumor tissue routinely obtainable in the clinical setting. As antagonizing nonā€”active PR with a c rogestin is therapeutically pointless, the with present methods provide new and critical information to guide ent of patients antiā€”progestins. Such a predictive diagnostic test would e (1) consistent methods to support therapeutic decisionā€”making with respect to DNA and other antiā€”progestins, (2) guide selection of individual ts and patient populations that are likely to respond to treatment, and (3) exclude those individual patients that are least likely to respond or beneļ¬t from an anti-progestin treatment.
In one aspect, a method for identiļ¬cation and treatment of a subset of with an anti- progesterone receptor (PR) positive tumors most susceptible to treatment progestin such as onapristone (DNA) is provided. Progesterone receptor positive tumors exhibiting a dense, focal PR r distribution pattern, as described herein, are more susceptible to treatment with antiā€”progestins such as onapristone. Results from in vitro homogeneous, experimental models are not necessarily predictive of the ties of naturally-occurring heterogeneous tumors.
In another aspect, a method of inhibiting the growth of a tumor susceptible to growth inhibition by antiā€”progestins is provided. A tissue sample suspected of being tumorigenic or cancerous can be obtained from a patient. Progesterone receptor positive cells in the tissue sample can be ļ¬ed. The degree of distribution of the progesterone tissue sample can then receptor foci in nuclei of the progesterone positive cells from the be determined and an rogestin can be administered to the patient if the degree of focal distribution in the tissue sample is greater than about 5% of the progesterone receptor positive cells.
These patients are more likely to beneļ¬t from treatment with an antiā€” progestin that inactivates activated progesterone foci (APP) (e.g., DNA) and ts further formation of APF than patients whose tumors do not express activated PR. The nonā€”activated form of the PR is typically seen as diffuse nuclear PR staining. vation of the APF by an anti-progestin may occur by any of a y of mechanisms, ing dissociation of the foci and inhibition of activation of the foci without substantially altering their structure. In one aspect, APF formation can be ted or prevented by an anti-progestin through several mechanisms. For example, onapristone may not allow the individual progesterone receptors to dimerize and prevent the PR from being phosphorylated at the ligand phosphorylation sites. The PR-ONA complex may bind weakly, or not at all, to its target DNA segment (PRES) and fail to induce the chromatin remodeling which is a necessary process for DNA transcription. In another example, other antiā€”progestins may allow the PR to dimerize and form complexes with co-activators or coā€”repressors which do not induce DNA transcription.
In this example, DNA binding may occur at the PRE, but transcription does not occur. Identiļ¬cation of APF may inform the decision of any antiā€”progestin treatment as long as the agent interferes with the PR pathway. In one aspect, identiļ¬cation of APF determines the status of the PR pathway as activated or not. For e, the use of mifepristone, or any progestin that complexes with PR and binds to the DNA, could be informed by the ļ¬cation of APF. The activity of other agents, ing those which would inhibit PR phosphorylation and thus interfere with PR tion, would be ted by the presence of APF in various cancers. Thus, identification of APF could be used to inform treatment recommendations for various classes of compounds which act by inhibiting the function of the PR.
Patient tumors that do not express activated PR foci (APP) may include those that are PRā€”negative by the conventional assay, or those that are PRā€”positive by the conventional assay. In one aspect, any cancer which exhibits APF is a candidate for treatment with such antiā€”progestins, including breast, brain, meningiomas, prostate, ovarian, endometrial, uterine leiomyoma, lung, and uterine cancers. Pulmonary leiomyomatosis which has yet to be formally classified as a cancerous condition would also be likely to benefit if APF is expressed in the abnormal tissue. In another aspect, benign tumors not manageable with standard treatment, but presenting APF, can be treated by an antiprogestin as the ce of APF indicates that the tumor is driven by aberrant activation of PR, i.e. by the progestin pathway. [0019A] Another aspect provides a method of treating patient with a tumor susceptible to growth inhibition by anti-progestins by ing a tissue sample suspected of being genic or cancerous from a t and exposing the tissue to an anti-progesterone receptor antibody. Progesterone receptor positive cells in the tissue sample can be identified. The degree of focal binding distribution of the progesterone receptor in nuclei of cells from the tissue can be determined. If the focal binding distribution is greater than about 5% of the progesterone receptor ve cells in the tissue sample, an anti-progestin is administered to the patient in a dosage range of about 10 to about 200 mg per day depending upon the potency, bioavailability, and safety profile of the antiprogestin. [0019B] In another aspect, the present invention provides the use of an antiprogestin composition in the preparation of a medicament for the treatment of a tumor susceptible to growth inhibition by one or more anti-progestin(s), wherein the tumor is identified as tible to growth inhibition by one or more anti-progestin(s) wherein a) a tissue sample suspected of being tumorigenic from a patient is exposed to an anti-progesterone receptor dy; b) flow cytometry is performed on the tissue sample to identify progesterone receptor ve cells; c) high magnification microscopy is used to fy activated progesterone receptor foci in the progesterone receptor ve cells; and the focal binding distribution of the progesterone receptor in nuclei of cells from the tissue is determined, wherein if the focal binding distribution is r than 5% of the progesterone receptor positive cells in the tissue sample with an A or AD binding pattern, then the tumor is determined to be susceptible to growth inhibition by one or more anti-progestin(s).
In another aspect, the tissue is a specimen of a tumor tissue ed from the group consisting of breast, brain, meningiomas, te, ovarian, endometrial, uterine leiomyoma, lung, and uterine tissue.
In another aspect, the presence or absence of focal bution is detected by fluorescence, a colorimetic reaction (e.g., an enzymatic reaction), imaged with a counter staining antibody (e.g., chromophore), radioactivity, and Western blot (e.g., differential phosphorylation of the PR).
In yet another , the anti-progestin is selected from the group consisting of onapristone, lonaprisan, mifepristone, PF-02413873, telapristone, lilopristone, ORG2058, asoprisnil, and ulipristal.
The presence of active terone receptor focal distribution is indicated by a degree of nuclear focal distribution of greater than about 5% of the progesterone receptor positive cells. In r aspect, a tumor may be heterogeneous with respect [Text continued on page 9] focal distribution and t an active binding pattern (A) with distinct terone receptor foci, a diffuse binding pattern (D) without distinct progesterone receptor foci, or of the tumor. a mixture of an A n and a D pattern (AD) in various areas In any of the foregoing aspects, when focal distribution (A or AD pattern) is present, the intensity or density of such focal distribution may be quantitated. For e, progesterone receptor dies may be radiolabeled, cently labeled, imaged with a counter staining antibody (chromophore), imaged with a colorimetic reaction (e. g., an enzymatic reaction), or labeled in another manner where the intensity of the label can be measured and quantiļ¬ed.
FIGURES FIGS. 1A and 1B shows ary histochemical brown nuclear staining patterns in human breast cancer samples derived from formalin-ļ¬xed and paraffinā€”embedded biopsies using antibodies directed to the progesterone receptor; FIGS. 2A and 2B show exemplary green nuclear staining patterns in human breast cancer samples derived from formalin-ļ¬xed and parafļ¬n-embedded biopsies using antibodies directed to the progesterone receptor; FIGS. 3A and 3B show exemplary immunohistochemical brown nuclear staining ns with HES background counterstaining in human breast cancer samples derived from formalinvfixed and paraffinā€”embedded biopsies using dies directed to the progesterone receptor; and shows the percent of breast cancer samples positive for PRā€”A and PRā€”B for three binding patterns, A, AD, and D.
DETAILED PTION Before describing several ary aspects described herein, it is to be understood that the invention is not limited to the details of construction or process steps set forth in the following description. The aspects described herein are capable of being practiced or being carried out in various ways.
