WO2013134546A1 - Methods and materials for treating cancer - Google Patents

Methods and materials for treating cancer Download PDF

Info

Publication number
WO2013134546A1
WO2013134546A1 PCT/US2013/029688 US2013029688W WO2013134546A1 WO 2013134546 A1 WO2013134546 A1 WO 2013134546A1 US 2013029688 W US2013029688 W US 2013029688W WO 2013134546 A1 WO2013134546 A1 WO 2013134546A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
scdl
mammal
polypeptide
inhibitor
Prior art date
Application number
PCT/US2013/029688
Other languages
French (fr)
Inventor
John A. Copland
Laura Ann MARLOW
Christina VON ROEMELING
Original Assignee
Mayo Foundation For Medical Education And Research
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 Mayo Foundation For Medical Education And Research filed Critical Mayo Foundation For Medical Education And Research
Priority to US14/383,385 priority Critical patent/US9233102B2/en
Publication of WO2013134546A1 publication Critical patent/WO2013134546A1/en
Priority to US14/961,444 priority patent/US20160152986A1/en
Priority to US15/692,491 priority patent/US10160972B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/451Non condensed piperidines, e.g. piperocaine having a carbocyclic group directly attached to the heterocyclic ring, e.g. glutethimide, meperidine, loperamide, phencyclidine, piminodine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • G01N33/57407Specifically defined cancers
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin
    • 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

