WO2014179678A1 - Méthodes de traitement du cancer et de troubles angioprolifératifs - Google Patents

Méthodes de traitement du cancer et de troubles angioprolifératifs Download PDF

Info

Publication number
WO2014179678A1
WO2014179678A1 PCT/US2014/036560 US2014036560W WO2014179678A1 WO 2014179678 A1 WO2014179678 A1 WO 2014179678A1 US 2014036560 W US2014036560 W US 2014036560W WO 2014179678 A1 WO2014179678 A1 WO 2014179678A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
polypeptide
pathway
tumor
fshr
Prior art date
Application number
PCT/US2014/036560
Other languages
English (en)
Inventor
John STRASSWIMMER
Original Assignee
Angiostrass Llc
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 Angiostrass Llc filed Critical Angiostrass Llc
Publication of WO2014179678A1 publication Critical patent/WO2014179678A1/fr

Links

Classifications

    • 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
    • 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
    • G01N2333/726G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • Basal cell carcinoma (BCC) of the skin a common malignancy in individuals of mixed European descent, is increasing in incidence in part due to an aging population and sun exposure habits. Thus, there is an increased need for effective treatments for BCC as well as other cancerous tumors.
  • the present disclosure encompasses the discovery that the presence of follicle- stimulating hormone (FSH) receptors (e.g., on or near tumor and/or noncancerous blood vessels) can be used as a basis to select a subject for treatment with a hedgehog pathway modulator.
  • FSH follicle- stimulating hormone
  • the present disclosure therefore provides, e.g., methods of selecting subjects for treatment with hedgehog pathway modulators (e.g., a Smo polypeptide modulator), and the use of hedgehog pathway modulators in treatment and/or prevention of tumors.
  • Combination therapy refers to those situations in which two or more different pharmaceutical agents are administered in overlapping regimens so that the subject is simultaneously exposed to both agents.
  • two or more different agents may be administered simultaneously or separately.
  • This administration in combination can include simultaneous administration of the two or more agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, two or more agents can be formulated together in the same dosage form and administered simultaneously. Alternatively, two or more agents can be simultaneously administered, wherein the agents are present in separate formulations.
  • a first agent can be administered just followed by one or more additional agents. In the separate administration protocol, two or more agents may be administered a few minutes apart, or a few hours apart, or a few days apart.
  • Characteristic portion As used herein, the term a "characteristic portion" of a substance, in the broadest sense, is one that shares some degree of sequence or structural identity with respect to the whole substance. In certain embodiments, a characteristic portion shares at least one functional characteristic with the intact substance.
  • a "characteristic portion" of a polypeptide or protein is one that contains a continuous stretch of amino acids, or a collection of continuous stretches of amino acids, that together are characteristic of a polypeptide or protein. In some embodiments, each such continuous stretch generally contains at least 2, 5, 10, 15, 20, 50, or more amino acids.
  • a characteristic portion of a substance is one that, in addition to the sequence and/or structural identity specified above, shares at least one functional characteristic with the relevant intact substance.
  • a characteristic portion may be biologically active.
  • Characteristic sequence is a sequence that is found in all members of a family of polypeptides or nucleic acids, and therefore can be used by those of ordinary skill in the art to define members of the family.
  • Hedgehog pathway As used herein, the terms “hedgehog pathway” and
  • Hh pathway activation transmits an Hh signal in cells and/or tissues. In some embodiments, Hh pathway activation results in changes in degree of downstream gene expression level and/or phenotypic changes.
  • Hedgehog pathway activator refers to any substance that activates or increases the activity of an Hh signaling pathway.
  • an Hh pathway activator is an Hh polypeptide, a Cdo polypeptide, a Boc polypeptide, a Gasl polypeptide, an Smo polypeptide, a Gli polypeptide, and/or a nucleic acid encoding such polypeptide.
  • an Hh pathway activator is a substance that activates the transcription, binding, activity or stability of an Hh polypeptide, a Cdo polypeptide, a Boc polypeptide, a Gasl polypeptide, an Smo polypeptide, a Gli polypeptide, and/or a nucleic acid encoding such polypeptide.
  • an Hh pathway activator is a substance that inhibits the transcription, binding, activity or stability of a Ptc polypeptide, an Hhip polypeptide, an SuFu polypeptide, and/or a nucleic acid encoding such polypeptide.
  • Hedgehog pathway inhibitor refers to any substance that inhibits or reduces the activity of an Hh signaling pathway.
  • an Hh pathway inhibitor is a Ptc polypeptide, an Hhip polypeptide, an SuFu polypeptide, and/or a nucleic acid encoding such polypeptide.
  • an Hh pathway inhibitor is a substance that inhibits the transcription, binding, activity or stability of an Hh polypeptide, a Cdo polypeptide, a Boc polypeptide, a Gasl polypeptide, an Smo polypeptide, a Gli polypeptide, and/or a nucleic acid encoding such polypeptide.
  • an Hh pathway inhibitor is a substance that activates the transcription, binding, activity or stability of a Ptc polypeptide, an Hhip polypeptide, an SuFu polypeptide, and/or a nucleic acid encoding such polypeptide.
  • Hedgehog (Hh) pathway modulator refers to any substance that modulates activity of an Hh pathway.
  • a hedgehog pathway modulator modulates the transcription, binding, activity and/or stability of an Hh pathway polypeptide and/or a nucleic acid encoding such polypeptide.
  • an Hh pathway modulator is an Hh pathway activator.
  • an Hh pathway modulator is an Hh pathway inhibitor.
  • Hedgehog pathway polypeptide refers to a polypeptide whose amino acid sequence includes at least one characteristic sequence of and/or shows at least 100%, 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80%, 79%, 78%, 77%, 76%, 75%, 74%, 73%, 72%, 71%, or 70% identity with a protein involved in a Hh pathway (e.g., Boc, Cdo, Gasl, Gli, Hh, Hhip, Ptc, Smo, or SuFu polypeptide).
  • a protein involved in a Hh pathway e.g., Boc, Cdo, Gasl, Gli, Hh, Hhip, Ptc, Smo, or SuFu polypeptide.
  • an Hh pathway polypeptide shares at least one characteristic sequence of and/or shows the specified degree of overall sequence identity with one of the Boc, Cdo, Gasl, Gli, Hh, Hhip, Ptc, Smo, or SuFu sequences set forth herein (each of which may be considered a "reference" Hh pathway polypeptide).
  • an Hh pathway polypeptide as described herein shares at least one biological activity with a reference Hh pathway polypeptide as set forth herein. In some such embodiments, the shared biological activity relates to Hh signaling pathway activation or inhibition.
  • Hedgehog (Hh) polypeptide As used herein, the term "hedgehog polypeptide"
  • Hh polypeptide refers to a polypeptide whose amino acid sequence includes at least one characteristic sequence of and/or shows at least 100%, 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80%, 79%, 78%, 77%, 76%, 75%, 74%, 73%, 72%, 71%, or 70% identity with a hedgehog polypeptide (e.g., Desert (Dhh), Sonic (Shh) or Indian (Ihh)).
  • a hedgehog polypeptide e.g., Desert (Dhh), Sonic (Shh) or Indian (Ihh)
  • Hh sequences from flies, vertebrates, and mammals are known in the art, such as those described herein; in some embodiments, a Hh polypeptide shares at least one characteristic sequence of and/or shows the specified degree of overall sequence identity with one of the Dhh, Shh, or Ihh sequences set forth herein (each of which may be considered a "reference" Hh polypeptide). In some embodiments, an Hh polypeptide as described herein shares at least one biological activity with a reference Hh polypeptide as set forth herein. In some such embodiments, the shared biological activity relates to Hh signaling pathway activation.
  • homology refers to the overall relatedness between polymeric molecules, e.g., between nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules.
  • polymeric molecules are considered to be “homologous” to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical.
  • polymeric molecules are considered to be "homologous" to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% similar.
  • Identity refers to the overall relatedness between polymeric molecules, e.g., between nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two nucleic acid sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes).
  • the length of a sequence aligned for comparison purposes is at least 30%>, at least 40%>, at least 50%>, at least 60%>, at least 70%>, at least 80%>, at least 90%), at least 95%, or substantially 100%) of the length of the reference sequence.
  • the nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4: 11-17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two nucleotide sequences can,
  • Polypeptide As used herein, a "polypeptide”, generally speaking, is a string of at least two amino acids attached to one another by a peptide bond. In some embodiments, a polypeptide may include at least 3-5 amino acids, each of which is attached to others by way of at least one peptide bond. Those of ordinary skill in the art will appreciate that polypeptides sometimes include "non-natural" amino acids or other entities that nonetheless are capable of integrating into a polypeptide chain, optionally.
  • Protein refers to a polypeptide (i.e., a string of at least two amino acids linked to one another by peptide bonds). Proteins may include moieties other than amino acids (e.g., may be glycoproteins, proteoglycans, etc.) and/or may be otherwise processed or modified. Those of ordinary skill in the art will appreciate that a
  • protein can be a complete polypeptide chain as produced by a cell (with or without a signal sequence), or can be a characteristic portion thereof. Those of ordinary skill will appreciate that a protein can sometimes include more than one polypeptide chain, for example linked by one or more disulfide bonds or associated by other means.
  • Polypeptides may contain L-amino acids, D- amino acids, or both and may contain any of a variety of amino acid modifications or analogs known in the art. Useful modifications include, e.g., terminal acetylation, amidation, methylation, etc.
  • proteins may comprise natural amino acids, non-natural amino acids, synthetic amino acids, and combinations thereof.
  • the term "peptide” is generally used to refer to a polypeptide having a length of less than about 100 amino acids, less than about 50 amino acids, less than 20 amino acids, or less than 10 amino acids.
  • Reference sample may include, but is not limited to, any or all of the following: a cell or cells, a portion of tissue, blood, serum, ascites, urine, saliva, and other body fluids, secretions, or excretions.
  • sample also includes any material derived by processing such a sample. Derived samples may include nucleotide molecules or polypeptides extracted from the sample or obtained by subjecting the sample to techniques such as amplification or reverse transcription of mR A, etc.
  • Subject refers to any organism upon which embodiments of the invention may be used or administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; insects; worms; etc.).
  • animals e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; insects; worms; etc.
  • Susceptible to An individual who is "susceptible to" a disease, disorder, or condition (e.g., a tumor) is at risk for developing the disease, disorder, or condition.
