EP4277613A1 - Pharmazeutische zusammensetzungen zur behandlung von brustkrebs und verfahren zur verwendung davon - Google Patents

Pharmazeutische zusammensetzungen zur behandlung von brustkrebs und verfahren zur verwendung davon

Info

Publication number
EP4277613A1
EP4277613A1 EP22740195.7A EP22740195A EP4277613A1 EP 4277613 A1 EP4277613 A1 EP 4277613A1 EP 22740195 A EP22740195 A EP 22740195A EP 4277613 A1 EP4277613 A1 EP 4277613A1
Authority
EP
European Patent Office
Prior art keywords
breast cancer
positive
treating
subject
her2
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP22740195.7A
Other languages
English (en)
French (fr)
Inventor
Mitchell Steiner
Ramesh Narayanan
Sunjoo Ahn
James Dalton
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Tennessee Research Foundation
Original Assignee
University of Tennessee Research Foundation
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
Priority claimed from US17/151,108 external-priority patent/US20210228529A1/en
Application filed by University of Tennessee Research Foundation filed Critical University of Tennessee Research Foundation
Publication of EP4277613A1 publication Critical patent/EP4277613A1/de
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • This invention relates to the treatment of androgen receptor-positive breast cancer in a subject, for example in a male or a female subject.
  • this invention provides methods of: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of estrogen receptor (ER), progesterone receptor (PR), and/or human epidermal growth factor receptor 2 (HER2); i) treating a subject suffering from triple negative breast cancer (TNBC); j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH
  • Breast cancer is a disease that kills over 45,000 women each year in the United States alone. Over 180,000 new cases of breast cancer are diagnosed annually, and it is estimated that one in eight women will develop breast cancer. These numbers indicate that breast cancer is one of the most dangerous diseases facing women today. Breast cancer occurs in men as well, but at a much lower incidence. Cancer research has been unable to determine the cause of breast cancer and has not found a suitable method of therapy or prevention. [0003] Genotyping has long been used to screen women who may be genetically predisposed to developing breast cancer. It is another diagnostic or prognostic tool that can be used to determine the availability of therapies.
  • BRCA breast cancer susceptibility genes
  • SERMs tamoxifen in 1999 and raloxifene in 2007, were approved for the primary prevention of breast cancer in patient populations that are high risk based on family history and/or genotype considerations.
  • hysterectomy or prophylactic mastectomy was often considered in these patients as a more definite preventative.
  • talazoparib (Talzenna) and olaparib (Lynparza)
  • PARP poly ADP ribose polymerase
  • alpelisib an inhibitor of phosphatidylinositol-3-kinase (PI3K) with inhibitory activity predominantly against PI3K ⁇ was approved for patients possessing certain gain-of-function mutations in the gene encoding the catalytic ⁇ -subunit of PI3K (PIK3CA).
  • PI3K inhibition by alpelisib treatment has been shown to induce an increase in estrogen receptor (ER) transcription in breast cancer cells.
  • ER estrogen receptor
  • the combination of alpelisib and fulvestrant demonstrated increased antitumor activity compared to either treatment alone in xenograft models derived from ER-positive, PIK3CA mutated breast cancer cell lines.
  • PIK3CA mutations are present in about 30-40% of breast cancer tumors and most prevalent in ER-positive patients.
  • the standard of care currently includes screening the tumor for the expression levels of the hormone receptors, estrogen receptor (ER) and progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) kinase.
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2 human epidermal growth factor receptor 2
  • a woman diagnosed with breast cancer may be treated preliminarily with surgery, chemotherapy (optional in some cases), and radiation before targeted therapy is initiated.
  • Hormone receptor-positive breast cancers are susceptible to hormone therapies (also referred to as endocrine therapies) with selective estrogen receptor modulators or SERMs (e.g., tamoxifen, toremifene, raloxifene), aromatase inhibitors or AI’s (e.g., anastrozole, letrozole, exemestane), or selective estrogen receptor degraders or SERDs (e.g., fulvestrant).
  • SERMs selective estrogen receptor modulators
  • SERMs e.g., tamoxifen, toremifene, raloxifene
  • AI aromatase inhibitors or AI’s
  • SERDs selective estrogen receptor degraders or SERDs
  • Hormone therapies such as gonadotropin- releasing hormone (GnRH) agonists (typically used in pre- and peri-menopausal women) and aromatase inhibitors (AI) (typically used in post-menopausal women or together with GnRH agonists in pre- or peri-menopausal women) block production of estrogens in the body, whereas SERMs and SERDs block the proliferative action of estrogens on the breast cancer cells. While the prognosis of most early-stage ER-positive breast cancer patients is relatively good compared to non-hormonal cancers, adjuvant hormone therapy failures do occur resulting in recurrence, including distant metastases (i.e, advanced breast cancer).
  • GnRH gonadotropin- releasing hormone
  • AI aromatase inhibitors
  • SERMs and SERDs block the proliferative action of estrogens on the breast cancer cells. While the prognosis of most early-stage ER-positive breast cancer
  • Metastatic or advanced breast cancer is often still ER-positive and still dependent on the ER axis for growth.
  • the treatment of advanced breast cancer is rapidly evolving from the use of an endocrine monotherapy such as SERM or AI or fulvestrant, to combinations of an endocrine therapy with recently approved kinase inhibitors, including the cyclin-dependent kinase 4/6 (CDK 4/6) inhibitors (palbociclib (approved 2015), ribociclib (approved 2017), or abemaciclib (approved 2017), trilaciclib, lerociclib), or mechanistic target of rapamycin (mTOR) inhibitor (everolimus (approved 2012)).
  • CDK 4/6 cyclin-dependent kinase 4/6
  • mTOR mechanistic target of rapamycin
  • HER2-positive breast cancers are susceptible to HER2 kinase inhibitors (e.g., trastuzumab, lapatinib, neratinib, and tucatinib) and are generally used in metastatic disease.
  • HER2 kinase inhibitors e.g., trastuzumab, lapatinib, neratinib, and tucatinib
  • Anti-angiogenic therapy was also approved in metastatic disease, but the FDA removed this for bevacizumab in 2011. Despite these multiple tiers of targeted treatments, patients often have or develop refractory forms of breast cancer. Examples of refractory breast cancer include primary tumors which are triple-negative (lacking ER, PR, HER2), hormone resistant (SERM-, SERD-, or AI-resistant), or kinase inhibitor resistant (e.g., inhibitors of CDK 4/6, mTor, and/or HER2), or metastatic breast cancer tumors.
  • primary tumors which are triple-negative (lacking ER, PR, HER2), hormone resistant (SERM-, SERD-, or AI-resistant), or kinase inhibitor resistant (e.g., inhibitors of CDK 4/6, mTor, and/or HER2), or metastatic breast cancer tumors.
  • steroidal androgens biotransform into estrogens (via aromatase pathway), they increase cell proliferation and mammary carcinogenesis risk.
  • the steroidal androgen receptor agonists testosterone, fluoxymesterone, and calusterone were used in advanced breast cancer. These agents suffered from side effects such as excessive virilization, cross-reactivity with the estrogen receptor, and aromatization to estrogens.
  • the use of steroidal androgens in advanced breast cancer pre-dates the screening of breast cancers for hormone and kinase receptors. Recently, it was found that the AR is expressed in 50-90% of breast tumors, providing a mechanism to use androgens as targeted therapy for AR-positive breast cancers.
  • TNBC Triple Negative Breast Cancer
  • Topoisomerases I and II are normal host enzymes that are found in the nucleus of mammalian cells and are required for normal DNA replication and cellular division. The enzymes create and then repair single stranded nicks in cellular DNA. The nicks allow for the untangling and relaxation of supercoiled double stranded DNA, so that replication can proceed.
  • topoisomerase reseals the relaxed double helix. Topoisomerase activity is particularly increased in rapidly dividing and in cancer cells. If topoisomerase is inhibited, the accumulation of DNA breaks results in inhibition of DNA replication and cell death. It represents an appropriate, but nonselective target for anticancer therapy.
  • CBSI based inhibition of tubulin dynamics causes apoptotic cell death due to arrest in G2/M phase of mitosis.
  • Deng et al. have recently characterized sabizabulin in a variety of in vitro and in vivo models of TNBC and confirmed potent and high efficacy tumor growth inhibition in primary and metastatic tumors, and prevention of metastasis to distance organs including liver, lung, spleen, and brain in taxane-sensitive (MDA-MB-231) and taxane-resistant models of TNBC including patient-derived xenografts.
  • TNBC tumor necrosis factor 1
  • PD-L1 programmed death ligand 1
  • a novel approach to treating TNBC was approved in which antibodies blocking programmed death ligand 1 (PD-L1) were employed in PD-LI positive patients.
  • PD-L1 blocking programmed death ligand 1
  • One such antibody is atezolizumab (Tecentriq; approved for TNBC in 2019) and another is pembrolizumab (Keytruda; approved for TNBC in 2020).
  • AR has been considered anti-proliferative and beneficial in hormone receptor positive breast cancers.
  • TNBC data demonstrates that the presence of AR and androgen synthesizing enzymes is associated with lower proliferation, lower tumor grade, better overall survival, and more favorable clinical outcomes as compared to those patients with TNBC not expressing AR.
  • PSA prostate specific antigen
  • ER-positive breast cancers For example, continued treatment of ER-positive breast cancers with ER antagonists or aromatase inhibitors (AI) results in resistance due to mutations in the ER ligand binding domain (LBD).
  • ER antagonists or aromatase inhibitors AI
  • LBD ER ligand binding domain
  • Clinical studies have estimated that over 30% of breast cancers treated with tamoxifen become refractory and recur as a resistant cancer and over 40% of recurrent breast cancers express mutated ER.
  • Treatment emergent mutant ERs have escaped inhibition of the hormonal axis fail to respond to endocrine therapy and, consequently, these patients will need to be treated with chemotherapeutic agents.
  • Such cancers require new non- or less- toxic effective endocrine therapies.
  • SARMs are compounds which demonstrate AR-mediated tissue selective activity.
  • SARMs are non-aromatizable, generally demonstrate no activity at other steroidal receptors including ER and PR, and are non-virilizing. Further, SARMs may be beneficial in refractory breast cancer patients due to their hypermyoanabolic effects that should improve their tolerance of high-dose chemotherapy. Further, SARMs have beneficial osteoblastic and anti-osteoclastic effects in bones that may decrease the risk of metastasis to the bones or may decrease risk of osteoporosis during endocrine and/or chemotherapies.
  • New innovative approaches are urgently needed at both the basic science and clinical levels to develop compounds which are useful for: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR- positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH)
  • this invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a selective androgen receptor modulator (SARM) compound and a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor, wherein said SARM compound is represented by a structure of formula I:
  • X is a bond, O, CH 2 , NH, S, Se, PR, NO, or NR; G is O or S; T is OH, OR, -NHCOCH 3 , or NHCOR; R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F, CHF 2 , CF 3 , CF 2 CF 3 , aryl, phenyl, halogen, alkenyl, or OH; R 1 is CH 3 , CH 2 F, CHF 2 , CF 3 , CH 2 CH 3 , or CF 2 CF 3 ; R 2 is H, F, Cl, Br, I, CH 3 , CF 3 , OH, CN, NO 2 , NHCOCH 3 , NHCOCF 3 , NHCOR, alkyl, arylalkyl, OR, NH 2 , NHR, N(R) 2 , or SR; R 3 is H, F, Cl, Br,
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof, and a CDK 4/6 inhibitor
  • the CDK 4/6 inhibitor is palbociclib, ribociclib, trilaciclib, lerociclib or abemaciclib.
  • the CDK 4/6 inhibitor is palbociclib.
  • the CDK 4/6 inhibitor is abemaciclib.
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and palbociclib, [00021]
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and ribociclib
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and trilaciclib, [00023]
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and trilaciclib,
  • the breast cancer is an AR- positive breast cancer, ER-positive breast cancer, triple negative breast cancer, HER2-positive breast cancer, advanced breast cancer, refractory breast cancer, metastatic breast cancer, or breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), ribociclib (Kisqali), abemaciclib (Vorzenio), trilaciclib, lerociclib), alpelisib (Piqray) (an inhibitor of phosphatidylinositol-3- kinase subunit alpha (PI3K ⁇ )
  • SERM selective estrogen receptor modulator
  • the breast cancer has failed treatment with a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor.
  • CDK 4/6 cyclin-dependent kinase 4/6
  • the subject in the method of the invention is resistant or non-responsive to the CDK 4/6 inhibitor.
  • the CDK 4/6 inhibitor is palbociclib (Ibrance), ribociclib (Kisqali), trilaciclib, lerociclib or abemaciclib (Vorzenio).
  • the CDK 4/6 inhibitor is palbociclib (Ibrance).
  • the CDK 4/6 inhibitor is abemaciclib (Vorzenio).
  • the composition of the invention as described herein re- sensitizes said breast cancer to treatment with CDK 4/6 inhibitors.
  • the composition of the invention as described herein overcomes estrogen endocrine resistance.
  • the composition of the invention as described herein overcomes resistance to estrogen endocrine and CDK 4/6 inhibitor co-therapy.
  • the estrogen endocrine therapy includes at least one of tamoxifen, toremifene, raloxifene, exemestane, letrozole, anastrozole, and fulvestrant.
  • the CDK 4/6 inhibitor is at least one of palbociclib (Ibrance), ribociclib (Kisqali), trilaciclib, lerociclib, and abemaciclib (Vorzenio).
  • Figure 1A shows MDA-MB-231 cell expression of AR following transfection.
  • Figure 1B shows the IC 50 in AR-positive MDA-MB- 231 cells.
  • Figure 1C- Figure 1J show the effects of DHT, Formula IX, bicalutamide and the (R) enantiomer of Formula IX on percent (%) cell survival.
  • Figure 1C, Figure 1E, Figure 1G and Figure 1I cells were treated in charcoal stripped FBS.
  • Figure 1D, Figure 1F, Figure 1H and Figure 1J cells were treated in full serum).
  • Figure 2A- Figure 2H illustrate that DHT and Formula IX inhibit HCC-38 triple negative breast cancer cell growth.
  • Figure 2A shows HCC-38 cell expression of AR following transfection.
  • Figure 2B shows the IC 50 in AR-positive HCC-38 cells.
  • Figure 2A- Figure 2H show the effects of DHT, Formula IX and Bicalutamide on percent (%) cell survival.
  • Figure 2C, Figure 2E and Figure 2G cells were treated in charcoal stripped FBS.
  • Figure 2D, Figure 2F and Figure 2H cells were treated in full serum).
  • Figure 3A- Figure 3E illustrate that the effect of DHT and Formula IX on MDA- MB-231 cells was reversed by bicalutamide.
  • Figure 3A- Figure 3D show the effects of DHT or Formula IX in the presence or absence of bicalutamide, on percent (%) cell survival.
  • Figure 3A and Figure 3C cells were treated in charcoal stripped FBS.
  • Figure 3B and Figure 3D cells were treated in full serum).
  • Figure 3E shows IC 50 values in AR-positive cells in the presence or absence of pretreatment with bicalutamide.
  • Figure 4A- Figure 4Q illustrate that AR agonists inhibit triple negative breast cancer cell growth.
  • Figure 4A, Figure 4B, Figure 4E, Figure 4F, Figure 4G, Figure 4H, Figure 4K, Figure 4L, Figure 4M, Figure 4N, Figure 4O and Figure 4P show effect of AR agonists on percent (%) cell survival.
  • Figure 4C and Figure 4D show the effect of AR antagonist on percent (%) cell survival.
  • Figure 4I and Figure 4J show the effect of AR non-binder on percent (%) cell survival.
  • Figure 4A, Figure 4C, Figure 4E, Figure 4G, Figure 4I, Figure 4M and Figure 4O cells were treated in charcoal stripped FBS.
  • Figure 4B, Figure 4D, Figure 4F, Figure 4H, Figure 4J, Figure 4L, Figure 4N and Figure 4P cells were treated in full serum.
  • Figure 4Q shows EC 50 and IC 50 values in AR-positive cells.
  • Figure 5 illustrates that growth inhibitory ligands are AR agonists in MDA-MB-231 cells.
  • Figure 6A- Figure 6E illustrate that growth inhibitory effects in MDA-MB-231 cells are selective to AR.
  • Figure 6A and Figure 6B show the expression of ER ⁇ or ER ⁇ in MDA- MB-231 cells following transfection, respectively.
  • Figure 6C, Figure 6D and Figure 6E show the effects of estradiol (E2) or ICI 182,780 (ICI) on percent (%) cell survival.
  • E2 estradiol
  • ICI ICI 182,780
  • Figure 6C cells were treated in charcoal stripped serum.
  • Figure 6D and Figure 6E cells were treated in full serum.
  • Figure 7 shows DHT alters the morphology of MDA-MB-231 cells.
  • Figure 8 illustrates the effect of Formula VIII on steroid receptor transactivation (agonist mode).
  • Figure 9 depicts a dose response curve of PR activity (antagonist mode) for compound of Formula VIII, Formula IX, R-enantiomer of Formula IX and RU486.
  • FIG. 10A- Figure 10B demonstrate that SARM (Formula VIII) inhibits MDA-MB- 231-AR tumor growth.
  • Body weight (10A) and tumor size (10B) were measured for 35 days in intact female nude mice having 150-200 mm 3 tumors from MDA-MB-231-AR triple negative breast cancer cells and then orally administered vehicle ( ) or 30 mg/kg of Formula VIII (•).
  • Figure 11 demonstrates that SARM (Formula VIII) inhibits MDA-MB-231-AR tumor growth. Tumor size in mm 3 (left pane) and % change in tumor size (middle pane), as well as tumor weight (right pane) were measured after 35 days in intact female nude mice having 150-200 mm 3 tumors from MDA-MB-231-AR triple negative breast cancer cells and then receiving oral administration of vehicle or 30 mg/kg of Formula VIII.
  • Figure 12 demonstrates the morphology of MDA-MB-231 breast cancer cells stably transfected with AR (MDA-MB-231-AR cells).
  • AR agonists, DHT, Formula IX, and Formula VIII altered the morphology into a more anchored phenotype compared to vehicle, bicalutamide or an inactive isomer of Formula IX. This may be indicative of a less metastatic breast cancer phenotype.
  • Figure 13A- Figure 13C demonstrate binding and transactivation of the indicated ligands to HEK-293 (13A) or MDA-MB-231 (13B & 13C) cells.
  • DHT, Formula IX and Formula VIII are agonists of AR in breast cancer cells.
  • Figure 14 demonstrates anti-proliferative activity of DHT and SARMs in MDA-MB- 231 breast cancer cells stably transfected with AR.
  • MDA-MB-231 cells stably transfected with AR using lentivirus were treated with the indicated ligands for 6 days and the number of cells counted using Coulter counter.
  • Figure 15 presents microarray results showing that activated AR (AR activated by compound of Formula VIII) suppressed the expression of more genes than it induced in MDA- MB-231-AR xenograft breast cancer cells.
  • Figure 16 depicts validation of microarray results.
  • Figure 17 illustrates that Formula VIII inhibited the growth of MCF-7-AR triple positive xenograft.
  • Figure 18 presents inhibition of uterus weight gain in estrogen supplemented animals treated with Formula VIII, demonstrating the ability of a SARM to counteract estrogenic stimuli in vivo.
  • Figure 19 shows that the AR expression pattern in response to an AR-agonist (Formula VIII) is similar to that observed in prostate cancer cells.
  • Figure 20 depicts validation of microarray results.
  • Figure 21 demonstrates up-regulation of JNK phosphorylation in MCF7-AR tumors using Formula VIII.
  • Figure 22 shows inhibition of triple negative breast cancer (TNBC) growth using Formulae VIII and IX.
  • TNBC triple negative breast cancer
  • Formula VIII and Formula IX demonstrated ⁇ 85% TGI at all doses tried (5, 10 mg per kg for Formula VIII; 5, 10, 30 mg per kg for Formula IX) in the TNBC model using MDA-MB-231-AR cells in nude mice.