As used herein, the s ing a tumor" and "treatment of a tumor" mean to inhibit the replication of tumor cells, inhibit the spread of the tumor, decrease tumor size, lessen or reduce the number of tumor cells in the body, or ameliorate or alleviate the symptoms of the disease caused by the tumor, decrease the growth of the tumor (increase the time it takes the tumor to progress) or improve the survival of the patient when death is due to the cancer or secondary effects of the cancer. The term also includes treatment of cancer. Tumors include both cancers and nonā€”cancerous tumors.
The treatment is considered therapeutic if there is a decrease in mortality and/or morbidity, improvement of tumor-related symptoms, or there is a decrease in disease burden as may be sted by reduced numbers of tumor cells in the body, decreased tumor size or improvement in the time to progression, improvement of progression free survival or improvement of disease free survival.
As used herein, the term ā€œAPPā€”active anti-progestinā€ and its equivalents refer to an antiā€”progestin drug which exhibits an ability to dissolve or dissociate activated PR foci (APF) in the nuclei of cells or t the formation of APF in the nuclei of cells, indicating that its mechanism of action is via the PR activation pathway of the cell.
The terms ā€œAPP-positiveā€, ā€œPR foci positiveā€, ā€œactivated PRā€, ā€œPRs in a functional stateā€ and the like refer to the presence of progesterone receptor aggregates in the nuclei of cells.
The term ā€œfocal distributionā€ refers to the distribution of ā€œfociā€ (i.e., aggregation of progesterone receptors) in the nuclei of terone positive cells. ed or hyperspeckled pattern are terms that can be used referring to steroid nuclear receptor foci n in biology.
The term ā€œdegree of focal distributionā€ refers to the relative amount of PR foci t in the nuclei of progesterone positive cells. The degree of focal distribution can be determined quantitatively or qualitatively.
For example, the use of a colorimetric, enzymatic, or radiolabeled ligand such as a progesterone receptor dy, can be used to bind to progesterone receptors in cell nuclei. The degree of focal distribution can be determined quantitatively, for example, by ing color intensity, ļ¬‚uorescence or quantifying the level of radioactivity emitted by the labeled dy. The degree of focal distribution can determined qualitatively by comparing the intensity of binding between a control sample and a labeled sample using a light cope at an riate magniļ¬cation or techniques including, but not limited to, DNA microarray, n proļ¬ling, radiolabeling, or other surrogates for measuring APF.
The term ā€œ diffuse patternā€ refers to a ļ¬nely granular pattern which is indicative of the absence of focal distribution.
The term ā€œprogestinā€ refers to a natural or synthetic progestational substance that mimics some or all of the actions of progesterone, also referred to as progesterone receptor modulators (PRM) or selective progesterone receptor modulators (SPRM).
The term ā€œantiā€”progestinā€ refers to a substance that inhibits the formation, transport, or action of or inactivates progestational agents, including, but not limited to, onapristone, lonaprisan, mifepristone, PFā€”024l3873, telapristone, istone, ORG2058, asoprisnil, and ulipristal. A PRM or SPRM may have some antiā€”progestin properties, and be considered an anti-progestin or a progestin depending on the t of use.
The term ā€œantibodyā€ or ā€œantibodiesā€ refers to a protein which is capable of speciļ¬cally binding to an n and includes any substance, or group of substances, which has a speciļ¬c binding afļ¬nity for an antigen to the exclusion of other substances. lly, the term ā€œantibodyā€ es polyclonal dies, monoclonal antibodies, antibodies derived from humans or animals, humanized antibodies (e. g., nding portions derived from a human, binding portions derived from an animals) and nts thereof.
The terms ā€œantiā€”PR-Aā€ and ā€œantiā€”PRā€”Bā€ antibodies refer to antibodies directed to isoforms of the progesterone receptor ā€” PRā€”A and PRā€”B respectively. Antiā€” PRā€”ABā€ refers to an antibody capable of binding to both PRā€”A and PRā€”B. Speciļ¬c antibodies suitable for use in accordance with aspects herein include, but are not limited to, PgR636 and PgR1294 (M. Press, et a1. (Steroids (2002) 67:799ā€”813)), Novacastra clone 16, clone SAN27, clone 1A6, Dako clone PgR636, Ventana, clone 1E2, Novus Biologicals Progesterone Receptor [p Serl62] dy Clone 32; Novus Biologicals Progesterone Receptor [p Serl90] Antibody Clone EP1516Y, Novus Biologicals Progesterone Receptor [p Ser294] Antibody Clone 608, Abcam Progesterone Receptor [p Ser400] Antibody Ref ab60954, and Genetex Progesterone Receptor [p Ser554] Antibody Ref. GTXl 18987.
The term ā€œadministerā€ refers to providing a drug or drugs, prescribing one or more drugs, or placing one or more drugs on a formulary. The term ā€œprovidingā€ refers to dispensing the drug directly to patient through any le route of stration (e.g., oral, injection, intravenous, intramuscular, and transdermal etc.) or providing instructions to a patient to do the same.
One aspect provides a method of inhibiting the growth of a tumor susceptible to growth inhibition by anti-progestins by obtaining a tissue suspected of being tumorigenic from a patient and determining the degree of focal bution of antiā€” progesterone receptor in nuclei of cells from the tissue. If the degree of focal distribution is greater than about 5%, an ant-progestin (e.g., onapristone, lonaprisan, istone, PFā€”024l3873, telapristone, lilopristone, ORG2058, asoprisnil, and ulipristal) can be administered to the patient.
While the role of PR, progestins and antiā€”progestins in breast and other cancers has previously been d, the results have been inconclusive leading to difļ¬culties in sing and treating patients. Multiple models have shown the numerous and x interactions of s, s, cancer type, carcinogens, and tumor environment among other factors. Without being bound by theory, the PR may be pathologically activated with altered physiological properties affecting the activation potential of the ligand resulting in abnormal or uncontrolled stimulation of cell growth and proliferation. r, the most commonly studied models originate from a small number of original tumors, and therefore do not accurately represent the physiological variability between tumor types or the tumors of different patients. That is, the limited number of cancer models is insufļ¬cient to cover the xity of heterogenic cancers in a human tion.
Studies of the formation of PR foci have been used to test compounds for their ability to induce PR translocation from the cytoplasm to the s in genetically engineered cell lines. These assays, such as the Thermo Scientiļ¬c PR (Progesterone Receptor) RedistributionĀ® Assay, use image analysis and ļ¬‚uorescence microscopy to quantitate nuclear accumulation of PR in the presence of the test compound. In contrast, aspects provided herein are designed for analysis of PR foci in y tumor tissue, irrespective of the presence of a PR ligand or a drug. In one aspect, the exemplary s described herein relate to the presence of PR foci in the nuclei of cells in naturally-occurring tumors indicating an anomaly that can be used to t the efļ¬cacy in that patient of an anti-progestin that has PR antagonist properties. In another aspect, the characterization of constitutively activated PR in the clinic has now been found to indicate that tumors and cancers are susceptible to treatment with anti-progestins, including onapristone.