  • an individual who is susceptible to a disease, disorder, or condition does not display any symptoms of the disease, disorder, or condition.
  • an individual who is susceptible to a disease, disorder, or condition has not been diagnosed with the disease, disorder, and/or condition.
  • an individual who is susceptible to a disease, disorder, or condition is an individual who displays conditions associated with development of the disease, disorder, or condition.
  • a risk of developing a disease, disorder, and/or condition is a population-based risk.
  • Symptoms are reduced: According to the present invention, "symptoms are reduced” when one or more symptoms of a particular disease, disorder or condition is reduced in magnitude (e.g., intensity, severity, etc.) or frequency. For purposes of clarity, a delay in the onset of a particular symptom is considered one form of reducing the frequency of that symptom. It is not intended that the present invention be limited only to cases where the symptoms are eliminated. The present invention specifically contemplates treatment such that one or more symptoms is/are reduced (and the condition of the subject is thereby "improved"), albeit not completely eliminated.
  • Target cell or target tissue refers to any cell, tissue, or organism that is affected by a condition described herein and to be treated, or any cell, tissue, or organism in which an FSH receptor is expressed.
  • target cells, target tissues, or target organisms include those cells, tissues, or organisms in which there is a detectable tumor.
  • target cells, target tissues, or target organisms include those cells, tissues or organisms that display a disease- associated pathology, symptom, or feature.
  • a target cell or target tissue is a tumor or vascular tissue involved in a vascular disorder.
  • therapeutic regimen refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. It may include a treatment or series of treatments designed to achieve a particular effect, e.g., reduction or elimination of a tumor and/or detrimental condition or disease such as cancer.
  • the treatment may include administration of one or more compounds either simultaneously, sequentially or at different times, for the same or different amounts of time. Alternatively, or additionally, the treatment may include exposure to radiation, chemotherapeutic agents, hormone therapy, or surgery.
  • a “treatment regimen” may include genetic methods such as gene therapy, gene ablation or other methods known to reduce expression of a particular gene or translation of a gene-derived mRNA.
  • Therapeutic agent refers to any agent that, when administered to a subject, has a therapeutic effect and/or elicits a desired biological and/or pharmacological effect.
  • therapeutically effective amount refers to an amount of an agent (e.g., a hedgehog modulator) that confers a therapeutic effect on the treated subject, at a reasonable benefit/risk ratio applicable to any medical treatment.
  • the therapeutic effect may be objective (i.e., measurable by some test or marker) or subjective (i.e., subject gives an indication of or feels an effect).
  • the "therapeutically effective amount” refers to an amount of a therapeutic agent or composition effective to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therapeutic or preventative effect, such as by ameliorating symptoms associated with the disease, preventing or delaying the onset of the disease, and/or also lessening the severity or frequency of symptoms of the disease.
  • a therapeutically effective amount is commonly administered in a dosing regimen that may comprise multiple unit doses.
  • a therapeutically effective amount (and/or an appropriate unit dose within an effective dosing regimen) may vary, for example, depending on route of administration, on combination with other pharmaceutical agents.
  • the specific therapeutically effective amount (and/or unit dose) for any particular patient may depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific pharmaceutical agent employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and/or rate of excretion or metabolism of the specific fusion protein employed; the duration of the treatment; and like factors as is well known in the medical arts.
  • treatment refers to any administration of a substance (e.g., provided compositions) that partially or completely alleviates, ameliorates, relieves, inhibits, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms, features, and/or causes of a particular disease, disorder, and/or condition (e.g., a tumor).
  • a substance e.g., provided compositions
  • Such treatment may be of a subject who does not exhibit signs of the relevant disease, disorder and/or condition and/or of a subject who exhibits only early signs of the disease, disorder, and/or condition.
  • such treatment may be of a subject who exhibits one or more established signs of the relevant disease, disorder and/or condition.
  • treatment may be of a subject who has been diagnosed as suffering from the relevant disease, disorder, and/or condition. In some embodiments, treatment may be of a subject known to have one or more susceptibility factors that are statistically correlated with increased risk of development of the relevant disease, disorder, and/or condition.
  • the present disclosure encompasses the discovery that the presence of FSH receptors on or near cancerous or noncancerous tumor blood vessels indicates that a hedgehog pathway modulator (e.g., a hedgehog pathway activator or inhibitor, e.g., a Smo polypeptide inhibitor) can be used to treat a tumor.
  • a hedgehog pathway modulator e.g., a hedgehog pathway activator or inhibitor, e.g., a Smo polypeptide inhibitor
  • the disclosure provides, among other things, methods of selecting a subject for treatment, and methods of treating a subject, with a hedgehog pathway modulator based on the presence of FSH receptors on or near cancerous or noncancerous tumor blood vessels.
  • the present disclosure provides methods of treating noncancerous vascular disorders using a hedgehog pathway modulator.
  • FSH is a gonadotrophin hormone synthesized and secreted by gonadotropes in the anterior pituitary gland.
  • FSH is a heterodimeric glycoprotein hormone consisting of two noncovalently linked subunits designated alpha and beta.
  • the subunits are 92 amino acids and 111 amino acids, respectively, and each has two N-linked glycosylation sites.
  • FSH has several biological functions in mammals. In males, for example, FSH, in combination with testosterone is required for the initiation and maintenance of qualitatively and quantitatively normal spermatogenesis. In females, FSH is necessary for selection and growth of ovarian follicles and for the production of estrogens from androgen substrate.
  • FSH is part of the hypothalamo-pituitary-ovarian axis, a classic endocrine closed loop biofeedback system, in which the gonadotrophins (e.g., FSH and luteinizing hormone (LH)) stimulate ovarian hormone production (e.g., estrogen), which in turn exerts a negative feedback effect on the gonadotrophins, to maintain a regulated system.
  • gonadotrophins e.g., FSH and luteinizing hormone (LH)
  • ovarian hormone production e.g., estrogen
  • FSH acts by binding to specific FSH receptors localized primarily in Sertoli cells of the testis and in granulosa cells of the ovary.
  • the FSH receptor belongs to the family of G protein-coupled receptors (GPCR), which are complex membrane-associated receptors characterized by seven-transmembrane spanning domains. The intracellular portion of the FSH receptor is coupled to the G protein Gs and adenylyl cyclase.
  • GPCR G protein-coupled receptors
  • FSH receptors are also expressed on the surface of blood vessels of a wide range of tumors (e.g., tumors of the prostate, breast, colon, pancreas, urinary bladder, kidney, lung, liver, stomach, testis, and ovary) of all grades, including early Tl tumors, as well as
  • noncancerous blood vessels such as placental endothelial cells (see, e.g., Radu et al, New England J. Med. 363: 1621-1630 (2010)).
  • the present disclosure encompasses the finding that the Hh pathway is involved in signaling through the FSH receptor, and that expression of FSH receptors on the surface of tumor blood vessels can be used to select subjects for treatment with an Hh pathway modulator (e.g., a Smo polypeptide inhibitor), e.g., to treat tumors.
  • an Hh pathway modulator e.g., a Smo polypeptide inhibitor
  • the present disclosure encompasses the use of Hh pathway modulators to treat noncancerous vascular disorders, e.g., hemangiomas or arteriovenous malformations.
  • Hedgehog Pathway e.g., a Smo polypeptide inhibitor
  • hedgehog (Hh) pathway is well known (see, e.g., U.S. Pat. No. 6,277,566 Bl;
  • the vertebrate family of Hh genes includes three members that exist in mammals, known as Desert (Dhh), Sonic (Shh) and Indian (Ihh) Hedgehogs, all of which encode secreted proteins. These various Hh proteins consist of a signal peptide, a highly conserved N-terminal region, and a more divergent C-terminal domain. Biochemical studies have shown that autoproteolytic cleavage of the Hh precursor protein proceeds through an internal thioester intermediate that subsequently is cleaved in a nucleophilic substitution. It is likely that the nucleophile is a small lipophilic molecule that becomes covalently bound to the C-terminal end of the N-peptide, tethering it to the cell surface. As a result of the tethering, a high local concentration of N-terminal Hh peptide is generated on the surface of Hh producing cells. It is this N-terminal peptide that mediates short- and long-range Hh signaling activities.
  • Smoothened (Smo) encodes a transmembrane protein of 1024 amino acids, which acts as a transducer of the Hh signal.
  • Smo protein has 7 hydrophobic membrane-spanning domains, an extracellular amino-terminal region, and an intracellular carboxy-terminal region.
  • Smo bears some similarity to G protein-coupled receptors and is most homologous to the Frizzled (Fz) family of serpentine proteins (Alcedo et al., Cell 86: 221 (1996)).
  • Activation of the pathway is initiated through binding of Hh polypeptide to Ptc.
  • Hh binding to Ptc alters the interaction of Smo and Ptc, reversing the repression of Smo by Ptc (see Zhu et al., Genes Dev. 17: 1240 (2003)).
  • Smo activation initates a cascade that leads to the translocation of an active form of the transcription factor Gli to the nucleus of the cell.
  • Three Gli proteins known in vertebrates are Glil, Gli2 and Gli3 (Kinzler et al, Science 236:70-73 (1987); Bai et al, Dev.
  • Gli proteins share high homology in the zinc finger domain, but have limited homology outside of this region (Matise et al.,
  • Glil is a transcriptional activator, while Gli2 and Gli3 are bifunctional and can function as transcriptional activators or, when proteolytically processed, transcriptional repressors (Dai et al., J. Biol. Chem. 12:8143-8152 (1999)).
  • Gli is prevented from entering the nucleus through interactions with cytoplasmic proteins, including Fused (Fu) and Suppressor of fused (Sufu) (see Methot et al, Dev. 127:4001- 4010 (2000)); Chen et al, Mol. Cell. Biol.
  • Hh pathway proteins include Gasl, Cdo, and Boc, which promote Hh pathway signaling (see Allen et al, Dev. Cell 20:775-787 (2011)), and Hhip, which inhibits Hh pathway signaling (see Beachy et al, Genes Dev. 24:2001-2012 (2010)).
  • Methods and compositions described herein include, for example, modulators of a hedgehog signaling pathway (e.g., modulators of one or more Hh pathway polypeptides).
  • Nucleic acids encoding Hh pathway polypeptides are known. According to the present disclosure, modulators of such nucleic acids (and polypeptides) are useful in the treatment of gynecologic disorders.
  • such nucleic acids have or include nucleotide sequences as set forth in SEQ ID NO: l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, or 27, or characteristic sequence elements thereof or therein.