  • Figure 23 demonstrates inhibition of triple negative breast cancer using Formulae VIII and IX. The tumor weights were likewise reduced for all doses of Formula VIII and Formula IX.
  • FIG. 25A - Figure 25E depict antagonism by SARM regarding the ability of estradiol to activate ER target genes in MCF-7-AR cells.
  • Figure 25B and Figure 25D show that adding AR (as opposed to Green Fluorescent Protein (GFP) as seen in Figures 25A and 25C) to MCF-7-AR cells increases the effects of estradiol (when unopposed) on the ER target genes PR and PS2, respectively.
  • Adding AR to MCF-7-AR cells suppressed the activation of these ER targets in the presence of SARM alone or SARM + estradiol (E2) as compared to GFP transfected cells (i.e. no AR; Figure 25A and Figure 25C).
  • Figure 25E shows that AR target genes are enhanced by SARM even in the presence of estradiol.
  • Figure 26A and Figure 26B depict immunohistochemistry of two regions of the same BR-0001 tumor, a triple negative breast cancer (TNBC). They show that AR expression is consistent throughout this formalin-fixed, paraffin-embedded (FFPE) tissue stained with AR antibody (AR N20 from SCBT).
  • Figure 26C depicts immunohistochemistry staining of an AR-negative TNBC FFPE tumor as a negative control.
  • Figure 27A- Figure 27C depict BR-0001 tumor xenograft growth inhibition by Formula IX compared to enzalutamide (Enza) or vehicle in terms of breast cancer tumor volume (Figure 27A and Figure 27B) and weight (Figure 27C) with time.
  • Figure 27A provides results for Experiment 1
  • Figure 27B provides results for Experiment 2
  • Figure 27C provides results for Experiment 2.
  • BR-0001 TNBC fragments of 1 mm 3 (approximately) were implanted subcutaneously in NOD scid gamma (NSG) mice. Once the tumors reach 100-200 mm 3 , the animals were randomized and treated with vehicle, 10 mg/kg/day Formula IX or enzalutamide orally. Tumor volume was measured thrice weekly. Animals were sacrificed and tumors were weighed.
  • Figure 28A- Figure 28B depict immunohistochemistry of BR-0001 tumors from animals treated with vehicle or Formula IX and stained for Ki-67. Ki-67 was reduced in tumors of animals treated with Formula IX. Quantification of Ki-67 indicated an approximately 50% reduction in Ki-67 staining in 2 weeks of treatment. Tumors from experiment 2 were fixed in formalin and paraffin embedded. Slides were cut and stained with Ki-67 antibody (Figure 28A), Ki-67 staining was reduced in tumors of animals treated with Formula IX. Ki-67 positive cells in each slide (total of 200 cells per view) were counted and represented as % stained cells ( Figure 28B). As a reference, inset into the graphics are bars which are 200 microns ( ⁇ m) in length.
  • Figure 29 depicts Z-scores of 50 genes (PAM50) used to identify BR-0001.
  • PAM50 is a set of 50 genes used to classify breast cancers.
  • PAM50 gene expression data indicated that the BR-0001 tumor belonged to basal-like breast cancer (BLBC) subtype of TNBC.
  • the expression (Z-score) of 50 genes required to classify the breast cancer is given here.
  • Figure 30A and Figure 30B depict gene expression data which is compared to the genes published (Pietenpol group) as useful to classify the Basal-Like Breast Cancer (BLBC) into subclassification.
  • BR-0001 belonged to luminal androgen receptor (LAR) and mesenchymal stem-like (MSL) subtypes.
  • the six TNBC subtypes according to the Pietenpol group include two basal-like (BL1 and BL2), an immunomodulatory (IM), a mesenchymal (M), a mesenchymal stem–like (MSL), and a luminal androgen receptor (LAR) subtype.
  • Figure 31 depicts gene expression changes in BR-0001 tumors treated with Formula IX.
  • Figure 32 depicts reduced tumor growth of ER-positive, PR-positive, HER2-positive and AR-positive tumors composed of HCI-007 cells using Formula IX.
  • Figure 33A and Figure 33B depict potent tumor growth reduction using Formula IX in xenografts composed of HCI-013 cells.
  • HCI-013 phenotype is a triple positive and also expresses AR.
  • Figure 33A tumor volume changes (%) and
  • Figure 33B tumor weight (g).
  • Figure 34A- Figure 34E depict that AR agonists inhibited proliferation of ER- and AR- positive breast cancer cells.
  • Figure 34A depicts that Formula IX inhibited the proliferation of ZR-75-1 cells.
  • FIG. 34B depicts that Formula IX inhibited proliferation of MCF-7 cells expressing AR.
  • Figure 34C depicts that breast cancer fibroblasts treated with AR agonists secreted factors that inhibited MCF-7-GFP cells lacking supplemented AR.
  • fibroblasts obtained from a breast cancer patient were cultured in growth medium and were treated in triplicates with vehicle, 10 nM DHT, 1 ⁇ M enzalutamide, or 1 ⁇ M Formula IX. Medium was changed, and the cells were re-treated on days 4 and 7. Medium was collected, pooled from triplicates, and stored in -80°C. After 10 days of treatment, cells were fixed, and cell viability was measured using SRB. MCF-7 cells stably transfected with GFP (MCF-7-GFP) were plated in growth medium. Twenty-four hours after plating, cells were fed with the conditioned medium obtained from patient-derived fibroblasts as indicated above. Cells were fed for 10 days with conditioned medium, with medium changed on days 4 and 7.
  • FIG. 34D depicts that AR ligands did not inhibit growth of ER-negative AR-positive HCI-9 PDX.
  • Figure 34E depicts that HCI-13 ER- ⁇ was resistant to ER antagonists fulvestrant and tamoxifen (right pane) compared to wt-ER- ⁇ (left pane).
  • ER- ⁇ from HCI-13 was cloned into pCR3.1 vector. Wildtype ER- ⁇ and HCI-13 ER- ⁇ , ERE-LUC, and CMV-LUC were transfected into COS-1 cells using lipofectamine. Cells were treated 24 hours after transfection with vehicle, 0.1 nM estradiol, 10 nM fulvestrant or 1 ⁇ M tamoxifen in combination with 0.1 nM estradiol. Twenty four hours after treatment cells were harvested and luciferase assay was performed.
  • ER antagonists in wt- ER- ⁇ were significantly different than vehicle-treated wt-ER- ⁇ as depicted by * p ⁇ 0.05.
  • Figure 35A- Figure 35D depict that AR agonists inhibited proliferation and growth of wildtype and mutant ER and AR -positive xenografts.
  • Figure 35A depicts that protein from HCI PDX (HCI-7, 9, or 13) tumor fragments was extracted and fractionated on a SDS-PAGE, and Western blotted for the AR. AR was also quantified at mRNA level and expressed as fold change from LNCaP prostate cancer cell AR (numbers provided under the blot).
  • Figures 35B depict that Formula IX inhibited HCI-7 tumor growth.
  • mice were randomized and treated with vehicle (DMSO:PEG-300 (15%:85%)), Formula IX (10 mpk p.o.), or enzalutamide (30 mpk p.o.). Tumor volume was measured weekly. At sacrifice, tumors were removed, weighed ( Figure 35C), and stored for further analysis.
  • Figure 36B and Figure 36C depict that AR agonist (Formula IX) inhibited growth of HCI-13 PDX.
  • FIG. 36C Tumor volume was measured weekly. At sacrifice, tumors were removed, weighed (Figure 36C), and stored for further analysis.
  • Figure 36D- Figure 36G depict that AR agonists, but not AR- or ER- antagonists, inhibited ER-target genes in HCI-13 ex vivo sponge culture.
  • Tissues were treated with vehicle, 10 nM DHT, 1 ⁇ M Formula IX, 1 ⁇ M enzalutamide, or 100 nM fulvestrant for three days.
  • RNA was extracted from the tissues and expression of genes was measured by real time PCR and normalized to GAPDH.
  • Figure 36H- Figure 36J depict the effect of Formula IX on ER- positive breast cancer patient specimens.
  • Tissues were treated with vehicle, 1 ⁇ M Formula IX, or 100 nM fulvestrant for three days.
  • RNA was extracted from the tissues and expression of genes was measured by real time PCR and normalized to GAPDH.
  • Table in Figure 36K denotes the fold difference in the expression of AR and ER at the mRNA level compared to HCI-13 tumors.
  • Figure 37H depicts that the GSEA KEGG pathway analysis provided ERBB2 (ERBB is abbreviated from erythroblastic oncogene B; also frequently called HER2 (from human epidermal growth factor receptor 2) or HER2/neu) pathway as one of the highly correlated pathway with Formula IX treatment (bottom four rows in the left column (vehicle treated) are downregulated genes (blue in the original color) whereas most of the rows are upregulated genes (red in the original color); in contrast, Formula IX treated column (right) is just the opposite).
  • ERBB2 erythroblastic oncogene B
  • HER2 from human epidermal growth factor receptor 2
  • HER2/neu HER2/neu
  • FIG. 38A- Figure 38H depict that ChIP-Sequencing showed rearrangement of ER and AR binding to the DNA.
  • ChIP chromatin immunoprecipitation
  • Heatmap of significantly different peaks (q ⁇ 0.05 for ER and corresponding AR peaks) is shown.
  • the top enriched motifs are shown in Figure 38H.
  • Figure 38B shows representative peaks from KLK3 regulatory regions from ER and AR ChIP-Seq.
  • Figure 38C shows Principal Component Analysis (PCA) plot of vehicle- and Formula IX -treated samples that corresponds to ER-ChIP peaks.
  • Figure 38D depicts that ChIP assay was performed with AR or ER antibody in HCI-13 specimens treated with vehicle or Formula IX and real time PCR was performed with the primers and Taqman probe to the specified regions.
  • Figure 38E depicts pie charts showing the distribution of ER enrichment in Formula IX -treated HCI-13 samples.
  • FIG. 38F depicts Venn diagrams showing the overlap between depleted FOXA1RE and ERE regions and enriched ARE, GRE, and FOXA1RE.
  • Figure 38G depicts that SRC-1 interacted with both AR and ER in response to Formula IX. Protein extracts from HCI-13 tumor samples treated with vehicle or Formula IX were immunoprecipitated with AR or ER antibodies and Western blot for SRC-1 was performed.
  • AR-androgen receptor ; ER-estrogen receptor; ChIP- chromatin immunoprecipitation; ARE-androgen response elements; ERE-estrogen response element; GRE-glucocorticoid response elements; SRC-1- steroid receptor coactivator-1, FOXA1RE- Forkhead box A1 response element.
  • Figure 38H depicts up-regulated motifs (ER).
  • Figure 39 depicts colocalization of AR and ER- ⁇ in luminal B breast cancer specimens.
  • Figure 40 depicts representative ChIP Seq peaks in the regulated regions of genes.
  • FIG. 41A- Figure 41E depict a phospho-proteome analysis of HCI-13 PDX.
  • Arrays were probed with a total of 174 antibodies targeting a wide range of protein kinases and their activation via phosphorylation. Arrays were stained with an anti-rabbit or anti-mouse biotinylated secondary antibody. The signals were amplified and a streptavidin-conjugated IRDye680 were used as secondary signal detection agents. Images were acquired and quantified.
  • Figure 41D- Figure 41E depict that activation of PKC overcame inhibition by Formula IX. HCI-13 tissues fragments were cultured on gelatin sponges and were treated with 100 nM phorbol 12-myristate 13-acetate (PMA) or 100 ng/mL EGF 30 minutes before addition of 1 ⁇ M Formula IX.
  • PMA phorbol 12-myristate 13-acetate
  • Figures 43A and Figure 43B present baseline [ 18 F]-16 ⁇ -fluoro-5 ⁇ - dihydrotestosterone (FDHT) SUVmax (FDHT uptake) versus AR.
  • FIG. 44A depicts that median baseline FDHT-SUVmax was 2.93 (range 1-4.38) for 7 patients with CB (defined as complete response (CR), partial response (PR), or stable disease (SD) as determined by RECIST criteria) at 12 weeks after therapy and 2.15 (0.96-3.77) for 4 patients with progressive disease (PD).
  • CB complete response
  • PR partial response
  • SD stable disease
  • Figure 44B depicts that the change in FDHT uptake from baseline to six weeks (SUVmax change from baseline to six weeks) declined for those with CB at 12 weeks whereas those with PD did not.
  • Figures 45A and 45B present that Formula IX (SARM) re-sensitized human breast cancer models to CDK 4/6 inhibition in models of CDK 4/6 resistance.
  • Data were presented as mean values ⁇ s.e.m.
  • Figure 45B depicts proliferation of Palb R cells in response to AR agonist (Formula IX 100 nM) and Palb (125 nM), alone or in combination. Data represented the mean ⁇ s.e.m. of four replicate cell culture wells per condition.
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject.
  • this invention provides methods of: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI)
  • the subject is a male. In one embodiment, the subject is a female.
  • a method for treating a subject suffering from breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating a subject suffering from metastatic breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat metastatic breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating a subject suffering from refractory breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat refractory breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating a subject suffering from AR-positive breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR- positive breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • the AR-positive breast cancer is ER, PR and HER2-positive.
  • the AR-positive breast cancer is ER, PR and HER2-negative.
  • the AR-positive breast cancer is ER-positive, and PR and HER2-negative. In another embodiment, the AR-positive breast cancer is ER and PR-positive, and HER2- negative. In yet another embodiment, the AR-positive breast cancer is ER and HER2-positive, and PR-negative. In still another embodiment, the AR-positive breast cancer is ER-negative, and PR and HER2-positive. In a further embodiment, the AR-positive breast cancer is ER and PR-negative, and HER2-positive. In still a further embodiment, the AR-positive breast cancer is ER and HER2-negative, and PR-positive. In one embodiment, the AR-positive breast cancer is ER-negative. In another embodiment, the AR-positive breast cancer is ER-positive.
  • a method for treating a subject suffering from AR-positive refractory breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive refractory breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from AR-positive metastatic breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive metastatic breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from AR-positive and ER-positive breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive metastatic breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from ER-positive breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat ER- positive breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • the ER-positive breast cancer is AR-positive.
  • the ER-positive breast cancer is AR-negative.
  • ER-positive breast cancer is triple positive (ER, PR, HER2) breast cancer. In another embodiment, ER- positive breast cancer is not triple positive breast cancer.
  • a method for treating a subject suffering from triple negative breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N- oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat triple negative breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating a subject suffering from AR-positive triple negative breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to AR-positive treat triple negative breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from advanced breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat advanced breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), ribociclib (Kisqali), abemaciclib (Vorzenio), trilaciclib, lerociclib), alpelisib (Piqray) (an inhibitor of phosphatidylinositol-3-kinase subunit alpha (PI3K ⁇ )), mTOR inhibitor (everolimus), poly ADP ribose polymerase (PARP) inhibitor (olaparib (Lynparza) or
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • this invention provides a method for treating a subject suffering from HER2-positive breast cancer, comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat HER2- positive breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • the HER2-positive breast cancer is HER2-positive refractory breast cancer.
  • the HER2-positive breast cancer is HER2-positive metastatic breast cancer. In one embodiment, the HER2-positive breast cancer is ER-negative. In another embodiment, the HER2-positive breast cancer is ER-positive. In one embodiment, the HER2-positive breast cancer is PR-positive. In another embodiment, the HER2-positive breast cancer is PR-negative. In one embodiment, the HER2-positive breast cancer is AR- positive. In another embodiment, the HER2-positive breast cancer is AR-negative. [00092] In certain embodiment, the HER2-positive breast cancer is ER-positive, PR- positive, and AR-positive. In another embodiment, the HER2-positive breast cancer is ER- positive, PR-negative, and AR-positive.
  • the HER2-positive breast cancer is ER-positive, PR-negative, and AR-negative. In other embodiment, the HER2- positive breast cancer is ER-positive, PR-positive, and AR-negative. In another embodiment, the HER2-positive breast cancer is ER-negative, PR-negative, and AR-positive. In another embodiment, the HER2-positive breast cancer is ER-negative, PR-positive, and AR-positive. In another embodiment, the HER2-positive breast cancer is ER-negative, PR-positive, and AR- negative. In certain embodiment, the HER2-positive breast cancer is ER-negative, PR- negative, and AR-negative. In certain embodiment, the HER2-positive breast cancer is triple- positive HER2 breast cancer.
  • this invention provides a method for treating a subject suffering from ER mutant expressing breast cancer, comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N- oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat ER mutant expressing breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • the ER mutant expressing breast cancer is Y537S mutation expressing breast cancer.
  • the ER mutant expressing breast cancer is D351Y mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is E380Q mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is V422del mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is S432L mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is G442A mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is S463P mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L469V mutation expressing breast cancer.
  • the ER mutant expressing breast cancer is L536R mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L536H mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L536P mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L536Q mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is Y537N mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is Y537C mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is Y537D mutation expressing breast cancer.
  • the ER mutant expressing breast cancer is D538G mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is E542G mutation expressing breast cancer. In one embodiment, ER mutant expressing breast cancer refers to mutants of ER-alpha. [00096] In a certain embodiment, the ER mutant expressing breast cancer is as described in Cancer Cell 2018, 33, 173–186, or in Nat Rev Cancer 2018, 18(6):377-388, which are incorporated herein by reference. In one embodiment, ER mutant expressing breast cancer refers to mutants of ER-alpha.
  • this invention provides a pharmaceutical composition
  • a selective androgen receptor modulator (SARM) compound and a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor wherein said SARM compound is represented by a structure of formula I: wherein X is a bond, O, CH 2 , NH, S, Se, PR, NO, or NR; G is O or S; T is OH, OR, -NHCOCH 3 , or NHCOR; R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F, CHF 2 , CF 3 , CF 2 CF 3 , aryl, phenyl, halogen, alkenyl, or OH; R 1 is CH 3 , CH 2 F, CHF 2 , CF 3 , CH 2 CH 3 , or CF 2 CF 3 ; R2 is H, F, Cl, Br, I, CH 3 , CF 3 ,
  • the SARM compound in the composition of the invention is represented by a structure of formula II: wherein X is a bond, O, CH 2 , NH, Se, PR, or NR; G is O or S; T is OH, OR, -NHCOCH 3 , or NHCOR; Z is NO 2 , CN, COR, COOH or CONHR; Y is I, CF 3 , Br, Cl, or Sn(R) 3 ; Q is CN, alkyl, halogen, N(R) 2 , NHCOCH 3 , NHCOCF 3 , NHCOR, NHCONHR, NHCOOR, OCONHR, CONHR, NHCSCH 3 , NHCSCF 3 , NHCSR, NHSO 2 CH 3 , NHSO 2 R, OR, COR, OCOR, OSO 2 R, SO 2 R, or SR; or Q together with the benzene ring to which it is attached is a fused ring system represented by structure
  • the invention provides a pharmaceutical composition comprising Formula IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof, and a CDK 4/6 inhibitor, [000101]
  • the CDK 4/6 inhibitor is palbociclib, ribociclib, trilaciclib, lerociclib, or abemaciclib.
  • the CDK 4/6 inhibitor is palbociclib.
  • the CDK 4/6 inhibitor is ribociclib.
  • the CDK 4/6 inhibitor is trilaciclib.
  • the CDK 4/6 inhibitor is lerociclib.
  • the CDK 4/6 inhibitor is abemaciclib.
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and palbociclib, [000103]
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and ribociclib, [000104]
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and trilaciclib, [000105]
  • the invention provides a pharmaceutical composition comprising compound IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a
  • the pharmaceutical composition of the invention can be used for treatment of breast cancer.
  • the invention provides a method for treating a subject suffering from breast cancer, comprising administering to said subject a pharmaceutical composition of the invention as described herein.