Onapristone, (e.g., (88,1 lR,l3R,14S,l7S)ā€”llā€”[4~(dimethylamino)phenyl]ā€” 17ā€”hydroxy-17ā€”(3ā€”hydroxypropyl)ā€”l3ā€”methylā€”l,2,6,7,8,1 1,12,14,15,16ā€” decahydrocyclopenta[a]phenanthrenā€”3-one) has the following chemical structure: Other rogestins include: progestational 3ā€”(6,6ā€”ethy1eneā€”17B- hydroxyā€”3ā€”0xoā€”17Aā€”pregnaā€”4-eneā€”17Aā€”YL)pr0pionic acid Gā€”lactones, 3~(6,6-ethy1eneā€” 17.beta.-hydroxy0X0ā€”17.alpha.ā€”pregnaā€”4ā€”ene-17.a1pha.-y- 1)propionic acid gamma.- lactone and the following: Mifepristone (108,1 1 S, 148,1 5 S,17R)ā€”17-[4-(dimethylamino)pheny1]ā€”14ā€”hydroxyā€”1 5-methy1ā€”14ā€”(prop- 1 ~ynā€”1ā€”y1)tetracyclo[8.7.0.0"{2,7} .O"{ 1 heptadecaā€”1,6-dienā€”5 ā€”one /ā€˜ /' ff ā€œMd/"V g x' l 0 {479C K.ā€ w ,/C ā€œ113/" 3/ W - X ā€˜_4_./"\-\R , aā€œ /ā€™Lā€œ // 1 ā€˜1ā€œ r .. ,4ā€™ x _/ n x ,, Lilopristone (1 1-beta,17ā€”beta,17(2))ā€”ropeny1);estraā€”4,9ā€”dienā€”3ā€”one,1 1ā€”(4ā€”(dimethylamino)phenyl)ā€”1 7ā€” hydroxyā€”17ā€”(3 -hydr0xyā€”1-p;1 1B~[4-(Dimethylamin0)pheny1]-17Bā€”hydroxy-17ā€”[(Z)~3 ā€” hydroxyā€” 1 ~propenyl]estraā€”4,9ā€”dien-3 -0ne [0049} 8 (8R,9S, 1 OR, 1 3 S,14S,16R,17S)-16ā€”ethy1-17ā€”(2ā€”hydroxyacety1)ā€”13-methy1-2,6, 7,8,9,10,1 1 ,12,14,15,16,17ā€”dodecahydrOā€”1Hā€”cyclopenta[a]phenanthrenā€”3~0ne 0xā€œ. .ā€˜f H m I /\"k /"'~ ā€˜1ā€™ -r& w: m.ā€˜1 ā€˜x " ā€œx. ā€™ 1 1. 0ā€; 7:31;ā€œ?N.ā€˜ #5333? ā€œI Q ā€xxx", Lonaprisan (88,1 1R,13S,14S,17S)(4-acety1pheny1)~17~hydroxy~13ā€”methy1ā€”17-(1,1,2, 2,2-pentaļ¬‚u0roethy1)ā€”1,2,6,7,8,1 1,12,14,15,16ā€”decahydrocyclopenta[a]phenanthrenā€”3- Asoprisnil (8 S, 1 1 R, 1 3 S,14S,17S)ā€”1 1-[4-[(E)-hydroxyimin0methy1]phenyl]ā€”17ā€”methoxy-17ā€” (methoxymethyl)ā€”13-methylā€”1,2,6,7,8,1 1,12,14,15,16ā€” decahydrocyclopenta[a]phenanthrenā€”3-0ne Ulipristal (8 S, 1 1 R, 1 3 S, 1 4S, 17R)ā€”17ā€”acety1-1 1-[4ā€”(dimethylamino)pheny1]ā€”17ā€”hydroxyā€”1 3 lā€” 1,2,6,7,8,1 1,12,14,15,16ā€”decahydrocyc10penta[a]phenanthrenā€”3ā€”one ,f"\ 42Ā¢ā€œ\,, a ā€˜35 L l W / ix ā€1" '\,._,xā€˜"g" 'J l 1' W x,Ā» /ā€™ ,xā€˜1 ifā€ K55ā€, 0 \xā€˜w 3873 4-[3-Cyclopropylā€”1ā€”(rnesylmethyl)ā€”5ā€”methylā€”1H-pyrazolā€”4ā€”yl]oxy,ā€”2,6- dimethylbenzonitrile /ā€™/\ā€™s :"~\ itQ Preļ¬x \ l 'N..._ (ā€f/":\QI// i lā€˜ In another aspect, focal PR binding provides a more sensitive and tive test than currently-used conventional PR assays. Patients classiļ¬ed in conventional PR assays as PRā€”negative as well as those that are conventionally PRā€” positive may test ve for focal PR nuclear binding and therefore be candidates for treatment with antiā€”progestins such as onapristone. Thus, a patient previously identified candidate for as PR negative using previous methods would not have been considered a treatment with antiā€”progestins such as onapristone. The presence of PR foci in patients conventionally tested as PR~negative would explain the apparently anomalous result that onapristone is active in some of these patients. Aspects described herein will therefore make hormonal treatment potentially available to a greater number of patients with that are classiļ¬ed as ā€œtriple cancer, ing potentially those patients with breast cancer negativeā€ (i.e., negative for estrogen receptor (ER), PR and Her2). ary suitable immunohistochemical methods for use in aspects described herein are described by M. Press, et al. (Steroids (2002) 67:799ā€”813) and M.
Nadji (Anatomic Pathol. (2005) 123121ā€”27) hereby incorporated by reference in their entirety. By way of example, primary cancer tissue specimens for analysis may be prepared as parafļ¬n sections or ļ¬ne needle aspiration smears of the cancer tissue as is known in the art for conventional PR assays. If parafļ¬n sections are used, the parafļ¬n is ļ¬rst melted by heating the slides, and dewaxed with xylene. Slides are then rehydrated in sing grades of ethanol and exposed to an antibody, ably a monoclonal antibody that speciļ¬cally binds to PRā€”A, PRā€”B, or both. g of the antibody is then detected using any one of the methods known in the art for detection of antibody binding, examples of which are described below.
One exemplary suitable method for detection of binding of an antibody to its target is a colorimetric assay, lly an enzymatic colorimetric assay. One such method employs dase to produce a colored stain visible under the light microscope.
Endogenous peroxidase in the tissue specimen is blocked using en peroxide and endogenous biotin is blocked using a biotinā€”blocking reagent prior to incubation with the antibody or antibodies. If the primary antibody is a mouse antibody, it is uently bound to a biotinylated use globulin. Streptavidin-peroxidase conjugate is added to bind the enzyme to the antibodyā€”target complex. Color is developed by addition of diaminobenzidine and cupric sulfate. The tissue specimen may be counterstained with fast green to increase visibility of the dase stain.
Alternatively, a fluorescence method may be used to detect antibody binding to PRā€”A, PR-B or both. In this case, a ļ¬‚uorescentlyā€”labeled y antibody under a ļ¬‚uorescence microscope. may be bound to the PR target and detected directly However, antibody to the PR a method ing binding of an led y followed by binding a ļ¬‚uorescentlyā€”labeled secondary (e.g., use immunoglobulin) antibody to the primary antibody may reduce nonā€”speciļ¬c ļ¬‚uorescence. Any ļ¬‚uorescent label known for use in immunohistochemical assays may be used in the aspects described herein, for example FITC (ļ¬‚uorescein isothiocyanate); ļ¬‚uorescein FITC 520 nm green Alexa 488 515 nm green phycoerythrin PE 565 nm yellow; rythrinā€”Texas Red ECD 620 nm red; phycoerythrin-cyanineS PC5 665 nm deep red; Peridinin chlorophyll PerCP 670 nm deep red; rythrinā€”cyanine 5.5 PC5.5 703 nm far red; phycoerythrin- cyanine 7 PC7 755 far red; E allophycocyanin APC 660 nm deep red; Allophycocyaninā€” cyanine 7 7.