  • useful nucleic acids show at least 100%, 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82% ,81%, 80%, 79%, 78%, 77%, 76%, 75%, 74%, 73%, 72%, 71%, or 70% overall sequence identity with one or more of SEQ ID NO: l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, or 27.
  • useful nucleic acids include at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or more contiguous residues found in SEQ ID NO: l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, or 27.
  • useful nucleic acids are generated in vitro; in some embodiments, useful nucleic acids are generated in vivo. In some embodiments, useful nucleic acids are generated using genetic engineering techniques (e.g., for production and/or mutagenesis of a reference sequence). To give but a few examples, in some
  • nucleic acid variants are generated using techniques such as site directed mutagenesis, random chemical mutagenesis, Exonuclease III deletion procedures, and standard cloning techniques.
  • useful nucleic acids are generating using chemical synthesis and/or modification procedures.
  • nucleic acids that are "variants" with respect to a reference nucleic acid are well known in the art. These include, for example, procedures in which nucleic acid sequences obtained from natural isolates are modified to generate nucleic acids that encode polypeptides having characteristics that enhance their value in industrial or laboratory applications. In such some embodiments of such procedures, a large number of variant sequences having one or more nucleotide differences with respect to the sequence obtained from the natural isolate are generated and characterized. Typically, these nucleotide differences result in amino acid changes with respect to the polypeptides encoded by the nucleic acids from the natural isolates.
  • a reference nucleic acid e.g., a naturally-occurring or other reference nucleic acid
  • variants can be created using error prone PCR (see, e.g., Leung et al, Technique 1 : 11-15, 1989; and Caldwell et al, PCR Methods Applic. 2:28-33, 1992).
  • error prone PCR PCR is performed under conditions where the copying fidelity of the DNA polymerase is low, such that a high rate of point mutations is obtained along the entire length of the PCR product.
  • nucleic acids to be mutagenized are mixed with PCR primers, reaction buffer, MgCl 2 , MnCl 2 , Taq polymerase, and an appropriate concentration of dNTPs for achieving a high rate of point mutation along the entire length of the PCR product.
  • the reaction can be performed using 20 fmoles of nucleic acid to be mutagenized, 30 pmole of each PCR primer, a reaction buffer comprising 50 mM KC1, 10 mM Tris HC1 (pH 8.3), and 0.01% gelatin, 7 mM MgCl 2 , 0.5 mM MnCl 2 , 5 units of Taq polymerase, 0.2 mM dGTP, 0.2 mM dATP, 1 mM dCTP, and 1 mM dTTP.
  • PCR can be performed for 30 cycles of 94 °C for 1 min, 45 °C for 1 min, and 72 °C for 1 min. However, it will be appreciated that these parameters can be varied as appropriate.
  • the mutagenized nucleic acids are then cloned into an appropriate vector and the activities of the polypeptides encoded by the mutagenized nucleic acids are evaluated.
  • Variants can also be created using oligonucleotide directed mutagenesis to generate site-specific mutations in any cloned DNA of interest.
  • Oligonucleotide mutagenesis is described in, for example, Reidhaar-Olson et al, Science 241 :53-57 (1988). Briefly, in such procedures a plurality of double stranded oligonucleotides bearing one or more mutations to be introduced into the cloned DNA are synthesized and inserted into the cloned DNA to be mutagenized. Clones containing the mutagenized DNA are recovered, and the activities of the polypeptides they encode are assessed.
  • Assembly PCR involves the assembly of a PCR product from a mixture of small DNA fragments. A large number of different PCR reactions occur in parallel in the same vial, with the products of one reaction priming the products of another reaction. Assembly PCR is described in, for example, U.S. Pat. No. 5,965,408.
  • Still another method of generating variants is sexual PCR mutagenesis. In sexual PCR mutagenesis, forced homologous recombination occurs between DNA molecules of different, but highly related, DNA sequence in vitro as a result of random fragmentation of the DNA molecule based on sequence homology. This is followed by fixation of the crossover by primer extension in a PCR reaction.
  • variants can also be created by in vivo mutagenesis.
  • random mutations in a nucleic acid sequence are generated by propagating the sequence in a bacterial strain, such as an E. coli strain, which carries mutations in one or more of the DNA repair pathways.
  • a bacterial strain such as an E. coli strain
  • Such "mutator" strains have a higher random mutation rate than that of a wild- type strain. Propagating a DNA sequence in one of these strains will generate random mutations within the DNA.
  • Mutator strains suitable for use for in vivo mutagenesis are described in, for example, PCT Publication No. WO 91/16427.
  • Variants can also be generated using cassette mutagenesis.
  • cassette mutagenesis a small region of a double stranded DNA molecule is replaced with a synthetic oligonucleotide "cassette" that differs from the native sequence.
  • the oligonucleotide often contains a completely and/or partially randomized native sequence.
  • Recursive ensemble mutagenesis can also be used to generate variants.
  • Recursive ensemble mutagenesis is an algorithm for protein engineering (i.e., protein mutagenesis) developed to produce diverse populations of phenotypically related mutants whose members differ in amino acid sequence. This method uses a feedback mechanism to control successive rounds of combinatorial cassette mutagenesis.
  • Recursive ensemble mutagenesis is described in, for example, Arkin et al, Proc. Natl. Acad. Sci., USA 89:7811-7815 (1992).
  • variants are created using exponential ensemble mutagenesis.
  • Exponential ensemble mutagenesis is a process for generating combinatorial libraries with a high percentage of unique and functional mutants, wherein small groups of residues are randomized in parallel to identify, at each altered position, amino acids which lead to functional proteins.
  • Exponential ensemble mutagenesis is described in, for example,
  • variants are created using shuffling procedures wherein portions of a plurality of nucleic acids that encode distinct polypeptides are fused together to create chimeric nucleic acid sequences that encode chimeric polypeptides as described in, for example, U.S. Pat. Nos.
  • nucleic acids for use in accordance with the present disclosure comprise naturally-occurring nucleotide residues.
  • nucleic acids for use in accordance with the present disclosure include one or more nucleotide "analogs".
  • a nucleotide analog is a nucleotide (i.e., an entity that is incorporated into a nucleic acid polymer without significantly disrupting the structure and/or function of that polymer) whose chemical structure differs from that of reference naturally-occurring ribonucleic or deoxyribonucleic acid residues adenine, guanine, cytosine, thymine, and uracil.
  • a nucleotide analog differs from its reference nucleotide at the base moiety, sugar moiety, and/or phosphate backbone.
  • a nucleotide analog contributes to one or more altered features in a nucleic acid polymer into which it is incorporated as compared with a comparable nucleic acid polymer containing its reference nucleotide rather than the analog.
  • such analog-containing polymer shows improved , stability, hybridization, and/or solubility.
  • base moiety alterations found in nucleotide analogs include deoxyuridine for deoxythymidine and 5-methyl-2'-deoxycytidine or 5-bromo-2'- deoxycytidine for deoxycytidine.
  • sugar moiety alterations found in nucleotide analogs include modification of the 2' hydroxyl of the ribose sugar to form 2'-0- methyl or 2'-0-allyl sugars.
  • deoxyribose phosphate backbone alterations found in nucleotide analogs include morpholino nucleic acids, in which each base moiety is linked to a six-membered, morpholino ring, or peptide nucleic acids, in which the
  • deoxyphosphate backbone is replaced by a pseudopeptide backbone and the four bases are retained (see, e.g., Summerton et al, Antisense Nucleic Acid Drug Dev. 7: 187-195 (1997);
  • a Hh pathway polynucleotide or variant for use in accordance with the present disclosure includes alterations to codon(s) to optimize for expression in a particular host cell.
  • a Hh pathway polynucleotide or variant can include one or more altered codons as described in, e.g., Grosjean et al, Gene 18: 199-209 (1982).
  • methods and compositions described utilize a modulator of one or more Hh pathway polypeptides (e.g., Boc, Cdo, Gasl, Gli, Hh, Hhip, Ptc, Smo, or SuFu polypeptides).
  • Hh pathway polypeptides e.g., Boc, Cdo, Gasl, Gli, Hh, Hhip, Ptc, Smo, or SuFu polypeptides.
  • such polypeptides are useful in the treatment of gynecologic disorders.
  • such polypeptides useful in the practice of the present disclosure have or include amino acid sequences as set forth in SEQ ID NO:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, or 28, or characteristic sequence elements thereof or therein.
  • useful polypeptides show at least 100%, 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82% ,81%, 80%, 79%, 78%, 77%, 76%, 75%, 74%, 73%, 72%, 71%, or 70% overall sequence identity with one or more of SEQ ID NO:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, or 28.
  • useful polypeptides include at least about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, or 150 or more contiguous amino acid residues found in SEQ ID NO:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, or 28.
  • a useful polypeptide differs from its reference polypeptide
  • the difference is a conservative or nonconservative substitution of one or more amino acid residues.
  • Conservative substitutions are those that substitute a given amino acid in a polypeptide by another amino acid of similar characteristics. Typical conservative substitutions are the following replacements: replacement of an aliphatic amino acid, such as alanine, valine, leucine, and isoleucine, with another aliphatic amino acid; replacement of a serine with a threonine or vice versa; replacement of an acidic residue, such as aspartic acid and glutamic acid, with another acidic residue; replacement of a residue bearing an amide group, such as asparagine and glutamine, with another residue bearing an amide group; exchange of a basic residue, such as lysine and arginine, with another basic residue; and replacement of an aromatic residue, such as phenylalanine and tyrosine, with another aromatic residue.
  • an aliphatic amino acid such as alanine, valine, leucine, and isoleucine
  • useful Hh pathway polypeptides include a substituent group on one or more amino acid residues. Still other useful polypeptides are associated with (e.g., fused, linked, or coupled to) another moiety (e.g., a peptide or molecule). For example, useful Hh pathway polypeptides can be fused, linked, or coupled to an amino acid sequence (e.g., a leader sequence, a secretory sequence, a proprotein sequence, a second polypeptide, or a sequence that facilitates purification, enrichment, or stabilization of the polypeptide).
  • a polypeptide includes a targeting agent, e.g., a targeting agent described herein.
  • Hh pathway polypeptides can be recombinantly produced by utilizing a host cell system engineered to express a nucleic acid encoding a Hh pathway polypeptide (e.g., a nucleic acid described herein).
  • a Hh pathway polypeptide can be produced by activating an endogenous gene (e.g., a nucleic acid encoding a Hh pathway polypeptide present endogenous ly in a cell).
  • a Hh pathway polypeptide can be partially or fully prepared by chemical synthesis.
  • a Hh pathway polypeptide can be purified from natural sources.
  • Hh pathway polypeptide is recombinantly produced
  • any expression system can be used.
  • Known expression systems include, without limitation, for example, egg, baculovirus, plant, yeast, or mammalian cells.