  • the pharmaceutical composition comprises a selective androgen receptor modulator (SARM) compound and a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor, wherein said SARM compound is represented by a structure of formula I:
  • X is a bond, O, CH 2 , NH, S, Se, PR, NO, or NR; G is O or S; T is OH, OR, -NHCOCH 3 , or NHCOR; R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F, CHF 2 , CF 3 , CF 2 CF 3 , aryl, phenyl, halogen, alkenyl, or OH; R 1 is CH 3 , CH 2 F, CHF 2 , CF 3 , CH 2 CH 3 , or CF 2 CF 3 ; R 2 is H, F, Cl, Br, I, CH 3 , CF 3 , OH, CN, NO 2 , NHCOCH 3 , NHCOCF 3 , NHCOR, alkyl, arylalkyl, OR, NH 2 , NHR, N(R) 2 , or SR; R 3 is H, F, Cl, Br,
  • the SARM compound is represented by a structure of formula II: wherein X is a bond, O, CH 2 , NH, Se, PR, or NR; G is O or S; T is OH, OR, -NHCOCH 3 , or NHCOR; Z is NO 2 , CN, COR, COOH or CONHR; Y is I, CF 3 , Br, Cl, or Sn(R) 3 ; Q is CN, alkyl, halogen, N(R) 2 , NHCOCH 3 , NHCOCF 3 , NHCOR, NHCONHR, NHCOOR, OCONHR, CONHR, NHCSCH 3 , NHCSCF 3 , NHCSR, NHSO 2 CH 3 , NHSO 2 R, OR, COR, OCOR, OSO 2 R, SO 2 R, or SR; or Q together with the benzene ring to which it is attached is a fused ring system represented by structure
  • the pharmaceutical composition comprises Formula IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof, and a CDK 4/6 inhibitor, [000114]
  • the CDK 4/6 inhibitor is palbociclib, ribociclib, trilaciclib, lerociclib, or abemaciclib.
  • the CDK 4/6 inhibitor is palbociclib.
  • the CDK 4/6 inhibitor is ribociclib.
  • the CDK 4/6 inhibitor is trilaciclib.
  • the CDK 4/6 inhibitor is lerociclib. In some embodiments, the CDK 4/6 inhibitor is abemaciclib.
  • the pharmaceutical composition comprises Formula IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and palbociclib, [000116] In some embodiments of the method of the invention, the pharmaceutical composition comprises Formula IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and ribociclib, [000117] In some embodiments of the method of the invention, the pharmaceutical composition comprises Formula IX, or an optical isomer, a racemic mixture, a pharmaceutically acceptable salt, a pharmaceutical product, a hydrate, an N-oxide, or a crystal thereof and trilaciclib, [000118] In some embodiments of
  • the breast cancer is AR-positive metastatic breast cancer. In certain embodiments, the breast cancer is AR-positive refractory breast cancer. [000122] In some embodiments, the ER-positive breast cancer is AR-positive and ER-positive breast cancer, or AR-negative and ER-positive breast cancer.
  • the AR-positive breast cancer is ER-negative; ER-negative, PR-negative, and HER2-negative; ER-negative, PR-negative, and HER2-positive; ER- negative, PR-positive, and HER2-negative; ER-negative, PR-positive, and HER2-positive; ER-positive, PR-negative, and HER2-negative; ER-positive, PR-negative, and HER2-positive; or ER-positive, PR-positive, and HER2- positive.
  • the breast cancer has failed treatment with a selective estrogen receptor modulator (SERM).
  • SERM selective estrogen receptor modulator
  • the SERM is tamoxifen, toremifene, or raloxifene.
  • the breast cancer has failed treatment with a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor.
  • the subject is resistant or non- responsive to the CDK 4/6 inhibitor.
  • the CDK 4/6 inhibitor is palbociclib (Ibrance), ribociclib (Kisqali), trilaciclib, lerociclib or abemaciclib (Vorzenio).
  • the CDK 4/6 inhibitor is palbociclib (Ibrance).
  • the CDK 4/6 inhibitor is ribociclib.
  • the CDK 4/6 inhibitor is trilaciclib. In some embodiments, the CDK 4/6 inhibitor is lerociclib. In some embodiments, the CDK 4/6 inhibitor is abemaciclib. [000126] In some embodiments of the method of the invention, the composition of the invention as described herein re-sensitizes said breast cancer to treatment with CDK 4/6 inhibitors. In some embodiments, the CDK 4/6 inhibitor is at least one of palbociclib (Ibrance), ribociclib (Kisqali), trilaciclib, lerociclib, and abemaciclib (Vorzenio). In certain embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance).
  • the CDK 4/6 inhibitor is ribociclib (Kisqali). In some embodiments, the CDK 4/6 inhibitor is trilaciclib. In some embodiments, the CDK 4/6 inhibitor is lerociclib. In some embodiments, the CDK 4/6 inhibitor is abemaciclib. [000127] In some embodiments of the method of the invention, the composition of the invention as described herein overcomes estrogen endocrine resistance. In some embodiments, the estrogen endocrine therapy includes at least one of tamoxifen, toremifene, raloxifene, exemestane, letrozole, anastrozole, and fulvestrant. In some embodiments, the estrogen endocrine therapy includes tamoxifen.
  • the estrogen endocrine therapy includes toremifene. In some embodiments, the estrogen endocrine therapy includes raloxifene. In some embodiments, the estrogen endocrine therapy includes exemestane. In some embodiments, the estrogen endocrine therapy includes letrozole. In some embodiments, the estrogen endocrine therapy includes anastrozole. In some embodiments, the estrogen endocrine therapy includes fulvestrant. [000128] It is another aspect of the invention that the composition of the invention as described herein overcomes resistance to estrogen endocrine and CDK 4/6 inhibitor co- therapy.
  • the CDK 4/6 inhibitor is at least one of palbociclib (Ibrance), ribociclib (Kisqali), trilaciclib, lerociclib, and abemaciclib (Vorzenio).
  • the CDK 4/6 inhibitor is palbociclib (Ibrance).
  • the CDK 4/6 inhibitor is ribociclib (Kisqali).
  • the CDK 4/6 inhibitor is trilaciclib.
  • the CDK 4/6 inhibitor is lerociclib.
  • the CDK 4/6 inhibitor is abemaciclib.
  • the estrogen endocrine therapy includes at least one of tamoxifen, toremifene, raloxifene, exemestane, letrozole, anastrozole, and fulvestrant.
  • the estrogen endocrine therapy includes tamoxifen.
  • the estrogen endocrine therapy includes toremifene.
  • the estrogen endocrine therapy includes raloxifene.
  • the estrogen endocrine therapy includes exemestane.
  • the estrogen endocrine therapy includes letrozole.
  • the estrogen endocrine therapy includes anastrozole.
  • the estrogen endocrine therapy includes fulvestrant.
  • the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy includes tamoxifen.
  • the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy includes toremifene.
  • the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy includes raloxifene.
  • the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy includes exemestane. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy includes letrozole. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy includes fulvestrant.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes tamoxifen.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes toremifene.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes raloxifene.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes exemestane.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes letrozole. In some embodiments, the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes fulvestrant. [000131] In some embodiments, where the composition of the invention as described herein overcomes resistance to estrogen endocrine and CDK 4/6 inhibitor co-therapy, the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes tamoxifen.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes toremifene. In some embodiments, the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes raloxifene. In some embodiments, the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes exemestane. In some embodiments, the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes letrozole. In some embodiments, the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes fulvestrant.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes tamoxifen.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes toremifene.
  • the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes raloxifene.
  • the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes exemestane.
  • the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes letrozole.
  • the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes fulvestrant. [000133] In some embodiments, where the composition of the invention as described herein overcomes resistance to estrogen endocrine and CDK 4/6 inhibitor co-therapy, the CDK 4/6 inhibitor is abemaciclib (Vorzenio) and the estrogen endocrine therapy includes tamoxifen. In some embodiments, the CDK 4/6 inhibitor is abemaciclib (Vorzenio) and the estrogen endocrine therapy includes toremifene.
  • the CDK 4/6 inhibitor is abemaciclib (Vorzenio) and the estrogen endocrine therapy includes raloxifene. In some embodiments, the CDK 4/6 inhibitor is abemaciclib (Vorzenio) and the estrogen endocrine therapy includes exemestane. In some embodiments, the CDK 4/6 inhibitor is abemaciclib (Vorzenio) and the estrogen endocrine therapy includes letrozole. In some embodiments, the CDK 4/6 inhibitor is abemaciclib (Vorzenio) and the estrogen endocrine therapy includes anastrozole.
  • the CDK 4/6 inhibitor is abemaciclib (Vorzenio) and the estrogen endocrine therapy includes fulvestrant.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes tamoxifen.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes toremifene.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes raloxifene.
  • the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes exemestane. In some embodiments, the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes letrozole. In some embodiments, the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is ribociclib (Kisqali) and the estrogen endocrine therapy includes fulvestrant.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes tamoxifen.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes toremifene.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes raloxifene.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes exemestane.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes letrozole.
  • the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is trilaciclib and the estrogen endocrine therapy includes fulvestrant. [000136] In some embodiments, where the composition of the invention as described herein overcomes resistance to estrogen endocrine and CDK 4/6 inhibitor co-therapy, the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes tamoxifen. In some embodiments, the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes toremifene. In some embodiments, the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes raloxifene.
  • the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes exemestane. In some embodiments, the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes letrozole. In some embodiments, the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is lerociclib and the estrogen endocrine therapy includes fulvestrant. [000137] In some embodiments, where the composition of the invention as described herein overcomes resistance to estrogen endocrine and CDK 4/6 inhibitor co-therapy, the CDK 4/6 inhibitor is abemaciclib and the estrogen endocrine therapy includes tamoxifen.
  • the CDK 4/6 inhibitor is abemaciclib and the estrogen endocrine therapy includes toremifene. In some embodiments, the CDK 4/6 inhibitor is abemaciclib and the estrogen endocrine therapy includes raloxifene. In some embodiments, the CDK 4/6 inhibitor is abemaciclib and the estrogen endocrine therapy includes exemestane. In some embodiments, the CDK 4/6 inhibitor is abemaciclib and the estrogen endocrine therapy includes letrozole. In some embodiments, the CDK 4/6 inhibitor is abemaciclib and the estrogen endocrine therapy includes anastrozole. In some embodiments, the CDK 4/6 inhibitor is abemaciclib and the estrogen endocrine therapy includes fulvestrant.
  • the breast cancer has failed treatment with an mTOR inhibitor.
  • the mTOR inhibitor is everolimus, sirolimus, temsirolimus, or ridafarolimus.
  • the method of the invention further prolongs the survival of the subject suffering from breast cancer or prolongs the progression-free survival of the subject suffering from breast cancer.
  • the composition of the invention is administered intravenously, intraarterially, intramuscularly, subcutaneously, orally, or topically. In some embodiments, the composition of the invention is administered orally.
  • the selective androgen receptor modulator is dosed from 1 mg to 50 mg per day.
  • the selective androgen receptor modulator is dosed per day from about 1 mg to about 5 mg, or from about 5 mg to about 50 mg, or from about 5 mg to about 10 mg, or from about 5 mg to about 15 mg, or from about 5 mg to about 20 mg, or from about 5 mg to about 30 mg, or from about 10 mg to about 50 mg, or from about 10 mg to about 40 mg, or from about 10 mg to about 30 mg, or from about 10 mg to about 20 mg, or from about 15 mg to about 50 mg, or from about 20 mg to about 50 mg, or from about 25 mg to about 50 mg, or from about 30 mg to about 50 mg, or from about 30 mg to about 40 mg.
  • the selective androgen receptor modulator is dosed about 9 mg per day or 18 mg per day.
  • the term “treating” may refer to treating, delaying the progression, preventing the recurrence or treating the recurrence. In one embodiment, the term “treating” refers to a reduction in morbidity, mortality, or a combination thereof, in association with breast cancer.
  • the term “preventing” may refer to preventing the initial occurance of a disorder, reducing risk factors, minimize the disability or potential health threat of a disorder.
  • breast cancer may refer to breast cancer; advanced breast cancer; metastatic breast cancer; AR-positive breast cancer; ER-positive breast cancer; AR- positive breast cancer with or without expression of ER, PR and/or HER2; triple-positive breast cancer (ER, PR and HER2-positive), AR-positive breast cancer with or without expression of ER; ER-positive breast cancer with or without expression of AR; AR-positive and ER-positive breast cancer; refractory breast cancer; AR-positive refractory breast cancer; ER-positive refractory breast cancer; AR-positive metastatic breast cancer; ER-positive metastatic breast cancer; breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI) (letrozole, anastrozole, exemestane), cyclin- dependent kina
  • SERM selective estrogen receptor modulator
  • breast cancer refers to a condition characterized by anomalous rapid proliferation of abnormal cells in one or both breasts of a subject.
  • the abnormal cells often are referred to as “neoplastic cells,” which refers to, in some embodiments, transformed cells that can form a solid tumor.
  • tumor in some embodiments, refers to an abnormal mass or population of cells (i.e. two or more cells) that result from excessive or abnormal cell division, whether malignant or benign, and pre- cancerous and cancerous cells.
  • Malignant tumors are distinguished from benign growths or tumors in that, in addition to uncontrolled cellular proliferation, they can invade surrounding tissues and can metastasize.
  • neoplastic cells may be identified in one or both breasts only and not in another tissue or organ, in one or both breasts and one or more adjacent tissues or organs (e.g., lymph node), or in a breast and one or more non-adjacent tissues or organs to which the breast cancer cells have metastasized.
  • adjacent tissues or organs e.g., lymph node
  • non-adjacent tissues or organs to which the breast cancer cells have metastasized.
  • the term “metastasis”, in some embodiments, refers to a process in which cancer cells travel from one organ or tissue to another non-adjacent organ or tissue. Cancer cells in the breast(s) can spread to tissues and organs of a subject, and conversely, cancer cells from other organs or tissue can invade or metastasize to a breast.
  • Cancerous cells from the breast(s) may invade or metastasize to any other organ or tissue of the body.
  • Breast cancer cells often invade lymph node cells and/or metastasize to the liver, brain and/or bone and spread cancer in these tissues and organs.
  • the term “invasion”, in some embodiments, refers to the spread of cancerous cells to adjacent surrounding tissues.
  • advanced breast cancer refers to cancer that has spread to other places in the body and usually cannot be cured or controlled with current treatment.
  • AR-positive breast cancer may refer to breast cancer wherein at least a portion of the cancer cells express at least the androgen receptor (AR).
  • the term "ER-positive breast cancer” may refer to breast cancer wherein at least a portion of the cancer cells express at least the estrogen receptor (ER).
  • the term “triple negative breast cancer” may refer to breast cancer cells that do not have estrogen receptors (ER), progesterone receptors (PR), or large amounts of HER2/neu protein.
  • Triple negative breast cancer may also be referred to herein as "ER- negative PR-negative HER2/neu-negative breast cancer”.
  • the term “triple positive breast cancer” may refer to breast cancer cells that express estrogen receptors (ER), progesterone receptors (PR), and large amounts of HER2/neu (HER2) protein.
  • HER2-positive breast cancer may refer to breast cancers wherein at least a portion of the cancer cells express elevated levels of HER2 protein (HER2 (from human epidermal growth factor receptor 2) or HER2/neu) which promotes rapid growth of cells.
  • the term “ER mutant expressing breast cancer” may refer to breast cancers that express estrogen receptor alpha (ER- ⁇ ) with therapy resistance conferring mutations. Often these mutations are located within the ligand binding domain of ER- ⁇ , are treatment emergent, and/or confer resistance to certain or all endocrine therapies such as SERMs, AIs, SERDs, and/or GnRH agonists.
  • the term “Y537S ER mutant expressing breast cancer” may refer to breast cancers that express estrogen receptor alpha (ER- ⁇ ) with the point mutation Y537S.
  • a method for treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat, prevent, suppress or inhibit metastasis in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for prolonging the survival of a subject with breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to prolong the survival of a subject with breast cancer.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for slowing the progression of breast cancer in a subject comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to slow the progression of breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for prolonging progression-free survival of a subject with breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N- oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to prolong progression-free survival of a subject with breast cancer.
  • the subject is a female subject.
  • the subject is a male subject.
  • breast cancer of this invention refers to in one embodiment to ER-positive metastatic breast cancer; In another embodiment to ER-positive refractory breast cancer; In another embodiment to ER-positive PR-positive HER2-negative breast cancer; In another embodiment to AR-positive ER-positive breast cancer; In another embodiment to AR- positive ER-positive refractory breast cancer; In another embodiment to AR-positive ER- positive metastatic breast cancer; In another embodiment to triple positive breast cancer; In another embodiment to advanced ER-positive breast cancer; In another embodiment to AR- positive; In another embodiment to ER-positive breast cancer; and in another embodiment to breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6
  • SERM selective estrogen receptor
  • a method for lowering biomarker levels in a subject with breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to lower the biomarker level in said subject.
  • the method comprises administering a compound of Formulae I-XIV of this invention.
  • biomarker may refer to a substance used as an indicator of a process, event, or condition.
  • a biomarker can be a biomolecule such as a nucleic acid molecule (e.g. microRNA, genomic DNA, etc.), a protein, a polysaccharide, and the like. Biomarkers include tumor antigens and tumor markers. In one embodiment, a biomarker indicates the presence of cancer, e.g., breast cancer. In one embodiment, a biomarker may be used to determine the efficacy of treatment. In one embodiment, a biomarker may be used to determine the progression of a condition, e.g., breast cancer. [000162] The MUC-1 associated antigen, or CA 27.29, is a cancer antigen highly associated with breast cancer.
  • CA27.29 biomarker refers to a biomarker for breast cancer.
  • CA27.29 is a biomarker for advanced breast cancer.
  • PSA prote-specific antigen
  • CTX biomarker and “NTX biomarker” are the C-telopeptide and N-telopeptide of collagen type I, respectively, which are used as biomarkers of bone turnover.
  • NTX and CTX biomarkers may be sensitive indicators of the presence of bone metastases in breast cancer patients.
  • a method of this invention lowers CA27.29 biomarker in a subject.
  • a method of this invention lowers PSA in a subject.
  • a method of this invention lowers CTX biomarker in a subject.
  • a method of this invention lowers NTX biomarker in a subject.
  • a method of this invention maintains the level of CA27.29 in a subject.
  • a method of this invention maintains the level of PSA in a subject.
  • a method of this invention maintains the level of CTX biomarker in a subject.
  • a method of this invention maintains the level of NTX biomarker.
  • the subject has breast cancer.
  • the subject has advanced breast cancer.
  • the subject has refractory breast cancer.
  • the subject has AR-positive breast cancer.
  • the subject has ER-positive breast cancer.
  • this invention is directed to a method of treating breast cancer in a subject, comprising a step of determining the 18 F-16 ⁇ -fluoro-5 ⁇ -dihydrotestosterone ( 18 F- DHT) tumor uptake and identifying said subject as having AR-positive breast cancer based on 18 F-DHT tumor uptake, followed by administering to said AR-positive breast cancer subject a selective androgen receptor modulator (SARM) compound.
  • SARM selective androgen receptor modulator
  • the selective androgen receptor modulator compound is a compound of formula I-XIV.
  • said tumor is metastatic breast cancer tumor.
  • said tumor is an ER-positive metastatic breast cancer tumor.
  • said tumor is an ER-positive metastatic breast cancer tumor that has failed FDA approved hormonal and/or kinase treatments.
  • the AR-positive breast cancer is ER-positive. In another embodiment, the AR-positive breast cancer is metastatic.
  • the breast cancer is any of refractory breast cancer; AR-positive breast cancer; AR-positive refractory breast cancer; AR-positive metastatic breast cancer; AR-positive and ER-positive breast cancer; AR-positive breast cancer with or without expression of estrogen receptor (ER), progesterone receptor (PR), and/or human epidermal growth factor receptor 2 (HER2); triple negative breast cancer (TNBC); advanced breast cancer; breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin- releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), ribociclib (Kisqali), abemaciclib (Vorzenio), trilaciclib, lerociclib), alpelis
  • the breast cancer is estrogen receptor positive (ER+) metastatic breast cancer.
  • the compound of this invention is an antagonist.