  • This document relates to methods and materials involved in treating cancer, for example, renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
  • this document provides methods and material for using one or more inhibitors of a stearoyl-Coenzyme A desaturase 1 (SCDl) polypeptide to treat cancer.
  • SCDl stearoyl-Coenzyme A desaturase 1
  • This document provides methods and materials for treating cancer, for example, renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
  • this document provides methods and material for using one or more inhibitors of an SCDl polypeptide to treat renal cell carcinoma (e.g., clear cell renal cell carcinoma (ccRCC)) or to increase the efficacy of a renal cell carcinoma treatment.
  • SCDl polypeptides are examples of an SCDl polypeptide to treat renal cell carcinoma (e.g., clear cell renal cell carcinoma (ccRCC)) or to increase the efficacy of a renal cell carcinoma treatment.
  • ccRCC clear cell renal cell carcinoma
  • one or more inhibitors of an SCDl polypeptide can be used to reduce the number of cancer cells within a mammal (e.g., a human). In some cases, one or more inhibitors of an SCDl polypeptide can be used to increase the efficacy of a cancer treatment.
  • one or more inhibitors of an SCDl polypeptide can be used to increase the efficacy of a renal cell carcinoma treatment (e.g., treatment with Nexavar ® , Sutent ® , Torisel ® , Afmitor ® , and interleukin-2).
  • a renal cell carcinoma treatment e.g., treatment with Nexavar ® , Sutent ® , Torisel ® , Afmitor ® , and interleukin-2).
  • one aspect of this document features a method for reducing the number of renal cell carcinoma cells within a mammal.
  • the method comprises, or consists essentially of, administering, to the mammal, an inhibitor of an SCDl polypeptide under conditions wherein the number of viable renal cell carcinoma cells present within the mammal is reduced.
  • the mammal can be a human.
  • the administration can be an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration.
  • the inhibitor can be A939572, MK-8245, CVT-11127, MF-152, or HYR-061.
  • one or more inhibitors of an SCDl polypeptide can be administered with one or more inhibitors of a mTor polypeptide.
  • Non-limiting examples of such inhibitors include sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RADOOl), and ridaforolimus (AP-23573).
  • this document features a method for reducing the number of renal cell carcinoma cells within a mammal.
  • the method comprises, or consists essentially of, administering, to the mammal, a composition under conditions wherein the number of viable renal cell carcinoma cells present within the mammal is reduced, wherein the composition comprises the ability to reduce SCDl mRNA expression or SCDl polypeptide expression.
  • the mammal can be a human.
  • the administration can be an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration.
  • the composition can comprise a nucleic acid construct having the ability to express an shRNA directed against SCDl nucleic acid.
  • this document features a method for reducing the number of cancer cells overexpressing an SCDl polypeptide within a mammal.
  • the method comprises, or consists essentially of, administering, to the mammal, an inhibitor of an SCDl polypeptide under conditions wherein the number of viable cancer cells overexpressing an SCDl polypeptide present within the mammal is reduced.
  • cancers include renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
  • this document features a method for reducing the number of cancer cells overexpressing an SCDl polypeptide within a mammal.
  • the method comprises, or consists essentially of, administering, to the mammal, a composition under conditions wherein the number of viable cancer cells overexpressing an SCDl polypeptide present within the mammal is reduced, wherein the composition comprises the ability to reduce SCDl mRNA expression or SCDl polypeptide expression.
  • cancers include renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
  • this document features a method for identifying a mammal having cancer cells responsive to treatment with an inhibitor of an SCDl polypeptide.
  • the method comprises, or consists essentially of, (a) detecting the presence of cancer cells expressing an elevated level of an SCDl mRNA or an SCDl polypeptide, and (b) classifying the mammal has having cancer cells responsive to treatment with the inhibitor of an SCDl polypeptide.
  • the method can comprise measuring SCDl mRNA expression using real time PCR.
  • the method can comprise measuring SCDl polypeptide expression using an immunohistochemical technique.
  • the method can comprise measuring SCDl polypeptide expression using a Western blot analysis.
  • Non- limiting examples of cancers include renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
  • this document features a method for reducing the number of cancer cells within a mammal.
  • the method comprises, or consists essentially of, administering, to said mammal, an inhibitor of an SCDl polypeptide and an inhibitor of an mTor polypeptide under conditions wherein the number of viable cancer cells present within said mammal is reduced.
  • the inhibitor of an mTor polypeptide can be sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RAD001), or ridaforolimus (AP-23573).
  • the mammal is a human.
  • the administration is an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration.
  • the inhibitor of an SCDl polypeptide is A939572, MK- 8245, CVT-11127, MF-152, or HYR-061.
  • cancer cells include one or more of ovarian cancer, breast cancer, prostate cancer, colon cancer, renal cancer, pancreatic cancer, bladder cancer, liver cancer, lung cancer, thyroid cancer, and melanoma.
  • Figure 1A is a graph plotting SCD1 mRNA levels in ccRCC tissue and matched normal tissue across stages I-IV.
  • Figure IB contains photographs of representative ccRCC tissue and matched normal tissue stained for SCD1 polypeptide expression.
  • Figure 1C is a graph plotting SCD1 mRNA levels in normal renal epithelial cell lines (347N, 355N, 359N, 360N, 365N, and 366N) versus ccRCC cell lines.
  • Figure ID contains photographs of a Western blot analysis of SCD1 polypeptide expression by normal cell lines and ccRCC cell lines.
  • Figures 2A and 2D shows the knockdown of SCD1 in ccRCC as shown by decrease in both (A) mRNA and (D) protein expression using two separate lentiviral constructs shSCD780 and shSCD1200.
  • Figures 2B and 2C show proliferation in (B) A498 and Cakil ccRCC cell lines and (C) NRE samples of NT versus shSCD lentiviral infected cells.
  • Figure 2D contains photographs of an immunoblot for Poly-ADP ribose polymerase (PARP) cleavage and SCD1 expression in A498 and Cakil cell lines.
  • PARP Poly-ADP ribose polymerase
  • Figure 3A is a bar graph showing proliferation for SCDl and PARP cleavage in Cakil and A498 NT versus shSCD with or without OA-BSA supplementation.
  • Figure 3B contains photographs of a Western blot analysis for SCDl and PARP cleavage in Cakil and A498 NT versus shSCD with or without OA-BSA supplementation.
  • Figure 3C contains photographs of a phase-contrast microscopy representative ccRCC cell (Cakil) confluence at day 5 of proliferation assay with different treatment conditions.
  • Figure 4 Treatment of ccRCC cells with a small molecule SCDl inhibitor, A939572, inhibits cell growth and induces apoptosis.
  • Figure 4A is a line graph showing cell proliferative response to dose out of A939572 in Cakil, A498, Caki2, and ACFiN ccRCC cell lines.
  • Figure 4B is a bar graph displaying ccRCC proliferation rescue with OABSA in A939572 treated ccRCC cell lines.
  • Figure 4C contains photographs of a Western blot analysis for PARP cleavage in A939572 treated vs. control, as well as OA- BSA rescue in ccRCC cell lines.
  • Figure 4D contains representative phase contrast images of A939572 treated ccRCC cells (A498) +/-OA-BSA rescue at day 5.
  • Figure 5 Inhibition of SCDl activity in ccRCC induces cell death mediated by endoplasmic reticulum stress response.
  • Figure 5A contains photographs of a Western blot analysis for expression of ER stress markers: BiP, CHOP, and spliced XBP1 in response to A939572 treatment or lentiviral silencing of SCDl in Cakil and A498.
  • Figure 5B provides bar graphs showing QPCR analysis of ER stress gene expression in Cakil and A498 cells treated with A939572 or shSCD lentivirus +/-OA-BSA rescue.
  • Figures 5C provides bar graphs showing relative luciferase activity of ER stress p5xATF6-GL3 (UPR) luciferase reporter transfected in Cakil and A498 cells treated with A939572 or shSCD lentivirus +/-OA-BSA supplementation.
  • URR ER stress p5xATF6-GL3
  • Figure 6 Treatment of ccRCC cells with SCDl inhibitor in combination with the mTOR inhibitor Temsirolimus synergistically inhibits tumor cell growth in vivo.
  • Figure 6B contains photographs of IHC of tissue harvested from treatment groups stained for Ki67 and CC3 (quantitated by N-score), CD31 (quantitated by I-score), and phospho- mTOR (quantitated by H-score).
  • Figure 6C contains photographs of Western blot and quantitation of CHOP expression in all four treatment groups.
  • Figure 6D is an illustration of proposed SCD1 activity in ccRCC model: inhibition of SCD 1 blocks desaturation of SFA resulting in an accumulation of SFA species which trigger the ER stress response.
  • Figures 7A-D are line graphs comparing cell number to dose of A939572 or Gemicitabine in MiaPaca and pancreatic cancer cells.
  • Figure 7E contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 8A-B are line graphs comparing cell number to dose of A939572 or
  • Figure 8C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 9A-C are line graphs comparing cell number to dose of A939572 or Temodar in A375 AND Mela 11 melanoma cancer cells.
  • Figure 9D contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 10A-B are line graphs comparing cell number to dose of A939572 or Capecitabine in CaCo2 and HT29 colon cancer cells.
  • Figure IOC contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 11A-B are line graphs comparing cell number to dose of A939572 or cisplatin in T24 and HT1276 bladder cancer cells.
  • Figure 11C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 12A-B are line graphs comparing cell number to dose of A939572 or cisplatin in BCJ4T bladder cancer cells.
  • Figure 12C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 13A-B are line graphs comparing cell number to dose of A939572 or
  • Taxol in KTC3 and FF1 anaplastic thyroid cancer cells contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 14A-B are line graphs comparing cell number to dose of A939572 or Taxol in A549 and CaLu-1 lung cancer cells.
  • Figure 14C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 15A-B are line graphs comparing cell number to dose of A939572 or Taxol in OVCA420 and HOVTax2res ovarian cancer cells.
  • Figure 15C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figure 16A is a line graph comparing cell number to dose of A939572 in MCF-7 (ER+/PR+), MDA-231 (triple negative) and T47D (PR+) breast cancer cells.
  • Figure 16B contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figure 17A is a line graph comparing cell number to dose of A939572 in DU-145 prostate cancer cells.
  • Figure 17B contains photographs of Western Blot and quantitation of SCDl and beta-actin expression.
  • Figure 18A is a bar graph illustrating SCD1 protein expression in various cancer cell lines.
  • Figure 18B contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figure 19A is a bar graph illustrating SCD1 protein expression in various cancer cell lines.
  • Figure 19B contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
  • Figures 20-22 provide structures for exemplary SCD1 inhibitors.
  • This document provides methods and materials for treating cancer, for example, for example, renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
  • this document provides methods and material for using one or more inhibitors of an SCD1 polypeptide to treat cancer (e.g., clear cell renal cell carcinoma (ccRCC)) or to increase the efficacy of a cancer treatment.
  • cancer e.g., clear cell renal cell carcinoma (ccRCC)
  • ccRCC clear cell renal cell carcinoma
  • one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be administered to a mammal (e.g., a human) having cancer (e.g., renal cancer) under conditions wherein the number of cancer cells within the mammal is reduced.
  • a mammal e.g., a human
  • cancer e.g., renal cancer
  • one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be administered to a mammal (e.g., a human) having renal cancer (e.g., ccRCC) under conditions wherein the number of renal cancer cells within the mammal is reduced.
  • An SCDl polypeptide can be a human SCDl polypeptide having the amino acid sequence set forth in GenBank ® Accession No. 000767 (GI No. 21431730) or a human SCDl polypeptide encoded by nucleic acid having the nucleic acid sequence set forth in GenBank ® Accession No. AF097514.1 (GI No. 4808600).
  • Examples of inhibitors of an SCDl polypeptide include, without limitation, inhibitory anti-SCDl polypeptide antibodies, siRNA molecules, shRNA molecules, nucleic acid vectors designed to express siRNA or shRNA molecules, anti-sense molecules, and small molecule antagonists such as A939572 (Biofine International Inc., Urvashi et al, Mol.
  • an inhibitor of an SCDl polypeptide can be an inhibitor described elsewhere (Igal, Carcinogenesis, 31(9): 1509-1515 (2010); Oballa, J. Med. Chem., 54:5082-5096 (2011); Li et al., Bioorganic & Medicinal Chemistry Letters, 19:5214-5217 (2009); Uto et al, Eur. J. Med. Chem., 46: 1892-1896 (2011); Uto et al, Eur. J. Med. Chem., 45:4788-4796 (2010); Liu, G.
  • compositions and methods described herein include, but are not limited to, the following:
  • Breast cancers including, for example ER + breast cancer, ER " breast cancer, her2 " breast cancer, her2 + breast cancer, stromal tumors such as fibroadenomas, phyllodes tumors, and sarcomas, and epithelial tumors such as large duct papillomas; carcinomas of the breast including in situ (noninvasive) carcinoma that includes ductal carcinoma in situ (including Paget's disease) and lobular carcinoma in situ, and invasive (infiltrating) carcinoma including, but not limited to, invasive ductal carcinoma, invasive lobular carcinoma, medullary carcinoma, colloid (mucinous) carcinoma, tubular carcinoma, and invasive papillary carcinoma; and miscellaneous malignant neoplasms.
  • in situ (noninvasive) carcinoma that includes ductal carcinoma in situ (including Paget's disease) and lobular carcinoma in situ
  • invasive (infiltrating) carcinoma including, but not limited to, invasive ductal carcinoma, invasive lobular carcinoma,
  • breast cancers can include luminal A, luminal B, basal A, basal B, and triple negative breast cancer, which is estrogen receptor negative (ER ⁇ ), progesterone receptor negative, and her2 negative (her2 ⁇ ).
  • the breast cancer may have a high risk Oncotype score;
  • lung cancers including, for example, bronchogenic carcinoma, e.g., squamous cell, undifferentiated small cell, undifferentiated large cell, and adenocarcinoma; alveolar and bronchiolar carcinoma; bronchial adenoma;
  • bronchogenic carcinoma e.g., squamous cell, undifferentiated small cell, undifferentiated large cell, and adenocarcinoma
  • alveolar and bronchiolar carcinoma bronchial adenoma
  • sarcoma lymphoma; chondromatous hamartoma; and mesothelioma
  • cancers of the kidney e.g., adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, and leukemia
  • cancers of the bladder and urethra e.g., squamous cell carcinoma, transitional cell carcinoma, and adenocarcinoma
  • cancers of the prostate e.g., adenocarcinoma, and sarcoma
  • cancer of the testis e.g., seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, and lipoma
  • testis e.g., seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma,
  • liver cancers including, for example, hepatoma, e.g., hepatocellular carcinoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hepatocellular adenoma; and hemangioma;
  • hepatoma e.g., hepatocellular carcinoma
  • cholangiocarcinoma e.g., hepatocellular carcinoma
  • hepatoblastoma hepatoblastoma
  • angiosarcoma hepatocellular adenoma
  • hemangioma hemangioma
  • gynecological cancers including, for example, cancers of the uterus, e.g., endometrial carcinoma; cancers of the cervix, e.g., cervical carcinoma, and pre tumor cervical dysplasia; cancers of the ovaries, e.g., ovarian carcinoma, including serous cystadenocarcinoma, epithelial cancer, mucinous
  • cystadenocarcinoma unclassified carcinoma, granulosa thecal cell tumors, Sertoli Leydig cell tumors, dysgerminoma, and malignant teratoma; cancers of the vulva, e.g., squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, and melanoma; cancers of the vagina, e.g., clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma, and embryonal rhabdomyosarcoma; and cancers of the fallopian tubes, e.g., carcinoma;
  • skin cancers including, for example, malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and
  • adrenal gland cancers including, for example, neuroblastoma.
  • one or more (e.g., one, two, three, four, or more) inhibitors of an SCDl polypeptide can be used as described herein to treat cancer, including renal cancer, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, and thyroid cancers as well as melanoma.
  • a human having cancer can be administered one or more inhibitors of an SCDl polypeptide under conditions that result in reduced tumor size or stable disease.
  • one or more (e.g., one, two, three, four, or more) inhibitors of an SCDl polypeptide can be used as described herein to increase the efficacy of a cancer treatment.
  • one or more (e.g., one, two, three, four, or more) inhibitors of an SCDl polypeptide (or a pharmaceutically acceptable salt form thereof) can be administered in combination with (i.e., before, during, or after) administration of a pain relief agent (e.g., a nonsteroidal anti-inflammatory drug such as celecoxib or rofecoxib), an antinausea agent, or an additional anticancer agent (e.g., paclitaxel, docetaxel, doxorubicin, daunorubicin, epirubicin, fluorouracil, melphalan, cis-platin, carboplatin,