  • a Hh pathway polypeptide suitable for use in methods described herein are produced in mammalian cells.
  • Non-limiting examples of mammalian cells include BALB/c mouse myeloma line (NSO/1, ECACC No: 85110503); human retinoblasts (PER.C6, CruCell, Leiden, The Netherlands); monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al, J. Gen Virol, 36:59,1977); human fibrosarcoma cell line (e.g., HT1080); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells +/-DHFR (CHO, Urlaub and Chasin, Proc. Natl.
  • BALB/c mouse myeloma line NSO/1, ECACC No: 85110503
  • human retinoblasts PER.C6, CruCell, Leiden, The Netherlands
  • monkey kidney CV1 line transformed by SV40 COS-7
  • Hh pathway activators useful in the methods described herein include an Hh, Cdo,
  • Boc, Gasl, Smo, or Gli polypeptide a nucleic acid encoding an Hh, Cdo, Boc, Gasl, Smo, or Gli polypeptide; any substance that increases level and/or activity of an Hh, Cdo, Boc, Gasl, Smo, or Gli polypeptide; any substance that increases level and/or activity of a nucleic acid encoding a Cdo, Boc, Gasl, Smo, or Gli polypeptide; any substance that decreases level or activity of a Ptc, Hhip, and/or SuFu polypeptide; and/or any substance that decreases level or activity of a nucleic acid encoding a Ptc, Hhip, and/or SuFu polypeptide.
  • an Hh pathway activator is or includes an Hh, Cdo, Boc,
  • an Hh pathway activator is or includes a Ptc, Hhip, and/or SuFu polypeptide having an inhibiting mutation (e.g., a difference of one or more amino acid residues relative to a reference Ptc, Hhip, and/or SuFu polypeptide and having a lower level of expression and/or activity as compared to such reference polypeptide).
  • Hh pathway inhibitors useful in the methods described herein include a Ptc, Hhip, or SuFu polypeptide; a nucleic acid encoding a Ptc, Hhip, or SuFu polypeptide; any substance that increases level and/or activity of a Ptc, Hhip, and/or SuFu polypeptide, any substance that increases level and/or activity of a nucleic acid encoding a Ptc, Hhip, and/or SuFu polypeptide; any substance that decreases level or activity of an Hh, Cdo, Boc, Gasl, Smo, or Gli polypeptide; and/or any substance the decreases level of activity of a nucleic acid encoding an Hh, Cdo, Boc, Gasl, Smo, or Gli polypeptide.
  • an Hh pathway inhibitor is or includes a Ptc, Hhip, and/or
  • an Hh pathway inhibitor is or includes an Hh, Cdo, Boc, Gasl , Smo, and/or Gli polypeptide having an inhibiting mutation (e.g., a difference of one or more amino acid residues relative to a reference Hh, Cdo, Boc, Gasl, Smo, and/or Gli polypeptide and a lower level of expression and/or activity as compared to such reference polypeptide).
  • Hh pathway inhibitors useful in the methods described herein include steroidal alkaloids, such as cyclopamine, and derivatives thereof; other small molecules such as SANT-1, SANT-2, SANT-3, and SANT-4 (Chen et al, Proc. Natl. Acad. Sci., USA 99: 14071-14076 (2002)); arsenical agents such as arsenic trioxide (ATO); steroidal alkaloids and derivatives thereof, including, for example, cyclopamine and jervine (see, e.g., Chen et al, Genes Devel. 16:2743-2748 (2002); and U.S. Pat. No. 6,432,970); and triparanol (see, e.g., U.S. Pat. No. 6,432,970).
  • Additional nonlimiting Hh pathway inhibitors include those described in
  • Nonlimiting examples of Smo polypeptide inhibitors useful in the methods described herein include vismodegib (ErivedgeTM, Genentech, Inc., San Francisco, CA); BMS- 833923 (also known as XL139; Siu et al, J. Clin. Oncol. 28: 15s suppl abstr 2501 (2010), and National Institute of Health Clinical Trial Identifier No. NCT006701891); LDE-225 (Pan et al, ACS Med. Chem. Lett. 1 : 130-134 (2010)); LEQ-506 (National Institute of Health Clinical Trial Identifier No. NCTOl 106508); PF-04449913 (National Institute of Health Clinical Trial
  • NCTOl 108094 NCTOl 108094
  • l-piperazinyl-4-arylphthalazines or analogues thereof (Lucas et al., Bioorg. Med. Chem. Lett. 20:3618-22 (2010)).
  • Gli inhibitors useful in methods described herein include
  • GANT 58 and GANT 61 (Lauth et al, Proc. Natl. Acad. Sci. U.S.A. 104:8455-8460 (2007)); arsenic trioxide (Beauchamp et al., J. Clin. Invest. 121 : 148-160 (2011); and Kim et al., Proc. Acad. Sci. U.S.A. 107: 13432-13437 (2010)); and HPI1/2/3 (Hyman et al, Proc. Acad. Sci.
  • hedgehog inhibitors can be used to treat a subject who has FSH receptors expressed on tumor and/or noncancerous blood vessels (e.g., to treat a cancerous tumor, a noncancerous tumor, and/or an angioproliferative disorder).
  • methods include measuring a level of FSH receptors on tumor and/or noncancerous blood vessels of a subject and if the level meets a predetermined level, selecting the subject for treatment.
  • FSH receptors on tumor and/or noncancerous blood vessels can be evaluated using methods known in the art.
  • Methods of detecting FSH receptors include, e.g., antibody based assays such as enzyme immunoassays (EIA), radioimmunoassays (RIA), and Western blot analysis.
  • EIA enzyme immunoassays
  • RIA radioimmunoassays
  • Western blot analysis Western blot analysis.
  • a variety of methods can be used to determine the level of FSH receptors.
  • these methods include contacting a binding agent that selectively binds to an FSH receptor, such as an antibody, with a sample, to evaluate the level of FSH receptor in the sample.
  • Antibodies can be polyclonal, or more preferably, monoclonal.
  • An exemplary antibody that can be used for detection of FSH receptor is FSHR-323 (see, e.g., Vannier et al, Biochemistry 35: 1358-1366 (1996)).
  • an intact antibody, or a fragment thereof e.g., Fab or F(ab') 2
  • the binding agent includes a detectable label.
  • a binding agent can direct labeled by coupling (i.e., physically linking) a detectable substance, as well as indirectly labeled by reactivity with a detectable substance (e.g., a detection agent).
  • a binding agent is labeled with a detection agent and used to detect or image cells.
  • the detection agent can be used to qualitatively or quantitatively analyze the location and/or the amount of a binding agent at a particular locus.
  • the detection agent can also be used to image a binding agent, and/or a cell or tissue target of a binding agent using standard methods.
  • Nonlimiting examples of detection agents include, without limitation, fluorescent compounds, various enzymes, prosthetic groups, luminescent materials, bioluminescent materials, fluorescent emitting metal atoms, (e.g., europium (Eu)), radioactive isotopes (described below), quantum dots, electron-dense reagents, and haptens.
  • the detection reagent can be detected using various means including, but are not limited to, spectroscopic,
  • Nonlimiting exemplary fluorescent detection agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-l-napthalenesulfonyl chloride, phycoerythrin, and the like.
  • a detection agent can also be a detectable enzyme, such as alkaline phosphatase, horseradish peroxidase, beta-galactosidase, acetylcholinesterase, glucose oxidase and the like.
  • a binding agent is derivatized with a detectable enzyme, it can be detected by adding additional reagents that the enzyme uses to produce a detectable reaction product.
  • a binding agent can also be derivatized with a prosthetic group (e.g., streptavidin/biotin and avidin/biotin).
  • a binding agent can be derivatized with biotin and detected through indirect measurement of avidin or streptavidin binding.
  • fluorescent compounds that can be used as detection reagents include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride, and phycoerythrin.
  • Luminescent materials include, e.g., luminol
  • bioluminescent materials include, e.g., luciferase, luciferin, and aequorin.
  • a detection agent can also be a radioactive isotope, such as, but not limited to, ⁇ -, ⁇ -, or ⁇ -emitters; or ⁇ - and ⁇ -emitters. Radioactive isotopes can be used in diagnostic or therapeutic applications.
  • radioactive isotopes include, but are not limited to, iodine ( 131 I or 125 I), yttrium ( 90 Y), lutetium ( 177 Lu), actinium ( 225 Ac), praseodymium ( 142 Pr or 143 Pr), astatine ( 211 At), rhenium ( 186 Re or 187 Re), bismuth ( 212 Bi or 213 Bi), indium ( m In), technetium ( 99m Tc), phosphorus ( 32 P), rhodium ( 188 Rh), sulfur ( 35 S), carbon ( 14 C), tritium ( 3 H), chromium ( 51 Cr), chlorine ( 36 C1), cobalt ( 57 Co or 58 Co), iron ( 59 Fe), selenium ( 75 Se), and gallium ( 67 Ga).
  • Detection methods can be used to detect FSH receptor in a biological sample in vitro as well as in vivo.
  • In vitro techniques for detection include, e.g., enzyme linked immunosorbent assays (ELISAs), immunoprecipitations, immunofluorescence, enzyme immunoassay (EIA), radioimmunoassay (RIA), and Western blot analysis (as are described in "Methods in Immunodiagnosis", 2nd Edition, Rose and Bigazzi, eds. John Wiley & Sons, 1980; Campbell et al., "Methods and Immunology", W. A. Benjamin, Inc., 1964; and Oellerich, M. 1984, J. Clin. Chem. Clin. Biochem., 22:895-904).
  • FSH receptors are detected in a biological sample from a subject.
  • Biological samples include, but are not limited to, cells, tissue biopsy, whole blood, plasma, serum, sputum, cerebrospinal fluid, pleural fluid, urine and the like.
  • Biological samples can be isolated from a subject according to standard methods well known to those of skill in the art, such as by biopsy or venipuncture.
  • a biological sample can be pretreated as necessary by dilution in an appropriate buffer solution or concentrated, if desired. Any of a number of standard aqueous buffer solutions, employing one of a variety of buffers, such as phosphate, Tris, or the like, at physiological pH can be used. Additional exemplary methods are described in, e.g., Siraj et al, J. Cell. Mol. Med. 16:2010-2016 (2012).
  • in vivo techniques for detection include, e.g., introducing into a subject a labeled binding agent, e.g., antibody.
  • a labeled binding agent can associate the label with an FSH receptor-bearing cell and/or blood vessel, which is then detected and localized using an appropriate detection method.
  • detection method is Single Photon Emission Computed Tomography (SPECT) or Positron Emission Tomography (PET). Additional methods are described in, e.g., WO2009/103741.
  • level of expression of FSH receptors on tumor and/or noncancerous blood vessels of a subject is compared to a reference level (e.g., a level of FSH receptors on tumor and/or noncancerous blood vessels of one or more additional subjects, and/or a record of levels of FSH receptors expressed on a population of tumors, e.g., from a population of subjects).
  • a reference level e.g., a level of FSH receptors on tumor and/or noncancerous blood vessels of one or more additional subjects, and/or a record of levels of FSH receptors expressed on a population of tumors, e.g., from a population of subjects.
  • level of FSH receptors on tumor and/or noncancerous blood vessels is quantitated, e.g., using standard techniques.
  • a subject having level of FSH receptor expression above a reference level is selected for treatment with an Hh pathway modulator (e.g., an Smo polypeptide inhibitor) described herein.
  • an Hh pathway modulator e.g., an Smo polypeptide inhibitor
  • a subject having a higher level of tumor and/or noncancerous blood vessel FSH receptor expression is preferentially selected for treating with an Hh pathway modulator.
  • a subject having any detectable level of FSH receptor expression on tumor and/or noncancerous blood vessels is selected for treatment with an Hh pathway modulator.
  • a subject having a level of FSH receptor expression on tumor and/or noncancerous blood vessels that is at least 10%, 20%>, 30%>, 40%>, 50%>, 60%>, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or higher, than a reference level is selected for treatment with an Hh pathway modulator.
  • a selected subject e.g., a subject expressing detectable level of FSH receptors on tumor and/or noncancerous blood vessels and/or expressing a level of FSH receptors above a reference level