  • the compound of this invention is an agonist.
  • the compound of this invention is a partial agonist/partial antagonist.
  • a compound of this invention is an AR agonist.
  • a compound is an AR antagonist.
  • a compound is a partial AR agonist and AR antagonist.
  • a compound of this invention is a PR agonist.
  • a compound is a PR antagonist.
  • a compound is a partial PR agonist and PR antagonist.
  • a compound of this invention is an AR agonist and a PR antagonist.
  • the SARM compounds of this invention may be useful, in some embodiments, for: a) treatment, prevention, delaying onset of, increasing time to first skeletal related event (SRE), suppression or inhibition of, or the reduction of the risk of developing a skeletal-related event (SRE), such as pathological bone fractures, surgery of the bone, radiation of the bone, spinal cord compression, new bone metastasis, and/or bone loss in a subject; b) treatment, prevention, suppression or inhibition of, or the reduction of the risk of developing a variety of hormone- related conditions in a subject, for example for increasing libido; and/or for c) improving quality of life in a subject.
  • SRE first skeletal related event
  • SRE skeletal-related event
  • Osteoporosis is a systemic skeletal disease, characterized by low bone mass and deterioration of bone tissue, with a consequent increase in bone fragility and susceptibility to fracture.
  • the condition affects more than 25 million people and causes more than 1.3 million fractures each year, including 500,000 spine, 250,000 hip and 240,000 wrist fractures annually.
  • Hip fractures are the most serious consequence of osteoporosis, with 5-20% of patients dying within one year, and over 50% of survivors being incapacitated.
  • the elderly are at greatest risk of osteoporosis, and the problem is therefore predicted to increase significantly with the aging of the population.
  • this invention provides for the use of a compound as herein described, or its prodrug, analog, isomer, metabolite, derivative, pharmaceutically acceptable salt, pharmaceutical product, polymorph, crystal, impurity, N-oxide, hydrate or any combination thereof, for: a) treating a bone related disorder; b) preventing a bone related disorder; c) suppressing a bone related disorder; d) inhibiting a bone related disorder; e) increasing a strength of a bone of a subject; f) increasing a bone mass in a subject; g) use for osteoclastogenesis inhibition; and/or h) use for osteoblastogenesis stimulation.
  • this invention provides for the use of a compound as herein described, or its prodrug, analog, isomer, metabolite, derivative, pharmaceutically acceptable salt, pharmaceutical product, polymorph, crystal, impurity, N-oxide, hydrate or any combination thereof, for: a) accelerating bone repair; b) treating bone disorders; c) treating bone density loss; d) treating low bone mineral density (BMD); e) treating reduced bone mass; f) treating metabolic bone disease; g) promoting bone growth or regrowth; h) promoting bone restoration; i) promoting bone fracture repair; j) promoting bone remodeling; k) treating bone damage following reconstructive surgery including of the face, hip, or joints; l) enhancing of bone strength and function; m) increasing cortical bone mass; n) increasing trabecular connectivity; o) preventing, inhibiting or delaying metastasis to the bone; and/or p) preventing, inhibiting or delaying the growth of metastatic tumors of the bone
  • the bone related disorder is a genetic disorder, or in another embodiment, is induced as a result of a treatment regimen for a given disease.
  • the compounds as herein described are useful in treating a bone- related disorder that arises as a result of cancer metastasis to bone, or in another embodiment, as a result of androgen-deprivation therapy, for example, given in response to prostate carcinogenesis in the subject.
  • “estrogen-deprivation therapy” may refer to therapy which is given in response to breast cancer in a subject.
  • Known treatments include treatment with GnRH agonists, SERMs, SERDs, or aromatase inhibitors (AI).
  • the compounds as herein described are useful in treating a bone-related disorder that arises as a result of cancer metastasis to bone, or in another embodiment, as a result of estrogen- deprivation therapy, for example, given in response to breast cancer in the subject. Menopause can also be induced using GnRH agonists such as gosarelin (Zoladex) which maintains endogeneous estrogens at low levels via inhibition of the hypothalamus-pituitary-gonadal axis.
  • the bone-related disorder is a loss of bone mineral density (BMD).
  • the bone-related disorder is osteoporosis.
  • the bone-related disorder is osteopenia.
  • the bone-related disorder is increased bone resorption.
  • the bone-related disorder is bone fracture.
  • the bone-related disorder is bone frailty.
  • the bone-related disorder is any combination of osteoporosis, osteopenia, increased bone resorption, bone fracture, bone frailty and loss of BMD.
  • Each disorder represents a separate embodiment of the present invention.
  • “Osteoporosis” refers, in one embodiment, to a thinning of the bones with reduction in bone mass due to depletion of calcium and bone protein.
  • osteoporosis is a systemic skeletal disease, characterized by low bone mass and deterioration of bone tissue, with a consequent increase in bone fragility and susceptibility to fracture.
  • bone strength is abnormal, in one embodiment, with a resulting increase in the risk of fracture.
  • osteoporosis depletes both the calcium and the protein collagen normally found in the bone, in one embodiment, resulting in either abnormal bone quality or decreased bone density.
  • bones that are affected by osteoporosis can fracture with only a minor fall or injury that normally would not cause a bone fracture.
  • the fracture can be, in one embodiment, either in the form of cracking (as in a hip fracture) or collapsing (as in a compression fracture of the spine).
  • the spine, hips, and wrists are common areas of osteoporosis-induced bone fractures, although fractures can also occur in other skeletal areas. Unchecked osteoporosis can lead, in another embodiment, to changes in posture, physical abnormality, and decreased mobility.
  • the osteoporosis results from androgen deprivation.
  • the osteoporosis follows androgen deprivation.
  • the osteoporosis results from estrogen-deprivation therapy.
  • the osteoporosis follows estrogen-deprivation therapy.
  • the osteoporosis is primary osteoporosis. In another embodiment, the osteoporosis is secondary osteoporosis. In another embodiment, the osteoporosis is postmenopausal osteoporosis. In another embodiment, the osteoporosis is juvenile osteoporosis. In another embodiment, the osteoporosis is idiopathic osteoporosis. In another embodiment, the osteoporosis is senile osteoporosis. In another embodiment, osteoporosis can predispose a breast cancer patient to metastasis to the bones and/or predispose the patients toward the development of a skeletally related event.
  • the primary osteoporosis is type I primary osteoporosis. In another embodiment, the primary osteoporosis is type II primary osteoporosis. Each type of osteoporosis represents a separate embodiment of the present invention.
  • the bone-related disorder is treated with a compound as herein described, or a combination thereof.
  • other bone-stimulating compounds can be provided to the subject, prior to, concurrent with or following administration of a compound or compounds as herein described.
  • such a bone stimulating compound may comprise natural or synthetic materials.
  • the bone stimulating compound may comprise a bone morphogenetic protein (BMP), a growth factor, such as epidermal growth factor (EGF), a fibroblast growth factor (FGF), a transforming growth factor (TGF, an insulin growth factor (IGF), a platelet-derived growth factor (PDGF) hedgehog proteins such as sonic, indian and desert hedgehog, a hormone such as follicle stimulating hormone, parathyroid hormone, parathyroid hormone related peptide, activins, inhibins, follistatin, frizzled, frzb or frazzled proteins, BMP binding proteins such as chordin and fetuin, a cytokine such as IL-3, IL-7, GM- CSF, a chemokine, such as eotaxin, a collagen, osteocalcin, osteonectin and others, as will be appreciated by one skilled in the art.
  • BMP bone morphogenetic protein
  • a growth factor such as epidermal growth factor (EGF), a fibro
  • compositions for use in treating a bone disorder of this invention may comprise a compound or compounds as herein described, an additional bone stimulating compound, or compounds, and osteogenic cells.
  • an osteogenic cell may be a stem cell or progenitor cell, which may be induced to differentiate into an osteoblast.
  • the cell may be an osteoblast.
  • nucleic acids which encode bone-stimulating compounds may be administered to the subject, which is to be considered as part of this invention.
  • this invention provides for the treatment, prevention, suppression or inhibition of, or the reduction of the risk of developing a skeletal-related event (SRE), such as bone fractures, surgery of the bone, radiation of the bone, spinal cord compression, new bone metastasis, bone loss, or a combination thereof in a subject with cancer, comprising administering a compound as herein described and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N- oxide, or any combination thereof.
  • SRE skeletal-related event
  • the invention relates, inter alia, to treatment of an SRE with the compound of Formulae I-XIV of this invention: (a) in a subject with prostate cancer undergoing or having undergone androgen deprivation therapy (ADT); or (b) in a subject with breast cancer undergoing or having undergone estrogen-deprivation therapy.
  • the skeletal-related events treated using the methods provided herein and/or utilizing the compositions provided herein are fractures, which in one embodiment, are pathological fractures, non-traumatic fractures, vertebral fracture, non- vertebral fractures, morphometric fractures, or a combination thereof.
  • fractures may be simple, compound, transverse, greenstick, or comminuted fractures.
  • fractures may be to any bone in the body, which in one embodiment, is a fracture in any one or more bones of the arm, wrist, hand, finger, leg, ankle, foot, toe, hip, collar bone, or a combination thereof. In breast cancer, metastasis occurs most often to the hip and vertebrae.
  • the skeletal-related is fractures to the hip and/or vertebrae.
  • the methods and/or compositions provided herein are effective in treatment, prevention, suppression, inhibition or reduction of the risk of skeletal- related events such as pathologic fractures, spinal cord compression, hypercalcemia, bone- related pain, or their combination.
  • the skeletal-related events sought to be treated using the methods provided herein and/or utilizing the compositions provided herein comprise the necessity for bone surgery and/or bone radiation, which in some embodiments, is for the treatment of pain resulting in one embodiment from bone damage, or nerve compression.
  • the skeletal-related events sought to be treated using the methods provided herein and/or utilizing the compositions provided herein comprise spinal cord compression, or the necessity for changes in antineoplastic therapy, including changes in hormonal therapy, in a subject.
  • skeletal-related events sought to be treated using the methods provided herein and/or utilizing the compositions provided herein comprise treating, suppressing, preventing, reducing the incidence of, or delaying progression or severity of bone metastases, or bone loss.
  • bone loss may comprise osteoporosis, osteopenia, or a combination thereof.
  • skeletal-related events may comprise any combination of the embodiments listed herein. [000191]
  • the methods provided herein and/or utilizing the compositions provided herein are effective in reducing metastases to the bone, such as in terms of number of foci, the size of foci, or a combination thereof.
  • a method of preventing or inhibiting cancer metastasis to bone in a subject comprising the step of administering to the subject a composition comprising toremifene, raloxifene, tamoxifen or an analogue, functional derivative, metabolite or a combination thereof, or a pharmaceutically acceptable salt thereof.
  • metabolites may comprise ospemifene, fispemifene or their combination.
  • the cancer is prostate cancer.
  • the cancer is breast cancer.
  • the skeletal-related events are a result of cancer therapy.
  • the skeletal-related events are a result of hormone deprivation therapy, while in another embodiment, they are a product of androgen deprivation therapy (ADT), and in another embodiment they are a product of estrogen-deprivation therapy
  • ADT androgen deprivation therapy
  • the term “libido” may refer to sexual desire, or as defined in Example 9.
  • the term “quality of life” may refer to the focuses on the health and life of a subject suffering from a condition or disease, for example suffering from breast cancer, post treatment until the end of life. It covers the physical, psychosocial, and economic issues faced by the subject, beyond the diagnosis and treatment phases.
  • the term “quality of life” may also be referred to herein as "survivorship".
  • survivorship includes issues related to the ability to get health care and follow-up treatment, late effects of treatment, second cancers, and quality of life. Family members, friends, and caregivers are also considered part of the survivorship experience.
  • the methods of this invention are useful to a subject, which is a human.
  • the subject is male.
  • the subject is female.
  • the methods as described herein may be useful for treating either males or females, females may respond more advantageously to administration of certain compounds, for certain methods.
  • males may respond more advantageously to administration of certain compounds, for certain methods.
  • the compound of this invention which is effective at: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin
  • SERM selective estrogen receptor modulator
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject, for example a female subject. Accordingly, this invention provides methods for: a) treating AR-positive breast cancer in a subject; b) treating metastatic AR-positive breast cancer, or advanced AR-positive breast cancer; c) treating refractory AR- positive breast cancer; d) treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer; e) prolonging progression-free survival of a subject suffering from breast cancer; f) treating a subject suffering from ER-positive breast cancer; g) treating a subject suffering from metastatic ER-positive breast cancer; h) treating a subject suffering from refractory ER-positive breast cancer; i) treating a subject suffering from AR-positive ER- positive breast cancer; j) treating a subject suffering from AR-positive ER-positive refractory breast cancer; k) treating a subject suffering from AR-positive ER-positive metastatic breast cancer;
  • this invention provides methods for: a) treating a subject suffering from HER2-positive breast cancer; b) treating a subject suffering from HER2- positive refractory breast cancer; c) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e) treating a subject suffering from HER2-positive and ER-positive breast cancer; f) treating a subject suffering from HER2-positive and PR-positive breast cancer; g) treating a subject suffering from HER2-positive and PR-negative breast cancer; h) treating a subject suffering from HER2-positive and AR-positive breast cancer; i) treating a subject suffering from HER2-positive and AR-negative breast cancer; j) treating a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject suffering from HER2-positive, HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject
  • X is a bond, O, CH 2 , NH, S, Se, PR, NO or NR; G is O or S; T is OH, OR, -NHCOCH 3 , or NHCOR; R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F, CHF 2 , CF 3 , CF 2 CF 3 , aryl, phenyl, halogen, alkenyl or OH; R 1 is CH 3 , CH 2 F, CHF 2 , CF 3 , CH 2 CH 3 , or CF 2 CF 3 ; R 2 is H, F, Cl, Br, I, CH 3 , CF 3 , OH, CN, NO 2 , NHCOCH 3 , NHCOCF 3 , NHCOR, alkyl, arylalkyl, OR, NH 2 , NHR, N(R) 2 , or SR; R 3 is H, F, Cl, Br, I,
  • the subject is a female subject. In one embodiment, the subject is a male subject.
  • G in Formula I is O. In another embodiment, X in Formula I is O. In another embodiment, T in Formula I is OH. In another embodiment, R 1 in Formula I is CH 3 . In another embodiment, Z in Formula I is NO 2 . In another embodiment, Z in Formula I is CN. In another embodiment, Y in Formula I is CF 3 . In another embodiment, Y in Formula I is Cl. In another embodiment, Q in Formula I is CN. In another embodiment, Q in Formula I is halogen. In another embodiment, Q in Formula I is F. In another embodiment, Q in Formula I is Cl. In another embodiment, Q in Formula I is NHCOCH 3 .
  • Q in Formula I is CN and R 2 is F. In another embodiment, Q in Formula I is Cl and R 2 is F. In another embodiment, Q in Formula I is in the para position. In another embodiment, Z in Formula I is in the para position. In another embodiment, Y in Formula I is in the meta position. [000200]
  • the substituents Z, Y and R 3 can be in any position of the ring carrying these substituents (hereinafter “A ring”). In one embodiment, the substituent Z is in the para position of the A ring. In another embodiment, the substituent Y is in the meta position of the A ring. In another embodiment, the substituent Z is in the para position of the A ring and substituent Y is in the meta position of the A ring.
  • the substituents Q and R2 can be in any position of the ring carrying these substituents (hereinafter “B ring”).
  • the substituent Q is in the para position of the B ring.
  • the substituent R 2 is in the meta position of the B ring.
  • the substituent Q is CN and is in the para position of the B ring.
  • the substituents R 2 and R 3 are not limited to one particular substituent, and can be any combination of the substituents listed above.
  • the compound of this invention which is effective at: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin),
  • SERM selective estrogen receptor
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject, for example a female subject. Accordingly, this invention provides methods for: a) treating AR-positive breast cancer in a subject; b) treating metastatic AR-positive breast cancer, or advanced AR-positive breast cancer; c) treating refractory AR- positive breast cancer; d) treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer; e) prolonging progression-free survival of a subject suffering from breast cancer; f) treating a subject suffering from ER-positive breast cancer; g) treating a subject suffering from metastatic ER-positive breast cancer; h) treating a subject suffering from refractory ER-positive breast cancer; i) treating a subject suffering from AR-positive ER- positive breast cancer; j) treating a subject suffering from AR-positive ER-positive refractory breast cancer; k) treating a subject suffering from AR-positive ER-positive metastatic breast cancer;
  • this invention provides methods for: a) treating a subject suffering from HER2-positive breast cancer; b) treating a subject suffering from HER2- positive refractory breast cancer; c) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e) treating a subject suffering from HER2-positive and ER-positive breast cancer; f) treating a subject suffering from HER2-positive and PR-positive breast cancer; g) treating a subject suffering from HER2-positive and PR-negative breast cancer; h) treating a subject suffering from HER2-positive and AR-positive breast cancer; i) treating a subject suffering from HER2-positive and AR-negative breast cancer; j) treating a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject suffering from HER2-positive, HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject
  • the subject is a female subject. In one embodiment, the subject is a male subject.
  • G in Formula II is O. In another embodiment, X in Formula II is O. In another embodiment, T in Formula II is OH. In another embodiment, R 1 in Formula II is CH 3 . In another embodiment, Z in Formula II is NO 2 . In another embodiment, Z in Formula II is CN. In another embodiment, Y in Formula II is CF 3 . In another embodiment, Y in Formula II is halogen. In another embodiment, Y in Formula II is Cl. In another embodiment, Q in Formula II is CN. In another embodiment, Q in Formula II is halogen. In another embodiment, Q in Formula II is Cl. In another embodiment, Q in Formula II is F.
  • Q in Formula II is NHCOCH 3 .
  • Q in Formula II is in the para position.
  • Z in Formula II is in the para position.
  • Y in Formula II is in the meta position.
  • G in Formula II is O, T is OH, R 1 is CH 3 , X is O, Z is CN, Y is CF 3 or halogen and Q is CN or F.
  • G in Formula II is O, T is OH, R 1 is CH 3 , X is O, Z is NO 2 , Y is CF 3 and Q is NHCOCH 3 , F or Cl.
  • the substituents Z and Y can be in any position of the ring carrying these substituents (hereinafter “A ring”). In one embodiment, the substituent Z is in the para position of the A ring. In another embodiment, the substituent Y is in the meta position of the A ring. In another embodiment, the substituent Z is in the para position of the A ring and substituent Y is in the meta position of the A ring. [000208]
  • the substituent Q can be in any position of the ring carrying this substituent (hereinafter “B ring”). In one embodiment, the substituent Q is in the para position of the B ring. In another embodiment, the substituent Qis CN and is in the para position of the B ring.
  • the compound of this invention which is effective at: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin
  • SERM selective estrogen receptor modul
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject, for example a female subject. Accordingly, this invention provides methods for: a) treating AR-positive breast cancer in a subject; b) treating metastatic AR-positive breast cancer, or advanced AR-positive breast cancer; c) treating refractory AR- positive breast cancer; d) treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer; e) prolonging progression-free survival of a subject suffering from breast cancer; f) treating a subject suffering from ER-positive breast cancer; g) treating a subject suffering from metastatic ER-positive breast cancer; h) treating a subject suffering from refractory ER-positive breast cancer; i) treating a subject suffering from AR-positive ER- positive breast cancer; j) treating a subject suffering from AR-positive ER-positive refractory breast cancer; k) treating a subject suffering from AR-positive ER-positive metastatic breast cancer;
  • this invention provides methods for: a) treating a subject suffering from HER2-positive breast cancer; b) treating a subject suffering from HER2- positive refractory breast cancer; c) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e) treating a subject suffering from HER2-positive and ER-positive breast cancer; f)reating a subject suffering from HER2-positive and PR-positive breast cancer; g) treating a subject suffering from HER2-positive and PR-negative breast cancer; h) treating a subject suffering from HER2-positive and AR-positive breast cancer; i) treating a subject suffering rom HER2-positive and AR-negative breast cancer; j) treating a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject suffering from a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating
  • the subject is a female subject. In one embodiment, the subject is a male subject.