  • a pain relief agent e.g., a nonsteroidal anti-inflammatory drug such as celecoxib or rofecoxib
  • an antinausea agent e.g., an anticancer agent
  • an additional anticancer agent e.g., paclitaxel, docetaxel, dox
  • cyclophosphamide mitomycin, methotrexate, mitoxantrone, vinblastine, vincristine, ifosfamide, teniposide, etoposide, bleomycin, leucovorin, taxol, herceptin, avastin, cytarabine, dactinomycin, interferon alpha, streptozocin, prednisolone, irinotecan, sulindac, 5 -fluorouracil, capecitabine, oxaliplatin/5 FU, abiraterone, letrozole,
  • the anticancer agent is paclitaxel or docetaxel. In other embodiments, the anticancer agent is cisplatin or irinotecan.
  • a human having ccRCC can be administered one or more inhibitors of an SCD1 polypeptide under conditions that result in reduced tumor size or stable disease.
  • one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be used as described herein to increase the efficacy of a renal cell carcinoma treatment.
  • renal cell carcinoma treatments include, without limitation, treatment with Nexavar ® , Sutent ® , Torisel ® , Afinitor ® , or interleukin-2.
  • one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be used as described herein can be used in combination with one or more (e.g., one, two, three, four, or more) inhibitors of mammailian target of rapamycin (mTor) polypeptide.
  • mTor inhibitors include:
  • sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RAD001),
  • the method comprises administering, to the mammal, an inhibitor of an SCD1 polypeptide and an inhibitor of an mTor polypeptide under conditions wherein the number of viable cancer cells present within said mammal is reduced.
  • the one or more mTor inhibitor can include a standard of care drug for a particular cancer cell type.
  • an SCD1 inhibitor can be administered with pacliltaxel and/or platin (cisplatin, carboplatin, or oxaliplatin) for the treatment of ovarian cancer.
  • the following standard of care drugs can be combined with an SCD1 inhibitor for the following cancers:
  • Hormonally responsive breast - aromatase inhibitors such as letrazole and/or antiestrogens such as tamoxifen
  • Thyroid - paclitaxel and/or cisplatin Thyroid - paclitaxel and/or cisplatin
  • the combination of one or more inhibitors of an SCD1 polypeptide and one or more inhibitors of mTor exhibit a synergistic response.
  • the one or more inhibitors of an SCD1 polypeptide can be administered before, during, or after administration of the one or more inhibitors of mTor.
  • An inhibitor of an SCD1 polypeptide can also be administered to a subject in combination with surgical methods to treat cancers, e.g., resection of tumors.
  • the inhibitor can be administered to the individual prior to, during, or after the surgery.
  • the inhibitor can be administered parenterally, intravenous or injected into the tumor or surrounding area after tumor removal.
  • one or more of the inhibitors of an SCD1 polypeptide provided herein can be formulated into a pharmaceutical composition that can be administered to a mammal (e.g., rat, dog, horse, cat, mouse, rabbit, pig, cow, monkey, or human).
  • a mammal e.g., rat, dog, horse, cat, mouse, rabbit, pig, cow, monkey, or human.
  • A939572 or a pharmaceutically acceptable salt thereof can be in a mammal (e.g., rat, dog, horse, cat, mouse, rabbit, pig, cow, monkey, or human).
  • A939572 or a pharmaceutically acceptable salt thereof can be in a mammal
  • pharmaceutically acceptable carrier refers to any pharmaceutically acceptable solvent, suspending agent, or other pharmacologically inert vehicle.
  • Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties.
  • Typical pharmaceutically acceptable carriers include, without limitation, water, saline solutions, dimethyl sulfoxide, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose), fillers (e.g., lactose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g., starch or sodium starch glycolate), and wetting agents (e.g., sodium lauryl sulfate).
  • binding agents e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose and other sugars, gelatin, or calcium sulfate
  • lubricants e.g., starch, polyethylene glycol, or sodium acetate
  • disintegrates e.g., starch or sodium starch glycolate
  • wetting agents e.g.,
  • pharmaceutically acceptable salt refers to the relatively non-toxic, inorganic and organic acid addition salts of a compound provided herein. These salts can be prepared in situ during the final isolation and purification of a compound provided herein, or by separately reacting the compound in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, laurylsulphonate salts, and amino acid salts, and the like.
  • sulfate bisulfate
  • phosphate nitrate
  • acetate valerate
  • oleate palmitate
  • stearate laurate
  • benzoate lactate
  • phosphate tosylate
  • citrate maleate
  • fumarate succinate
  • tartrate naphthylate
  • mesylate glucoheptonate
  • lactobionate lactobionate
  • laurylsulphonate salts
  • a compound provided herein may contain one or more acidic functional groups and, thus, is capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases.
  • pharmaceutically acceptable salts refers to the relatively non-toxic inorganic and organic base addition salts of a compound provided herein. These salts can likewise be prepared in situ during the final isolation and purification of the compound, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al, supra).
  • ccRCC cell lines RWV366T and KD265T (16) (both stage IV ccRCC patient tissue derived), A498, Cakil, Caki2, and ACHN (ATCC) and K347N, K355N, K359N, K360N, K365N, and K366N normal renal tissue derived mortal cells (NRE) were cultured in DMEM medium (Cellgro) containing 5%FBS (Hyclone) and lxpenicillin- streptomycin (Invitrogen) at 37°C in humidified conditions with 5%C0 2 .
  • DMEM medium Cellgro
  • FBS Hyclone
  • Invitrogen lxpenicillin- streptomycin
  • Cells were plated (0.5 or lxl0 5 /well) in 24-well plates for proliferation or treatment assays, in triplicate. Cells were trypsinized (0.25%) and counted using a Coulter Particle Counter at specified time intervals. For SCD1 rescue assays, oleic acid- albumin was added to media at 5 ⁇ . Drug stocks were prepared in
  • DMSO diluting 2x growth medium 1 : 1 in 1.5% Seaplaque®GTG® agarose, with 500 cells/plate in 60mm culture dishes.
  • Colonies were stained with Giemsa (LabChem Inc.) and counted after 3 wks. Cell images were obtained with an 01ympusIX71 microscope at 20x magnification.
  • Cakil and A498 cells were transfected using
  • RNAqueous Midi Kit An R Aqueous Midi Kit was utilized to extract and purify RNA from cell lines.
  • Human tissue RNA was prepared using using TRIzol® per manufacturer's protocol followed by purification using the RNAqueous Midi Kit.
  • the O.D. 260/280 ratio of the mRNA was at least 1.8 and the 18s/28s bands were verified on a 1% agarose gel.
  • cDNA was prepared from purified RNA samples using High Capacity cDNA Reverse
  • TaqMan®Fast Universal PCR Master Mix and TaqMan®FAMTM dye-labeled probes including POLR2A (Hs00172187_ml) (normalization control), SCD1 (Hs01682761_ml), HSPA5 (Hs99999174_ml), CEBPp (CEBPB Hs00270923_sl), GADD45A (Hs00169255_ml), DDIT3 (Hs01090850_ml), and HERPUD1 (HsOl 124269 ml) were combined with prepared cDNA samples to analyze relative mRNA expression via qPCR.
  • POLR2A Hs00172187_ml
  • SCD1 Hs01682761_ml
  • HSPA5 Hs99999174_ml
  • CEBPp CEBPB Hs00270923_sl
  • GADD45A Hs00169255_ml
  • DDIT3 Hs01090850_ml
  • HERPUD1 H
  • Fold change values were compared between normal and tumor, non-target scrambled lentiviral and target lentiviral infected, and DMSO vs. A939572 treated samples using the AACt method (Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc.
  • Gene array expression analysis was performed using Affymetrix Human Genome U133 Plus 2.0 Array chip. The details of the data processing and methodology were previously described in (Tun HW, Marlow LA, von Roemeling CA, Cooper SJ, Kreinest P, Wu K, et al. Pathway signature and cellular differentiation in clear cell renal cell carcinoma. PLoS One. 2010;5:el0696). Gene expression data was deposited at Gene Expression Omnibus (Accession#GSE41485). Pathway analysis was performed using IP A (Ingenuity® Systems).
  • Protein extracts, electrophoresis, and membrane transfers were prepared as previously described (Copland JA, Marlow LA, Kurakata S, Fujiwara K, Wong AK, Kreinest PA, et al. Novel highaffinity PPARgamma agonist alone and in combination with paclitaxel inhibits human anaplastic thyroid carcinoma tumor growth via p21WAFl/CIPl . Oncogene. 2006;25:2304-17).
  • a Supersignal chemiluminescent kit was used to perform detection. IHC and ICC Analysis
  • TMA Formalin fixed, paraffin-embedded tissue microarray
  • the TMAs were mounted on slides from paraffin-embedded blocks according to IHC procedure and samples were blocked with Diluent that contained Background Reducing Components (Dakocytomation) for 30 min and then probed for SCD1, Ki67, Caspase-3, CD31, phospho-mTOR, DDIT3, and XBPl .
  • ICC preparation and staining was performed as previously described (Cooper SJ, Von Roemeling CA, Kang KH, Marlow LA, Grebe SK, Menefee ME, et al. Reexpression of tumor suppressor, sFRPl, leads to antitumor synergy of combined HDAC and methyltransferase inhibitors in
  • intensity (I)-scores were calculated by dividing signal intensity by area
  • nuclear (N)-scores were calculated by dividing % positive nuclei by total nuclei examined per area. Cases where insufficient tumor tissue presented were excluded from the study. 20x images were obtained using Scanscope XT and Imagescope software. RWV366T cell line validation was carried out as previously
  • A498 cells were subcutaneously implanted in athymic nu/nu mice at lxlO 6 cells/mouse in 50%Matrigel. Tumors reached ⁇ 50 mm 3 prior to treatment, which was carried out for 4 wks.
  • A939572 was administered via oral feeding using strawberry flavored Kool-Aid® in sterilized H 2 0 (0.2g/mL) vehicle at 30 mg/kg in a 50 ⁇ dose twice daily/mouse.
  • Temsirolimus was solubilized in 30% ethanol/saline and administered via intraperitoneal injection at 10 mg/kg in a 50 ⁇ dose once every 72 hrs/mouse. Tumor volumes were calculated using the formula 0.5236(L*W*H) and body weight were measured every 3 days.
  • Genomic DNA was extracted from both RWV366T patient primary tissue and matching cell line using PurelinkTM Genomic DNA mini kit. Sixteen STR markers were PCR amplified using fluorescently labeled primers from ABI, and were analyzed using ABI 3130. Peak sizes were calculated versus a co-injected size standard using Gene Marker.
  • Example 1 - SCDl Polypeptide is Upregulated in ccRCC and is Involved
  • SCDl mRNA and SCDl polypeptide expression also were determined in established ccRCC cell lines and normal renal epithelial cells. SCDl mRNA levels and SCDl polypeptide levels were elevated in established ccRCC cell lines when compared to normal renal epithelial cells ( Figures 1C and ID).
  • RWV366T is a newly established patient derived ccRCC cell line, whose patient and renal origins were validated by STR analysis and IHC for renal markers (data not shown).
  • the first lentiviral construct was designed to express an shRNA designated SCD780.
  • the sequence of SCD780 was as follows: 5'-CTACGGCTCTTTCTGATCATT-3' (SEQ ID NO: l).
  • the second lentiviral construct was designed to express an shRNA designated SCD1200.
  • the sequence of SCD1200 was as follows: 5 '-CGTCCTTATGACAAGAACATT-3 ' (SEQ ID NO:2).
  • a non-target lentiviral construct was designed as a control.
  • a proliferation assay was performed to determine if reduced SCD1 expression preferentially reduced the ability of established ccRCC cell lines to proliferate as compared to normal kidney cells.
  • Treatment of established ccRCC cell lines (Cakil and A498) with lentiviral constructs designed to express SCD780 or SCD1200 resulted in reduced proliferation as compared to the levels of proliferation observed with normal kidney cells (K359N and K360N) treated with the lentiviral constructs ( Figures 2B and 2C).
  • Example 2 - Oleic Acid Reverses Effects of Decreased SCD1 Expression in Tumor Cells
  • OA oleic acid
  • OA-BSA a cell culture stable form of OA conjugated to albumin from bovine serum
  • A939572 was dosed out in four ccRCC cell lines- Cakil, A498, Caki2, and ACHN, and demonstrated a significant dose-dependent decrease in proliferation at day 5 (IC50s of 65 nM, 50 nM, 65 nM, and 6 nM, respectively) (Figure 4A).
  • IC50s of 65 nM, 50 nM, 65 nM, and 6 nM, respectively
  • Figure 4A Molecular target specificity was confirmed by addition of OA-BSA to the growth inhibitory assay, with IC50 doses applied to all four cell lines versus DMSO+BSA control. Addition of OABSA prevented A939572 mediated growth inhibition which was comparable to control groups in all four cell lines ( Figure 4B).
  • Activating transcription factor 6 is a key bZIP transcription factor that mediates part of the UPR stress response.
  • ATF6 is proteolytically cleaved into the activated transcription factor allowing it to transcribe several downstream mediators in the ER stress response pathway including XBP1, BiP,
  • HSP90B1 heat shock protein 90kDa beta
  • CHOP CHOP
  • Tumor cells may therefore be prone to elevated levels of ER stress requiring the induction of protective factors such as SCD1 in order to preserve cell viability.
  • Targeting ER protective constituents presents another potential route for therapeutic intervention not only in ccRCC, but likely in other cancers as well.
  • both TKIs were dosed in combination with A939572 up to approximately the IC50 dose for each drug in the Cakil and the A498 cell lines. No synergy was noted in either Cakil or A498 cell proliferative responses with combinatorial treatment. Temsirolimus (Tern) when dosed out in the four ccRCC cell lines yielded a limited reduction in cell proliferation, and no dose response could be determined. Combinatorial treatments were therefore done using a fixed dose of Tern (O.
  • Colony formation assay of A498 cells grown in soft agar treated with mono and combination doses of 5nM A939572 and 5nM Tern reflected synergistic effects observed in combination growth assays performed in 2-D culture and provided the rationale for in vivo analysis of combinatorial therapy.
  • Athymic nude (nu/nu) mice bearing A498 ccRCC xenografts were treated with A939572 and Tern individually or in combination over the course of four weeks, and tumor volume (mrm) was recorded (Figure 6A).
  • A939572 and Tern monotherapy generated similar growth responses with approximately 20-30% reductions in tumor volume (vs. placebo control) being observed upon study completion, with values reaching statistical significance only within the last week of treatment.
  • the combination group yielded over a 60% decrease in tumor volume (vs. placebo control) by study completion with significant reductions recorded after approximately 1 week of treatment. All of the animals maintained a healthy weight throughout the course of the treatment ( Figure 6A), however those in both the A939572 and the Combo group exhibited increased blinking, and slight mucosal discharge from the eyes after the first week of treatment.
  • Phosphorylated mTOR was inspected as a marker for temsirolimus activity, and decreased expression was confirmed in both the Tem and the Combo groups as compared to the Placebo and A939572 groups.
  • ER stress was examined via western blot of total protein extractions prepared from randomly selected tumor tissue samples representing each treatment group, and resulting quantitative expression was normalized to respective Pactin controls.
  • Increased expression of CHOP was confirmed in all samples treated with A939572 (A939572 and Combo) (Figure 6C) confirming that inhibition of SCD1 in ccRCC contributes to ER stress in vivo.
  • Figure 6D A proposed mechanism is summarized in Figure 6D.
  • Temsirolimus has been previously reported to decrease SCD1 expression in breast cancer cells. Inhibition of mTOR in ccRCC could indirectly mediate ER stress through decrease of SCD1, thereby explaining our observations. No significant increase in CHOP expression was seen in any placebo samples, confirming specificity of ER stress induction as a result of drug treatment.
  • a number of cancer cell lines were tested to determine whether SCD1 protein expression correlates with growth inhibition of an SCD1 inhibitor in human cancer cell lines.
  • SNU449 liver cancer cells express SCDl protein and are growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 8).
  • An estimated IC 50 concentration occurred around 100 nM.
  • Sorafenib is FDA approved for liver cancer treatment and is effective between 1 - 10 micromolar concentrations.
  • A375 melanoma cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 9). An estimated IC50 concentration occurred around 50 nM. Mela 11 melanoma cells do not express SCDl and were not growth inhibited. Standard of care, Temodar, dose responsively inhibits growth in A375 cells but not Mela 11.
  • Caco2 and HT29 colon cancer cells express SCDl protein and are growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 10).
  • T24 and HT1376 bladder cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 11).
  • SCDl inhibitor A939572 (see Figure 11).
  • Cisplatin The standard of care for bladder cancer, Cisplatin, has minimal growth inhibitory effects on these two cell lines.
  • KTC3 thyroid cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 13). Taxol is growth inhibitory in KTC3 cells but has minimal growth inhibitory effects on FF1 cells.
  • A549 nonsmall cell lung cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 while Calu-1 lung cancer cells do not express SCDl and are not growth inhibited by A939572 (see Figure 14). Taxol is growth inhibitory in A549 but was not tested in Calu-1 cells.
  • OVCA420 and HOV TAX2 ovarian cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 while Calu-1 lung cancer cells do not express SCDl and were not growth inhibited by A939572 (see Figure 15).
  • Taxol is growth inhibitory in HOV Tax2 cells but was not tested in OVCA420 cells.
  • MCF-7 ER+/PR+
  • MDA-231 triple negative
  • T47D PR+
  • DU-145 and LNCAP prostate cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 17).