  • an Hh pathway modulator e.g., an Smo polypeptide inhibitor
  • a therapeutically effective amount of an Hh pathway modulator for a selected subject is lower than a corresponding amount for a subject not having a detectable level of FSH receptors on tumor and/or noncancerous blood vessels and/or not expressing a level of FSH receptors above a reference level.
  • an Hh pathway modulator is administered to a selected subject at a reduced concentration, at a reduced therapeutic regimen, and/or in combination with a reduced level of one or more additional therapies, as compared with a corresponding concentration, regimen, and/or level of additional therapies administered to a subject not having a detectable level of FSH receptors on tumor and/or noncancerous blood vessels and/or not expressing a level of FSH receptors above a reference level.
  • a selected subject exhibits a higher level of response to treatment than a subject not having a detectable level of FSH receptors on tumor and/or noncancerous blood vessels and/or not expressing a level of FSH receptors above a reference level.
  • Level of response can be determined, e.g., by comparing one or more parameters of effectiveness relative to a control.
  • a suitable control is a baseline measurement, such as a measurement in the same individual prior to initiation of a treatment described herein, or a measurement in a control individual (or multiple control individuals) in the absence of a treatment described herein.
  • level of effectiveness is a level to which a treatment partially or completely alleviates, ameliorates, relieves, inhibits, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms, features, and/or causes of a particular disease, disorder, and/or condition described herein (e.g., a cancerous tumor, a noncancerous tumor, and/or an angioproliferative disorder).
  • effectiveness of treatment is determined by measuring tumor size and/or tumor grade before and/or after treatment.
  • a level of response to treatment for a selected subject is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or higher, as compared to a level of response to treatment for a subject not having a detectable level of FSH receptors on tumor and/or noncancerous blood vessels and/or not expressing a level of FSH receptors above a reference level.
  • treatment of a selected subject results in a greater reduction in tumor grade level as compared to a reduction in tumor grade level for a treated subject not having a detectable level of FSH receptors on tumor and/or noncancerous blood vessels and/or not expressing a level of FSH receptors above a reference level.
  • An Hh pathway modulator e.g., an Hh pathway activator or inhibitor
  • a targeting agent such as an agent to localize an Hh pathway modulator to a specific target cell or target tissue (e.g., a tumor).
  • targeting agents include, but are not limited to, nucleic acids (e.g., R A and DNA), polypeptides (e.g., receptor ligands, signal peptides, avidin, Protein A, and antigen binding proteins), polysaccharides, biotin, hydrophobic groups, hydrophilic groups, drugs, and any organic molecules that bind to target cells or target tissues (e.g., receptors on target cells or target tissues).
  • nucleic acids e.g., R A and DNA
  • polypeptides e.g., receptor ligands, signal peptides, avidin, Protein A, and antigen binding proteins
  • polysaccharides e.g., biotin, hydrophobic groups, hydrophilic groups, drugs, and any organic molecules that bind to target cells or target tissues (e.g., receptors on target cells or target tissues).
  • Targeting agents can be associated with Hh pathway modulators in any of a number of ways.
  • polypeptide targeting agents can be coupled to or fused to an Hh pathway polypeptide.
  • a targeting agent is associated (e.g., covalently or noncovalently bound) to an Hh pathway modulator with either short (e.g., direct coupling), medium (e.g., using small-molecule bifunctional linkers such as SPDP (Pierce Biotechnology, Inc., Rockford, 111.)), or long (e.g., PEG bifunctional linkers (Nektar Therapeutics, Inc., San Carlos, Calif.)) linkages.
  • short e.g., direct coupling
  • medium e.g., using small-molecule bifunctional linkers such as SPDP (Pierce Biotechnology, Inc., Rockford, 111.)
  • long e.g., PEG bifunctional linkers (Nektar Therapeutics, Inc., San Carlos, Calif.) linkages.
  • targeting agents are or comprise antigen binding proteins or antibodies or binding portions thereof.
  • Antibodies can be generated to allow for specific targeting of antigens or immunogens (e.g., target cell or target tissue specific antigens).
  • Such antibodies include, but are not limited to, polyclonal antibodies; monoclonal antibodies or antigen binding fragments thereof; modified antibodies such as chimeric antibodies, reshaped antibodies, humanized antibodies, or fragments thereof (e.g., Fv, Fab', Fab, F(ab')2); or biosynthetic antibodies, e.g., single chain antibodies, single domain antibodies (DAB), Fvs, or single chain Fvs (scFv) (see, e.g., in Harlow et al, Using Antibodies: A Laboratory Manual: Portable Protocol I. Cold Spring Harbor Laboratory (Dec. 1, 1998); Zola, Monoclonal
  • Antibodies Preparation and Use of Monoclonal Antibodies and Engineered Antibody
  • Antibody attachment can be performed by any known method e.g., through standard covalent binding to free amine groups (see, e.g., Torchilin et al., Hybridoma 6:229-240 (1987); Torchilin et al, Biochim. Biophys. Acta 1511 :397-411 (2001); Masuko et al, Biomacromol. 6:800-884 (2005)).
  • a targeting agent is or comprises a nucleic acid (e.g., RNA or
  • nucleic acid targeting agents are designed to hybridize by base pairing to a particular nucleic acid (e.g., chromosomal DNA, mRNA, or ribosomal RNA).
  • nucleic acid targeting agents bind a ligand on a target cell or target tissue.
  • a nucleic acid can bind human nerve growth factor (Binkley et al., Nuc. Acids Res. 23:3198-205 (1995)).
  • Nucleic acids that bind ligands can be identified by known methods, such as SELEX procedures (see, e.g., U.S. Pat. Nos.
  • targeting agents can be or comprise aptamers, for example that bind to particular sequences.
  • Hh pathway modulators e.g., Hh pathway activators or inhibitors
  • a disease or disorder described herein e.g., a tumor, e.g., in a subject.
  • the route and/or mode of administration of an Hh pathway modulator described herein can vary depending upon the desired results.
  • dosage regimens can be adjusted to provide the desired response, e.g., a therapeutic response.
  • Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intracerebral, intrathecal, intravaginal, transdermal, rectal, by inhalation, or topical, particularly to the ears, nose, eyes, or skin.
  • the mode of administration is left to the discretion of the practitioner.
  • Hh pathway modulator described herein can be formulated as a
  • physiologically acceptable excipients can be, e.g., liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • physiologically acceptable excipients can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like.
  • physiologically acceptable excipients are sterile when administered to an animal.
  • the physiologically acceptable excipient should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid excipients, particularly for injectable solutions.
  • Suitable physiologically acceptable excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • suitable excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • suitable physiologically acceptable excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, gly
  • compositions if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Liquid carriers can be used in preparing solutions, suspensions, emulsions, syrups, and elixirs.
  • An Hh pathway modulator described herein can be suspended in a pharmaceutically acceptable liquid carrier such as water, an organic solvent, a mixture of both, or
  • liquid carrier can contain other suitable ingredients
  • liquid carriers for oral and parenteral administration include water (particular containing additives described herein, e.g., cellulose derivatives, including sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g., glycols) and their derivatives, and oils (e.g., fractionated coconut oil and arachis oil).
  • the carrier can also be an oily ester such as ethyl oleate and isopropyl myristate.
  • the liquid carriers can be in sterile liquid form for administration.
  • the liquid carrier for pressurized compositions can be halogenated hydrocarbon or other pharmaceutically acceptable propellant.
  • compositions for intravenous administration can comprise a sterile isotonic aqueous buffer.
  • the compositions can also include a solubilizing agent.
  • Compositions for intravenous administration can optionally include a local anesthetic such as lidocaine to lessen pain at the site of the injection.
  • the ingredients can be supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water- free concentrate in a hermetically sealed container such as an ampule or sachette indicating the quantity of active agent.
  • an Hh pathway modulator described herein is administered by infusion, it can be dispensed, for example, with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an Hh pathway modulator described herein is administered by injection, an ampule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • An Hh pathway modulator described herein can be administered rectally or vaginally in the form of a conventional suppository.
  • Suppository formulations can be made using methods known to those in the art from traditional materials, including cocoa butter, with or without the addition of waxes to alter the suppository's melting point, and glycerin.
  • Water- soluble suppository bases such as polyethylene glycols of various molecular weights, can also be used.
  • an Hh pathway modulator described herein that is effective for treating a disease or disorder described herein, e.g., a tumor, can be determined using standard clinical techniques known to those with skill in the art.
  • in vitro or in vivo assays can optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed can also depend on the route of administration, the condition, the seriousness of the condition being treated, as well as various physical factors related to the individual being treated, and can be decided according to the judgment of a health-care practitioner.
  • compositions described herein can be administered as single administrations or as multiple administrations. Such compositions can be administered at regular intervals, depending on the nature, severity and extent of the subject's condition.
  • a therapeutically effective amount of a therapeutic agent e.g., an Hh pathway modulator
  • is administered periodically at regular intervals e.g., once every year, once every six months, once every five months, once every three months, bimonthly (once every two months), monthly (once every month), biweekly (once every two weeks), or weekly).
  • a therapeutically effective amount is largely determined based on the total amount of the therapeutic agent contained in pharmaceutical compositions described herein. Generally, a therapeutically effective amount is sufficient to achieve a meaningful benefit to a subject (e.g., treating, modulating, curing, preventing and/or ameliorating a tumor). For example, a therapeutically effective amount can be an amount sufficient to achieve a desired therapeutic and/or prophylactic effect, such as an amount sufficient to treat a tumor or the symptoms thereof. Generally, the amount of a therapeutic agent (e.g., an Hh pathway modulator) administered to a subject in need thereof will depend upon the characteristics of the subject.
  • a therapeutic agent e.g., an Hh pathway modulator
  • Such characteristics include the condition, disease severity, general health, age, sex and body weight of the subject.