Both onal and polyclonal antibodies may be useful in aspects described herein. A non-exhaustive list of suitable monoclonal antibodies is described by M. Press, et al. supra, including two antibodies which are resistant to formalin ļ¬xation and parafļ¬n embedding (PgR636 and PgR1294). Speciļ¬c antibodies suitable for use in accordance with aspects herein include, but are not limited to, PgR636 and PgRl294 (M.
Press, et a1. (Steroids (2002) 67:799ā€”813)), Novacastra clone 16, clone SAN27, clone 1A6, Dako clone PgR636, Ventana, clone 1E2, Novus Biologicals Progesterone Receptor [p Serl62] dy Clone 1064-132; Novus Biologicals terone Receptor [p Serl90] Antibody Clone EP1516Y, Novus icals Progesterone Receptor [p Ser294] Antibody Clone 608, Abcam Progesterone Receptor [p Ser400] Antibody Ref ab60954, and Genetex Progesterone or [p Ser554] Antibody Ref. GTXl 18987.
In one aspect, binding of the antibody to PR is detected by observation of the stained slide under a light microscope or ļ¬‚uorescence microscope as appropriate.
Magniļ¬cation is typically about 200X or 400X to evaluate, for example, the percentage of cells positive for binding to an antibody. However, to e sensitivity for detection of APF it may be desirable to evaluate the slides at SOOXā€”lOOOX to facilitate study of subnuclear structures.
Samples that are apparently PR negative by microscopy may be evaluated by ļ¬‚ow cytometry to detect positive samples below the threshold of light or ļ¬‚uorescence microscopy. If ļ¬‚ow cytometry indicates rare positive cells, high magniļ¬cation X800ā€” XlOOO microscopy may be used to study subnuclear structures and fy activated progesterone receptor foci (APF). However, if the ve cells detected by ļ¬‚ow cytometry are too rare to be reliably detected by microscopy for analysis of APF, a cenceā€”activated cell sorter (FACS) can be used to separate positive cells from the cells in suspension based on their ļ¬‚uorescence (e.g., Sony Cell Sorter SH800, Siemens Immulite 2000). As positive cells are concentrated but not damaged by this process, the reliability and ility of successfully visualizing APF on subsequent microscopic evaluation is substantially increased.
The ce or absence of APF in individual tumor cell nuclei may be detected visually under a light or ļ¬‚uorescence microscope, or by any other appropriate In one aspect, visual means means, such as ļ¬‚uorescence or colorimetric measurements. for ion will be used. The s of staining may be quantitated by noting presence or absence of APF, or by counting the number or tage of positive cells.
Alternatively, speciļ¬c characteristics of the staining may be quantitated. For example, detection may include notation of r or not focal binding in the form of APF is anied by diffuse nuclear staining, tation of positive cells by number or percentage, and/or quantitation of intensity or number/density of APF. Quantitation of APF density may be determined as the average number of foci/cell, or using an arbitrary scale (e. g., ā€œfewā€, ā€œmoderateā€ or ā€œmanyā€). Intensity may similarly be determined using 1ā€”5. In another an arbitrary scale, e.g., low/medium/high or a cal scale such as aspect, the results of the analysis of the patientā€™s tumor tissue will be compared to positive and/or negative controls.
In one aspect, a tumor tissue en is judged as APP-positive when lā€” 100%, 5-100%, 25ā€”100% or 50100% of the nuclei of progesterone positive cells in the specimen exhibit APF. In yet another aspect, the therapeutic efficacy of an APFā€”active antiā€”progestin may also be correlated with the intensity of APF staining or with the number or density of APF, these parameters may also be used to determine the ivity of the tumor to treatment with the APF~active antiā€”progestin. In general, and without being bound by theory, the sensitivity of a tumor to treatment with APF-active antiā€” progestin will increase with increasing number or percentage of positive cells, increasing intensity of APF and/or increasing number of APF in the cells of the tumor tissue specimen.
In further aspects, methods for determining the sensitivity of a tumor to APPā€”active anti-progestins may be either manual (e.g., visual detection using a ļ¬‚uorescence microscope) or they may be automated or semiā€”automated using methods for rapid scanning, detection and quantitation of colorimetricallyā€” or ļ¬‚uorescentIyā€”labeled tissue specimens. For example, a fully automated scanning and is system may be developed and used in n aspects. While manual selection of speciļ¬c regions of the tumor to be analyzed may be used in one aspect, (e.g., InScapeĀ® immunohistochemistry 3 Giralda system ((e.g., InScapeĀ® immunohistochemistry system (Quest Diagnostics APF in Farms Madison, NJ , an automated system for ng and analysis of cell nuclei can be used to provide automated specimen scanning and analysis of the antigenā€”speciļ¬c immunohistochemistry stained specimen. In another aspect, image recognition can be used to create a digital image of the entire stained tissue section; An antigenā€”speciļ¬c computer algorithm can be used to analyze the results of the digital image representing the whole specimen. In yet another aspect, the software can conļ¬gured to distinguish foci from diffuse background staining in the nucleus, and measure ļ¬‚uorescence intensity and size of foci on a cell-by-cell or clusterā€”byā€”cluster basis, repeating the s for each cell or cluster over the entire specimen. These automated s can, in certain aspects, result in improved accuracy by ming a on that is not possible ly, with reduced cost. Full automation can also make the test accessible to pert medical s.
In one aspect, the decision whether to treat the patient based on the results of the diagnostic assay is based on the number/percentage, intensity and/or density of APF when they are present. Without being bound by theory, it is anticipated that the efficacy of treatment with an APP-active anti-progestin will increase with increasing number or percentage of positive cells, sing intensity of APF and/or increasing number of APF in the cells of the tumor tissue en. Based on these parameters the medical practitioner may also determine the dosing, timing and length of treatment.
Accordingly, another aspect relates to use of an APF~active antiā€”progestin for treating an APF-positive tumor.
The tumor to be identiļ¬ed or treated ing to the above methods may include any ous or nonā€”cancerous tumor in which APF occur, and in which the presence of APF can be determined. Such cancers or tumors e breast cancer, lung, uterine cancer, uterine leiomyoma, ovarian cancer, te cancer, brain, and angiomas.
Benign tumors which can be identified or treated according to certain aspects include meningiomas, 70% of which express PR by conventional is.
The APF-active anti-progestin of the foregoing methods may be any anti~ progestin drug having the ability to inactivate APF (for example by dissolving or dissociating the aggregates or preventing formation of APF or forming ve APF).
Such drugs e onapristone (DNA), but others with a similar mechanism of action are also suitable for use in aspects bed herein.
Another aspect provides methods of identifying a tumor susceptible to growth inhibition by rogestins by obtaining a tissue suspected of being tumorigenic or cancerous from a patient and exposing the tissue to an antiā€”progesterone receptor antibody. Progesterone positive cells in the tissue sample can be identified. The degree of focal distribution of the progesterone receptor in nuclei of the progesterone positive cells from the tissue sample can be determined and an antiprogestin can be administered to the patient if the degree of focal bution in the tissue sample is greater than about % of the progesterone receptor positive cells.
In yet another aspect, a method of treating a t with a tumor susceptible to growth inhibition by anti-progestins is provided. The method comprises obtaining a tissue sample suspected of being tumorigenic from a patient and exposing the tissue to an antiā€”progesterone receptor antibody. The progesterone receptor positive cells in the tissue sample can be ļ¬ed and the focal binding distribution of the progesterone receptor in nuclei of cells from the tissue can be determined. If the focal g distribution is greater than 5% A or AD binding pattern of the progesterone receptor positive cells in the tissue , an antiā€”progestin is administered to the patient in a dosage range of about 10 to about 200 mg per day depending upon the potency, bioavailability, and safety proļ¬le of the antiā€”progestin.