Abstract

This document provides methods and materials for treating cancers including renal cancer (e.g., renal cell carcinoma) as well as ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, and thyroid cancers and melanoma. For example, methods and material for using one or more inhibitors of an SCDl polypeptide to treat renal cell carcinoma (e.g., clear cell renal cell carcinoma (ccRCC)) or to increase the efficacy of a renal cell carcinoma treatment are provided. In addition, this document provides methods and materials for using elevated SCDl expression levels in diseased tissues as an indication that an SCDl inhibitor can be used as an appropriate therapeutic to ameliorate the disease.

Description

METHODS AND MATERIALS FOR TREATING CANCER
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application Serial No.
61/607,961, filed on March 7, 2012, which is incorporated by reference in its entirety herein.
STATEMENT AS TO FEDERALLY FUNDED RESEARCH
This invention was made with government support under CA104505, CA136665, and CA104505-05 SI awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND
1. Technical Field
This document relates to methods and materials involved in treating cancer, for example, renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma. For example, this document provides methods and material for using one or more inhibitors of a stearoyl-Coenzyme A desaturase 1 (SCDl) polypeptide to treat cancer.
2. Background Information
The incidence and deaths caused by renal cell carcinoma are increasing in the United States. Indeed, mortality from renal cell carcinoma has increased over 37% since 1950.
SUMMARY
This document provides methods and materials for treating cancer, for example, renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma. For example, this document provides methods and material for using one or more inhibitors of an SCDl polypeptide to treat renal cell carcinoma (e.g., clear cell renal cell carcinoma (ccRCC)) or to increase the efficacy of a renal cell carcinoma treatment. As described herein, SCDl polypeptides are
overexpressed in certain cancer cells and are involved in the survival or proliferation of cancer cells. For example, reducing expression of renal cell carcinoma cells can result in reduced proliferation of renal cell carcinoma cells with minimal or no reduction in proliferation of normal kidney cells. In some cases, one or more inhibitors of an SCDl polypeptide can be used to reduce the number of cancer cells within a mammal (e.g., a human). In some cases, one or more inhibitors of an SCDl polypeptide can be used to increase the efficacy of a cancer treatment. For example, one or more inhibitors of an SCDl polypeptide can be used to increase the efficacy of a renal cell carcinoma treatment (e.g., treatment with Nexavar®, Sutent®, Torisel®, Afmitor®, and interleukin-2).
In general, one aspect of this document features a method for reducing the number of renal cell carcinoma cells within a mammal. The method comprises, or consists essentially of, administering, to the mammal, an inhibitor of an SCDl polypeptide under conditions wherein the number of viable renal cell carcinoma cells present within the mammal is reduced. The mammal can be a human. The administration can be an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration. The inhibitor can be A939572, MK-8245, CVT-11127, MF-152, or HYR-061. In another embodiment, one or more inhibitors of an SCDl polypeptide can be administered with one or more inhibitors of a mTor polypeptide. Non-limiting examples of such inhibitors include sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RADOOl), and ridaforolimus (AP-23573).
In another aspect, this document features a method for reducing the number of renal cell carcinoma cells within a mammal. The method comprises, or consists essentially of, administering, to the mammal, a composition under conditions wherein the number of viable renal cell carcinoma cells present within the mammal is reduced, wherein the composition comprises the ability to reduce SCDl mRNA expression or SCDl polypeptide expression. The mammal can be a human. The administration can be an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration. The composition can comprise a nucleic acid construct having the ability to express an shRNA directed against SCDl nucleic acid.
In another aspect, this document features a method for reducing the number of cancer cells overexpressing an SCDl polypeptide within a mammal. The method comprises, or consists essentially of, administering, to the mammal, an inhibitor of an SCDl polypeptide under conditions wherein the number of viable cancer cells overexpressing an SCDl polypeptide present within the mammal is reduced. Non- limiting examples of cancers include renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
In another aspect, this document features a method for reducing the number of cancer cells overexpressing an SCDl polypeptide within a mammal. The method comprises, or consists essentially of, administering, to the mammal, a composition under conditions wherein the number of viable cancer cells overexpressing an SCDl polypeptide present within the mammal is reduced, wherein the composition comprises the ability to reduce SCDl mRNA expression or SCDl polypeptide expression. Non- limiting examples of cancers include renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
In another aspect, this document features a method for identifying a mammal having cancer cells responsive to treatment with an inhibitor of an SCDl polypeptide. The method comprises, or consists essentially of, (a) detecting the presence of cancer cells expressing an elevated level of an SCDl mRNA or an SCDl polypeptide, and (b) classifying the mammal has having cancer cells responsive to treatment with the inhibitor of an SCDl polypeptide. The method can comprise measuring SCDl mRNA expression using real time PCR. The method can comprise measuring SCDl polypeptide expression using an immunohistochemical technique. The method can comprise measuring SCDl polypeptide expression using a Western blot analysis. Non- limiting examples of cancers include renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma.
In a further aspect, this document features a method for reducing the number of cancer cells within a mammal. The method comprises, or consists essentially of, administering, to said mammal, an inhibitor of an SCDl polypeptide and an inhibitor of an mTor polypeptide under conditions wherein the number of viable cancer cells present within said mammal is reduced. In some cases, the inhibitor of an mTor polypeptide can be sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RAD001), or ridaforolimus (AP-23573). In certain cases, the mammal is a human. In some cases, the administration is an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration. In some cases, the inhibitor of an SCDl polypeptide is A939572, MK- 8245, CVT-11127, MF-152, or HYR-061. Non-limiting examples of cancer cells include one or more of ovarian cancer, breast cancer, prostate cancer, colon cancer, renal cancer, pancreatic cancer, bladder cancer, liver cancer, lung cancer, thyroid cancer, and melanoma.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
DESCRIPTION OF THE DRAWINGS
Figure 1A is a graph plotting SCD1 mRNA levels in ccRCC tissue and matched normal tissue across stages I-IV. Figure IB contains photographs of representative ccRCC tissue and matched normal tissue stained for SCD1 polypeptide expression. Figure 1C is a graph plotting SCD1 mRNA levels in normal renal epithelial cell lines (347N, 355N, 359N, 360N, 365N, and 366N) versus ccRCC cell lines. Figure ID contains photographs of a Western blot analysis of SCD1 polypeptide expression by normal cell lines and ccRCC cell lines.
Figures 2A and 2D shows the knockdown of SCD1 in ccRCC as shown by decrease in both (A) mRNA and (D) protein expression using two separate lentiviral constructs shSCD780 and shSCD1200. Figures 2B and 2C show proliferation in (B) A498 and Cakil ccRCC cell lines and (C) NRE samples of NT versus shSCD lentiviral infected cells. Figure 2D contains photographs of an immunoblot for Poly-ADP ribose polymerase (PARP) cleavage and SCD1 expression in A498 and Cakil cell lines. Figure 3. Anti-pro liferative and apoptotic induction via loss of SCDl expression can be rescued with addition of oleic acid (OA-BSA). Figure 3A is a bar graph showing proliferation for SCDl and PARP cleavage in Cakil and A498 NT versus shSCD with or without OA-BSA supplementation. Figure 3B contains photographs of a Western blot analysis for SCDl and PARP cleavage in Cakil and A498 NT versus shSCD with or without OA-BSA supplementation. Figure 3C contains photographs of a phase-contrast microscopy representative ccRCC cell (Cakil) confluence at day 5 of proliferation assay with different treatment conditions.
Figure 4. Treatment of ccRCC cells with a small molecule SCDl inhibitor, A939572, inhibits cell growth and induces apoptosis. Figure 4A is a line graph showing cell proliferative response to dose out of A939572 in Cakil, A498, Caki2, and ACFiN ccRCC cell lines. Figure 4B is a bar graph displaying ccRCC proliferation rescue with OABSA in A939572 treated ccRCC cell lines. Figure 4C contains photographs of a Western blot analysis for PARP cleavage in A939572 treated vs. control, as well as OA- BSA rescue in ccRCC cell lines. Figure 4D contains representative phase contrast images of A939572 treated ccRCC cells (A498) +/-OA-BSA rescue at day 5.
Figure 5. Inhibition of SCDl activity in ccRCC induces cell death mediated by endoplasmic reticulum stress response. Figure 5A contains photographs of a Western blot analysis for expression of ER stress markers: BiP, CHOP, and spliced XBP1 in response to A939572 treatment or lentiviral silencing of SCDl in Cakil and A498. Figure 5B provides bar graphs showing QPCR analysis of ER stress gene expression in Cakil and A498 cells treated with A939572 or shSCD lentivirus +/-OA-BSA rescue. Figures 5C provides bar graphs showing relative luciferase activity of ER stress p5xATF6-GL3 (UPR) luciferase reporter transfected in Cakil and A498 cells treated with A939572 or shSCD lentivirus +/-OA-BSA supplementation.
Figure 6. Treatment of ccRCC cells with SCDl inhibitor in combination with the mTOR inhibitor Temsirolimus synergistically inhibits tumor cell growth in vivo. Figures 6A is a line graph illustrating in vivo tumor growth analysis and animal weight of A498 ccRCC subcutaneous xenografts in female athymic nude mice treated with A939572 and Temsirolimus alone or in combination versus placebo control (n=10 per group). Figure 6B contains photographs of IHC of tissue harvested from treatment groups stained for Ki67 and CC3 (quantitated by N-score), CD31 (quantitated by I-score), and phospho- mTOR (quantitated by H-score). Average group scores +/- the standard error are reported for each stain. Figure 6C contains photographs of Western blot and quantitation of CHOP expression in all four treatment groups. Figure 6D is an illustration of proposed SCD1 activity in ccRCC model: inhibition of SCD 1 blocks desaturation of SFA resulting in an accumulation of SFA species which trigger the ER stress response.
Figures 7A-D are line graphs comparing cell number to dose of A939572 or Gemicitabine in MiaPaca and pancreatic cancer cells. Figure 7E contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 8A-B are line graphs comparing cell number to dose of A939572 or
Sorafenib in SNU449 liver cancer cells. Figure 8C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 9A-C are line graphs comparing cell number to dose of A939572 or Temodar in A375 AND Mela 11 melanoma cancer cells. Figure 9D contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 10A-B are line graphs comparing cell number to dose of A939572 or Capecitabine in CaCo2 and HT29 colon cancer cells. Figure IOC contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 11A-B are line graphs comparing cell number to dose of A939572 or cisplatin in T24 and HT1276 bladder cancer cells. Figure 11C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 12A-B are line graphs comparing cell number to dose of A939572 or cisplatin in BCJ4T bladder cancer cells. Figure 12C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 13A-B are line graphs comparing cell number to dose of A939572 or
Taxol in KTC3 and FF1 anaplastic thyroid cancer cells. Figure 13C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 14A-B are line graphs comparing cell number to dose of A939572 or Taxol in A549 and CaLu-1 lung cancer cells. Figure 14C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression. Figures 15A-B are line graphs comparing cell number to dose of A939572 or Taxol in OVCA420 and HOVTax2res ovarian cancer cells. Figure 15C contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figure 16A is a line graph comparing cell number to dose of A939572 in MCF-7 (ER+/PR+), MDA-231 (triple negative) and T47D (PR+) breast cancer cells. Figure 16B contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figure 17A is a line graph comparing cell number to dose of A939572 in DU-145 prostate cancer cells. Figure 17B contains photographs of Western Blot and quantitation of SCDl and beta-actin expression.
Figure 18A is a bar graph illustrating SCD1 protein expression in various cancer cell lines. Figure 18B contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figure 19A is a bar graph illustrating SCD1 protein expression in various cancer cell lines. Figure 19B contains photographs of Western Blot and quantitation of SCD1 and beta-actin expression.
Figures 20-22 provide structures for exemplary SCD1 inhibitors.
DETAILED DESCRIPTION
This document provides methods and materials for treating cancer, for example, for example, renal cell carcinoma, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, thyroid cancers, and melanoma. In some embodiments, this document provides methods and material for using one or more inhibitors of an SCD1 polypeptide to treat cancer (e.g., clear cell renal cell carcinoma (ccRCC)) or to increase the efficacy of a cancer treatment.
As described herein, one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be administered to a mammal (e.g., a human) having cancer (e.g., renal cancer) under conditions wherein the number of cancer cells within the mammal is reduced. In some embodiments, one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be administered to a mammal (e.g., a human) having renal cancer (e.g., ccRCC) under conditions wherein the number of renal cancer cells within the mammal is reduced.
An SCDl polypeptide can be a human SCDl polypeptide having the amino acid sequence set forth in GenBank® Accession No. 000767 (GI No. 21431730) or a human SCDl polypeptide encoded by nucleic acid having the nucleic acid sequence set forth in GenBank® Accession No. AF097514.1 (GI No. 4808600). Examples of inhibitors of an SCDl polypeptide include, without limitation, inhibitory anti-SCDl polypeptide antibodies, siRNA molecules, shRNA molecules, nucleic acid vectors designed to express siRNA or shRNA molecules, anti-sense molecules, and small molecule antagonists such as A939572 (Biofine International Inc., Urvashi et al, Mol. Cancer Res., 9: 1551 (2011); Bristol-Myers Squibb R&D, Roongta et al., Mol. Cancer Res., 9(11): 1551-61 (2011)), MK-8245 (Merck Research Laboratories, Oballa et al, J. Med. Chem., 54(14):5082-96 (2011)), CVT-11127, MF-152 (Merck, Li et al, Bioorganic & Medicinal Chemistry Letters, 19:5214 (2009)), LCF369, CVT-11,563, CVT-12,012, DSR-4029, and GSK993 (Uto et al, Eur. J. Med. Chem., 45:4788-4796 (2010)), MF-438 (Leger, S. et al, Bioorg Med Chem Lett. 20(2):499-502 (2010)), and HYR-061
(Medchem Express, Koltun et al., Bioorganic & Medicinal Chemistry Letters,
19(7):2048-2052 (2009), and Xin et al., Bioorganic & Medicinal Chemistry Letters, 18(15):4298-4302 (2008)). In some cases, an inhibitor of an SCDl polypeptide can be an inhibitor described elsewhere (Igal, Carcinogenesis, 31(9): 1509-1515 (2010); Oballa, J. Med. Chem., 54:5082-5096 (2011); Li et al., Bioorganic & Medicinal Chemistry Letters, 19:5214-5217 (2009); Uto et al, Eur. J. Med. Chem., 46: 1892-1896 (2011); Uto et al, Eur. J. Med. Chem., 45:4788-4796 (2010); Liu, G. Expert Opin Ter Pat, 19(9): 1169-91 (2009); Powell, D.A., Bioorg Med Chem Lett. 20(22):6366-9 (2010), Mason P, et al, PLoS ONE 7(3): 33823 (2012), and Roongta et al, Mol. Cancer Res., 9: 1551-1561 (2011)).
Cancers that may be treated by an inhibitor of an SCDl polypeptide,
compositions and methods described herein include, but are not limited to, the following:
Breast cancers, including, for example ER+ breast cancer, ER" breast cancer, her2" breast cancer, her2+ breast cancer, stromal tumors such as fibroadenomas, phyllodes tumors, and sarcomas, and epithelial tumors such as large duct papillomas; carcinomas of the breast including in situ (noninvasive) carcinoma that includes ductal carcinoma in situ (including Paget's disease) and lobular carcinoma in situ, and invasive (infiltrating) carcinoma including, but not limited to, invasive ductal carcinoma, invasive lobular carcinoma, medullary carcinoma, colloid (mucinous) carcinoma, tubular carcinoma, and invasive papillary carcinoma; and miscellaneous malignant neoplasms. Further examples of breast cancers can include luminal A, luminal B, basal A, basal B, and triple negative breast cancer, which is estrogen receptor negative (ER~), progesterone receptor negative, and her2 negative (her2~). In some embodiments, the breast cancer may have a high risk Oncotype score;
lung cancers, including, for example, bronchogenic carcinoma, e.g., squamous cell, undifferentiated small cell, undifferentiated large cell, and adenocarcinoma; alveolar and bronchiolar carcinoma; bronchial adenoma;
sarcoma; lymphoma; chondromatous hamartoma; and mesothelioma;
genitourinary tract cancers, including, for example, cancers of the kidney, e.g., adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, and leukemia; cancers of the bladder and urethra, e.g., squamous cell carcinoma, transitional cell carcinoma, and adenocarcinoma; cancers of the prostate, e.g., adenocarcinoma, and sarcoma; cancer of the testis, e.g., seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, and lipoma;
liver cancers, including, for example, hepatoma, e.g., hepatocellular carcinoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hepatocellular adenoma; and hemangioma;
gynecological cancers, including, for example, cancers of the uterus, e.g., endometrial carcinoma; cancers of the cervix, e.g., cervical carcinoma, and pre tumor cervical dysplasia; cancers of the ovaries, e.g., ovarian carcinoma, including serous cystadenocarcinoma, epithelial cancer, mucinous