  • Z in Formula III is NO 2 . In another embodiment, Z in Formula III is CN. In another embodiment, Y in Formula III is CF 3 . In another embodiment, Y in Formula III is Cl. In another embodiment, Y in Formula III is halogen. In another embodiment, Q in Formula III is CN. In another embodiment, Q in Formula III is halogen. In another embodiment, Q in Formula III is F. In another embodiment, Q in Formula III is Cl. In another embodiment, Q in Formula III is NHCOCH 3 . In another embodiment, Z is CN, Y is CF 3 or halogen, and Q is CN or F.
  • Z is NO 2
  • Y is CF 3
  • Q is NHCOCH 3 , F or Cl.
  • the compound of this invention which is effective at: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, torem
  • SERM selective estrogen receptor modulator
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject, for example a female subject. Accordingly, this invention provides methods for: a) treating AR-positive breast cancer in a subject; b) treating metastatic AR-positive breast cancer, or advanced AR-positive breast cancer; c) treating refractory AR- positive breast cancer; d) treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer; e) prolonging progression-free survival of a subject suffering from breast cancer; f) treating a subject suffering from ER-positive breast cancer; g) treating a subject suffering from metastatic ER-positive breast cancer; h) treating a subject suffering from refractory ER-positive breast cancer; i) treating a subject suffering from AR-positive ER- positive breast cancer; j) treating a subject suffering from AR-positive ER-positive refractory breast cancer; k) treating a subject suffering from AR-positive ER-positive metastatic breast cancer;
  • this invention provides methods for: a) treating a subject suffering from HER2-positive breast cancer; b) treating a subject suffering from HER2- positive refractory breast cancer; c) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e) treating a subject suffering from HER2-positive and ER-positive breast cancer; f) treating a subject suffering from HER2-positive and PR-positive breast cancer; g) treating a subject suffering from HER2-positive and PR-negative breast cancer; h) treating a subject suffering from HER2-positive and AR-positive breast cancer; i) treating a subject suffering from HER2-positive and AR-negative breast cancer; j) treating a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject suffering from HER2- positive, ER-positive, PR-positive breast cancer; k) treating a subject suffering from HER2- positive, ER-
  • the subject is a female subject. In one embodiment, the subject is a male subject.
  • G in Formula IV is O. In another embodiment, X in Formula IV is O. In another embodiment, T in Formula IV is OH. In another embodiment, R 1 in Formula IV is CH 3 . In another embodiment, Z in Formula IV is NO 2 . In another embodiment, Z in Formula IV is CN. In another embodiment, Y in Formula IV is CF 3 . In another embodiment, Y in Formula IV is halogen. In another embodiment, Y in Formula IV is Cl. In another embodiment, Q 1 in Formula II is CN. In another embodiment, Q 1 in Formula IV is F. In another embodiment, Q 1 in Formula IV is Cl.
  • Q 1 in Formula II is NHCOCH 3 .
  • Q 1 in Formula IV is in the para position.
  • Z in Formula IV is in the para position.
  • Y in Formula IV is in the meta position.
  • G in Formula IV is O, T is OH, R 1 is CH 3 , X is O, Z is NO 2 or CN, Y is CF 3 or halogen and Q 1 is CN, F, Cl, or NHCOCH 3 .
  • the substituents Z and Y can be in any position of the ring carrying these substituents (hereinafter “A ring”). In one embodiment, the substituent Z is in the para position of the A ring.
  • the substituent Y is in the meta position of the A ring.
  • the substituent Z is in the para position of the A ring and substituent Y is in the meta position of the A ring.
  • the substituents Q 1 and Q 2 can be in any position of the ring carrying these substituents (hereinafter “B ring”).
  • the substituent Q 1 is in the para position of the B ring.
  • the substituent is Q 2 is H.
  • the substituent Q 1 is in the para position of the B ring and the substituent is Q 2 is H.
  • the substituent Q 1 is CN and is in the para position of the B ring, and the substituent is Q 2 is H.
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject, for example a female subject. Accordingly, this invention provides methods for: a) treating AR-positive breast cancer in a subject; b) treating metastatic AR-positive breast cancer, or advanced AR-positive breast cancer; c) treating refractory AR- positive breast cancer; d) treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer; e) prolonging progression-free survival of a subject suffering from breast cancer; f) treating a subject suffering from ER-positive breast cancer; g) treating a subject suffering from metastatic ER-positive breast cancer; h) treating a subject suffering from refractory ER-positive breast cancer; i) treating a subject suffering from AR-positive ER- positive breast cancer; j) treating a subject suffering from AR-positive ER-positive refractory breast cancer; k) treating a subject suffering from AR-positive ER-positive metastatic breast cancer;
  • this invention provides methods for: a) treating a subject suffering from HER2-positive breast cancer; b) treating a subject suffering from HER2- positive refractory breast cancer; c) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e) treating a subject suffering from HER2-positive and ER-positive breast cancer; f) treating a subject suffering from HER2-positive and PR-positive breast cancer; g) treating a subject suffering from HER2-positive and PR-negative breast cancer; h) treating a subject suffering from HER2-positive and AR-positive breast cancer; i) treating a subject suffering from HER2-positive and AR-negative breast cancer; j) treating a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject suffering from HER2- positive, ER-positive, PR-positive breast cancer; k) treating a subject suffering from HER2- positive, ER-
  • Q is CN, alkyl, halogen, N(R) 2 , NHCOCH 3 , NHCOCF 3 , NHCOR, NHCONHR, NHCOOR, OCONHR, CONHR, NHCSCH 3 , NHCSCF 3 , NHCSR, NHSO 2 CH 3 , NHSO 2 R, OR, COR, OCOR, OSO 2 R, SO 2 R or SR; or Q together with the benzene ring to which it is attached is a fused ring system represented by structure A, B or C: ; R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F, CHF 2 , CF 3 , CF 2 CF 3 , aryl, phenyl, halogen, alkenyl or OH; and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any
  • the subject is a female subject. In one embodiment, the subject is a male subject.
  • Q in Formula V or VI is CN. In one embodiment, Q in Formula V or VI is halogen. In one embodiment, Q in Formula V or VI is F. In one embodiment, Q in Formula V or VI is Cl. In one embodiment, Q in Formula V or VI is NHCOCH 3 .
  • the compound of this invention which is effective at: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin
  • SERM selective estrogen receptor modul
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject, for example a female subject. Accordingly, this invention provides methods for: a) treating AR-positive breast cancer in a subject; b) treating metastatic AR-positive breast cancer, or advanced AR-positive breast cancer; c) treating refractory AR- positive breast cancer; d) treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer; e) prolonging progression-free survival of a subject suffering from breast cancer; f) treating a subject suffering from ER-positive breast cancer; g) treating a subject suffering from metastatic ER-positive breast cancer; h) treating a subject suffering from refractory ER-positive breast cancer; i) treating a subject suffering from AR-positive ER- positive breast cancer; j) treating a subject suffering from AR-positive ER-positive refractory breast cancer; k) treating a subject suffering from AR-positive ER-positive metastatic breast cancer;
  • this invention provides methods for: a) treating a subject suffering from HER2-positive breast cancer; b) treating a subject suffering from HER2- positive refractory breast cancer; c) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e) treating a subject suffering from HER2-positive and ER-positive breast cancer; f) treating a subject suffering from HER2-positive and PR-positive breast cancer; g) treating a subject suffering from HER2-positive and PR-negative breast cancer; h) treating a subject suffering from HER2-positive and AR-positive breast cancer; i) treating a subject suffering from HER2-positive and AR-negative breast cancer; j) treating a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e)
  • the subject is a female subject. In one embodiment, the subject is a male subject.
  • SERM selective estrogen receptor modulator
  • this invention provides methods for: a) treating AR-positive breast cancer in a subject; b) treating metastatic AR-positive breast cancer, or advanced AR-positive breast cancer; c) treating refractory AR- positive breast cancer; d) treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer; e) prolonging progression-free survival of a subject suffering from breast cancer; f) treating a subject suffering from ER-positive breast cancer; g) treating a subject suffering from metastatic ER-positive breast cancer; h) treating a subject suffering from refractory ER-positive breast cancer; i) treating a subject suffering from AR-positive ER- positive breast cancer; j) treating a subject suffering from AR-positive ER-positive refractory breast cancer; k) treating a subject suffering from AR-positive ER-positive metastatic breast cancer; l) treating a subject suffering from AR-positive and ER-positive breast cancer; m) treating a subject suffering from AR-positive ER-positive breast cancer;
  • this invention provides methods for: a) treating a subject suffering from HER2-positive breast cancer; b) treating a subject suffering from HER2- positive refractory breast cancer; c) treating a subject suffering from HER2-positive metastatic breast cancer; d) treating a subject suffering from HER2-positive and ER-negative breast cancer; e) treating a subject suffering from HER2-positive and ER-positive breast cancer; f) treating a subject suffering from HER2-positive and PR-positive breast cancer; g) treating a subject suffering from HER2-positive and PR-negative breast cancer; h) treating a subject suffering from HER2-positive and AR-positive breast cancer; i) treating a subject suffering from HER2-positive and AR-negative breast cancer; j) treating a subject suffering from HER2- positive, ER-positive, PR-positive, and AR-positive breast cancer; k) treating a subject suffering from HER2- positive, ER-positive, PR-positive breast cancer; k) treating a subject suffering from HER2- positive, ER-
  • the subject is a female subject. In one embodiment, the subject is a male subject. [000235] In one embodiment, the methods of this invention make use of a compound of Formula VIII. In one embodiment, the methods of this invention make use of a compound of Formula IX. In one embodiment, the methods of this invention make use of a compound of Formula X. In one embodiment, the methods of this invention make use of a compound of Formula XI. In one embodiment, the methods of this invention make use of a compound of Formula XII. In one embodiment, the methods of this invention make use of a compound of Formula XIII. In one embodiment, the methods of this invention make use of a compound of Formula XIV.
  • the methods of the present invention comprise administering an analog of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering a derivative of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering an isomer of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering a metabolite of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering a pharmaceutically acceptable salt of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering a pharmaceutical product of the compound of Formulae I-XIV.
  • the methods of the present invention comprise administering a hydrate of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering an N-oxide of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering a polymorph of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering a crystal of the compound of Formulae I-XIV. In another embodiment, the methods of the present invention comprise administering a prodrug of the compound of Formulae I-XIV.
  • the methods of the present invention comprise administering a combination of any of an analog, derivative, metabolite, isomer, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, polymorph, crystal or prodrug of the compound of Formulae I-XIV.
  • the methods of this invention comprise administering a compound of Formulae I-XIV.
  • the methods of this invention comprise administering a compound of Formula I.
  • the methods of this invention comprise administering a compound of Formula II.
  • the methods of this invention comprise administering a compound of Formula III.
  • the methods of this invention comprise administering a compound of Formula IV.
  • the methods of this invention comprise administering a compound of Formula V.
  • the methods of this invention comprise administering a compound of Formula VI. In another embodiment, the methods of this invention comprise administering a compound of Formula VII. In another embodiment, the methods of this invention comprise administering a compound of Formula VIII. In another embodiment, the methods of this invention comprise administering a compound of Formula IX. In another embodiment, the methods of this invention comprise administering a compound of Formula X. In another embodiment, the methods of this invention comprise administering a compound of Formula XI. In another embodiment, the methods of this invention comprise administering a compound of Formula XII. In another embodiment, the methods of this invention comprise administering a compound of Formula XIII. In another embodiment, the methods of this invention comprise administering a compound of Formula XIV.
  • the compounds of the present invention are useful for: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER- positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist
  • SERM selective estrogen receptor modulator
  • the compounds of the present invention offer a significant advance over steroidal androgen treatment since treatment of breast cancer with these compounds will not be accompanied by serious side effects, inconvenient modes of administration, or high costs and still have the advantages of oral bioavailability, lack of cross-reactivity with other steroid receptors, lack of aromatizability, and long biological half-lives.
  • this invention relates to the treatment of androgen receptor- positive breast cancer in a subject.
  • this invention provides methods of: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR-positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast cancer that has failed selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI)
  • the term “isomer” includes, but is not limited to, optical isomers and analogs, structural isomers and analogs, conformational isomers and analogs, and the like. As used herein, the term “isomer” may also be referred to herein as an "enantiomer” having all of the qualities and properties of an "isomer”. [000242] In one embodiment, this invention encompasses the use of various optical isomers of the selective androgen receptor modulator. It will be appreciated by those skilled in the art that the selective androgen receptor modulators of the present invention contain at least one chiral center.
  • the selective androgen receptor modulators used in the methods of the present invention may exist in, and be isolated in, optically-active or racemic forms. Some compounds may also exhibit polymorphism. It is to be understood that the present invention encompasses any racemic, optically-active, polymorphic, or stereoisomeric form, or any combination thereof, which form possesses properties useful in the treatment of androgen- related conditions described herein.
  • the selective androgen receptor modulators are the pure (R)-isomers.
  • the selective androgen receptor modulators are the pure (S)-isomers.
  • the selective androgen receptor modulators are a mixture of the (R) and the (S) isomers.
  • the selective androgen receptor modulators are a racemic mixture comprising an equal amount of the (R) and the (S) isomers. It is well known in the art how to prepare optically-active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
  • the invention includes “pharmaceutically acceptable salts” of the compounds of this invention, which may be produced, by reaction of a compound of this invention with an acid or base.
  • the invention includes pharmaceutically acceptable salts of amino-substituted compounds with organic and inorganic acids, for example, citric acid and hydrochloric acid.
  • the invention also includes N-oxides of the amino substituents of the compounds described herein.
  • Pharmaceutically acceptable salts can also be prepared from the phenolic compounds by treatment with inorganic bases, for example, sodium hydroxide.
  • esters of the phenolic compounds can be made with aliphatic and aromatic carboxylic acids, for example, acetic acid and benzoic acid esters.
  • Suitable pharmaceutically acceptable salts of the compounds of Formulae I-XIV may be prepared from an inorganic acid or from an organic acid.
  • examples of inorganic salts of the compounds of this invention are bisulfates, borates, bromides, chlorides, hemisulfates, hydrobromates, hydrochlorates, 2-hydroxyethylsulfonates (hydroxyethanesulfonates), iodates, iodides, isothionates, nitrates, persulfates, phosphate, sulfates, sulfamates, sulfanilates, sulfonic acids (alkylsulfonates, arylsulfonates, halogen substituted alkylsulfonates, halogen substituted arylsulfonates), sulfonates and thiocyanates.
  • examples of organic salts of the compounds of this invention may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which are acetates, arginines, aspartates, ascorbates, adipates, anthranilates, algenates, alkane carboxylates, substituted alkane carboxylates, alginates, benzenesulfonates, benzoates, bisulfates, butyrates, bicarbonates, bitartrates, citrates, camphorates, camphorsulfonates, cyclohexylsulfamates, cyclopentanepropionates, calcium edetates, camsylates, carbonates, clavulanates, cinnamates, dicarboxylates, digluconates, dodecylsulfonates, dihydrochlorides, decan
  • the salts may be formed by conventional means, such as by reacting the free base or free acid form of the product with one or more equivalents of the appropriate acid or base in a solvent or medium in which the salt is insoluble or in a solvent such as water, which is removed in vacuo or by freeze drying or by exchanging the ions of an existing salt for another ion or suitable ion-exchange resin.
  • This invention further includes derivatives of the selective androgen receptor modulators.
  • derivatives includes but is not limited to ether derivatives, acid derivatives, amide derivatives, ester derivatives and the like.
  • this invention further includes hydrates of the selective androgen receptor modulators.
  • hydrate includes but is not limited to hemihydrate, monohydrate, dihydrate, trihydrate and the like.
  • This invention further includes metabolites of the selective androgen receptor modulators.
  • metabolite means any substance produced from another substance by metabolism or a metabolic process.
  • pharmaceutical products of the selective androgen receptor modulators means a composition suitable for pharmaceutical use (pharmaceutical composition), as defined herein.
  • pharmaceutical composition suitable for pharmaceutical use (pharmaceutical composition), as defined herein.
  • This invention further includes prodrugs of the selective androgen receptor modulators.
  • prodrug means a substance which can be converted in vivo into a biologically active agent by such reactions as hydrolysis, esterification, de-esterification, activation, salt formation and the like.
  • This invention further includes crystals of the selective androgen receptor modulators.
  • this invention provides polymorphs of the selective androgen receptor modulators.
  • crystal means a substance in a crystalline state.
  • polymorph refers to a particular crystalline state of a substance, having particular physical properties such as X-ray diffraction, IR spectra, melting point, and the like.
  • a method for treating a subject suffering from breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating a subject suffering from metastatic breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat metastatic breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from refractory breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat refractory breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from AR-positive breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • the AR-positive breast cancer is ER, PR and HER2-positive.
  • the AR-positive breast cancer is ER, PR and HER2-negative.
  • the AR-positive breast cancer is ER-positive, and PR and HER2-negative. In another embodiment, the AR-positive breast cancer is ER and PR-positive, and HER2- negative. In yet another embodiment, the AR-positive breast cancer is ER and HER2-positive, and PR-negative. In still another embodiment, the AR-positive breast cancer is ER-negative, and PR and HER2-positive. In a further embodiment, the AR-positive breast cancer is ER and PR-negative, and HER2-positive. In still a further embodiment, the AR-positive breast cancer is ER and HER2-negative, and PR-positive. In one embodiment, the AR-positive breast cancer is ER-negative. In another embodiment, the AR-positive breast cancer is ER-positive.
  • a method for treating a subject suffering from AR-positive refractory breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive refractory breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from AR-positive metastatic breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive metastatic breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from ER-positive breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat ER-positive breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from AR-positive and ER-positive breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive metastatic breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from ER-positive refractory breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat ER-positive refractory breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from ER-positive metastatic breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat ER-positive metastatic breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • an ER-positive breast cancer is AR-positive.
  • an ER-positive breast cancer is AR-negative.
  • a method for treating a subject suffering from advanced breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat advanced breast cancer in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating a subject suffering from AR-positive and ER-positive breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive and ER-positive refractory breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from AR-positive and ER-negative breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat AR-positive and ER-negative metastatic breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from triple negative breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat triple negative breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • SERM selective estrogen receptor modulator
  • GnRH
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method is provided for treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer, comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat, prevent, suppress or inhibit metastasis in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating and/or preventing skeletal related events in a subject suffering comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat and/or prevent skeletal related events in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for improving libido in a subject comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N- oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to improve libido in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for improving quality of life in a subject comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to quality of life in the subject.
  • the subject is a female subject. In another embodiment, the subject is a male subject.
  • a method for treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer comprising the step of administering to the subject a selective androgen receptor modulator of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat, prevent, suppress or inhibit metastasis in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • a method for treating a subject suffering from HER2-positive breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N- oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat HER2-positive breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • the HER2-positive breast cancer is HER2-positive refractory breast cancer.
  • the HER2-positive breast cancer is HER2-positive metastatic breast cancer.
  • the HER2-positive breast cancer is ER-negative. In another embodiment, the HER2-positive breast cancer is ER-positive. In one embodiment, the HER2-positive breast cancer is PR-positive. In another embodiment, the HER2-positive breast cancer is PR-negative. In one embodiment, the HER2-positive breast cancer is AR- positive. In another embodiment, the HER2-positive breast cancer is AR-negative. [000277] In certain embodiment, the HER2-positive breast cancer is ER-positive, PR- positive, and AR-positive. In another embodiment, the HER2-positive breast cancer is ER- positive, PR-negative, and AR-positive. In another embodiment, the HER2-positive breast cancer is ER-positive, PR-negative, and AR-negative.