In another , the degree of focal bution can be determined by suitable method as discussed herein including immunochemical, ļ¬‚uorescence, DNA rray, protein ng, radiolabeling, or other surrogates for measuring APP.
In another aspect, the tumor tissue is selected from the group ting of breast, meningiomas, prostate, ovarian, endometrial, uterine leiomyoma, lung, and uterine tissue.
In yet another aspect, the anti-progestin is selected from the group consisting of onapristone, lonaprisan, mifepristone, PFā€”024l3873, telapristone, lilopristone, ORG2058, asoprisnil, and ulipristal.
In another aspect, the degree of focal distribution is determined by identifying the binding pattern of progesterone or in the nuclei of progesterone positive tissue cells. Heterogeneous tumors include cells which may have active progesterone receptor foci or inactive progesterone receptor foci. Therefore, there may be cellular regions containing active foci as shown by distinct clumps in the cellular nuclei, and cellular regions which exhibit a more diffuse pattern.
For example, Figure 1 depicts two exemplary binding patterns from brown nuclear staining obtained with antiā€”progesterone antibodies in human breast cancer samples formalin-ļ¬xed and parafļ¬nā€”embedded tissue samples obtained from biopsies of breast cancer ts. Figure 1A shows a diffuse, granular pattern (D) indicative of cells which are not likely to be susceptible to treatment with anti-progestins. In contrast, Figure 1B shows a mottled binding pattern (A) indicative of cells which are likely to be WO 52652 susceptible to treatment with rogestins. A mixed pattern exhibits both A and D patterns and is termed AD.
In another aspect, the antiā€”progesterone antibody is ed from the antiā€”PR-B antibody, and a mixture of antiā€”PR-A group consisting of antiā€”PRā€”A antibody, and antiā€”PR~B dies, and bispecific antiā€”PR AB antibodies.
In yet another aspect, the antiā€”progestin is administered in an amount from to about 200 mg per day depending upon the potency, bioavailability, and safety proļ¬le of the antiā€”progestin.. Without being bound by theory, it is believed that by identifying patients with tumors that are susceptible to treatment with progestins, a lower dose of the anti-progestin may be used resulting in a lower risk of toxic side effects.
Thus, than 5% focal a lower dosage range can be used for patients exhibiting r bution of the progesterone receptor. In one aspect, the A or AD classification could result in different doses, while D pattern would indicate that treatment with an antiā€” tin treatment is not warranted.
In yet another , methods for screening antitumor drugs for the ability to inactivate APF are provided. These s are useful, for example, to identify additional antiā€”progestins which may be candidates for use in treating of sitive tumors according to the methods described herein. In one aspect, the method provides a method of screening a drug candidate for the ability to decrease focal distribution of the cells in a tumor. At progesterone receptor in the nuclei of progesterone receptor positive least two tumor tissue specimens from the same tumor can be obtained. One tumor tissue specimen can be exposed to a drug ate. The tumor tissue specimens can then be exposed to anti-progesterone receptor antibodies and the degree of focal distribution of cells from the progesterone receptors in the nuclei of the progesterone receptor positive tumor tissue specimens can be determined. If the focal distribution of the progesterone receptor in the tumor tissue specimen exposed to the drug candidate is decreased compared to tumor tissue specimens not exposed to the drug candidate, the drug candidate is e of decreasing focal distribution of the progesterone receptor in progesterone receptor positive cells of the tumor.
Another aspect provides a system for classifying a tumor susceptible for treatment with an antiā€”progestin, sing a tissue sample and at least one antibody or antibody g nt capable of detecting the progesterone receptor. The dy or antibody g fragment can be used to determine the degree of focal distribution of the progesterone receptor in the progesterone receptor ve nuclei of cells from a tumor tissue en. In another aspect, the tumor is susceptible to treatment with an antiā€”progestin if the degree of focal distribution in the cell nuclei of the progesterone positive cells is greater than about 5%.
In another aspect, detecting a decrease in able staining of the APF is an indication of APF inactivating activity of the antitumor drug. Detecting no substantial decrease in detectable staining of the APF is an tion of lack of APF inactivation of the antitumor drug.
In another aspect, an APF-active antiā€”progestin may be used in combination with additional hormonal treatment that does not act by an APF inactivation mechanism (e.g., antiestrogens) to achieve improved therapeutic y as compared to either agent alone. Alternatively, an APFā€”active rogestin may be used in combination with one or more conventional chemotherapeutic agents which are negative for APF activity in the screening assay to achieve improved therapeutic efficacy as compared to either agent alone (e. g., everolimus, trastuzumab, TMlā€”D, antiā€”HER2 drugs, bevacizumab, or chemotherapy with agents such as paclitaxel, docetaxel, taxanes, bicin, liposomal doxorubicin, pegylated liposomal doxorubicin, anthracyclines, anthracenediones, carboplatin, cisplatin, S-FU, gemcitabine and cyclophosphamide). For example, everolimus is an mTor inhibitor that is indicated in combination with an aromatase inhibitor and may, in the future, be indicated in combination with an antiā€” progestin.
In yet another aspect, detecting the presence of focal distribution of the antibody to progesterone receptors in the nuclei may be used as an indication that the tumor of a patient previously treated with an antitumor drug, which has become resistant to that drug, is still ive to an APF-active antiā€”progestin such as onapristone. In one aspect, the method can be adapted to determine r chemoresistance of a tumor resulting from previous chemotherapy can be reversed by treatment with an APFā€”active anti-progestin. Reversal of such chemoresistance may be based on the ent mechanisms of action of the previous chemotherapy and the APPā€”active antiā€”progestin.
Another aspect is directed to a system for classifying a tumor susceptible for treatment with an antiā€”progestin. The system comprises a tissue sample and at least one antibody or antibody binding fragment capable of detecting the progesterone receptor n the antibody or antibody binding fragment is used to determine the degree of focal bution of the progesterone receptor in the nuclei of cells from a tumor tissue specimen and wherein the tumor is susceptible to treatment with an anti-progestin if the degree of focal distribution is greater than about 5%.
Tumor specimens from patients with breast cancer (invasive ductal carcinoma) and endometrial cancer were selected from the archives of Oscar Lambret Cancer Center (Lille, France), anatomical pathological department. ts had previously provided consent for the use of their tissues for research purposes. Samples of breast or endometrial tumor tissues which had been ļ¬xed in 4% formalin ļ¬xative and embedded in parafļ¬n were obtained. lmmunohistochemistry (IHC) was performed on 3ā€”4 um sections of the archival breast or trial tumor s. The sections were deparafļ¬nized, hydrated and washed in working buffer (0.05 mol/L Tris/HCI, 0.15 mol/L NaCl, 0.05% Tween 20, pH 7.6, Dako, k, code S3006). Antigen retrieval was carried out with the Dako Target Retrieval Solution (modiļ¬ed citrate buffer, pH 6.1, Dako, Denmark, code 81699) in a water bath at 98Ā°C for 20 min. Then, the sections were d with the Dako Peroxydase Block solution to block endogenous peroxides at room temperature (RT) for min (Dako EnVisionĀ® +/HRP Mouse (DAB+) Kit, Dako, Denmark, code K4007), washed and incubated with the primary antibodies at the appropriate l dilutions at RT for 60 min in a humidiļ¬ed chamber (Table 1). Following a 5ā€”min. wash with working buffer, the Dako Labelled Polymer (Dako EnVisionĀ® +/HRP Mouse (DAB+) Kit, Dako, Denmark, code K4007) was used for the detection of the primary antibody binding at RT for 30 min. Chromogen (DAB) was then used with Substrateā€”Batch at room temperature for 5ā€”10 min and the sections were lightly counterstained with Gillā€™s hematoxylin.