cystadenocarcinoma, unclassified carcinoma, granulosa thecal cell tumors, Sertoli Leydig cell tumors, dysgerminoma, and malignant teratoma; cancers of the vulva, e.g., squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, and melanoma; cancers of the vagina, e.g., clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma, and embryonal rhabdomyosarcoma; and cancers of the fallopian tubes, e.g., carcinoma;
skin cancers, including, for example, malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and
adrenal gland cancers, including, for example, neuroblastoma.
In some cases, one or more (e.g., one, two, three, four, or more) inhibitors of an SCDl polypeptide can be used as described herein to treat cancer, including renal cancer, ovarian, breast, prostate, colon, pancreatic, bladder, liver, lung, and thyroid cancers as well as melanoma.
For example, a human having cancer can be administered one or more inhibitors of an SCDl polypeptide under conditions that result in reduced tumor size or stable disease. In some cases, one or more (e.g., one, two, three, four, or more) inhibitors of an SCDl polypeptide can be used as described herein to increase the efficacy of a cancer treatment. In some embodiments (e.g., when compositions comprising one or more (e.g., one, two, three, four, or more) inhibitors of an SCDl polypeptide are administered in conjunction with another anticancer agent), one can create a synergistic effect among the agents administered and thereby improve the outcome for a patient. In some
embodiments, one or more (e.g., one, two, three, four, or more) inhibitors of an SCDl polypeptide (or a pharmaceutically acceptable salt form thereof) can be administered in combination with (i.e., before, during, or after) administration of a pain relief agent (e.g., a nonsteroidal anti-inflammatory drug such as celecoxib or rofecoxib), an antinausea agent, or an additional anticancer agent (e.g., paclitaxel, docetaxel, doxorubicin, daunorubicin, epirubicin, fluorouracil, melphalan, cis-platin, carboplatin,
cyclophosphamide, mitomycin, methotrexate, mitoxantrone, vinblastine, vincristine, ifosfamide, teniposide, etoposide, bleomycin, leucovorin, taxol, herceptin, avastin, cytarabine, dactinomycin, interferon alpha, streptozocin, prednisolone, irinotecan, sulindac, 5 -fluorouracil, capecitabine, oxaliplatin/5 FU, abiraterone, letrozole,
5aza/romidepsin, or procarbazine). In certain embodiments, the anticancer agent is paclitaxel or docetaxel. In other embodiments, the anticancer agent is cisplatin or irinotecan.
For example, a human having ccRCC can be administered one or more inhibitors of an SCD1 polypeptide under conditions that result in reduced tumor size or stable disease. In some cases, one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be used as described herein to increase the efficacy of a renal cell carcinoma treatment. Examples of such renal cell carcinoma treatments include, without limitation, treatment with Nexavar®, Sutent®, Torisel®, Afinitor®, or interleukin-2.
In some cases, one or more (e.g., one, two, three, four, or more) inhibitors of an SCD1 polypeptide can be used as described herein can be used in combination with one or more (e.g., one, two, three, four, or more) inhibitors of mammailian target of rapamycin (mTor) polypeptide. Non-limiting examples of mTor inhibitors include:
sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RAD001),
ridaforolimus (AP-23573).
Accordingly, provided herein is a method for reducing the number of cancer cells within a mammal, wherein the method comprises administering, to the mammal, an inhibitor of an SCD1 polypeptide and an inhibitor of an mTor polypeptide under conditions wherein the number of viable cancer cells present within said mammal is reduced. In some embodiments, the one or more mTor inhibitor can include a standard of care drug for a particular cancer cell type. For example, an SCD1 inhibitor can be administered with pacliltaxel and/or platin (cisplatin, carboplatin, or oxaliplatin) for the treatment of ovarian cancer. In some embodiments, the following standard of care drugs can be combined with an SCD1 inhibitor for the following cancers:
Lung - paclitaxel
Colon - capecitabine
Breast
Metastatic breast - capecitabine, paclitaxel, and/or gemcitabine
Hormonally responsive breast - aromatase inhibitors such as letrazole and/or antiestrogens such as tamoxifen
HER2 positive - Herceptin
Melanoma - temodar, and/or BRAF inhibitors Prostate - abiraterone
Bladder - gemcitabine and/or paclitaxel
Thyroid - paclitaxel and/or cisplatin
Pancreatic - gemcitabine
Liver - sorafanib
In some embodiments, the combination of one or more inhibitors of an SCD1 polypeptide and one or more inhibitors of mTor exhibit a synergistic response. In some embodiments, the one or more inhibitors of an SCD1 polypeptide can be administered before, during, or after administration of the one or more inhibitors of mTor.
An inhibitor of an SCD1 polypeptide can also be administered to a subject in combination with surgical methods to treat cancers, e.g., resection of tumors. The inhibitor can be administered to the individual prior to, during, or after the surgery. The inhibitor can be administered parenterally, intravenous or injected into the tumor or surrounding area after tumor removal.
Typically, one or more of the inhibitors of an SCD1 polypeptide provided herein can be formulated into a pharmaceutical composition that can be administered to a mammal (e.g., rat, dog, horse, cat, mouse, rabbit, pig, cow, monkey, or human). For example, A939572 or a pharmaceutically acceptable salt thereof can be in a
pharmaceutically acceptable carrier or diluent. A "pharmaceutically acceptable carrier" refers to any pharmaceutically acceptable solvent, suspending agent, or other pharmacologically inert vehicle. Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties. Typical pharmaceutically acceptable carriers include, without limitation, water, saline solutions, dimethyl sulfoxide, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose), fillers (e.g., lactose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g., starch or sodium starch glycolate), and wetting agents (e.g., sodium lauryl sulfate).
The term "pharmaceutically acceptable salt" refers to the relatively non-toxic, inorganic and organic acid addition salts of a compound provided herein. These salts can be prepared in situ during the final isolation and purification of a compound provided herein, or by separately reacting the compound in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, laurylsulphonate salts, and amino acid salts, and the like. (See, for example, Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66: 1-19.)
In some embodiments, a compound provided herein may contain one or more acidic functional groups and, thus, is capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases. The term "pharmaceutically acceptable salts" in these instances refers to the relatively non-toxic inorganic and organic base addition salts of a compound provided herein. These salts can likewise be prepared in situ during the final isolation and purification of the compound, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al, supra).
The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims. EXAMPLES
General Materials and Methods
Cell Culture
ccRCC cell lines: RWV366T and KD265T (16) (both stage IV ccRCC patient tissue derived), A498, Cakil, Caki2, and ACHN (ATCC) and K347N, K355N, K359N, K360N, K365N, and K366N normal renal tissue derived mortal cells (NRE) were cultured in DMEM medium (Cellgro) containing 5%FBS (Hyclone) and lxpenicillin- streptomycin (Invitrogen) at 37°C in humidified conditions with 5%C02.
Proliferation, Treatment, and Rescue Assays
Cells were plated (0.5 or lxl05/well) in 24-well plates for proliferation or treatment assays, in triplicate. Cells were trypsinized (0.25%) and counted using a Coulter Particle Counter at specified time intervals. For SCD1 rescue assays, oleic acid- albumin was added to media at 5 μΜ. Drug stocks were prepared in
DMSO. Monotherapeutic treatment identified drug dose-response. Combinatorial dosing ranged up to the IC50 for each inhibitor. Temsirolimus dosing was performed as described in the text. Soft agar cultures were prepared by diluting 2x growth medium 1 : 1 in 1.5% Seaplaque®GTG® agarose, with 500 cells/plate in 60mm culture dishes.
Colonies were stained with Giemsa (LabChem Inc.) and counted after 3 wks. Cell images were obtained with an 01ympusIX71 microscope at 20x magnification.
Lentivirus
MISSION shRNA pLKO. l constructs were used to make self-inactivating shRNA lentiviruses for human SCD1 (clones: NM_005063.3-1200slcl [shSCD1200],
NM_005063.3-780slcl [shSCD780]), and a non-target (NT) random scrambled sequence control (SHC002). Trans fection reagents Lipofectamine 2000 and ViraPower were used to generate lentiviruses using HEK293FT viral progenitor cells. ccRCC and NRE cells were incubated with lentivirus plus 5μg/mL polybrene for 24 hrs prior to clonal selection with Puromycin. Transfections and Luciferase Assays
For transient transfection, Cakil and A498 cells were transfected using
Lipofectamine 2000. Cells treated with DMSO vs. A939572 or infected using shSCD780 lentiviral constructs vs. NT control were harvested after 48hrs using Promega's Dual Luciferase assay kit per the manufacturer's protocol and luciferase activity was measured using a Veritas Luminometer; reported as relative luminescence. RNA Isolation and Quantitative PCR
An R Aqueous Midi Kit was utilized to extract and purify RNA from cell lines. Human tissue RNA was prepared using using TRIzol® per manufacturer's protocol followed by purification using the RNAqueous Midi Kit. The O.D. 260/280 ratio of the mRNA was at least 1.8 and the 18s/28s bands were verified on a 1% agarose gel. cDNA was prepared from purified RNA samples using High Capacity cDNA Reverse
Transcriptase Kit per manufacturer's instruction. TaqMan®Fast Universal PCR Master Mix and TaqMan®FAMTM dye-labeled probes including POLR2A (Hs00172187_ml) (normalization control), SCD1 (Hs01682761_ml), HSPA5 (Hs99999174_ml), CEBPp (CEBPB Hs00270923_sl), GADD45A (Hs00169255_ml), DDIT3 (Hs01090850_ml), and HERPUD1 (HsOl 124269 ml) were combined with prepared cDNA samples to analyze relative mRNA expression via qPCR. Fold change values were compared between normal and tumor, non-target scrambled lentiviral and target lentiviral infected, and DMSO vs. A939572 treated samples using the AACt method (Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc.
2008;3: 1101-8).
Gene Array Expression Analysis
Gene array expression analysis was performed using Affymetrix Human Genome U133 Plus 2.0 Array chip. The details of the data processing and methodology were previously described in (Tun HW, Marlow LA, von Roemeling CA, Cooper SJ, Kreinest P, Wu K, et al. Pathway signature and cellular differentiation in clear cell renal cell carcinoma. PLoS One. 2010;5:el0696). Gene expression data was deposited at Gene Expression Omnibus (Accession#GSE41485). Pathway analysis was performed using IP A (Ingenuity® Systems).
Western Blot Analysis
Protein extracts, electrophoresis, and membrane transfers were prepared as previously described (Copland JA, Marlow LA, Kurakata S, Fujiwara K, Wong AK, Kreinest PA, et al. Novel highaffinity PPARgamma agonist alone and in combination with paclitaxel inhibits human anaplastic thyroid carcinoma tumor growth via p21WAFl/CIPl . Oncogene. 2006;25:2304-17). Primary antibodies included SCD1, PARP, DDIT3, BiP, sXBPl, and β-actin. A Supersignal chemiluminescent kit was used to perform detection. IHC and ICC Analysis
Formalin fixed, paraffin-embedded tissue microarray (TMA) of patient ccRCC tumor and matched normal tissues and TMA of combinatorial in vivo mouse tumor tissue. The TMAs were mounted on slides from paraffin-embedded blocks according to IHC procedure and samples were blocked with Diluent that contained Background Reducing Components (Dakocytomation) for 30 min and then probed for SCD1, Ki67, Caspase-3, CD31, phospho-mTOR, DDIT3, and XBPl . ICC preparation and staining was performed as previously described (Cooper SJ, Von Roemeling CA, Kang KH, Marlow LA, Grebe SK, Menefee ME, et al. Reexpression of tumor suppressor, sFRPl, leads to antitumor synergy of combined HDAC and methyltransferase inhibitors in
Chemoresistant cancers. Mol Cancer Ther. 2012). Stain scoring was done using algorithms generated with Imagescope software created by a histologist. H-scores were calculated based upon signal intensity (0-3+) using the formula:
[(l+%xl)+(2+%x2)+(3+%x3)], intensity (I)-scores were calculated by dividing signal intensity by area, and nuclear (N)-scores were calculated by dividing % positive nuclei by total nuclei examined per area. Cases where insufficient tumor tissue presented were excluded from the study. 20x images were obtained using Scanscope XT and Imagescope software. RWV366T cell line validation was carried out as previously
described (Cooper SJ, Von Roemeling CA, Kang KH, Marlow LA, Grebe SK, Menefee ME, et al. Reexpression of tumor suppressor, sFRPl, leads to antitumor synergy of combined HDAC and methyltransferase inhibitors in chemoresistant cancers. Mol Cancer Ther. 2012).
In Vivo Analysis
A498 cells were subcutaneously implanted in athymic nu/nu mice at lxlO6 cells/mouse in 50%Matrigel. Tumors reached ~50 mm3 prior to treatment, which was carried out for 4 wks. A939572 was administered via oral feeding using strawberry flavored Kool-Aid® in sterilized H20 (0.2g/mL) vehicle at 30 mg/kg in a 50 μΐ dose twice daily/mouse. Temsirolimus was solubilized in 30% ethanol/saline and administered via intraperitoneal injection at 10 mg/kg in a 50 μΐ dose once every 72 hrs/mouse. Tumor volumes were calculated using the formula 0.5236(L*W*H) and body weight were measured every 3 days.
DNA isolation and STR Analysis
Genomic DNA was extracted from both RWV366T patient primary tissue and matching cell line using PurelinkTM Genomic DNA mini kit. Sixteen STR markers were PCR amplified using fluorescently labeled primers from ABI, and were analyzed using ABI 3130. Peak sizes were calculated versus a co-injected size standard using Gene Marker.
Statistical Analysis
Data values are presented as either percentage or fold change ±s.d. unless otherwise specified. Fold change values 1.5< are considered statistically significant. Treatment group comparisons were analyzed using two-tailed paired Student's t-test with p<0.05 being considered statistically significant. Statistically significant results are indicated by asterisk (*). Drug synergy statistics are indicated via combination index (CI) determined using CalcuSyn® as described in the text.
Example 1 - SCDl Polypeptide is Upregulated in ccRCC and is Involved
in Tumor Cell Survival
Normal kidney tissue and matched ccRCC tissue samples were obtained, and the levels of SCDl mRNA and SCDl polypeptide expression were determined. SCDl mRNA levels and SCDl polypeptide levels were elevated in matched ccRCC samples when compared to normal samples (Figures 1 A and IB).
The levels of SCDl mRNA and SCDl polypeptide expression also were determined in established ccRCC cell lines and normal renal epithelial cells. SCDl mRNA levels and SCDl polypeptide levels were elevated in established ccRCC cell lines when compared to normal renal epithelial cells (Figures 1C and ID). RWV366T is a newly established patient derived ccRCC cell line, whose patient and renal origins were validated by STR analysis and IHC for renal markers (data not shown).
To determine the involvement of SCD1 expression in renal cell carcinoma proliferation, two separate lentiviral constructs were designed to express shRNA molecules having the ability to reduce SCD1 expression. The first lentiviral construct was designed to express an shRNA designated SCD780. The sequence of SCD780 was as follows: 5'-CTACGGCTCTTTCTGATCATT-3' (SEQ ID NO: l). The second lentiviral construct was designed to express an shRNA designated SCD1200. The sequence of SCD1200 was as follows: 5 '-CGTCCTTATGACAAGAACATT-3 ' (SEQ ID NO:2). A non-target lentiviral construct was designed as a control.
Treatment of established ccRCC cell lines (Cakil and A498) with lentiviral constructs designed to express SCD780 or SCD1200 resulted in reduced SCD1 mRNA expression levels (Figure 2A) and reduced SCD1 polypeptide expression levels (Figure 2D). The reduction in SCD1 expression in ccRCC cells revealed the induction of apoptosis as demonstrated by poly ADP ribose polymerase (PARP) cleavage (Figure 2D). Treatment of normal renal epithelial cells (K359N and K360N) with lentiviral constructs designed to express SCD780 or SCD1200 resulted in reduced SCD1 mRNA expression levels (Figure 2A).
A proliferation assay was performed to determine if reduced SCD1 expression preferentially reduced the ability of established ccRCC cell lines to proliferate as compared to normal kidney cells. Treatment of established ccRCC cell lines (Cakil and A498) with lentiviral constructs designed to express SCD780 or SCD1200 resulted in reduced proliferation as compared to the levels of proliferation observed with normal kidney cells (K359N and K360N) treated with the lentiviral constructs (Figures 2B and 2C).
These results demonstrate that inhibitors of SCD1 can be used to reduce the number of ccRCC cells present within a mammal, while having little or no effect on normal kidney cells. These results also demonstrate that loss of SCD1 in ccRCC cells can lead to apoptotic programmed cell death.
Example 2 - Oleic Acid Reverses Effects of Decreased SCD1 Expression in Tumor Cells As oleic acid (OA) is the principle product of SCDl mediated SFA dehydrogenation, a cell culture stable form of OA conjugated to albumin from bovine serum (OA-BSA) was utilized to perform rescue experimentation in order to confirm that decreased tumor cell growth and induction of cell death was due to lentiviral mediated suppression of SCD 1.
Media alone and BSA supplemented media served as control groups. Proliferation assay of NT control versus shSCD780 infected Cakil and A498 cells cultured in media with or without OABSA were counted after five days. Both Cakil and A498 shSCD780 cells exhibited significant decreases in growth when compared to controls; however the addition of OA-BSA rescued the proliferative capacity of these cells to near control rates (Figure 3A). Notably, addition of OABSA to Cakil NT cells marginally enhanced proliferation (Figure 3 A). SCDl knockdown by lentiviral infection was confirmed at the protein level (Figure 3B). In addition to growth rescue, supplementation with OA-BSA also decreased shSCD780 induced apoptosis as demonstrated by reduction in PARP cleavage shown by western blot (Figure 3B). Representative phase contrast images of ccRCC cells for each group are shown in Figure 3C.
Example 3 - Small Molecule Inhibition of SCDl Induces ccRCC Cell Death