  • the HER2- positive breast cancer is ER-positive, PR-positive, and AR-negative. In another embodiment, the HER2-positive breast cancer is ER-negative, PR-negative, and AR-positive. In another embodiment, the HER2-positive breast cancer is ER-negative, PR-positive, and AR-positive. In other embodiment, the HER2-positive breast cancer is ER-negative, PR-positive, and AR- negative. In certain embodiment, the HER2-positive breast cancer is ER-negative, PR- negative, and AR-negative.
  • a method for treating a subject suffering from ER mutant expressing breast cancer comprising the step of administering to the subject a compound of Formulae I-XIV of this invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat ER mutant expressing breast cancer in the subject.
  • the subject is a female subject.
  • the subject is a male subject.
  • the ER mutant expressing breast cancer is Y537S mutation expressing breast cancer.
  • the ER mutant expressing breast cancer is D351Y mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is E380Q mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is V422del mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is S432L mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is G442A mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is S463P mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L469V mutation expressing breast cancer.
  • the ER mutant expressing breast cancer is L536R mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L536H mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L536P mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is L536Q mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is Y537N mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is Y537C mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is Y537D mutation expressing breast cancer.
  • the ER mutant expressing breast cancer is D538G mutation expressing breast cancer. In a certain embodiment, the ER mutant expressing breast cancer is E542G mutation expressing breast cancer. In one embodiment, ER mutant expressing breast cancer refers to mutants of ER-alpha. [000280] In a certain embodiment, the ER mutant expressing breast cancer is as described in Cancer Cell 2018, 33, 173–186, or in Nat Rev Cancer. 2018 Jun;18(6):377-388, which are incorporated herein by reference. In one embodiment, ER mutant expressing breast cancer refers to mutants of ER-alpha.
  • the substituent R is defined herein as an alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F, CHF 2 , CF 3 , CF 2 CF 3 ; aryl, phenyl, halogen, alkenyl, or hydroxyl (OH).
  • An “alkyl” group refers to a saturated aliphatic hydrocarbon, including straight- chain, branched-chain and cyclic alkyl groups. In one embodiment, the alkyl group has 1-12 carbons. In another embodiment, the alkyl group has 1-7 carbons. In another embodiment, the alkyl group has 1-6 carbons. In another embodiment, the alkyl group has 1-4 carbons.
  • the alkyl group may be unsubstituted or substituted by one or more groups selected from halogen, hydroxy, alkoxy carbonyl, amido, alkylamido, dialkylamido, nitro, amino, alkylamino, dialkylamino, carboxyl, thio and thioalkyl.
  • a “haloalkyl” group refers to an alkyl group as defined above, which is substituted by one or more halogen atoms, e.g., by F, Cl, Br or I.
  • aryl group refers to an aromatic group having at least one carbocyclic aromatic group or heterocyclic aromatic group, which may be unsubstituted or substituted by one or more groups selected from halogen, haloalkyl, hydroxy, alkoxy carbonyl, amido, alkylamido, dialkylamido, nitro, amino, alkylamino, dialkylamino, carboxy or thio or thioalkyl.
  • Nonlimiting examples of aryl rings are phenyl, naphthyl, pyranyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyrazolyl, pyridinyl, furanyl, thiophenyl, thiazolyl, imidazolyl, isoxazolyl, and the like.
  • a “hydroxyl” group refers to an OH group.
  • An “alkenyl” group refers to a group having at least one carbon to carbon double bond.
  • a halo group refers to F, Cl, Br or I.
  • arylalkyl refers to an alkyl bound to an aryl, wherein alkyl and aryl are as defined above.
  • An example of an aralkyl group is a benzyl group.
  • the selective androgen receptor modulators provided herein are a new class of compounds, which suppress growth of AR-positive breast cancers.
  • the compounds of this invention have a tissue-selective myoanabolic activity profile of a nonsteroidal ligand for the androgen receptor.
  • compounds of the present invention are non-aromatizable, non-virilizing, and are not commonly cross-reactive with ER and PR.
  • the selective androgen receptor modulators (SARMs) of the present invention are beneficial to refractory breast cancer patients undergoing chemotherapy due to anabolism.
  • the appropriately substituted selective androgen receptor modulators of the present invention are useful for: a) treating a subject suffering from breast cancer; b) treating a subject suffering from metastatic breast cancer; c) treating a subject suffering from refractory breast cancer; d) treating a subject suffering from AR-positive breast cancer; e) treating a subject suffering from AR-positive refractory breast cancer; f) treating a subject suffering from AR-positive metastatic breast cancer; g) treating a subject suffering from AR-positive and ER-positive breast cancer; h) treating a subject suffering from AR- positive breast cancer with or without expression of ER, PR, and/or HER2; i) treating a subject suffering from triple negative breast cancer; j) treating a subject suffering from advanced breast cancer; k) treating a subject suffering from breast
  • a “refractory breast cancer” is a breast cancer that has not responded to treatment.
  • a “refractory breast cancer” is a breast cancer resistant to treatment.
  • refractory breast cancer is refractory metastatic breast cancer.
  • refractory breast cancer has not responded to treatment with anthracyclines, taxanes, capecitabine, ixabepilone, selective estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin-releasing hormone (GnRH) agonist (goserelin), aromatase inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), ribociclib (Kisqali), abemaciclib (Vorzenio), trilaciclib, lerociclib), alpelisib (Piqray) (an inhibitor of phosphatidylinositol-3-kinase subunit alpha (PI3K ⁇ )), mTOR inhibitor (everolimus), poly ADP ribose polymerase (PARP) inhibitor (o
  • a “triple negative breast cancer” is defined by lack of expression of estrogen, progesterone, and ErbB2 (also known as human epidermal growth factor receptor 2 (HER2)) receptors. This subgroup accounts for 15% of all types of breast cancer. This subtype of breast cancer is clinically characterized as more aggressive and less responsive to standard treatment and associated with poorer overall patient prognosis.
  • the methods of this invention are directed to treating a subject suffering from AR-positive breast cancer, regardless of grade, stage or prior treatments.
  • the methods of this invention are directed to treating a subject suffering from HER2-positive breast cancer, regardless of grade, stage or prior treatments.
  • the methods of this invention are first, second, third, or fourth line therapies for breast cancer.
  • a first line therapy refers to a medical therapy recommended for the initial treatment of a disease, sign or symptom.
  • a second line therapy therapy is given when initial treatment (first-line therapy) does not work, or stops working.
  • Third line therapy is given when both initial treatment (first-line therapy) and subsequent treatment (second-line therapy) does not work, or stop working, etc.
  • kinases are a group of enzymes that catalyze the transfer of a phosphate group from a donor, such as ADP or ATP, to an acceptor.
  • phosphorylation results in a functional change of the target protein (substrate) by changing enzyme activity, cellular location, or association with other protein kinases.
  • Kinases regulate the majority of cellular pathways, especially those involved in signal transduction.
  • deregulated kinase activity is a frequent cause of disease, in particular cancer, wherein kinases regulate many aspects that control cell growth, movement and death.
  • drugs that inhibit specific kinases are used to treat kinase-related diseases, including cancer.
  • HER2-positive breast cancers are susceptible to HER2 kinase inhibitors (e.g., trastuzumab and lapatinib) and are generally used in metastatic disease.
  • cell signaling receptors receptors for extracellular signaling molecules are collectively referred to as "cell signaling receptors". Many cell signaling receptors are transmembrane proteins on a cell surface; when they bind an extracellular signaling molecule (i.e., a ligand), they become activated so as to generate a cascade of intracellular signals that alter the behavior of the cell. In contrast, in some cases, the receptors are inside the cell and the signaling ligand has to enter the cell to activate them; these signaling molecules therefore must be sufficiently small and hydrophobic to diffuse across the plasma membrane of the cell.
  • Steroid hormones are one example of small hydrophobic molecules that diffuse directly across the plasma membrane of target cells and bind to intracellular cell signaling receptors. These receptors are structurally related and constitute the intracellular receptor superfamily (or steroid-hormone receptor superfamily). Steroid hormone receptors include but are not limited to progesterone receptors, estrogen receptors, androgen receptors, glucocorticoid receptors, and mineralocorticoid receptors. In one embodiment, the present invention is directed to androgen receptors. In one embodiment, the present invention is directed to androgen receptor agonists. In one embodiment, the present invention is directed to progesterone receptors. In one embodiment, the present invention is directed to progesterone receptor antagonists.
  • the receptors can be blocked to prevent ligand binding.
  • affinity If the affinity of a substance is greater than the original hormone, it will compete with the hormone and bind the binding site more frequently.
  • signals may be sent through the receptor into the cells, causing the cell to respond in some fashion. This is called activation. On activation, the activated receptor then directly regulates the transcription of specific genes.
  • the substance and the receptor may have certain attributes, other than affinity, in order to activate the cell. Chemical bonds between atoms of the substance and the atoms of the receptors may form. In some cases, this leads to a change in the configuration of the receptor, which is enough to begin the activation process (called signal transduction).
  • signal transduction the compounds of this invention inhibit the intratumoral expression of genes and pathways that promote breast cancer development through their actions on the AR.
  • a compound of this invention inhibits intratumoral expression of Muc1, SLUG, VCAM1, SPARC or MMP2, or any combination thereof.
  • Formula VIII inhibits gene expression that promotes breast cancer.
  • a receptor antagonist is a substance which binds receptors and inactivates them.
  • a selective androgen receptor modulator is a molecule that exhibits in vivo tissue selectivity, activating signaling activity of the androgen receptor (AR) in anabolic (muscle, bone, etc.) tissues to a greater extent than in the androgenic tissues.
  • the selective androgen receptor modulators of the present invention are useful in binding to and activating steroidal hormone receptors.
  • the SARM compound of the present invention is an agonist which binds the androgen receptor.
  • the compound has high affinity for the androgen receptor.
  • AR agonistic activity can be determined by monitoring the ability of the selective androgen receptor modulators to maintain and/or stimulate the growth of AR containing androgenic tissue such as prostate and seminal vesicles, as measured by weight, in castrated animals.
  • AR antagonistic activity can be determined by monitoring the ability of the selective androgen receptor modulators to inhibit the growth of AR containing tissue in intact animals or counter the effects of testosterone in castrated animals.
  • An androgen receptor (AR) is an androgen receptor of any species, for example a mammal. In one embodiment, the androgen receptor is an androgen receptor of a human.
  • the selective androgen receptor modulators bind reversibly to an androgen receptor of a human. In another embodiment, the selective androgen receptor modulators bind reversibly to an androgen receptor of a mammal.
  • the term “selective androgen receptor modulator” refers to, in one embodiment, a molecule that exhibits in vivo tissue selectivity, activating signaling activity of the androgen receptor in anabolic (muscle, bone, etc.) tissues to a greater extent than in the androgenic tissues.
  • a selective androgen receptor modulator selectively binds the androgen receptor.
  • a selective androgen receptor modulator selectively affects signaling through the androgen receptor.
  • the SARM is a partial agonist.
  • the SARM is a tissue- selective agonist, or in some embodiments, a tissue-selective antagonist.
  • a SARM of this invention exerts its effects on the androgen receptor in a tissue-dependent manner.
  • a SARM of this invention will have an IC 50 or EC50 with respect to AR, as determined using AR transactivation assays, as known in the art, or, in other embodiments, as described herein.
  • IC 50 refers, in some embodiments, to a concentration of the SARM which reduces the activity of a target (e.g., AR) to half-maximal level.
  • EC 50 refers, in some embodiments, to a concentration of the SARM that produces a half-maximal effect.
  • Figure 5 shows that compounds of this invention exhibit AR agonist activity in MDA-MB-231 cells transfected with AR.
  • contacting means that the selective androgen receptor modulators of the present invention are introduced into a sample containing the receptor in a test tube, flask, tissue culture, chip, array, plate, microplate, capillary, or the like, and incubated at a temperature and time sufficient to permit binding of the selective androgen receptor modulators to the receptor.
  • Methods for contacting the samples with the selective androgen receptor modulators or other specific binding components are known to those skilled in the art and may be selected depending on the type of assay protocol to be run. Incubation methods are also standard and are known to those skilled in the art.
  • the term “contacting” means that the selective androgen receptor modulators of the present invention are introduced into a subject receiving treatment, and the selective androgen receptor modulator is allowed to come in contact with the androgen receptor in vivo.
  • the term “treating” includes disorder remitative treatment.
  • the terms “reducing”, “suppressing” and “inhibiting” have their commonly understood meaning of lessening or decreasing.
  • progression means increasing in scope or severity, advancing, growing or becoming worse.
  • the term “recurrence” means the return of a disease after a remission.
  • the term “delaying” means stopping, hindering, slowing down, postponing, holding up or setting back.
  • the term “metastasis” refers to the transfer of a disease from one organ or part thereof to another not directly connected with it. Metastasis can occur for example as a result of transfer of malignant cells from one organ (for example breast) to other organs.
  • "treating" refers to reducing tumor growth by 75%, as demonstrated in, e.g., Example 8.
  • treating refers to reducing tumor growth by at least 75%.
  • treating refers to reducing tumor growth by at least 50%.
  • treating refers to reducing tumor growth by at least 25%.
  • treating refers to reducing tumor growth by 50-100%. In another embodiment, treating refers to reducing tumor growth by 70-80%. In another embodiment, treating refers to reducing tumor growth by 25-125%. [000311] In another embodiment, "treating" refers to reducing tumor weight by 50%, as demonstrated in, e.g., Example 8. In another embodiment, treating refers to reducing tumor weight by at least 50%. In another embodiment, treating refers to reducing tumor weight by at least 40%. In another embodiment, treating refers to reducing tumor weight by at least 30%. In another embodiment, treating refers to reducing tumor weight by at least 20%. In another embodiment, treating refers to reducing tumor weight by 25-75%. In another embodiment, treating refers to reducing tumor weight by 25-100%.
  • administering refers to bringing a subject in contact with a compound of the present invention.
  • administration can be accomplished in vitro, i.e., in a test tube, or in vivo, i.e., in cells or tissues of living organisms, for example humans.
  • the present invention encompasses administering the compounds of the present invention to a subject.
  • a compound of the present invention is administered to a subject once a week.
  • a compound of the present invention is administered to a subject twice a week.
  • a compound of the present invention is administered to a subject three times a week.
  • a compound of the present invention is administered to a subject four times a week. In another embodiment, a compound of the present invention is administered to a subject five times a week. In another embodiment, a compound of the present invention is administered to a subject daily. In another embodiment, a compound of the present invention is administered to a subject weekly. In another embodiment, a compound of the present invention is administered to a subject bi- weekly. In another embodiment, a compound of the present invention is administered to a subject monthly. [000314] In one embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator as the sole active ingredient.
  • methods for hormone therapy for treating breast cancer, for delaying the progression of breast cancer, and for preventing and treating the recurrence of breast cancer and/or breast cancer metastasis, which comprise administering the selective androgen receptor modulators in combination with one or more therapeutic agents.
  • SERM selective estrogen receptor modulator
  • GnRH gonadotropin-releasing hormone
  • AI aromatase inhibitor
  • PIC palbociclib
  • ribociclib ribociclib
  • PI3K ⁇ phosphatidylinositol-3-kinase subunit alpha
  • mTOR inhibitor everolimus
  • PARP poly ADP ribose polymerase
  • Additional therapeutic agents that may be administered in combination with a selective androgen receptor modulator compound of this invention include, but are not limited to: abemaciclib, Abitrexate ® (methotrexate), Abraxane® (paclitaxel albumin-stabilized nanoparticle formulation), ado-trastuzumab emtansine, adriamycin PFS (doxorubicin hydrochloride), adriamycin RDF (doxorubicin hydrochloride), Adrucil® (fluorouracil), Afinitor® (everolimus), alpelisib, anastrozole, Arimidex® (anastrozole), Aromasin® (exemestane), velumab, atezolizumb, bicalutamide, buparlisib, Caelyx® (pegylated liposomal doxorubicin), capecitabine, carboplatin, cisplatin, Clafen® (cycl
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with a selective estrogen receptor modulator.
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with a selective estrogen receptor degrader (fulvestrant).
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with a CDK4/6 inhibitor (palbociclib, ribociclib, abemaciclib, trilaciclib, lerociclib).
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with a PIK3A inhibitor (alpelisib, buparlisib, tapelisib).
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with a HER2 inhibitor (lapatinib, trastuzumab, neratinib).
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with a VEGF-A inhibitor (bevacizumab).
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with a chemotherapeutic agent.
  • the chemotherapeutic agent is a taxane. In another embodiment, the chemotherapeutic agent is an anthracycline. In one embodiment, the chemotherapeutic agent is an epothilone (ixabepilone).
  • the methods of the present invention comprise administering the selective androgen receptor modulator, in combination with an LHRH analog. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator, in combination with a reversible antiandrogen. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator, in combination with an antiestrogen.
  • the methods of the present invention comprise administering a selective androgen receptor modulator, in combination with an anticancer drug. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator, in combination with a 5-alpha reductase inhibitor. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with an aromatase inhibitor. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with a progestin. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with an agent acting through other nuclear hormone receptors.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with a selective estrogen receptor modulator (SERM).
  • SERM selective estrogen receptor modulator
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with a progestin or anti-progestin.
  • the methods of the present invention comprise administering a selective androgen receptor modulator, in combination with an estrogen.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with a PDE5 inhibitor.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with apomorphine.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with a bisphosphonate (pamidronate, zoledronic acid). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with a denosumab (Xgeva®). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with a growth factor inhibitor. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with one or more additional selective androgen receptor modulators (SARMs).
  • SARMs selective androgen receptor modulators
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Abitrexate® (methotrexate). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Abraxane® (paclitaxel albumin-stabilized nanoparticle formulation). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with ado-trastuzumab emtansine. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Adriamycin PFS® (doxorubicin hydrochloride).
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Adriamycin RDF ® (doxorubicin hydrochloride). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Adrucil® (fluorouracil). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Afinitor® (everolimus). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with anastrozole.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Arimidex® (anastrozole). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Aromasin® (exemestane). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with capecitabine. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Clafen® (cyclophosphamide). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with cyclophosphamide.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Cytoxan® (cyclophosphamide). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with docetaxel. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with doxorubicin hydrochloride. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Efudex® (fluorouracil).
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Ellence® (epirubicin hydrochloride). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with epirubicin hydrochloride. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with everolimus. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with exemestane. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Fareston® (toremifene).
  • Ellence® epirubicin hydrochloride
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with epirubicin hydrochloride.
  • the methods of the present invention comprise administering a selective androgen
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Evista® (raloxifene). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Faslodex® (fulvestrant). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Femara® (letrozole). [000318] In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Fluoroplex® (5-fluorouracil).
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with fluorouracil. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Folex® (methotrexate). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Folex PFS® (methotrexate). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with fulvestrant. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with gemcitabine hydrochloride.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Gemzar® (gemcitabine hydrochloride). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Herceptin® (trastuzumab). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Ibrance (palbociclib). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Kisquali (ribociclib).
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Verenzio (abemaciclib). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with trilaciclib. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with lerociclib. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with ixabepilone. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Ixempra® (ixabepilone).
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with lapatinib ditosylate. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with letrozole. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with methotrexate. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with methotrexate LPF (methotrexate). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Mexate® (methotrexate).
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Mexate-AQ® (methotrexate). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Neosar® (cyclophosphamide). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Nolvadex® (tamoxifen citrate). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with paclitaxel.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with paclitaxel albumin- stabilized nanoparticle formulation. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Perjeta® (pertuzumab). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with pertuzumab. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with tamoxifen citrate.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Taxol® (paclitaxel). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Taxotere® (docetaxel). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with trastuzumab. In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with oremifene.
  • the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Tykerb® (lapatinib ditosylate). In another embodiment, the methods of the present invention comprise administering a selective androgen receptor modulator of this invention, in combination with Xeloda® (capecitabine). [000319] In one embodiment, the methods of the present invention comprise administering a pharmaceutical composition (or pharmaceutical preparation, used herein interchangeably) comprising the selective androgen receptor modulator of the present invention and/or its analog, derivative, isomer, metabolite, pharmaceutical product, hydrate, N-oxide, polymorph, crystal, prodrug or any combination thereof; and a suitable carrier or diluent.