  • One of ordinary skill in the art will be readily able to determine appropriate dosages depending on these and other related factors.
  • both objective and subjective assays can optionally be employed to identify optimal dosage ranges.
  • therapeutically effective amount can be administered in a dosing regimen that can include multiple unit doses.
  • a pharmaceutical composition described herein is in unit dosage form, e.g., as a tablet, capsule, powder, solution, suspension, emulsion, granule, or suppository.
  • the pharmaceutical composition can be sub-divided into unit doses containing appropriate quantities of an Hh pathway modulator described herein.
  • the unit dosage form can be a packaged pharmaceutical composition, for example, packeted powders, vials, ampoules, pre-filled syringes or sachets containing liquids.
  • the unit dosage form can be, for example, a capsule or tablet itself, or it can be the appropriate number of any such compositions in package form.
  • Such unit dosage form can contain from about 1 mg/kg to about 250 mg/kg, and can be given in a single dose or in two or more divided doses.
  • an Hh pathway modulator consists of or comprises a nucleic acid encoding an Hh pathway polypeptide
  • the present disclosure includes methods of administering such nucleic acid to a subject.
  • a nucleic acid encoding an Hh pathway inhibitor e.g., Ptc, Hhip, or SuFu polypeptide
  • a subject to treat a disease or disorder described herein (e.g., a tumor).
  • a nucleic acid encoding an Hh pathway polypeptide is inserted into a viral vector for delivery to a subject.
  • retrovirus vectors can be used as a recombinant delivery system for transferring nucleic acids encoding Hh pathway polypeptides in vivo (see, e.g., Dropulic, Hum. Gene Ther. 22:649-57 (2011); and Kumar et al, Curr. Gene Ther. 11 : 144-53 (2011)).
  • Retroviruses useful in methods of the present disclosure include, but are not limited to, murine leukemia virus (MLV), human immunodeficiency virus (HIV), equine infectious anaemia virus (EIAV), mouse mammary tumour virus (MMTV), Rous sarcoma virus (RSV), Fujinami sarcoma virus (FuSV), FBR murine osteosarcoma virus (FBR MSV), Moloney murine sarcoma virus (Mo-MSV), Abelson murine leukemia virus (A-MLV), Avian myelocytomatosis virus-29 (MC29), Avian erythroblastosis virus (AEV) and all other retroviridiae including lentiviruses (see, e.g., Coffin et al, "Retroviruses", 1997 Cold Spring Harbor Laboratory Press Eds: J M Coffin, S M Hughes, H E Varmus, pp 758-763)).
  • a replication defective retrovirus can be packaged into virions that can be used to infect a target cell through the use of a helper virus by standard techniques (see, e.g., Current Protocols in Molecular Biology, Ausubel, F. M. et al. (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14).
  • adenovirus-derived vectors are used to deliver nucleic acids encoding Hh pathway polypeptides.
  • the genome of an adenovirus can be manipulated such that it encodes and expresses an Hh pathway polypeptide, but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle (see, e.g., Berkner et al. (1988)
  • Suitable adenoviral vectors useful in the methods of the present disclosure include those derived from the adenovirus strain Ad type 5 dl324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.).
  • an adeno-associated virus is used to deliver a nucleic acid encoding an Hh pathway polypeptide (see, e.g., Muzyczka et al. (1992) Curr.
  • AAV vectors include those that normally infect humans (e.g., serotypes 1, 2, 3A, 3B, 4, 5, and 6) or primates (e.g., serotypes 1 and 4).
  • non-viral methods are useful to deliver a nucleic acid encoding an Hh pathway polypeptide to a subject.
  • Such nonviral methods of gene transfer can exploit mechanisms normally used by mammalian cells for uptake and intracellular transport of macromolecules.
  • liposomal delivery systems, poly-lysine conjugates, and artificial viral envelopes can be used.
  • a nucleic acid encoding an Hh pathway polypeptide is entrapped in liposomes bearing positive charges on their surface (e.g., lipofectins).
  • a liposome can be conjugated to a targeting agent described herein (see, e.g., Mizuno et al. (1992) No Shinkei Geka 20:547-551).
  • Certain cationic polymers known to spontaneously bind to and condense nucleic acids into nanoparticles can also be used including, e.g., naturally occurring proteins, peptides, or derivatives, as well as synthetic cationic polymers such as polyethylenimine (PEI), polylysine (PLL), etc.
  • PKI polyethylenimine
  • PLL polylysine
  • Many useful polymers contain both chargeable amino groups, to allow for ionic interaction with negatively charged DNA phosphate, and a degradable region, such as a hydro lyzable ester linkage.
  • complexation agents can protect DNA against degradation, e.g., by nucleases, serum components, etc., and create a less negative surface charge, which may facilitate passage through hydrophobic membranes (e.g., cytoplasmic, lysosomal, endosomal, nuclear) of the cell.
  • Certain complexation agents facilitate intracellular trafficking events such as endosomal escape, cytoplasmic transport, and nuclear entry, and can dissociate from the nucleic acid.
  • Hh pathway polynucleotide can also be advantageously provided to a cell ex vivo, followed by administration of the living cell to the subject.
  • primary or secondary cells are genetically engineered to express an Hh pathway polypeptide.
  • Such cells can be obtained from a variety of tissues and include cell types which can be maintained propagated in culture.
  • primary and secondary cells include fibroblasts, endothelial cells, glial cells, and neural cells.
  • primary cells are obtained from an individual to whom a genetically engineered primary or secondary cells is to be administered.
  • Primary cells can also be obtained from a donor (other than the recipient) of the same species or another species (e.g., mouse, rat, rabbit, cat, dog, pig, cow, bird, sheep, goat, horse).
  • Primary or secondary cells can be transfected with a nucleic acid encoding an Hh pathway polypeptide.
  • a cell is transfected with an exogenous nucleic acid sequence that includes a nucleic acid encoding an Hh pathway polypeptide and an additional nucleic acid sequence (e.g., a regulatory sequence, e.g., a promoter, which causes expression, e.g., inducible expression or upregulation, of an endogenous Hh pathway sequence).
  • Transfected primary or secondary cells may also include DNA encoding a selectable marker that confers a selectable phenotype upon them, facilitating their identification and isolation.
  • Methods for treating disease by implanting a cell that has been modified to express a recombinant protein are also well known. See, for example, U.S. Pat. No. 5,399,346. Although use of human cells for ex vivo therapy is preferred in some embodiments, other cells such as bacterial cells may be implanted in a subject's vasculature, continuously releasing a therapeutic agent. See, for example, U.S. Pat. Nos. 4,309,776 and 5,704,910.
  • an Hh pathway modulator described herein is administered to a subject in combination with one or more additional therapies, e.g., to treat a tumor and/or an angioproliferative disorder, and/or one or more symptoms of a tumor or angioproliferative disorder.
  • an Hh pathway modulator can be administered in combination with vismodegib (ErivedgeTM, Genentech, San Francisco, CA).
  • combined administration of an Hh pathway modulator and a second agent results in an improvement in a disease or disorder described herein or a symptom thereof to an extent that is greater than one produced by either the Hh pathway modulator or the second agent alone.
  • the difference between the combined effect and the effect of each agent alone can be a statistically significant difference.
  • combined administration of an Hh pathway modulator and a second agent allows administration of the second agent at a reduced dose, at a reduced number of doses, and/or at a reduced frequency of dosage compared to a standard dosing regimen approved for the second agent.
  • approved standard regimen for ErivedgeTM is 150 mg once daily.
  • a therapeutically effective amount of ErivedgeTM can be a dosage of less than about 150 mg and/or a frequency of greater than about once daily.
  • kits can be provided in a kit.
  • the kit includes (a) a container that contains an Hh pathway modulator described herein (e.g., a pharmaceutical composition comprising an Hh pathway modulator) and, optionally (b) informational material.
  • the informational material can be descriptive, instructional, marketing or other material that relates to the methods described herein and/or the use of an Hh pathway modulator, e.g., for therapeutic benefit.
  • the informational material of the kits is not limited in its form.
  • the informational material can include information about production of an Hh pathway modulator, molecular weight of an Hh pathway modulator, concentration, date of expiration, batch or production site information, and so forth.
  • the informational material relates to methods of administering an Hh pathway modulator, e.g., in a suitable amount, manner, or mode of administration (e.g., a dose, dosage form, or mode of administration described herein).
  • the method can be a method of treating a subject having a tumor.
  • the informational material e.g., instructions
  • the informational material can also be provided in other formats, such as Braille, computer readable material, video recording, or audio recording.
  • the informational material of the kit is contact information, e.g., a physical address, email address, website, or telephone number, where a user of the kit can obtain substantive information about an Hh pathway modulator therein and/or their use in the methods described herein.
  • the informational material can also be provided in any combination of formats.
  • kits can include other ingredients, such as a solvent or buffer, a stabilizer, or a preservative.
  • a kit can also include other agents, e.g., a second or third agent, e.g., other therapeutic agents.
  • the components can be provided in any form, e.g., liquid, dried or lyophilized form.
  • the components can be substantially pure (although they can be combined together or delivered separate from one another) and/or sterile.
  • the liquid solution can be an aqueous solution, such as a sterile aqueous solution.
  • reconstitution generally is by the addition of a suitable solvent.
  • the solvent e.g., sterile water or buffer, can optionally be provided in the kit.
  • a kit can include one or more containers for an Hh pathway modulator and/or other agents.
  • a kit contains separate containers, dividers or compartments for an Hh pathway modulator and informational material.
  • an Hh pathway modulator can be contained in a bottle, vial, or syringe, and the informational material can be contained in a plastic sleeve or packet.
  • the separate elements of a kit are contained within a single, undivided container.
  • an Hh pathway modulator can be contained in a bottle, vial or syringe that has attached thereto the informational material in the form of a label.
  • a kit can include a plurality (e.g., a pack) of individual containers, each containing one or more unit dosage forms (e.g., a dosage form described herein) of an Hh pathway modulator.
  • Containers can include a unit dosage, e.g., a unit that includes an Hh pathway modulator.
  • a kit can include a plurality of syringes, ampules, foil packets, blister packs, or medical devices, e.g., each containing a unit dose.
  • the containers of kits can be air tight, waterproof (e.g., impermeable to changes in moisture or evaporation), and/or light-tight.
  • a kit can optionally include a device suitable for administration of an Hh pathway modulator, e.g., a syringe or other suitable delivery device.
  • a device can be provided preloaded with an Hh pathway modulator, e.g., in a unit dose, or can be empty, but suitable for loading.