A939572 was dosed out in four ccRCC cell lines- Cakil, A498, Caki2, and ACHN, and demonstrated a significant dose-dependent decrease in proliferation at day 5 (IC50s of 65 nM, 50 nM, 65 nM, and 6 nM, respectively) (Figure 4A). Molecular target specificity was confirmed by addition of OA-BSA to the growth inhibitory assay, with IC50 doses applied to all four cell lines versus DMSO+BSA control. Addition of OABSA prevented A939572 mediated growth inhibition which was comparable to control groups in all four cell lines (Figure 4B). In congruity with previous experimentation examining SCDl lentiviral knockdown models, A939572 induced apoptosis confirmed by PARP cleavage via western blot analysis in all four cell lines (Figure 4C). Addition of OA-BSA blocked apoptosis noted by lack of PARP cleavage (Figure 4C). Representative phase contrast cell images (Figure 4D) demonstrate marked reduction in confluence of A939572 treated ccRCC cells (day 5), which reflects decreased proliferation and induction of cell death as a result of treatment. OA-BSA supplemented cells display no visible alterations in phenotype. Thus, we have identified a specific small molecule SCD1 inhibitor that induces apoptotic cell death that can be rescued by oleic acid.
Example 4 - Treatment of ccRCC Cells with A939572 Induces Endoplasmic
Reticulum Stress
In order to determine the mechanism of decreased proliferation and induction of cell death associated with loss of SCD1 activity in ccRCC cells, gene array analysis was performed with Cakil, A498, Caki2, and ACFIN ccRCC cells treated for 24 hours with a 75 nM dose of A939572 compared to DMSO control. Gene expression data was analyzed using the Ingenuity® Systems (IP A) program and revealed increased expression of ER stress response genes associated with UPR.
Western blot of Cakil and A498 cells for protein expression of key ER stress markers including BiP (heat shock 70kDa protein, GRP78), CHOP (DNA damage inducible transcript 3, DDIT3), and spliced-XBPl (x-box binding protein 1, s-XBPl) revealed amplified expression in both drug treated (75nM) and shSCD780 lentiviral knockdown cells after 48 hours (Figure 5 A), confirming induction of ER stress upon loss of SCD1 activity or expression as implicated by the gene array analysis.
In order to validate the specificity of ER stress induction mediated by both A939572 and shSCD780, rescue assays were performed using OA-BSA in Cakil and A498 cells qPCR analysis of five ER stress genes identified in the gene array including BiP, CHOP, HERPUD1 (homocysteine-inducible, ER-stress inducible, ubiquitin-like-1), GADD45a (DNA damage inducible transcript 1, DDITl), and ΟΕΒΡβ (CCAAT/enhancer binding protein beta) were examined. In A939572 (SCDi) treated Cakil and A498 cells, all five ER stress related genes were expressed at significantly increased levels compared to DMSO+BSA control, and this elevated expression could be blocked with the addition of OA-BSA (Figure 5B). In shSCD780 lentiviral infected Cakil and A498 cells, all of the ER stress genes were significantly induced in the Cakil shSCD780 sample and 4 of the 5 were significantly induced in the A498 shSCD780 sample. Similar to the drug treated cells, OA-BSA successfully blocked shSCD780 induced expression of the ER stress genes (Figure 5B). Activating transcription factor 6 (ATF6) is a key bZIP transcription factor that mediates part of the UPR stress response. Upon stress induction ATF6 is proteolytically cleaved into the activated transcription factor allowing it to transcribe several downstream mediators in the ER stress response pathway including XBP1, BiP,
HSP90B1 (heat shock protein 90kDa beta), and CHOP (23). Cakil and A498 cells transfected with an ATF6 luciferase reporter (p5xATF6-GL3) were treated with a 75 nM dose of A939572 or were infected with shSCD780. Resulting luminescence was measured after 48 hours. Inhibiting SCD1 genetically or pharmacologically resulted in significant enhancement of luciferase activity as compared to DMSO and NT controls with Cakil A939572+BSA, Cakil shSCD780+BSA, A498 A939572+BSA, and A498 shSCD780+BSA cells expressing fold change inductions of 1.6, 1.7, 3.8, and 2.0 respectively (Figure 5C). The addition of OA-BSA significantly reduced reporter activation in response to A939572 and shSCD780, thereby confirming specificity of ATF6 stimulation by loss of SCD1 activity in ccRCC cells. Collectively, these data are indicative that SCD1 inhibition activates the UPR stress response. Tumor cells may therefore be prone to elevated levels of ER stress requiring the induction of protective factors such as SCD1 in order to preserve cell viability. Targeting ER protective constituents presents another potential route for therapeutic intervention not only in ccRCC, but likely in other cancers as well.
Example 5 - Combination of A939572 with Temsirolimus Synergistically Enhances
Tumor Cell Death
In order to target ccRCC using a multifaceted approach, synergy was examined through application of combinatorial treatment utilizing A939572 in congruence with a current FDA approved regimen for ccRCC treatment. These included the TKIs pazopanib and sunitinib, as well as the mTOR inhibitor temsirolimus.
After identifying appropriate cell proliferative dose responses for pazopanib and sunitinib in four ccRCC cell lines including A498, Cakil, Caki2, and ACFIN, both TKIs were dosed in combination with A939572 up to approximately the IC50 dose for each drug in the Cakil and the A498 cell lines. No synergy was noted in either Cakil or A498 cell proliferative responses with combinatorial treatment. Temsirolimus (Tern) when dosed out in the four ccRCC cell lines yielded a limited reduction in cell proliferation, and no dose response could be determined. Combinatorial treatments were therefore done using a fixed dose of Tern (O. lnM, InM, and lOnM) combined with a dose range of A939572 up to the IC50 in Cakil, A498, Caki2 and ACHN cells. Both drugs in combination yielded very strong synergy in all four cell lines as indicated by the combination index (CI) determined using CalcuSyn® based on the Chou-Talalay Method where CI values >1 represent an antagonistic effect and values <1 represent synergy, with lower values signifying enhanced synergy. Colony formation assay of A498 cells grown in soft agar treated with mono and combination doses of 5nM A939572 and 5nM Tern reflected synergistic effects observed in combination growth assays performed in 2-D culture and provided the rationale for in vivo analysis of combinatorial therapy.
Athymic nude (nu/nu) mice bearing A498 ccRCC xenografts were treated with A939572 and Tern individually or in combination over the course of four weeks, and tumor volume (mrm) was recorded (Figure 6A). A939572 and Tern monotherapy generated similar growth responses with approximately 20-30% reductions in tumor volume (vs. placebo control) being observed upon study completion, with values reaching statistical significance only within the last week of treatment. The combination group yielded over a 60% decrease in tumor volume (vs. placebo control) by study completion with significant reductions recorded after approximately 1 week of treatment. All of the animals maintained a healthy weight throughout the course of the treatment (Figure 6A), however those in both the A939572 and the Combo group exhibited increased blinking, and slight mucosal discharge from the eyes after the first week of treatment.
IHC analysis of tumors resected from each treatment group was analyzed for proliferation, angiogenesis, and cell death (Figure 6B). All treatment groups (A939572, Tern, and Combo) when compared to the placebo control exhibited decreased
proliferation as marked by reduction in percent positivity of nuclear Ki67 staining, with the combinatorial group demonstrating the most significant decline. Angiogenesis as examined by intensity of microvessel density demonstrated a slight decrease in both the Tern and the Combo groups; however the cumulative scores were not considered significant. Cell death as examined by cleaved caspase-3 (CC3) demonstrated significant increases in the Combo group when compared to all groups. A moderate increase in cell death was also seen in the A939572 and Tem groups compared to the placebo.
Phosphorylated mTOR was inspected as a marker for temsirolimus activity, and decreased expression was confirmed in both the Tem and the Combo groups as compared to the Placebo and A939572 groups. ER stress was examined via western blot of total protein extractions prepared from randomly selected tumor tissue samples representing each treatment group, and resulting quantitative expression was normalized to respective Pactin controls. Increased expression of CHOP was confirmed in all samples treated with A939572 (A939572 and Combo) (Figure 6C) confirming that inhibition of SCD1 in ccRCC contributes to ER stress in vivo. A proposed mechanism is summarized in Figure 6D. Interestingly, samples in the Tem group also exhibited induction of CHOP, although to a lesser extent when compared to A939572 and Combo groups. Temsirolimus has been previously reported to decrease SCD1 expression in breast cancer cells. Inhibition of mTOR in ccRCC could indirectly mediate ER stress through decrease of SCD1, thereby explaining our observations. No significant increase in CHOP expression was seen in any placebo samples, confirming specificity of ER stress induction as a result of drug treatment.
Example 6 - Inhibition of SCD1 Polypeptide in Various Cancer Cell Lines
A number of cancer cell lines were tested to determine whether SCD1 protein expression correlates with growth inhibition of an SCD1 inhibitor in human cancer cell lines.
Pancreatic Cancer
Cells (20,000/ml) were plated in 12 well cell culture plates, allowed to attach and treated with the indicated dose of SCD1 inhibitor (A939753) or standard of care
(gemcitabine). Cell number was counted using a Coulter Counter. As show in Figure 7, the data are expressed as percent of DMSO control. Each value represents triplicates. Western analysis for SCD1 protein expression was performed on each cell line with beta- actin as the loading control. The data indicated that MiaPaca cells express SCD1 and were growth inhibited in a dose dependent fashion while Pane cells expressed very low levels of SCDl and were growth inhibited at only high levels of SCDl inhibitor, A939572.
The following cell lines were studied using a similar method as that described above.
Liver Cancer
It was found that SNU449 liver cancer cells express SCDl protein and are growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 8). An estimated IC50 concentration occurred around 100 nM. Sorafenib is FDA approved for liver cancer treatment and is effective between 1 - 10 micromolar concentrations.
Melanoma
A375 melanoma cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 9). An estimated IC50 concentration occurred around 50 nM. Mela 11 melanoma cells do not express SCDl and were not growth inhibited. Standard of care, Temodar, dose responsively inhibits growth in A375 cells but not Mela 11.
Colon Cancer
Caco2 and HT29 colon cancer cells express SCDl protein and are growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 10).
Bladder Cancer
T24 and HT1376 bladder cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 11). The standard of care for bladder cancer, Cisplatin, has minimal growth inhibitory effects on these two cell lines.
BCJ4T bladder cancer cells do not express SCDl protein and were not growth inhibited by the SCDl inhibitor, A939572 (see Figure 12). The standard of care for bladder cancer, Cisplatin, has minimal growth inhibitory effects on these this cell line. Anaplastic Thyroid Cancer
KTC3 thyroid cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 13). Taxol is growth inhibitory in KTC3 cells but has minimal growth inhibitory effects on FF1 cells.
Lung Cancer
A549 nonsmall cell lung cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 while Calu-1 lung cancer cells do not express SCDl and are not growth inhibited by A939572 (see Figure 14). Taxol is growth inhibitory in A549 but was not tested in Calu-1 cells.
Ovarian Cancer
OVCA420 and HOV TAX2 ovarian cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 while Calu-1 lung cancer cells do not express SCDl and were not growth inhibited by A939572 (see Figure 15). Taxol is growth inhibitory in HOV Tax2 cells but was not tested in OVCA420 cells.
Breast Cancer
MCF-7 (ER+/PR+), MDA-231 (triple negative) and T47D (PR+) breast cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 16).
Prostate Cancer
DU-145 and LNCAP prostate cancer cells express SCDl protein and were growth inhibited in a dose dependent fashion by the SCDl inhibitor, A939572 (see Figure 17).
Quantitation (relative) of SCDl protein expression in different cancer cell lines
As shown in Figure 18, Western analysis was performed on SCDl and beta actin. Quantitation was performed by first normalizing to each respective beta-actin followed by normalization to A498 control. SNU449 cells appeared to have the highest SCDl protein expression while BCJ4, Melal l and PANC had the lowest protein levels. Protein levels appear to correlate with growth inhibition of the SCDl inhibitor.
As shown in Figure 19, Western analysis was also performed on SCDl and beta actin. Quantitation was performed by first normalizing to each respective beta-actin followed by normalization to A498 control. LN Cap cells appeared to have the highest SCDl protein expression while Calul, FFl and KTC3 cells had the lowest protein levels. Protein levels appear to correlate with growth inhibition of the SCDl inhibitor.
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method for reducing the number of cancer cells within a mammal, wherein said method comprises administering, to said mammal, an inhibitor of an SCDl polypeptide and an inhibitor of an mTor polypeptide under conditions wherein the number of viable cancer cells present within said mammal is reduced.
2. The method of claim 1, wherein said inhibitor of an mTor polypeptide is sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RADOOl), or ridaforolimus (AP- 23573).
3. The method of claim 1, wherein said mammal is a human.
4. The method of claim 1, wherein said administration is an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration.
5. The method of claim 1, wherein said inhibitor of an SCDl polypeptide is
A939572, MK-8245, CVT-11127, MF-152, or HYR-061.
6. The method of claim 1, wherein said cancer cells comprise one or more of ovarian cancer, breast cancer, prostate cancer, colon cancer, renal cancer, pancreatic cancer, bladder cancer, liver cancer, lung cancer, thyroid cancer, and melanoma.
7. A method for reducing the number of renal cell carcinoma cells within a mammal, wherein said method comprises administering, to said mammal, an inhibitor of an SCDl polypeptide under conditions wherein the number of viable renal cell carcinoma cells present within said mammal is reduced.
8. The method of claim 7, wherein said mammal is a human.
9. The method of claim 7, wherein said administration is an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration.
10. The method of claim 7, wherein said inhibitor is A939572, MK-8245, CVT- 11127, MF-152, or HYR-061.
11. The method of claim 7, wherein said mammal is further administered an inhibitor of a mTor polypeptide.
12. The method of claim 11, wherein said inhibitor is sirolimus (RAPAMUNE®), temsirolimus (CCI-779), everolimus (RAD001), or ridaforolimus (AP-23573).
13. A method for reducing the number of renal cell carcinoma cells within a mammal, wherein said method comprises administering, to said mammal, a composition under conditions wherein the number of viable renal cell carcinoma cells present within said mammal is reduced, wherein said composition comprises the ability to reduce SCDl mR A expression or SCDl polypeptide expression.
14. The method of claim 13, wherein said mammal is a human.
15. The method of claim 13, wherein said administration is an intratumoral, oral, intraperitoneal, intramuscular, or intravenous administration.
16. The method of claim 13, wherein said composition comprises a nucleic acid construct having the ability to express an shRNA directed against SCDl nucleic acid.
17. A method for reducing the number of cancer cells overexpressing an SCDl polypeptide within a mammal, wherein said method comprises administering, to said mammal, an inhibitor of an SCDl polypeptide under conditions wherein the number of viable cancer cells overexpressing an SCDl polypeptide present within said mammal is reduced.
18. The method of claim 17, wherein the cancer is selected from the group consisting of: renal cell carcinoma, ovarian cancer, breast cancer, prostate cancer, colon cancer, pancreatic cancer, bladder cancer, liver cancer, lung cancer, thyroid cancer, and melanoma.
19. The method of claim 18, wherein the cancer is renal cell carcinoma.
20. A method for reducing the number of cancer cells overexpressing an SCDl polypeptide within a mammal, wherein said method comprises administering, to said mammal, a composition under conditions wherein the number of viable cancer cells overexpressing an SCDl polypeptide present within said mammal is reduced, wherein said composition comprises the ability to reduce SCDl mR A expression or SCDl polypeptide expression.
21. The method of claim 20, wherein the cancer is selected from the group consisting of: renal cell carcinoma, ovarian cancer, breast cancer, prostate cancer, colon cancer, pancreatic cancer, bladder cancer, liver cancer, lung cancer, thyroid cancer, and melanoma.
22. The method of claim 21, wherein the cancer is renal cell carcinoma.
23. A method for identifying a mammal having cancer cells responsive to treatment with an inhibitor of an SCDl polypeptide, wherein said method comprises:
(a) detecting the presence of cancer cells expressing an elevated level of an SCDl mRNA or an SCDl polypeptide, and
(b) classifying said mammal has having cancer cells responsive to treatment with said inhibitor of an SCDl polypeptide.
24. The method of claim 23, wherein said method comprises measuring SCDl mRNA expression using real time PCR.
25. The method of claim 23, wherein said method comprises measuring SCDl polypeptide expression using an immunohistochemical technique.
26. The method of claim 23, wherein said method comprises measuring SCDl polypeptide expression using a Western blot analysis.
PCT/US2013/029688 2012-03-07 2013-03-07 Methods and materials for treating cancer WO2013134546A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/383,385 US9233102B2 (en) 2012-03-07 2013-03-07 Methods and materials for treating cancer
US14/961,444 US20160152986A1 (en) 2012-03-07 2015-12-07 Methods and materials for treating cancer
US15/692,491 US10160972B2 (en) 2012-03-07 2017-08-31 Methods and materials for treating cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261607961P 2012-03-07 2012-03-07
US61/607,961 2012-03-07