  • compositions means therapeutically effective amounts of the selective androgen receptor modulator together with suitable diluents, preservatives, solubilizers, emulsifiers, adjuvant and/or carriers.
  • a “therapeutically effective amount” as used herein refers to that amount which provides a therapeutic effect for a given condition and administration regimen.
  • compositions are liquids or lyophilized or otherwise dried formulations and include diluents of various buffer content (e.g., Tris-HCl, acetate, phosphate), pH and ionic strength, additives such as albumin or gelatin to prevent absorption to surfaces, detergents (e.g., Tween 20®, Tween 80®, Pluronic F68®, bile acid salts), solubilizing agents (e.g., glycerol, polyethylene glycerol), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimerosal ®, benzyl alcohol, parabens), bulking substances or tonicity modifiers (e.g., lactose, mannitol), covalent attachment of polymers such as polyethylene glycol to the protein, complexation with metal ions, or incorporation of the material into or onto particulate preparations of polymeric compounds such as polylactic acid, pol
  • compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance.
  • Controlled or sustained release compositions include formulation in lipophilic depots (e.g., fatty acids, waxes, oils).
  • the pharmaceutical compositions comprising the compounds of this invention make use in the methods of this inventionof a dosage of between 1 and 50 mg of a compound of this invention.
  • the dosage is 1 mg, 3 mg, 9 mg, 10 mg, 18 mg or 30 mg of the compound of this invention.
  • the pharmaceutical compositions comprising the compounds of this invention make use in the methods of this invention of a dosage of 1 mg of a compound of this invention.
  • the pharmaceutical compositions comprising the compounds of this invention make use in the methods of this invention of a dosage of 3 mg of a compound of this invention. In another embodiment, the pharmaceutical compositions comprising the compounds of this invention make use in the methods of this invention of a dosage of 9 mg of a compound of this invention. In another embodiment, the pharmaceutical compositions comprising the compounds of this invention make use in the methods of this invention of a dosage of 10 mg of a compound of this invention. In another embodiment, the pharmaceutical compositions comprising the compounds of this invention make use in the methods of this invention of a dosage of 18 mg of a compound of this invention. In another embodiment, the pharmaceutical compositions comprising the compounds of this invention make use in the methods of this invention of a dosage of 30 mg of a compound of this invention.
  • compositions coated with polymers e.g., poloxamers or poloxamines.
  • Other embodiments of the compositions of the invention incorporate particulate forms protective coatings, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral.
  • the pharmaceutical composition is administered parenterally, paracancerally, transmucosally, transdermally, intramuscularly, intravenously, intradermally, subcutaneously, intraperitoneally, intraventricularly, intravaginally, intracranially and intratumorally.
  • pharmaceutically acceptable carriers are well known to those skilled in the art and include, but are not limited to, 0.01-0.1 M and preferably 0.05 M phosphate buffer or about 0.8% saline. Additionally, such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non- aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's and fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, collating agents, inert gases and the like.
  • Controlled or sustained release compositions include formulation in lipophilic depots (e.g. fatty acids, waxes, oils). Also comprehended by the invention are particulate compositions coated with polymers (e.g.
  • compositions of the invention incorporate particulate forms, protective coatings, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral.
  • the pharmaceutical composition can be delivered in a controlled release system.
  • the agent may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng.14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med.321:574 (1989).
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity to the therapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984). Other controlled release systems are discussed in the review by Langer (Science 249:1527- 1533 (1990).
  • the pharmaceutical preparation can comprise the selective androgen receptor modulator alone, or can further include a pharmaceutically acceptable carrier, and can be in solid or liquid form such as tablets, powders, capsules, pellets, solutions, suspensions, elixirs, emulsions, gels, creams, or suppositories, including rectal and urethral suppositories.
  • Pharmaceutically acceptable carriers include gums, starches, sugars, cellulosic materials, and mixtures thereof.
  • the pharmaceutical preparation containing the selective androgen receptor modulator can be administered to a subject by, for example, subcutaneous implantation of a pellet; in a further embodiment, the pellet provides for controlled release of selective androgen receptor modulator over a period of time.
  • the preparation can also be administered by intravenous, intraarterial, or intramuscular injection of a liquid preparation, oral administration of a liquid or solid preparation, or by topical application. Administration can also be accomplished by use of a rectal suppository or a urethral suppository.
  • the pharmaceutical preparations of the invention can be prepared by known dissolving, mixing, granulating, or tablet-forming processes.
  • the selective androgen receptor modulators or their physiologically tolerated derivatives such as salts, esters, N-oxides, and the like are mixed with additives customary for this purpose, such as vehicles, stabilizers, or inert diluents, and converted by customary methods into suitable forms for administration, such as tablets, coated tablets, hard or soft gelatin capsules, aqueous, alcoholic or oily solutions.
  • suitable inert vehicles are conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders such as acacia, cornstarch, gelatin, with disintegrating agents such as cornstarch, potato starch, alginic acid, or with a lubricant such as stearic acid or magnesium stearate.
  • suitable oily vehicles or solvents are vegetable or animal oils such as sunflower oil or fish-liver oil. Preparations can be effected both as dry and as wet granules.
  • the selective androgen receptor modulators or their physiologically tolerated derivatives such as salts, esters, N-oxides, and the like are converted into a solution, suspension, or emulsion, if desired with the substances customary and suitable for this purpose, for example, solubilizers or other auxiliaries.
  • sterile liquids such as water and oils, with or without the addition of a surfactant and other pharmaceutically acceptable adjuvants.
  • Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil.
  • compositions which contain an active component are well understood in the art.
  • Such compositions can be prepared as aerosols of the active component delivered to the nasopharynx or as injectables, either as liquid solutions or suspensions; however, solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared.
  • the preparation can also be emulsified.
  • the active therapeutic ingredient is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient.
  • Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like or any combination thereof.
  • the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents which enhance the effectiveness of the active ingredient.
  • An active component can be formulated into the composition as neutralized pharmaceutically acceptable salt forms.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide or antibody molecule), which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed from the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides
  • organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • the selective androgen receptor modulators or their physiologically tolerated derivatives such as salts, esters, N-oxides, and the like are prepared and applied as solutions, suspensions, or emulsions in a physiologically acceptable diluent with or without a pharmaceutical carrier.
  • the active compound can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez- Berestein and Fidler (eds.), Liss, New York, pp.353-365 (1989); Lopez-Berestein, ibid., pp.317-327; see generally ibid).
  • the salts of the selective androgen receptor modulator will be pharmaceutically acceptable salts. Other salts may, however, be useful in the preparation of the compounds of the invention or of their pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound of the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulphuric acid, methanesulphonic acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • a pharmaceutically acceptable acid such as hydrochloric acid, sulphuric acid, methanesulphonic acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • the term “consisting essentially of” refers to a composition, whose only active ingredient is the indicated active ingredient, however, other compounds may be included which are for stabilizing, preserving, etc. the formulation, but are not involved directly in the therapeutic effect of the indicated active ingredient.
  • the term “consisting essentially of” may refer to components, which exert a therapeutic effect via a mechanism distinct from that of the indicated active ingredient. In some embodiments, the term “consisting essentially of” may refer to components, which exert a therapeutic effect and belong to a class of compounds distinct from that of the indicated active ingredient. In some embodiments, the term “consisting essentially of” may refer to components, which exert a therapeutic effect and belong to a class of compounds distinct from that of the indicated active ingredient, by acting via a different mechanism of action, for example, and representing an embodiment of this invention, polypeptides comprising T cell epitopes present in a composition may be specifically combined with polypeptides comprising B cell epitopes.
  • the term “consisting essentially of” may refer to components which facilitate the release of the active ingredient.
  • the term “consisting” refers to a composition, which contains the active ingredient and a pharmaceutically acceptable carrier or excipient.
  • the term “comprising” is intended to mean that the system includes the recited elements, but not excluding others which may be optional.
  • the phrase “consisting essentially of” it is meant a method that includes the recited elements but exclude other elements that may have an essential significant effect on the performance of the method. “Consisting of” shall thus mean excluding more than traces of other elements. Embodiments defined by each of these transition terms are within the scope of this invention.
  • the present invention provides combined preparations.
  • a combined preparation defines especially a "kit of parts" in the sense that the combination partners as defined above can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners i.e., simultaneously, concurrently, separately or sequentially.
  • the parts of the kit of parts can then, e.g., be administered simultaneously or chronologically staggered, that is at different time points and with equal or different time intervals for any part of the kit of parts.
  • the ratio of the total amounts of the combination partners in some embodiments, can be administered in the combined preparation.
  • the combined preparation can be varied, e.g., in order to cope with the needs of a patient subpopulation to be treated or the needs of the single patient which different needs can be due to a particular disease, severity of a disease, age, sex, or body weight as can be readily made by a person skilled in the art.
  • the term “a” or “one” or “an” refers to at least one.
  • the phrase “two or more” may be of any denomination, which will suit a particular purpose.
  • “about” may comprise a deviance from the indicated term of + 1%, or in some embodiments, - 1%, or in some embodiments, ⁇ 2.5%, or in some embodiments, ⁇ 5%, or in some embodiments, ⁇ 7.5%, or in some embodiments, ⁇ 10%, or in some embodiments, ⁇ 15%, or in some embodiments, ⁇ 20%, or in some embodiments, ⁇ 25%.
  • HCC 1937, HCC 1954, HCC 38, T47D-Kbluc, MDA-MB-453, and MDA-MB-231 cells were grown in RPMI-1640 medium containing 2 mM L-glutamine supplemented with 10% fetal bovine serum (FBS). Cells were maintained in a 5% CO 2 /95% air humidified atmosphere at 37 o C. MCF-7 cells were grown in Minimum Essential Medium supplemented with 10% FBS. [000345] Breast cancer tumors typically express AR 70-90% of the time, however breast cancer cell lines typically do not express AR. This makes development of a preclinical model for the study of androgen effects on breast cancer very difficult.
  • SRB Sulforhodamine B
  • the SRB assay was used to determine cell number during cytotoxocity experiments. The following protocol was used: 1. Cells were detached with 0.25% trypsin. 2. Experimental cultures were cultured in 96-well microtiter plates (200 uL growth medium per well; 1,000-200,000 cells per well). 3. Cultures were fixed with 50 uL 50% TCA (4 o C). (see cell fixation protocol for details). 4. Fixed cells were stained with 50 uL 0.4% (wt/vol) SRB in 1% acetic acid for 10 minutes. 5.
  • MDA-MB-231 and HCC-38 triple negative breast cancer cells were infected with 200 ⁇ L or 500 ⁇ L adenovirus containing LacZ (negative control) or AR, and were treated with various AR ligands (agonists: DHT and Formula IX, and antagonist: bicalutamide) or a non- AR binder that is structurally similar to Formula IX, R-enantiomer of Formula IX.
  • various AR ligands agonists: DHT and Formula IX, and antagonist: bicalutamide
  • a non- AR binder that is structurally similar to Formula IX, R-enantiomer of Formula IX.
  • EXAMPLE 2 REVERSAL OF EFFECT OF FORMULA IX ON GROWTH Materials and Methods: [000352] To determine if the growth inhibition observed with DHT and Formula IX in AR- positive cells is AR dependent, MDA-MB-231 cells were infected with adenovirus containing LacZ (negative control) or AR and were treated with AR agonists, DHT or Formula IX, in the presence or absence of the AR antagonist, bicalutamide. Cells were treated in charcoal stripped FBS ( Figures 3A and 3C or full serum ( Figures 3B and 3D for 3 days, fixed and stained with sulforhodamine blue (SRB) to measure cell viability. IC 50 values were calculated.
  • SRB sulforhodamine blue
  • EXAMPLE 3 EFFECT OF AR LIGANDS ON BREAST CANCER CELL GROWTH Materials and Methods: [000354] To determine if all AR ligands inhibit the growth of triple negative breast cancer cells, MDA-MB-231 cells were infected with adenovirus containing LacZ or AR and were treated with various AR ligands (agonists: DHT, Formula VIII, Formula IX, Formula X, Formula XIII, Formula XIV; antagonist: bicalutamide) and a non-AR-binder: R-enantiomer of Formula IX.
  • various AR ligands agonists: DHT, Formula VIII, Formula IX, Formula X, Formula XIII, Formula XIV; antagonist: bicalutamide
  • a non-AR-binder R-enantiomer of Formula IX.
  • Figure 5 shows that all AR ligands that elicited anti-proliferative activity are agonists in MDA-MB-231 cells transfected with AR and their agonist and growth inhibitory properties compare well.
  • growth inhibitory ligands are AR agonists in MDA- MB-231 cells transfected with AR.
  • EXAMPLE 5 ANALYSIS OF GROWTH INHIBITORY EFFECTS IN BREAST CANCER CELLS EXPRESSING ESTROGEN RECEPTOR Materials and Methods: [000359] To ensure that growth inhibitory effects in MDA-MB-231 cells are selective to AR, and to determine if the ligand dependent growth inhibitory effects are exclusive to AR and also to ensure that the effects are not artifacts of adenoviral infection, MDA-MB-231 triple negative breast cancer cells were infected with ER- ⁇ or ER- ⁇ adenovirus constructs and were treated with ER agonist: 17 ⁇ -estradiol (E2) or ER antagonist: ICI 182,780 (ICI) in charcoal stripped serum (Figure 6C) [000360] or full serum ( Figures 6D and 6E) for 3 days.
  • E2 17 ⁇ -estradiol
  • ER antagonist ICI 182,780
  • FIGS. 6A-6B show the presence or absence of ER ⁇ or ER ⁇ in MDA-MB-231 cells following transfection. These results show that the anti-proliferative effects observed with androgens is unique to ligand activated AR and not an artifact of adenovirus.
  • Figures 6C-6E show that over-expression of ER- ⁇ or ER- ⁇ in MDA-MB-231 cells failed to promote growth inhibition either in the presence of ER agonists or antagonists.
  • MDA-MB-231 cells were stably transfected with AR using lentivirus. Following transfection, cells were treated for 3 days with the indicated concentrations of DHT or bicalutamide. Live cells were visualized using a light-microscope and photographed. The cells were imaged at the same magnification and under the same microscopic conditions.
  • FIG. 7 shows that DHT altered the morphology of MDA-MB-231 cells into more anchorage dependent and differentiated cells, indicating that AR agonist-bound AR expressing breast cancer cells will have less invasive and migratory properties (e.g., less likely to metastasize).
  • DHT and SARMs alter the morphology of AR-positive MDA-MB-231 cells.
  • MDA-MB-231 cells were stably transfected with AR using lentivirus and were treated with vehicle or AR agonists at the indicated concentrations. At the end of 3 days of incubation, the cells were imaged under a microscope (40X).
  • Rat GR, MR, PR, ER- ⁇ and ER- ⁇ were individually cloned into a pCR3.1 vector backbone. Sequencing was performed to verify the absence of any mutations.
  • HEK-293 cells were plated at 90,000 cells per well of a 24 well plate in Dulbecco’s Minimal Essential Media supplemented with 5% charcoal-stripped FBS.
  • the cells were transfected using Lipofectamine (Invitrogen, Carlsbad, CA) with 0.25 ⁇ g GRE-LUC for GR, MR and PR and ERE-LUC for ER- ⁇ and ER- ⁇ , 0.5 ng CMV-LUC (renilla luciferase) and 12.5-25 ng of the respective expression vector for each receptor.
  • the cells were treated 24 h after transfection with Formula VIII in the absence (agonist mode) and presence (antagonist mode) of known agonists (17 ⁇ - estradiol for ER; dexamethasone for GR; aldosterone for MR; progesterone for PR) as controls. Luciferase assays were performed 48 h after transfection.
  • Transcriptional activation values are represented as firefly luciferase normalized to renilla luciferase.
  • a compound of Formula VIII at all the tested concentrations did not induce GR- or MR-mediated transactivation, whereas the known ligands (dexamethasone and aldosterone) induced the activities of GR or MR by 70- and 60-fold, respectively, at a concentration of 1 nM.
  • a compound of Formula VIII increased the transactivation of PR at 1 ⁇ M and 10 ⁇ M by 3 and 8 fold, respectively.
  • Progesterone activated PR by 23 folds at a 1 nM concentration, indicating that a compound of Formula VIII is greater than 10,000- fold weaker than the endogenous agonist for PR.
  • Formula VIII was about 1,000 fold weaker as a PR antagonist, than RU486.
  • Compounds of Formulae VIII and IX are specific for the AR and do not stimulate or inhibit receptor-mediated transactivation of ER ⁇ , ER ⁇ , GR, or MR. Unexpectedly, Formula VIII exhibited moderate potency partial agonist activity for PR, and potent PR partial antagonism (see Figure 9). Combined AR-agonism and PR-antagonism will be beneficial in certain breast cancers (e.g., PR-positive breast cancers).
  • TGI % tumor growth inhibition
  • EXAMPLE 9 EFFECT OF FORMULA IX IN WOMEN WITH METASTATIC OR ER AND/OR AR- POSITIVE REFRACTORY BREAST CANCER [000377]
  • This clinical trial assessed the safety and efficacy of 9 mg of the compound represented by the structure of Formula IX (Formula IX), in 22 post-menopausal women who have estrogen receptor (ER) positive metastatic breast cancer, and who have responded previously to adjuvant and/or salvage endocrine therapy.
  • the goal of this study was to determine the importance of the AR status as a therapeutic target in women with ER-positive metastatic breast cancer (MBC) that had previously responded to hormone therapy. The treatment was continued until disease progression (PD).
  • MBC ER-positive metastatic breast cancer
  • Subject Population Female subjects with ER-positive metastatic breast cancer who have previously been treated with up to 3 prior hormonal therapies for the treatment of breast cancer. Subjects must have been treated with and responded to previous adjuvant therapy for ⁇ 3 years or hormonal therapy for metastatic disease for ⁇ 6 months prior to progression. Details of subject selection criteria are presented below: [000382] To be eligible for participation in this study, subjects must meet all of the following criteria, including give voluntary, signed informed consent in accordance with institutional policies; be a woman that has been diagnosed with ER-positive metastatic breast cancer; and be clinically confirmed as postmenopausal. Subjects must have undergone the onset of spontaneous, medical or surgical menopause prior to the start of this study.
  • spontaneous menopause is defined as the natural cessation of ovarian function as indicated by being amenorrheic for at least 12 months. If the subject has been amenorrheic for ⁇ 6 months but ⁇ 12 months they must have a serum FSH concentration of ⁇ 50 mIU/mL and an 17 ⁇ -estradiol concentration of ⁇ 25 pg/mL; medical menopause is defined as treatment with a luteinizing hormone receptor hormone agonist; and surgical menopause is defined as bilateral oophorectomy).
  • Subjects with any of the following exclusion criteria will NOT be eligible for enrollment in this study: have triple negative breast cancer; have, in the judgment of the Investigator, a clinically significant concurrent illness or psychological, familial, sociological, geographical or other concomitant condition that would not permit adequate follow-up and compliance with the study protocol; have uncontrolled hypertension, congestive heart failure or angina; have Stage 4 chronic obstructive pulmonary disease (COPD); have positive screen for hepatitis B consisting of HBsAg (Hepatitis B Surface Antigen), unless subject was diagnosed > 10 years prior to enrollment and no evidence of active liver disease; have ALT/SGOT or AST/SGPT above 1.5 times the upper limit of normal (ULN); have positive screen for hepatitis A antibody IgM or HIV; have received chemotherapy for metastatic breast cancer within the 3 months prior to enrollment in the study or be expected to receive chemotherapy for metastatic breast cancer during the study; be currently taking testosterone, methyltestosterone, oxandrolone (Oxandrin®
  • Efficacy Endpoints [000392] The primary efficacy analysis was the clinical benefit in subjects with AR-positive breast cancer at 6 months as measured by a modified Response Evaluation Criteria In Solid Tumors (RECIST 1.1) classification. Key secondary endpoints of clinical benefit in all subjects and AR-negative subjects, as well as objective response rate, progression free survival, time to progression, duration of response, incidence of SREs, and time to first SRE in subsets based on AR status (i.e., all subjects, AR-positive subjects, and AR-negative subjects) was also assessed. Effects on CA 27-29, PSA, bone turnover markers, QOL, and libido were assessed as tertiary endpoints.