Negative controls were obtained by tution of the primary antibodies with isotype control mouse IgGl (Table 1) or with antibody diluent alone (wash buffer negative control) in the histochemical staining procedure.
Table 1. Antibodies used for immunohistoehemistry Antibody Clone Dilutions Host / Isotype Supplier Code against PR,Aform 16 1:100 (3.6 Mouse lgGl PGR-312ā€”LNovocastra ug/ml) CB 1:200 (1.8 PR, B form SAN27 1:100 (04 Mouse lgGlK PGRā€”Bā€”CE Novocastra ug/ml) 1:200 (0.2 rig/m1) PR,A/B forms 1A6 1:40 (1.2 Mouse IgGl PGR-L-CE , Novocastra rig/m1) 1:80 (0.6 pig/ml) PR, A/B forms 16SAN27 1:100 (2 ug/ml) Mouse IgGl stra PGR-ABā€”Lā€” 1:200 (l ug/ml) CE Negative control DAK- 1:25 (4 ug/ml) Mouse IgGl Dako X0931 G01 1:100 (1 ug/ml) 1:200 (0.5 rig/m1) Immunohistochemistry analysis was performed using a Zeiss Axioscope microscope, ed with an Imaging Model ROHS digital camera. lmmunoreactive s were classiļ¬ed as unequivocal brown labeling of tumor cell nuclei. The intensity of labeling was deļ¬ned as 0 for negative, + for weak, ++ for moderate and +++ for strong.
Example 2 12 breast cancer samples were analyzed with 3 different antibodies and 4 methods in lHC. 6 samples could be processed for further histoļ¬‚uorescence (II-IF) analysis.
Immunohistoļ¬‚uorescence was performed using a Zeiss ļ¬‚uorescent microscope equipped with a CCD camera and Smart Capture software, speciļ¬c for capture of ļ¬‚uorescent images. IHF was performed on 3ā€”4 um sections of the archival breast tumor tissues. The sections were deparaffinized, hydrated and washed in working buffer (0.05 mol/L Tris/HCl, 0.15 mol/L NaCl, 0.05% Tween 20, pH 7.6, Dako, Denmark, code S3006). Antigen retrieval was carried out with the Dako Target Retrieval bath at Solution (modiļ¬ed citrate buffer, pH 6.1, Dako, k, code 81699) in a water 98Ā°C for 20 min. Then, the sections were ted with the primary dies at the appropriate optimal dilutions at RT for 60 min in a black humidiļ¬ed chamber (Table 2). ing a 5-minute wash with g buffer, appropriate secondary antibody conjugated to Alexa Fluor 488 was used for the detection of the primary antibody binding at RT for 30 min mouse lgG (H+L), F(abā€™)2, Cell Signaling, USA, code 44088, dilution 121000 ; Antiā€”rabbit IgG (H+L), F(abā€™)2, Cell ing, USA, code 44128, dilution 1:1000). All slides were then washed and lipped using VectashieldĀ® HardSet Mounting Medium (Vector Labs, USA, code Hā€”1400) and stored refrigerate the dark until analysis, to preserve ļ¬‚uorescence. Negative controls were obtained by tution of the primary antibodies with isotype control mouse lgGl or rabbit serum (see lHC table) or with antibody diluent alone (wash buffer negative control) in the immunohistoļ¬‚uorescence staining ure.
All tumor samples were PR Positive for the three different antibodies. of 11 PR positive However, the analysis of the nuclear pattern was inconclusive in 6 out cases with the bispecific A and B antibody (1 case was PR negative with this antibody only). Six cases were subjected to IHF analysis with all of the antibodies. In two cases, the lHF procedure could not be performed with all antibodies because not enough tumor tissue ed available. The four cases could be analyzed with the PR B antibody.
The IHF analysis with the other antibodies (PRA and PRA + B) was inconclusive in one instance for characterizing the r pattern. The IHF PR nuclear distribution and binding ns observed were concordant with IHC.
Thereafter, a larger sample was analyzed in lHC with the Antiā€”PR A antibody, Antiā€”PR B antibody, or the mixture of both (called thereafter A+B). 75 breast s and 25 endometrial cancer samples were processed. For each labeled tumor sample, positive focal distribution was defined as the tage labeled tumor cells in the entire tumor , excluding necrotic areas.
The two basic patterns found are presented in Figure 1. These images show the ng of tissue samples with antiā€”PR antibodies using (THC). Figure 1A shows a brown, ļ¬nely granular, and diffuse D pattern. Figure 1B shows a mottled, clumped pattern representing a positive focal binding A pattern. Figure 2 shows the same samples processed using IHF. Figure 2A shows a diffuse D pattern similar to the IHC result in Figure 2A. Figure 2B shows a similar mottled, clumped, focal binding pattern as in Figure 2B. The diffuse D pattern of Figures 1A and 2A are similar to the results in ngineered cells that a ļ¬‚uorescent receptor when ed express no progesterone or no progesteroneā€”agonist is present (Amettā€”Manfield et A1, 2004, 1C Control, 1D, and 1E) and in normal human endometrial tissue and in endometrial cancer (Arnett-Manfield et A1, 2004, 1A, 1B, 1C, 1D, 1E, 1F).
The active A pattern observed in formalin ļ¬xed, paraffin embedded tumor tissue may differ from images obtained in fresh cells. This is expected because formalinā€” fixation and paraffin embedding tissue will result in changes to the cellular contents, thereby resulting in a different pattern of PR. Another difference relative to the research publications which utilized IHF, is related to the . In the research setting, a confocal cope (i.e. using two laser beams) provides high resolution and 3D images; thin slices of tissue samples (e.g., 2 microns) are utilized. The IHC pattern results from a al reaction that modifies the cellular content. In contrast for IHC, a ional wide-field microscope is used for reading the standard thicker tumor slices resolution. (e. g., 4 microns). The IHC que described results in some loss of 2012/058732 The IHF technique is less chemically aggressive for tumor tissues, in that it does not alter the microscopic cellular architecture. IHF requires specialized, equipment, a pathologist experienced with the technique, and is much more timeā€” consuming. IHF cannot be easily coupled with other pathology analyses such as standard histology that requires formalinā€”ļ¬xed paraffin embedded tissues. Thus, in one aspect, IHC may be used as a routine pathological laboratory procedure. In the developed IHC technique used herein, 4 micrometer tissue ns (a commonly used thickness for routine clinical analysis ) were used for all analysis.
Figures 3A and 3B are equivalent to s 1A and 1B with ound ng. The diffuse pattern observed in 5A, or in immunoļ¬‚uorescence, is darkened by the counterstaining. Likewise, 5B demonstrates gross nuclear anomalies. However, the even, diffuse pattern of 5A is still characteristic with 5A with homogeneous nuclei, while 1B translated in dysformed nuclei in SE.
Thus, two basic ns are found: a diffuse PR nuclear staining indicating an absence of activated PRs, or and heterogeneous staining where aggregates, called PR foci, can be recognized within the nucleus of the cells. PR foci are larger than ts of a diffuse pattern that are substantially smaller (see Figures).
Example 3 Three categories or ypes have been identiļ¬ed for use with aspects described herein and which are ed at higher magniļ¬cation (800X). In st, standard magnification (400X) is used in for conventional IHC PR status determination.
Categories (observed at high magnification) D : Diffuse Staining, no PR Foci (e.g., Figure 1A) AD : Area associating A and D cells, or heterogeneous distribution of PR foci with smaller sizes than A.