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/383,385 A-371-Of-International US9233102B2 (en) 2012-03-07 2013-03-07 Methods and materials for treating cancer
US14/961,444 Continuation US20160152986A1 (en) 2012-03-07 2015-12-07 Methods and materials for treating cancer

Publications (1)

Publication Number Publication Date
WO2013134546A1 true WO2013134546A1 (en) 2013-09-12

Family

ID=49117356

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/029688 WO2013134546A1 (en) 2012-03-07 2013-03-07 Methods and materials for treating cancer

Country Status (2)

Country Link
US (3) US9233102B2 (en)
WO (1) WO2013134546A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10973810B2 (en) 2017-01-06 2021-04-13 Yumanity Therapeutics, Inc. Methods for the treatment of neurological disorders
US11873298B2 (en) 2017-10-24 2024-01-16 Janssen Pharmaceutica Nv Compounds and uses thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9233102B2 (en) 2012-03-07 2016-01-12 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
EP4180041A1 (en) 2014-08-07 2023-05-17 Mayo Foundation for Medical Education and Research Compounds and methods for treating cancer
EP3429567B1 (en) 2016-03-16 2024-01-10 The J. David Gladstone Institutes Methods and compositions for treating obesity and/or diabetes and for identifying candidate treatment agents
CA3054572A1 (en) 2017-02-28 2018-09-07 Mayo Foundation For Medical Education And Research Compounds and methods for treating cancer
WO2018170316A1 (en) * 2017-03-16 2018-09-20 The Board Of Trustees Of The Leland Stanford Junior University Methods of identifying myc-driven and lipogenesis-dependent neoplasms and methods of treating the same
US11243207B2 (en) 2018-03-29 2022-02-08 Mayo Foundation For Medical Education And Research Assessing and treating cancer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009060053A1 (en) * 2007-11-09 2009-05-14 Smithkline Beecham Corporation 1, 2, 3-triazole derivatives for use as stearoyl-coa desaturase inhibitors