  • an Hh pathway modulator described herein is used to treat a disorder, disease, or condition associated with expression (e.g., abnormal expression) of an FSH receptor on a target cell or target tissue.
  • an Hh pathway modulator described herein is used to treat hyperproliferative, hyperplastic, metaplastic, dysplastic, and preneoplastic tissues, e.g., to treat a subject having, suffering from, or susceptible to, a condition associated with one or more of hyperproliferative, hyperplastic, metaplastic, dysplastic, and pre- neoplastic tissues.
  • such tissues and/or blood vessels associated with such tissues express FSH receptors.
  • hyperproliferative tissue is meant a neoplastic cell growth or proliferation, whether malignant or benign, including all transformed cells and tissues and all cancerous cells and tissues. Hyperproliferative tissues include, but are not limited to, precancerous lesions, abnormal cell growths, benign tumors, malignant tumors, and cancer.
  • hyperproliferative tissues include neoplasms, whether benign or malignant, located in the brain, prostate, colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, or urogenital tract.
  • neoplasms whether benign or malignant, located in the brain, prostate, colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, or urogenital tract.
  • tumor or tumor tissue refers to an abnormal mass of tissue that results from excessive cell division.
  • a tumor or tumor tissue comprises “tumor cells”, which are neoplastic cells with abnormal growth properties and no useful bodily function.
  • Tumors, tumor tissue, and tumor cells may be benign or malignant.
  • a tumor or tumor tissue can also comprise "tumor-associated non-tumor cells", such as vascular cells that form blood vessels to supply the tumor or tumor tissue.
  • Non-tumor cells can be induced to replicate and develop by tumor cells, for example, induced to undergo angiogenesis within or surrounding a tumor or tumor tissue.
  • malignancy refers to a non-benign tumor or a cancer.
  • cancer means a type of hyperproliferative disease that includes a malignancy characterized by deregulated or uncontrolled cell growth.
  • examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung
  • cancer of the peritoneum hepatocellular cancer
  • gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer, uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • cancer includes primary malignant cells or tumors (e.g., those whose cells have not migrated to sites in the subject's body other than the site of the original malignancy or tumor) and secondary malignant cells or tumors (e.g., those arising from metastasis, the migration of malignant cells or tumor cells to secondary sites that are different from the site of the original tumor).
  • primary malignant cells or tumors e.g., those whose cells have not migrated to sites in the subject's body other than the site of the original malignancy or tumor
  • secondary malignant cells or tumors e.g., those arising from metastasis, the migration of malignant cells or tumor cells to secondary sites that are different from the site of the original tumor.
  • the methods described herein can also be used to treat premalignant tissue and to prevent progression to a neoplastic or malignant state including, but not limited to, those disorders described above. Such uses are indicated in conditions known or suspected of preceding progression to neoplasia or cancer, in particular where non-neoplastic cell growth consisting of hyperplasia, metaplasia, or dysplasia has occurred (see, e.g., Robbins and Angell, Basic Pathology, 2d Ed., W.B. Saunders Co., Philadelphia, pp. 68-79 (1976)).
  • Hyperplasia is a form of controlled cell proliferation, involving an increase in cell number in a tissue or organ, without significant alteration in structure or function.
  • Hyperplastic disorders include, but are not limited to, angiofollicular mediastinal lymph node hyperplasia,
  • angiolymphoid hyperplasia with eosinophilia atypical melanocytic hyperplasia, basal cell hyperplasia, benign giant lymph node hyperplasia, cementum hyperplasia, congenital adrenal hyperplasia, congenital sebaceous hyperplasia, cystic hyperplasia, cystic hyperplasia of the breast, denture hyperplasia, ductal hyperplasia, endometrial hyperplasia, fibromuscular hyperplasia, focal epithelial hyperplasia, gingival hyperplasia, inflammatory fibrous hyperplasia, inflammatory papillary hyperplasia, intravascular papillary endothelial hyperplasia, nodular hyperplasia of prostate, nodular regenerative hyperplasia, pseudoepitheliomatous hyperplasia, senile sebaceous hyperplasia, and verrucous hyperplasia.
  • the methods described herein can also be used to treat metaplastic tissue
  • Metaplasia is a form of controlled cell growth in which one type of adult or fully differentiated cell substitutes for another type of adult cell.
  • Metaplastic disorders include, but are not limited to, agnogenic myeloid metaplasia, apocrine metaplasia, atypical metaplasia, autoparenchymatous metaplasia, connective tissue metaplasia, epithelial metaplasia, intestinal metaplasia, metaplastic anemia, metaplastic ossification, metaplastic polyps, myeloid metaplasia, primary myeloid metaplasia, secondary myeloid metaplasia, squamous metaplasia, squamous metaplasia of amnion, and symptomatic myeloid metaplasia.
  • Dysplasia can be a forerunner of cancer and is found mainly in the epithelia.
  • Dysplasia is a disorderly form of non-neoplastic cell growth, involving a loss in individual cell uniformity and in the architectural orientation of cells.
  • Dysplastic cells can have abnormally large, deeply stained nuclei, and exhibit pleomorphism.
  • Dysplasia can occur, e.g., in areas of chronic irritation or inflammation.
  • Dysplastic disorders include, but are not limited to, anhidrotic ectodermal dysplasia, anterofacial dysplasia, asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal dysplasia, cleidocranial dysplasia, congenital ectodermal dysplasia, craniodiaphysial dysplasia, craniocarpotarsal dysplasia, craniometaphysial dysplasia, dentin dysplasia, diaphysial dysplasia, ectodermal dysplasia, enamel dysplasia, encephalo-ophthalmic dysplasia, dysplasia epiphysialis hemimelia, dysplasia epiphysialis multiplex, dysplasia epiphysialis punctata, epithelial dys
  • oculovertebral dysplasia odontogenic dysplasia, ophthalmomandibulomelic dysplasia, periapical cemental dysplasia, polyostotic fibrous dysplasia, pseudoachondroplastic spondyloepiphysial dysplasia, retinal dysplasia, septo-optic dysplasia, spondyloepiphysial dysplasia, and
  • Additional pre-neoplastic tissue that can be treated by the methods described herein include, but are not limited to, benign dysproliferative disorders (e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia), leukoplakia, keratoses, Bowen's disease, Farmer's Skin, solar cheilitis, and solar keratosis.
  • benign dysproliferative disorders e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia
  • leukoplakia keratoses
  • Bowen's disease keratoses
  • Farmer's Skin Farmer's Skin
  • solar cheilitis solar cheilitis
  • a hedgehog modulator is used to treat an angioproliferative disorder.
  • angioproliferative disorder refers to any disease or condition characterized by abnormal vascular structures (e.g., structures not normally present in a healthy subject or malformed structures as compared to a healthy subject) and/or an abnormally high growth of vascular cells.
  • abnormal vascular structures e.g., structures not normally present in a healthy subject or malformed structures as compared to a healthy subject
  • proliferative vascular disorders include vascular tumors, hemangioma, and vascular malformations.
  • vascular malformations are disorders in which the vasculature is improperly formed, resulting in discolored areas, masses, benign growths, and lesions.
  • Non- limiting examples of vascular malformations include slow- flow vascular malformations (e.g., capillary malformation, venous malformation, and lymphatic malformation), fast-flow vascular malformations (e.g., arterial malformation, arteriovenous fistula, and arteriovenous
  • vascular malformation and combined-complex vascular malformations.
  • Methods of diagnosing vascular malformations are well known in the art (see, e.g. Buckmiller et al., Oral Diseases 16:405-418 (2010); Flors et al, RadioGraphics 31 :1321-1340 (2011)).
  • vascular malformations are treated using a combination therapy of a hedgehog modulator described herein and any known treatment for vascular malformations.
  • an angioproliferative disorder is a hemangioma.
  • Hemangiomas can occur anywhere in the body, but are typically found in or on the skin, particularly on the face and neck, or in the liver.
  • hemangioma include, but are not limited to, infantile hemangioma, congenital hemangioma, kaposiform hemangioendothelioma, infant syndromed hemangioma ("strawberry naevus”), senile hemangioma ("cherry hemangioma”), arteriovenous hemangioma ("cirsoid aneurysm”), verrucous hemangioma, and pyogenic granuloma.
  • Hemangiomas are also referred to as strawberry hemangioma, capillary
  • hemangioma and/or cavernous hemangioma.
  • the diagnosis and treatment of hemangiomas are known in the art (see, e.g., Mulliken et al, J. Plastic and Reconstructive Surgery 69:412-22 (1982); Greene, Clinics in Plastic Surgery 38: 1-5 (2011); Ernemann et al, Eur. J. Radiol. 75:2-11 (2010)).
  • Symptoms and complications of hemangioma can include, e.g., ulcerations (break down of the hemangioma), bleeding, occlusion, amblyopia (if the hemangioma is near or in the eye), psychosocial complications, alteration of the subject's appearance, attention and malicious reactions from others, and PHACES syndrome (in the case of segmented hemangiomas of the head and neck).
  • hemangiomas are treated using a combination therapy of a hedgehog modulator described herein and any known treatment for hemangiomas.
  • Base cell carcinoma nevus syndrome Online Mendelian Inheritance in Man #109400
  • ultrasonography revealed a small, retroflexed uterus with a small pedunculated leiomyoma and normal ovaries, bilaterally containing multiple pre-antral follicles.
  • ovarian failure follicle depletion or menopause
  • high gonadotropins which may be induced by radiation and/or medication such as chemotherapy.
  • This patient's prior lymphoma therapy could have produced such a scenario, but onset 23 years after completing treatment and the abrupt occurrence of amenorrhea after years of regular menses following initiation of vismodegib suggested this was unlikely.
  • CTTCCCCGAC CCAGGGAAAA AGGCCTGTCC TCTGGCGGTG GCCGCAGCTG TAGCCGCCCA
  • CTACCTCAAC CCTGTGGATG CATCACGATT CTCTAGTCCA CGTGTGACCC CACGACTGAG
  • CTCTGGCATC TCCCCATACT TCTCTAGCCG TCGCTCCAGC GAGGCTTCGC CTCTCGGTGG
  • ACTCCCTCAG CAAATGGAAG TTGTCCCCAA TGCCACCATC ATGAATGGCC ATCAACGGGA
  • CATGGAGTCC CAGCAGAACG CGGGCTTTGG TCTCATGCAG CCTCGGCCAC CCCTGGAACC
  • CAACACGGCC AGCCGTCACC GTGGAGTACG TTCTGGGCAA CAGCAGTTGT ATGCCAGGAC
  • CTTCCATCCT CCTGTACCAA TTGATGCCAG ACATCATGAG GGCCGTTACC ATTACGATCC
  • CTATGCAGAC ATTATTCCCT CAGCTGCCAC CGCCGGCACG GGGGCCATCC AC AT GGAATA
  • GAGGCCTATT CCAGGGATCC CTACGGTTCT GAACCCCGTC CAGGTCAGCT CCGGCCCTTC
  • GAAGGAGTTC GTGTGCAGGT GGCTGGACTG CTCAAGAGAG CAGAAACCCT TCAAAGCCCA GTATATGTTG GTAGTGCATA T GAGAAGAC A CACGGGCGAG AAGCCTCACA AATGCACTTT
  • GGCCAAGTAC GCGGCTGCCA CAGGAGGGCC GCCGCCGACG CCCCTGCCCA AC AT GGAGAG
  • GCACTTCCCC AGCGCCCTCC CGGACGACAG CAAAGTGCCC CACGGGCCCG GTGACTTTGA
  • CAGCATGCAG CACCCGGGGG CAGGCCGCCC CGGTCAGCAG ATGCTTGGGC AGATTAGTGC
  • TGACACAAGT CAGACCTGCA GGGTGAATGG TATCAAGATG GAGAT GAAAG GGCAGCCCCA
  • CAGTCAGCAA GACACGAAAG CTGGTTCATT CTCTATTTCA GACGCCAGCT GCCTGCTACA
  • GTCCATGAGC ACCACCAACA TGGCTATCGG GGAC AT GAGT TCTTTGCTGA CCTCCCTAGC
  • TTCACTCCAC TACGAAGGCC GTGCTTTGGA CATCACTACG TCTGACCGCG ACCGCAACAA
  • GCAGAACTGC AAAATGTGAG CCAGCATGTC GTCATGGAGG TGTCTGTGTT AGACCGAACA
  • GACGCGAGGA CAGGAGACTG GATATTTTCT GCTGTTTTAC AAGCCCCTGC GTCAGCAGAG
  • CAGAGAGCAC CAGCTCCACA AGGGACCTGC TCTCCCAGTT CTCCGACTCC AGCCTCCACT
  • CTGAGACTGA CCACGGCCTG TTTGAGGACC CCCACGTGCC TTTCCACGTC CGGTGTGAGA