] A Large Foci distributed in an heterogeneous manner (e.g., Figure 113) This classiļ¬cation (D, AD, and A) was evaluated on 100 additional cases (75 breast cancer and 25 endometrial cancer tissue samples). In some cases the samples for PR-A but not for PRā€” were positive for one PR isotype and not the other (e. g., positive Breast Cancer Samples (61 cases are analyzed for standard PR expression, 12 cases were PR negative for all antibodies, 2 cases had missing data).
Table 2 Breast Cancer Tumor Cells Positive for Indicated Antibody Number In Percentages Mean Min Max of Cases * Each antibody gives statistically similar data with the same average percent (31- 36%) of PR Positive cells and varying within the same range (5-95%).
* This is a ation that selects the highest percentage of PR A or PR B, as it of positive progesterone receptor cells was apparent that with the antibodies used, the rate was not the same for both antibodies in a same biopsy.
Table 3 Endometrial Cancer Cells Positive for Indicated Antibody R A Alone 21% Anti-PR B Alone C. Focal Distribution The n below be the frequencies of A, AD, D patterns and N (negative, no PR staining). All cases were ed at high magniļ¬cation (800X). Two trate that the breast evaluable. The data in table 4 cancer cases were not classiļ¬cation varies with the antibody (PRA or PRB) used, and that there is more variability among the antibodies for the AD pattern. This most likely s the inherent deregulation of the two PR receptors (A and B) in cancer tissue. In certain aspects, antibodies targeted at each of the PR isoforms may be used to e additional information for interpreting the results of the analysis. For example, a case may be ā€œDā€ with an antiā€”PR A antibody and ā€œADā€ with the second antiā€”PR B antibody. Based on the later classiļ¬cation of ā€œADā€, a treatment with a anti-progestin would be potentially appropriate. rly, a case may be ā€œAā€ with an antibody against PR A and ā€œADā€ with an dy against PR B, which could potentially require a different (higher) dose of the antiā€”progestin because of the greater degree of malignant cell growth indicated by the aberrant PR activity. Conventional IHC methods to determine PR cannot provide this information because they only indicate the presence or absence of hormone receptors (i.e., ER and PR). In one aspect, the activated PR foci pattern based on analysis with 1 or more te antibodies would provide additional ation for analyzing the ted PR foci pattern.
Table 4 ā€” PR Focal Distribution for Breast Cancer Cells Number AD Neg In Number of cases Of cases' 71 h In Percentages % ---u Anti-PRAandB 600 Example 4 In the data set outlined in the tables below, a given tumor sample could be APF negative for one dy and APF positive for another and show a different pattern for one antibody versus the other antibody. However, the results were generally concordant between PRā€”A and PR-B antibodies. This concordance is shown on the diagonal of the crossā€”tabulations that follow below. The concordance between the two sets of conditions is highlighted in the shaded text box of the table. These results rate that in certain s, more than one antibody would provide additional information to identify the APP nuclear bution pattern.
Table 5 below compares the APF patterns with the PR A antibody in relationship to the PR A+B antibody mixture in the breast cancer samples. A: Aggregated Pattern with large foci, AD: mix of A Cells and D cells, or heterogeneous ā€”medium size foci. D: diffuse pattern or absence of Activated PR. The columns classify the cases according to the indicated binding pattern using only the PR-A antibody while the rows classify the cases using PRā€”A + PRā€”B antibodies. The diagonal, highlighted row shows the number of concordant cases, i.e., cases with the same binding pattern using both methods. Other cells show dant results, i.e., cases with different binding patterns for each .
Table 5: Comparison of the APF patterns with PR A versus PR A+B Table 6: Breast cancer samples: Crossā€”tabulation of results obtained with an antiā€”PR B antibody (PR B) vs the mixture of antiā€”PR A and antiā€”PR B (PR A+B).
Aggregated Pattern with large foci, AD: mix of A Cells and D cells, or heterogeneous mediumā€”medium size foci. D: diffuse pattern or absence of Activated PR. The columns fy the cases according to the indicated g pattern using only the PR-B antibody while the rows classify the cases using PRā€”A + PRā€”B antibodies. The diagonal, highlighted row shows the number of concordant cases, i.e., cases with the same binding pattern using both methods. Other cells show discordant results, i.e., cases with different binding ns for each method.
Table 6: Comparison of the APF patterns with PR B versus PR A+B Breast Cancer Table 7: Breast cancer samples: Cross-tabulation of results obtained with an antiā€”PR B dy (PR B) vs an antibody antiā€”PR A (PR A). A: Aggregated Pattern with large foci, AD: mix of A Cells and D cells, or heterogeneous ā€”medium size foci. D: diffuse pattern or absence of Activated PR. The columns classify the cases according to the indicated binding pattern using only the PR B antibody while the rows classify the cases using PR A antibody. The al, highlighted row shows the number of concordant cases, i.e., cases with the same binding pattern using both methods. Other cells show dant results, i.e., cases with different binding patterns for each method.
Table 7 Comparison of the APP patterns with PR A versus PR B _-Breast Cancer PR A n- Endometrial Cancer Similar patterns of PR nuclear distribution are observed in endometrial cancer samples. lmportantly, normal ļ¬broblasts were found in biopsy samples and were noted to be PR positive. These normal ļ¬broblasts had a D PR nuclear distribution phenotype indicating that the PR in these normal cells were not activated, most likely because the patients are post menopausal and thus are not producing logic levels of progesterone. Therefore, the ļ¬broblasts are not exposed to endogenous progesterone. In st, cancer tissue was presenting activated form of PR (APF) even in absence of physiological progesterone as indicated by the ļ¬broblast pattern.
Table 8: Endometrial cancer samples: Crossā€”tabulation of results obtained with an anti-PR A antibody (PR A) vs the mixture of Antiā€”PR A and an antibody R B (PR A+B). A: Aggregated n with large foci, AD: mix of A Cells and D cells, or heterogeneous ā€”medium size foci. D: diffuse pattern or absence of Activated PR.
The columns classify the cases according to the indicated binding pattern using only the PR A antibody while the rows classify the cases using PR A and PR B antibodies. The al, highlighted row shows the number of concordant cases, i.e., cases with the same g pattern using both methods. Other cells show dant results, i.e., cases with different binding patterns for each method.
Table 8: Comparison of the APF patterns with PR A versus PR A+ B Endometrial Cancer T-otal PR A+B Table 9: Endometrial cancer samples: Crossā€”tabulation of results obtained Antiā€”PR with an anti-PR B antibody (PR B) vs the mixture of R A and an antibody D cells, or B (PR A+B). A: Aggregated Pattern with large foci, AD: mix of A Cells and of Activated PR. heterogeneous mediumā€”medium size foci. D: diffuse pattern or absence The columns classify the cases according to the indicated binding pattern using only PR B antibody while the rows classify the cases using PR A and PR B antibodies. diagonal, highlighted row shows the number of concordant cases, i.e., cases with the same binding pattern using both methods. Other cells ShOW discordant results, i.e., cases with ent g patterns for each .
Table 9: Comparison of the APF patterns with PR B versus PR A+B Table 10: Endometrial cancer samples: Crossā€”tabulation of results obtained with an antiā€”PR B dy (PR B) vs an antibody Antiā€”PR A (PR A). A: Aggregated Pattern with large foci, AD: mix of A Cells and D cells, or heterogeneous medium- medium size foci. D: diffuse pattern or absence of Activated PR. The columns classify the cases according to the ted binding pattern using only the PR B antibody while the rows classify the cases using PR A antibody. The diagonal, highlighted row shows the number of concordant cases, i.e., cases with the same binding pattern using both methods. Other cells show dant s, i.e., cases with different binding patterns for each method.