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030064950A1 (en) 2001-02-23 2003-04-03 Ntambi James M. Methods for reducing body fat and increasing lean body mass by reducing stearoyl-CoA desaturase 1 activity
US20050256068A1 (en) 2001-05-18 2005-11-17 Sirna Therapeutics, Inc. RNA interference mediated inhibition of stearoyl-CoA desaturase (SCD) gene expression using short interfering nucleic acid (siNA)
US20050043256A1 (en) 2001-07-30 2005-02-24 Isis Pharmaceuticals, Inc. Antisense modulation of stearoyl-CoA desaturase expression
BRPI0412343A (en) 2003-07-30 2006-09-05 Xenon Pharmaceuticals Inc pyridazine derivatives and their use as therapeutic agents
JP4838128B2 (en) 2003-07-30 2011-12-14 ゼノン・ファーマシューティカルズ・インコーポレイテッド Piperazine derivatives and their use as therapeutic agents
US7759348B2 (en) 2003-07-30 2010-07-20 Xenon Pharmaceuticals Inc. Pyridazine derivatives and their use as therapeutic agents
WO2005024603A2 (en) 2003-09-10 2005-03-17 The Board Of Regents Of The University Of Texas System Methods for detecting, diagnosing and treating human renal cell carcinoma
DK1778837T3 (en) 2004-08-09 2008-08-04 Cellzome Ag Treatment of neurodegenerative diseases using SCD4 inhibitors
JP4958785B2 (en) 2004-09-20 2012-06-20 ゼノン・ファーマシューティカルズ・インコーポレイテッド Heterocyclic derivatives and their use as stearoyl-CoA desaturase inhibitors
MX2007003327A (en) 2004-09-20 2007-06-05 Xenon Pharmaceuticals Inc Heterocyclic derivatives and their use as mediators of stearoyl-coa desaturase.
AU2005286790A1 (en) 2004-09-20 2006-03-30 Xenon Pharmaceuticals Inc. Bicyclic heterocyclic derivatives and their use as inhibitors of stearoyl-CoA-desaturase (SCD)
CN101084207A (en) 2004-09-20 2007-12-05 泽农医药公司 Heterocyclic derivatives and their use as stearoyl-coa desaturase inhibitors
CA2580855A1 (en) 2004-09-20 2006-03-30 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as stearoyl-coa desaturase inhibitors
WO2006121250A1 (en) 2005-05-10 2006-11-16 Amorepacific Corporation Pharmaceutical composition for the inhibition of lipogenesis comprising a regulator of prp19 expression
CA2618646A1 (en) 2005-06-03 2007-11-15 Xenon Pharmaceuticals Inc. Aminothiazole derivatives as human stearoyl-coa desaturase inhibitors
WO2006130986A1 (en) 2005-06-09 2006-12-14 Merck Frosst Canada Ltd. Azacyclohexane derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
CN101203244A (en) 2005-07-01 2008-06-18 诺瓦提斯公司 Compositions of rennin inhibitor and insulin secernent or insulin sensitizer
JP2009501733A (en) 2005-07-20 2009-01-22 メルク フロスト カナダ リミテツド Heteroaromatic compounds as inhibitors of stearoyl coenzyme A delta-9 desaturase
JP4657053B2 (en) 2005-07-29 2011-03-23 株式会社アドバンテスト Timing generator and semiconductor test apparatus
WO2007056846A1 (en) 2005-11-15 2007-05-24 Merck Frosst Canada Ltd. Azacyclohexane derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
JP2009519984A (en) 2005-12-20 2009-05-21 メルク フロスト カナダ リミテツド Aromatic heterocyclic compounds as stearoyl-coenzyme δ-9 desaturase inhibitors
EP2027121A1 (en) 2006-05-22 2009-02-25 Merck Frosst Canada Ltd. Cyclic amine derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
WO2007143823A1 (en) 2006-06-12 2007-12-21 Merck Frosst Canada Ltd. Azetidine derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
WO2007143824A1 (en) 2006-06-13 2007-12-21 Merck Frosst Canada Ltd. Azacyclopentane derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
WO2008017161A1 (en) 2006-08-09 2008-02-14 Merck Frosst Canada Ltd. Azacycloalkane derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
WO2008020435A2 (en) 2006-08-15 2008-02-21 Quark Pharmaceuticals, Inc Compositions and methods for treatment of mood disorders
KR20090040478A (en) 2006-08-24 2009-04-24 노파르티스 아게 2-(pyrazin-2-yl)-thiazole and 2-(1h-pyraz0l-3-yl)-thiazole derivatives as well as related compounds as stearoyl-coa desaturase (scd) inhibitors for the treatment of metabolic, cardiovascular and other disorders
US7893066B2 (en) 2006-10-05 2011-02-22 Gilead Palo Alto, Inc. Pyridol[2,3-B]pyrazinones for use as stearoyl CoA desaturase inhibitors
US20100004245A1 (en) 2006-10-20 2010-01-07 Merck Frosst Canada Ltd. Azacycloalkane derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
EP2099755A2 (en) 2006-11-20 2009-09-16 Glenmark Pharmaceuticals S.A. Acetylene derivatives as stearoyl coa desaturase inhibitors
TW200826936A (en) 2006-12-01 2008-07-01 Merck Frosst Canada Ltd Azacycloalkane derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
US8258160B2 (en) 2006-12-20 2012-09-04 Novartis Ag SCD1 inhibitors triazole and tetrazole compounds
GB0625604D0 (en) 2006-12-21 2007-01-31 Smithkline Beecham Corp Compounds
GB0625594D0 (en) 2006-12-21 2007-01-31 Smithkline Beecham Corp Compounds
GB0625654D0 (en) 2006-12-21 2007-01-31 Smithkline Beecham Corp Compounds
AR064965A1 (en) 2007-01-26 2009-05-06 Merck Frosst Canada Inc DERIVATIVES OF AZACICLOALCANS AS INHIBITORS OF ESTEAROIL - COENZIMA A DELTA -9 DESATURASA
US7842696B2 (en) 2007-06-21 2010-11-30 Forest Laboratories Holdings Limited Piperazine derivatives as inhibitors of stearoyl-CoA desaturase
AU2008285255B2 (en) 2007-08-08 2012-09-27 Thesan Pharmaceuticals, Inc. Phenoxy-pyrrolidine derivative and its use and compositions
GB0722075D0 (en) 2007-11-09 2007-12-19 Smithkline Beecham Corp Compounds
AU2008336224A1 (en) 2007-12-11 2009-06-18 Merck Frosst Canada Ltd Novel heteroaromatic compounds as inhibitors of stearoyl-coenzyme A delta-9 desaturase
WO2009129625A1 (en) 2008-04-22 2009-10-29 Merck Frosst Canada Ltd. Novel substituted heteroaromatic compounds as inhibitors of stearoyl-coenzyme a delta-9 desaturase
PE20100156A1 (en) 2008-06-03 2010-02-23 Boehringer Ingelheim Int NAFLD TREATMENT
JP2012501975A (en) 2008-09-08 2012-01-26 メルク カナダ インコーポレイテッド Aromatic heterocycles as inhibitors of stearoyl coenzyme Aδ-9 desaturase
WO2010037225A1 (en) 2008-10-02 2010-04-08 Merck Frosst Canada Ltd. Heteroaromatic compounds as inhibitors of stearoyl-coenzyme a delta-9 desaturase
JP2012505839A (en) 2008-10-17 2012-03-08 メルク カナダ インコーポレイテッド Azetidine derivatives as inhibitors of stearoyl-coenzyme A delta-9 desaturase
US20100160323A1 (en) 2008-12-23 2010-06-24 Alexander Bischoff NOVEL PIPERAZINE DERIVATIVES AS INHIBITORS OF STEAROYL-CoA DESATURASE
TW201036975A (en) 2009-01-07 2010-10-16 Boehringer Ingelheim Int Treatment for diabetes in patients with inadequate glycemic control despite metformin therapy
AR075204A1 (en) 2009-01-29 2011-03-16 Boehringer Ingelheim Int DPP-4 INHIBITORS AND PHARMACEUTICAL COMPOSITIONS THAT INCLUDE THEM, USEFUL TO TREAT METABOLIC DISEASES IN PEDIATRIC PATIENTS, PARTICULARLY MELLITUS DIABETES TYPE 2
CN102387795A (en) 2009-02-13 2012-03-21 贝林格尔.英格海姆国际有限公司 Antidiabetic medications comprising a dpp-4 inhibitor (linagliptin) optionally in combination with other antidiabetics
ES2545864T3 (en) 2009-02-17 2015-09-16 Merck Canada Inc. New spiranic compounds useful as stearoyl-coenzyme A delta-9 desaturase inhibitors
WO2010094126A1 (en) 2009-02-23 2010-08-26 Merck Frosst Canada Ltd. Heterocyclic derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
WO2010108268A1 (en) 2009-03-23 2010-09-30 Merck Frosst Canada Ltd. Heterocyclic compounds as inhibitors of stearoyl-coenzyme a delta-9 desaturase
AU2010264763B2 (en) 2009-06-26 2015-07-23 Noviogendix Research B.V. Molecular markers in kidney cancer
WO2011011872A1 (en) 2009-07-28 2011-02-03 Merck Frosst Canada Ltd. Novel spiro compounds useful as inhibitors of stearoyl-coenzyme a delta-9 desaturase
EP2475367A1 (en) 2009-09-10 2012-07-18 Centre National De La Recherche Scientifique NOVEL INHIBITORS OF STEAROYL-CoA-DESATURASE-1 AND THEIR USES
BR112012007234A2 (en) 2009-10-02 2016-04-05 Boehringer Ingelheim Int pharmaceutical combination comprising dpp-4 inhibitor and metformin, as well as their use and preparation process
KR20230021159A (en) 2009-11-27 2023-02-13 베링거 인겔하임 인터내셔날 게엠베하 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
MX2014004426A (en) * 2011-10-15 2014-07-09 Genentech Inc Scd1 antagonists for treating cancer.
US9233102B2 (en) 2012-03-07 2016-01-12 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009060053A1 (en) * 2007-11-09 2009-05-14 Smithkline Beecham Corporation 1, 2, 3-triazole derivatives for use as stearoyl-coa desaturase inhibitors

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHAN S.: "Targeting the mammalian target ofrapamycin (mTOR): a new approach to treating cancer", BRITISH JOURNAL OF CANCER, vol. 91, no. 8, 2004, pages 1420 - 1424, XP002523239, DOI: doi:10.1038/sj.bjc.6602162 *
FRITZ, VANESSA ET AL.: "Abrogation of De novo Lipogenesis by Stearoyl-CoA Desaturase 1 Inhibition Interferes with Oncogenic Signaling and Blocks Prostate Cancer Progression in Mice", MOLECULAR CANCER THERAPEUTICS, vol. 9, no. 6, 2010, pages 1740 - 1754 *
LUYIMBAZI, DAVID ET AL.: "Rapamycin Regulates Stearoyl CoA Desaturase 1 Expression in Breast Cancer", MOLECULAR CANCER THERAPEUTICS, vol. 9, no. 10, 2010, pages 2770 - 2784 *
ROONGTA URVASHI V. ET AL.: "Cancer Cell Dependence on Unsaturated Fatty Acids Implicates Stearoyl-CoA Desaturase as a Target for Cancer Therapy", MOLECULAR CANCER RESEARCH, vol. 9, no. 11, 2011, pages 1551 - 1561, XP055072211, DOI: doi:10.1158/1541-7786.MCR-11-0126 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10973810B2 (en) 2017-01-06 2021-04-13 Yumanity Therapeutics, Inc. Methods for the treatment of neurological disorders
US11873298B2 (en) 2017-10-24 2024-01-16 Janssen Pharmaceutica Nv Compounds and uses thereof

Also Published As

Publication number Publication date
US10160972B2 (en) 2018-12-25
US20160152986A1 (en) 2016-06-02
US20150045418A1 (en) 2015-02-12
US20170362595A1 (en) 2017-12-21
US9233102B2 (en) 2016-01-12

Similar Documents

Publication Publication Date Title
US10160972B2 (en) Methods and materials for treating cancer
JP2022017495A (en) Combination therapy for treating cancer
JP2021185210A (en) Small molecule trail gene induction by normal and tumor cells as an anticancer therapy
Soo et al. Metformin synergizes 5-fluorouracil, epirubicin, and cyclophosphamide (FEC) combination therapy through impairing intracellular ATP production and DNA repair in breast cancer stem cells
US20130142784A1 (en) Method of treating tumor resistant to herceptin or paclitaxel using foxm1 inhibitors and detecting same
Classen et al. Autophagy induced by ionizing radiation promotes cell death over survival in human colorectal cancer cells
US8759097B2 (en) Inhibition of dynamin related protein 1 to promote cell death
EP2981252A1 (en) Methods of treating diseases characterized by excessive wnt signalling
Chung et al. Platinum‐based combination chemotherapy triggers cancer cell death through induction of BNIP3 and ROS, but not autophagy
EP3220908B1 (en) Compositions and methods for treating endometriosis
US9492450B2 (en) Inhibition of dynamin related protein 1 to promote cell death
Walter et al. Inhibition of MDM2 via Nutlin-3A: a potential therapeutic approach for pleural mesotheliomas with MDM2-induced inactivation of wild-type P53
Ge et al. Ubiquitin carboxyl-terminal hydrolase isozyme L5 inhibits human glioma cell migration and invasion via downregulating SNRPF
Wang et al. Sanguinarine induces apoptosis in osteosarcoma by attenuating the binding of STAT3 to the single‐stranded DNA‐binding protein 1 (SSBP1) promoter region
Li et al. Gracillin shows potential efficacy against non-small cell lung cancer through inhibiting the mTOR pathway
Kang et al. Rebamipide attenuates Helicobacter pylori CagA-induced self-renewal capacity via modulation of β-catenin signaling axis in gastric cancer-initiating cells
Xing et al. Genome‐wide gain‐of‐function screening identifies EZH2 mediating resistance to PI3Kα inhibitors in oesophageal squamous cell carcinoma
Muhammad et al. Defective transcription elongation in human cancers imposes targetable proteotoxic vulnerability
US20090281166A1 (en) Treatment of Cancer by Inhibition of HSP27
US20220047546A1 (en) Combination cancer therapies
US11103518B2 (en) Prostate cancer treatment via synergistic inhibition of aryl hydrocarbon receptor (AhR) and SRC
KR101900278B1 (en) Method for Screening a Composition for Preventing or Treating MYC-AP4 Activated Cancers
Lin et al. PF-429242 exhibits anticancer activity in hepatocellular carcinoma cells via FOXO1-dependent autophagic cell death and IGFBP1-dependent anti-survival signaling
Barua The prognostic and therapeutic value of XBP1 signalling in ER-positive breast cancer
Zhao et al. m6A/HOXA10-AS/ITGA6 axis aggravates oxidative resistance and malignant progression of laryngeal squamous cell carcinoma through regulating Notch and Keap1/Nrf2 pathways

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: 13758514

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14383385

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13758514

Country of ref document: EP

Kind code of ref document: A1