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des procédés de sélection de sujets pour le traitement par des modulateurs de la voie hedgehog, tels que des activateurs ou des inhibiteurs de la voie hedgehog, et l'utilisation de tels modulateurs pour traiter des sujets.
PCT/US2014/036560 2013-05-03 2014-05-02 Méthodes de traitement du cancer et de troubles angioprolifératifs WO2014179678A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361818968P 2013-05-03 2013-05-03
US61/818,968 2013-05-03

Publications (1)

Publication Number Publication Date
WO2014179678A1 true WO2014179678A1 (fr) 2014-11-06

Family

ID=51843971

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/036560 WO2014179678A1 (fr) 2013-05-03 2014-05-02 Méthodes de traitement du cancer et de troubles angioprolifératifs

Country Status (1)

Country Link
WO (1) WO2014179678A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100316572A1 (en) * 2008-02-18 2010-12-16 Institut National De La Sante Et De La Recherche Medicale (Inserm) Use of fsh receptor ligands for diagnosis and therapy of cancer
US20110077256A1 (en) * 1999-09-16 2011-03-31 Guicherit Oivin M Mediators of hedgehog signaling pathways, compositions and uses related thereto
US20130045240A1 (en) * 2009-08-25 2013-02-21 Abraxis Bioscience, Llc Combination therapy with nanoparticle compositions of taxane and hedgehog inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110077256A1 (en) * 1999-09-16 2011-03-31 Guicherit Oivin M Mediators of hedgehog signaling pathways, compositions and uses related thereto
US20100316572A1 (en) * 2008-02-18 2010-12-16 Institut National De La Sante Et De La Recherche Medicale (Inserm) Use of fsh receptor ligands for diagnosis and therapy of cancer
US20130045240A1 (en) * 2009-08-25 2013-02-21 Abraxis Bioscience, Llc Combination therapy with nanoparticle compositions of taxane and hedgehog inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
INNAMORATI, G ET AL.: "Molecular Approaches To Target GPCRs in Cancer Therapy.", PHARMACEUTICALS, vol. 4, 25 March 2011 (2011-03-25), pages 567 - 589 *

Similar Documents

Publication Publication Date Title
US20050037439A1 (en) Differentially expressed genes involved in cancer, the polypeptides encoded thereby, and methods of using the same
US9453260B2 (en) Genetic products differentially expressed in tumors and use thereof
US10175244B2 (en) Dominant negative HSP110 mutant and its use in prognosing and treating cancers
WO2002092635A2 (fr) Compositions immunologiques et procedes d'etude et de traitement des cancers par l'expression des antigenes frizzled
US20090214517A1 (en) Compositions and methods of use for modulators of nectin 4, semaphorin 4b, igsf9, and kiaa0152 in treating disease
US9844541B2 (en) Methods of modulating follicle stimulating hormone activity
US20150344972A1 (en) Compositions comprising soluble cd84 or anti-cd84 antibodies and methods for diagnosing and treating b-cll
WO2005033133A2 (fr) Polynucleotides codant pour des nouveaux polypeptides erbb-2; trousses et methodes d'utilisation
US20060275314A1 (en) Transmembrane protein differentially expressed in cancer
WO2004074436A2 (fr) Procede d'utilisation d'un gpcr pour diagnostiquer et traiter le cancer du colon et du poumon
US20100028867A1 (en) LRRTM1 Compositions and Methods of Their Use for the Diagnosis and Treatment of Cancer
CA2518787A1 (fr) Anticorps contre un antigene specifique aux tumeurs
WO2014179678A1 (fr) Méthodes de traitement du cancer et de troubles angioprolifératifs
PT2190469E (pt) Polipéptidos e polinucleótidos, e utilizações dos mesmos como um alvo de fármacos para produzir fármacos e agentes biológicos
JP4416650B2 (ja) 新規なポリヌクレオチドおよびポリペプチド配列ならびにその使用
JP4530631B2 (ja) 新規タンパク質および癌の予防・治療剤
KR20050085881A (ko) 신규 단백질 및 그 용도
DK2190469T3 (en) POLYPEPTIDES AND POLYNUCLEOTIDES AND USES THEREOF AS a drug FOR THE PRODUCTION OF PHARMACEUTICAL AND BIOTECHNOLOGY PRODUCTS
JP2004501630A (ja) Gp286核酸およびポリペプチド
WO2004033643A2 (fr) Methodes d'utilisation d'une adn helicase dans le diagnostic et le traitement du cancer du colon et du poumon
MX2008003464A (en) Identification of tumor-associated antigens for diagnosis and therapy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14791172

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14791172

Country of ref document: EP

Kind code of ref document: A1