Table 10: ison of the APF patterns with PR B versus PR A Endometrial Cancer PRA n In one aspect, the use of antibodies directed to PRā€”A and PR-B or bi- speciļ¬c antibodies directed to PR-A and PR-B can be used together to identify the AD pattern PR nuclear distribution pattern where use of a single antibody (e.g., PRā€”A or PR- B) may not identify the AD pattern in certain cases.
In another aspect, the methods disclosed herein describe a PR nuclear pattern in cancer biopsies shown using, for example, IHC, and conļ¬rmed using fresh tissues and IHF. The diffuse n is found in normal cells/tissues that are not exposed to tins under experimental and physiological conditions. The diffuse r distribution pattern indicates that the PR of the tumor cells is not activated, and therefore treatment of the tumor with an antiprogestin is unlikely to be effective. In contrast, the 2012/058732 what is observed when mental models presence of the AD or A pattern is similar to or normal cells are exposed to progestins. These ns signal that PRS are activated and transcriptionally activate in some cells and that treatment with antiprogestins is likely to be effective in these cases.
Expression of these patterns (e.g., A and AD) is heterogeneous in tumors and across different samples, which is a characteristic of cancers. In contrast, the D phenotype is homogeneous, a pattern consistent with a lack of PR biologic function. The expression of PR and the phenotype we have described vary according to the expressed and the PR Isotype (A or B) and the antibody used (e.g., bispeciļ¬c AB, A only, B only mixture of A + B). This variability of the PR nuclear bution pattern is not unexpected in naturally occurring human cancers which are inherently heterogenous .
Example 5 The plot of shows the percent of breast cancer samples positive for PRā€”A and PRā€”B for the three g patterns, A, AD, and D. The s support the conclusion that conventional a positive progesterone or status determined by herein. methods does not correlate with the presence of PRF distribution as described Example 6 Table 11 Table 11 shows the percentage of ā€œAā€ binding n cells for tissue samples exhibiting both ā€œAā€™ and ā€œDā€ binding pattern cells. The column labeled ā€œAPRā€ indicates the overall pattern observed for the tissue sample while the ā€œA%ā€ column Each indicates the percentage of cells in the sample that exhibit the ā€œAā€ binding pattern. row shows the results for one case using both antiā€”PRā€”A and antiā€”PRā€”B antibodies or each antibody alone.
Table 11 Percentage of Cells expressing the APF pattern with ent antibodies m % of Cells with APF APR % of Cells with APF APR % of Cells with APF >>J>J> UUUU Ill -ā€” -ā€”-ā€” >> GOEH0101 HED >DO 40% >>DC] AD 5% Although the invention herein has been described with reference to particular embodiments, it is to be understood that these embodiments are merely illustrative of the principles and applications of the present invention. It will be apparent to those d in the art that various modiļ¬cations and variations can be made to the method and systems described herein without departing from the spirit and scope of the invention. Thus, it is intended that the present invention include modifications and variations that are within the scope of the appended claims and their equivalents.
Throughout the specification and claims, unless the context requires ise, the word ā€œcompriseā€ or variations such as ā€œcomprisesā€ or isingā€, will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.

Claims (7)

What is claimed is:
1. Use of an anti-progestin composition in the preparation of a medicament for the treatment of a tumor susceptible to growth inhibition by one or more antiprogestin (s), wherein the tumor is identified as susceptible to growth inhibition by one or more anti-progestin(s) wherein a) a tissue sample suspected of being tumorigenic from a patient is exposed to an anti-progesterone or antibody; b) flow cytometry is performed on the tissue sample to identify progesterone receptor positive cells; c) high magnification microscopy is used to identify ted progesterone or foci in the progesterone receptor positive cells; and d) the focal binding distribution of the progesterone receptor in nuclei of cells from the tissue is determined, wherein if the focal binding bution is greater than 5% of the progesterone receptor positive cells in the tissue sample with an A or AD binding pattern, then the tumor is ined to be susceptible to growth inhibition by one or more anti-progestin(s).
2. The use according to claim 1, wherein the rogestin composition is in a dosage range of about 10 to about 200 mg per day.
3. The use according to claim 1 or claim 2, wherein the tumor is selected from the group consisting of breast, brain, iomas, prostate, ovarian, endometrial, uterine leiomyoma, lung, and uterine tissues.
4. The use according to any one of claims 1-3, wherein the anti-progestin is selected from the group ting of onapristone, lonaprisan, mifepristone, PF-02413873, telapristone, lilopristone, ORG2058, asoprisnil, and ulipristal.
5. The use according to any one of claims 1-4, wherein the degree of focal distribution is determined by identifying the binding pattern of progesterone receptor in the cells of the tissue.
6. The use according to any one of claims 1-5, wherein the antibody is selected from the group ting of anti-PR-A dy, anti-PR-B antibody, a bispecific antibody directed to PR-A and PR-B, and a mixture of anti-PR-A and anti-PR-B antibodies.
7. The use according to any one of claims 1-6, wherein the high magnification microscopy is performed at X800 to X1000.
NZ717890A 2011-10-04 2012-10-04 Methods and Systems for Identifying and Treating Anti-Progestin Sensitive Tumors NZ717890B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161542931P 2011-10-04 2011-10-04
US61/542,931 2011-10-04
NZ623140A NZ623140B2 (en) 2011-10-04 2012-10-04 Methods and systems for identifying and treating anti-progestin sensitive tumors

Publications (2)

Publication Number Publication Date
NZ717890A NZ717890A (en) 2017-10-27
NZ717890B2 true NZ717890B2 (en) 2018-01-30

Family

ID=

Similar Documents

Publication Publication Date Title
AU2012318618B2 (en) Methods and systems for identifying and treating anti-progestin sensitive tumors
Vereide et al. Effect of levonorgestrel IUD and oral medroxyprogesterone acetate on glandular and stromal progesterone receptors (PRA and PRB), and estrogen receptors (ER-Ī± and ER-Ī²) in human endometrial hyperplasia
McGhan et al. Androgen receptor-positive triple negative breast cancer: a unique breast cancer subtype
Arnould et al. Accuracy of HER2 status determination on breast core-needle biopsies (immunohistochemistry, FISH, CISH and SISH vs FISH)
Mote et al. Progesterone receptor A predominance is a discriminator of benefit from endocrine therapy in the ATAC trial
Manojlovic-Gacic et al. Oncogene-induced senescence in pituitary adenomasā€”an immunohistochemical study
AU2006278456A1 (en) Predictive methods for cancer chemotherapy
WO2015157343A1 (en) Systems and methods for identifying progesterone receptor subtypes
Silva-Ortega et al. Proposal of a clinically relevant working classification of pituitary neuroendocrine tumors based on pituitary transcription factors
WO2015048804A2 (en) Protein biomarker and uses thereof
Drăghici et al. The immunoprofile of interstitial Cajal cells within adenomyosis/endometriosis lesions
EP2773962B1 (en) Assay for predictive biomarkers of anti-estrogen efficacy
Cowen et al. Oestrogen receptor in breast cancer: Prognostic studies using a new immunohistochemical assay
NZ717890B2 (en) Methods and Systems for Identifying and Treating Anti-Progestin Sensitive Tumors
NZ623140B2 (en) Methods and systems for identifying and treating anti-progestin sensitive tumors
Kean-Hooi Teoh MBChB et al. An analysis of predictive biomarkers in routine histopathological reporting of infiltrating ductal breast carcinoma in a tertiary hospital in Malaysia with a focus on limitations and directions for future development