  • Clinical benefit in a subject is defined as a complete response [CR], a partial response [PR] or stable disease [SD] as measured by modified RECIST 1.1, which is described in detail below. (Eisenhauer EA et al. New response evaluation criteria in solid tumors: revised RECIST guideline (version 1.1). European Journal of Cancer 45:228-247, 2009). [000394] For subjects with non-measurable (non-target) disease only at baseline, SD was defined as those with non-CR/non-PD combined response. The primary endpoint of the study was to assess the proportion of subjects with clinical benefit (PCB) at 6 months (CR+PR+SD) in subjects with AR-positive breast cancer.
  • the secondary efficacy endpoints include: • To assess the clinical benefit in all subjects with breast cancer treated with Formula IX. The clinical benefit is defined as the proportion of subjects with complete response [CR] + partial response [PR] + stable disease [SD] as measured by modified RECIST 1.1 (Eisenhauer EA et al. New response evaluation criteria in solid tumors: revised RECIST guideline (Version 1.1). European Journal of Cancer 45: 228-247, 2009). • For subjects with non-measurable (non-target) disease only at baseline, SD was defined as those with non-CR/non-PD combined response. • To assess objective response rate (ORR) in subjects with breast cancer treated with Formula IX.
  • ORR objective response rate
  • Objective response rate is defined as the proportion of subjects with a CR or PR at 6 months as measured by modified RECIST 1.1.
  • ORR is defined as the proportion of subjects with a CR at 6 months as measured by modified RECIST 1.1.
  • PFS progression free survival
  • TTP time to progression
  • liver enzymes ALT, AST and bilirubin
  • ALT, AST and bilirubin returned to baseline with no interruption of therapy and no increase in total bilirubin.
  • Formula IX demonstrated promise as a novel targeted therapy for AR-positive MBC. The primary endpoint was achieved, with 6/17 AR-positive patients meeting statistical threshold for success. Serum PSA appeared to be a surrogate marker for AR activity and disease response.
  • Measurable lesions must be at least 2 times the slice thickness or at least two times the size of the CT scan interval cut. [000407] Lesions seen on chest x-ray but not confirmed by CT or MRI scan are not acceptable as measurable lesions for this study. [000408] To be considered pathologically enlarged and measurable, a lymph node must be > 15 mm in short axis when assessed by CT scan (CT scan slice thickness recommended to be no greater than 5 mm). At baseline and in follow-up, only the short axis will be measured and followed. [000409] Measurable disease is defined as the presence of at least one measurable lesion. [000410] All measurements will be taken and recorded in millimeters using an electronic measurement method.
  • Non-measurable lesions are defined as any lesion(s) that are smaller than the criteria for measurable lesions stated above (non-nodal lesions with longest diameter ⁇ 10mm or pathological lymph nodes with ⁇ 10 mm to ⁇ 15 mm in short axis) or truly non measurable lesions (or sites of disease). Lesions considered to be truly non-measurable are bone lesions (lytic lesions or mixed lytic-blastic lesions without identifiable soft tissue components, and blastic lesions), leptomeningeal disease, ascites, pleural/pericardial effusions, lymphangitis cutis/pulmonis, inflammatory breast disease, abdominal masses not confirmed by imaging techniques, and cystic lesions.
  • Target lesions must be measurable lesions. [000413] All target lesions up to a maximum of two lesions per organ and five lesions in total, representative of all involved organs, will be selected/confirmed as target lesions, recorded and measured at baseline. [000414] Target lesions should be selected on the basis of their size (lesions with the longest diameter) and their suitability for accurate repetitive measurements by CT/MRI imaging techniques and be most representative of the subject’s tumor burden. [000415] Target lesions will be measured in one dimension by the size estimation of their diameter. A sum of the diameters (longest for non-nodal lesions and shortest for nodal lesions) for all target lesions will be calculated and reported for each time point.
  • Non-Target Lesions All other lesions (or sites of disease) and any measurable lesions that were not selected as target lesions should be identified as non-target lesions and indicated as present at baseline. [000417] Measurements of the non-target lesions may be performed, however the continued presence or absence as well as the disappearance or progression status of these lesions will be noted throughout follow-up assessments. New Lesions [000418] New lesions will be called at follow-up visits regardless of whether they occur in anatomic regions that were routinely subjected to follow-up, or in regions without disease at baseline and for which a follow-up scan is performed for clinical suspicion of new disease.
  • New lymph nodes need to have a minimum size of 10 mm in their shortest axis.
  • New non- nodal lesions need not to be measurable or to have a minimum size. Measurements of new lesions may be performed.
  • Response Criteria Definitions [000419] The following response criteria will be applied for target and non-target lesions: Target Lesion Response Criteria [000420] Complete Response (CR): Disappearance of all target lesions. Target lymph node lesions that become ⁇ 10 mm in their shortest diameter will be considered to be normal (non- pathologic) and their actual measurement will be recorded.
  • Partial Response At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of the diameters.
  • Stable Disease SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD taking as reference the smallest sum of diameters (nadir).
  • Progressive Disease At least a 20% increase in the sum of the diameters of target lesions taking as reference the smallest sum of diameters (nadir) recorded since the treatment started.
  • NE Not evaluable
  • NE can be applied if repeated measurements cannot be assessed for reasons such as inadequate or missing imaging.
  • Non-Target Lesion Response Criteria [000425] Complete Response (CR): Disappearance of all non-target lesions. All lymph nodes must be non-pathological in size ( ⁇ 10 mm short axis). Disappearance of bone lesions identified on bone scintigraphy.
  • Non-CR/Non-PD Persistence of one or more non-target lesions. Stability, decrease, or mild increase in uptake of bone lesions on bone scintigraphy.
  • PD Progressive Disease
  • NE Not Evaluable
  • Determination of an overall response for each time point is based on the combination of responses for target, non-target, and the presence or absence of new lesions using the algorithm outlined on tables C1 and C2 below.
  • Table C 1 Summary of Definitions of Response for Patients with Measurable (Target) Disease at Baseline
  • Table C2 Summary of Definitions of Response for Patients with Non-Measurable (Non-Target) Disease only at Baseline Response of Combined Lesion Types Non-Target Lesions New Lesions Combined Response CR No CR Non-CR/non-PD No Non-CR/non-PD NE No NE PD Yes or No PD Any Yes PD
  • SYNTHESIS OF (S) ENANTIOMER OF FORMULA VIII [000430] (2R)-1-Methacryloylpyrrolidin–2-carboxylic Acid.
  • the combined organic solution was dried over Na2SO4, filtered through Celite, and evaporated in vacuo to dryness.
  • This oil was purified by column chromatography using CH 2 Cl2/EtOAc (80:20) to give an oil which was crystallized from CH 2 Cl 2 /hexane to give 33.2 g (59.9%) of (S)-N-(4-cyano-3-(trifluoromethyl)phenyl)-3- (4-cyanophenoxy)-2-hydroxy-2-methylpropanamide as a colorless solid (a cotton type).
  • EXAMPLE 16 BINDING AND TRANSACTIVATION OF SARMS IN BREAST CANCER CELLS
  • HEK-293 or MDA-MB-231 cells were transfected with 0.25 ⁇ g GRE-LUC, 10 ng CMV- renilla LUC, and 25 ng CMV-hAR using lipofectamine. Twenty four hours after transfection, the cells were treated with DHT, compound of Formula VIII or compound of Formula IX and luciferase assay was performed 48 hrs after transfection.
  • DHT, compound of Formula VIII and compound of Formula IX were measured using an in vitro competitive radioligand binding assay with [17 ⁇ -methyl- 3 H]-mibolerone ([ 3 H]MIB), a known steroidal and high affinity AR ligand, and purified AR-LBD protein.
  • Results: [000457] DHT, compound of Formula VIII and Formula IX are agonists of AR in breast cancer cells as presented in Figure 13A-13C (HEK-293 cells in Figure 13A and MDA-MB- 231 cells in Figures 13B-13C).
  • RBAs relative binding affinities
  • bicalutamide were 1.0, 0.330, 0.314, and 0.016, respectively, demonstrating high affinity AR binding for the SARM compounds of this invention (data not shown).
  • AR agonists differentially regulate genes in ER-positive and ER-negative breast cancer cells.
  • MDA-MB-231 and MCF-7 cells infected with AR or GFP containing adenovirus were maintained in charcoal stripped serum containing medium for 3 days and were treated with DHT or Formula VIII. After overnight treatment, the cells were harvested, RNA isolated and real-time PCR for the indicated genes were performed.
  • EXAMPLE 18 GENE EXPRESSION ARRAY OF MDA-MB-231-AR XENOGRAFT [000460] RNA was extracted from MDA-MB-231-AR tumors (n 5/group) treated with vehicle or compound of Formula VIII. RNA was pooled and Affymetrix microarray was performed to determine the change in expression of gene signature.
  • the results presented in Figure 15 show that activation of AR in MDA-MB-231- AR xenografts suppressed the expression of more genes than it induced in these tumors. This pattern is unique in breast cancer cells and is different from gene expression results observed in prostate cancer cells, where more genes are induced than repressed (data not shown).
  • the results presented in Figure 16 validate the microarray results presented in Figure 15 by analyzing selected genes using realtime PCR TaqMan primers and probe in ABI 7900.
  • EXAMPLE 21 GENE EXPRESSION ANALYSIS OF MDA-MB-231-AR AND MCF-7-AR XENOGRAFTS FOLLOWING TREATMENT WITH FORMULA VIII AND FORMULA IX
  • Microarray Analysis was performed on RNA from MDA-MB-231-AR and MCF-7- AR tumors in order to identify and compare changes in gene expression in ER-negative (MDA- MB-231-AR; triple negative) an ER-positive (MCF-7-AR; triple positive) breast cancer tumors treated with a compound of Formula VIII (30 mg/kg/day p.o.
  • Table 7 below presents the sum totals of up-regulated and down-regulated genes in MDA-MB-231-AR and MCF-7-AR tumors.
  • Table 7 [000473] Of particular interest was that of the 1547 regulated genes identified in MCF-7- AR tumors and the 1536 regulated genes identified in MDA-MB-231-AR tumors, the subset of overlapping genes was only 245 genes. This result indicated that Formula VIII regulated distinct sets of genes in MCF-7-AR (ER-positive; triple positive) and MDA-MB-231-AR (ER- negative; triple negative) breast cancer cells.
  • Tables 8 and 9 below present genes involved in mammary tumorigenesis that were differentially regulated (by at least 2 fold) by Formula VIII in MDA-MB-231-AR tumors (Table 8) and MCF-7-AR tumors (Table 9). Indications of up-regulation or down-regulation are presented in the right-most column. [000475] Table 8: Breast cancer relevant genes modulated in MDA-MB-231-AR tumors
  • Table 9 Breast cancer relevant genes modulated in MCF-7-AR tumors
  • Well known androgen-dependent genes e.g., FKP5 and MUC 1 ; See Table 10 below were elevated, showing SARM penetration into the tumor.
  • Formula VIII and Formula IX demonstrated ⁇ 85% TGI at all doses tried (5, 10 mg per kg for Formula VIII; 5, 10, 30 mg per kg for Formula IX) in the triple negative breast cancer model using MDA-MB-231-AR cells in nude mice.
  • the results presented in Figure 23 demonstrate inhibition of triple negative breast cancer using Formulae VIII and IX. Tumor weights were likewise reduced for all doses of Formula VIII and Formula IX. Spleen enlargement (680 mg vs.200-300 mg for normal mice) was seen only in vehicle treated mice, possibly indicative of prevention by the SARMs of tumor metastasis to the spleen.
  • tumor size reached 100-200 mm 3
  • Tumor volume was measured thrice weekly. Tumor volume was calculated using the formula length ⁇ width ⁇ width ⁇ 0.5236.
  • Formula IX (lower trace) produced some tumor growth inhibition of this AR-positive TNBC tumor in each experiment whereas enzalutamide was indistinguishable from vehicle treatment ( Figures 26A and 26B).
  • Formula IX reduced tumor weight in experiment 2 by ⁇ 40%.
  • EXAMPLE 23 Ki-67 STAINING WAS REDUCED IN AR-POSITIVE TNBC TUMORS OF ANIMALS TREATED WITH FORMULA IX
  • Figures 28A-28B demonstrated an approximately 50% reduction in Ki-67 staining in 2 weeks of treatment. Tumors from replicate experiment 2 ( Figure 27B) were fixed in formalin and paraffin embedded. Slides were cut and stained with Ki-67 antibody.
  • Ki-67 positive cells total 200 cells were counted in each slide
  • Ki-67 staining was reduced in tumors of the animals treated with Formula IX.
  • EXAMPLE 24 GENE EXPRESSION STUDY AND ChIP-SEQ STUDY IN AR-POSITIVE TNBC TUMOR XENOGRAFTS Methods [000488] Chromatin Immunoprecipitation Assay (ChIP). Proteins were cross-linked by incubation with 1% formaldehyde (final concentration) at 37°C for 10 min. Tumors were homogenized using a probe hand-held homogenizer.
  • the cells were washed with 1 ⁇ PBS twice, scraped in 1 mL of PBS containing protease inhibitors ([1 mg each of aprotinin, leupeptin, antipain, benzamidine HCl, and pepstatin/ml], 0.2 mM phenylmethylsulfonyl fluoride, and 1 mM sodium vanadate), pelleted, and resuspended in SDS lysis buffer (1% SDS, 10 mM EDTA, 50 mM Tris-HCl [pH 8.1]).
  • protease inhibitors [1 mg each of aprotinin, leupeptin, antipain, benzamidine HCl, and pepstatin/ml], 0.2 mM phenylmethylsulfonyl fluoride, and 1 mM sodium vanadate
  • the cell extract was sonicated (Branson sonifier 250) in a cold room eight times for 10 s each at constant duty cycle, with an output of 3 and with incubation on ice after every sonication.
  • the debris was pelleted at 13,000 rpm for 10 min at 4°C, and the supernatant was diluted 10-fold with ChIP dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mM EDTA, 16.7 mM Tris HCl [pH 8.1], 167 mM NaCl).
  • ChIP dilution buffer 0.01% SDS, 1.1% Triton X-100, 1.2 mM EDTA, 16.7 mM Tris HCl [pH 8.1], 167 mM NaCl.
  • the proteins were precleared with 50 ⁇ L of 1:1 protein A-Sepharose beads in TE.
  • the beads were pelleted and washed three times with low-salt wash buffer (0.1% sodium dodecyl sulfate [SDS], 1% Triton X-100, 2 mM EDTA, 20 mM Tris HCl [pH 8.1], 0.15 M NaCl), and twice with 1 ⁇ TE (10 mM Tris HCl, 1 mM EDTA; pH 8.0).
  • DNA-protein complexes were obtained by extracting the beads with 50 ⁇ L of freshly prepared extraction buffer (1% SDS, 0.1 M NaHCO 3 ) three times. Cross-linking of the DNA protein complexes was reversed by incubating at 65°C for 6 h.
  • RNA Analysis and microarray Tumors were homogenized, RNA isolated, purified and submitted to the UTHSC MRC core facility for microarray analysis (ST2.0 array from Affymetrix). Results [000492] In the gene expression study described above, RNA was isolated from the BR-0001 TNBC tumors and the expression of genes in the entire genome was measured by microarray (Affymetrix, ST2.0 array).
  • TNBC triple-negative breast cancer
  • TNBC subtypes Based on 3,247 gene expression profiles from 21 breast cancer data sets, six TNBC subtypes, including 2 basal- like (BL1 and BL2), an immunomodulatory (IM), a mesenchymal (M), a mesenchymal stem– like (MSL), and a luminal androgen receptor (LAR) subtype from 587 TNBC samples with unique gene expression patterns and ontologies, were discovered (Brian D. Lehman et al., J. Clin. Invest. 2011, 121(7), 2750-2767). Cell line models representing each of the TNBC subtypes also displayed different sensitivities to targeted therapeutic agents. Gene expression data was compared to the genes published (Pietenpol group) to classify the BLBC into sub- classification.
  • IM immunomodulatory
  • M mesenchymal
  • MSL mesenchymal stem– like
  • LAR luminal androgen receptor
  • Figures 30A-30B depict that Pietenpol classification of TNBC suggests that the BR-0001 tumor is LAR and MSL subtypes.
  • EXAMPLE 25 GENE EXPRESSION CHANGES IN AR-POSITIVE TNBC XENOGRAFT TUMORS
  • Figure 31 demonstrates that in BR-0001 tumors Formula IX up-regulated gene expression. Approximately 4200 genes were up-regulated by Formula IX compared to vehicle, while approximately 1170 genes were down-regulated by Formula IX compared to vehicle.
  • Formula IX recruited AR to 176 promoters (-5 kb to +1 kb). 20% of the promoters occupied by the AR in response to Formula IX also had the gene up-regulated by Formula IX.
  • Subjects will be randomized to receive either Formula IX 9 mg or 18 mg given PO daily for up to 24 months. Each dose arm will be treated independently and each assessed for efficacy using Simon’s two-stage (optimal) design (Simon R. Optimal two- stage designs for Phase 2 clinical trials. Controlled Clinical Trials 1989; 10: 1-10). Subjects will be randomized in a 1:1 fashion to one of the two dose arms. [000497] Randomization will be stratified by subjects presenting with bone only metastases and all other subjects, and further by setting of immediately preceding therapy (adjuvant setting or metastatic setting) in order to balance the proportion of subjects with these presenting features in each dose arm.
  • immediately preceding therapy adjuvant setting or metastatic setting
  • CBR clinical benefit response
  • the first stage in each study arm will be assessed among the first 18 evaluable subjects. If at least 3/18 subjects achieve CB (defined as CR, PR, or SD) at week 24, the arm will proceed to the second stage of recruitment up to a total of 44 evaluable subjects per arm. Otherwise, the arm will be discontinued for lack of efficacy.
  • CB defined as CR, PR, or SD
  • Statistical significance i.e., rejection of the null hypothesis of an unacceptably low CBR of ⁇ 10% in favor of the alternative hypothesis that indicates the higher rate, ⁇ 30%, is more likely, will be declared if at least 9/44 subjects achieve CB at week 24 in that arm.
  • Subjects who are not centrally confirmed AR+ may remain on the trial, but will not be part of the primary efficacy analysis – these subjects will contribute to secondary and tertiary analyses.
  • Subjects on the 18 mg treatment arm who experience an adverse event (AE) with Grade ⁇ 3 intensity may have a dose reduction from 18 mg to 9 mg per day or a drug interruption based on the medical judgment of the Investigator and after confirmation by the study Medical Monitor.
  • the drug interruption may last for a period of up to 5 days after which the subject must be rechallenged with study drug (18 mg or 9 mg) or discontinued from the study.
  • Subjects on the 9 mg treatment arm who experience an AE with Grade ⁇ 3 intensity (NCI-CTCAE 4.0) and/or intolerance may have a drug interruption based on the medical judgment of the Investigator and after confirmation by the study Medical Monitor. The drug interruption may last for a period of up to 5 days after which the subject must be rechallenged with study drug (9 mg) or discontinued from the study.
  • subjects will be analyzed in the treatment arm in which they are initially dosed.
  • Study Duration The study duration is estimated at 3 years.
  • Description of Agent or Intervention Three (3) Formula IX 3.0 mg softgels for a 9 mg daily dose or six (6) Formula IX 3.0 mg softgels for an 18 mg daily dose will be taken PO with water at approximately the same time each day, with or without food.
  • Potential Benefits Based on the trial of Example 9, Formula IX 9 mg once daily has been studied in 22 postmenopausal women with metastatic ER+ BC who have previously responded to hormonal therapy. The primary endpoint was assessed in 17 AR-positive subjects. Six of these 17 subjects demonstrated CB (SD) at six months. In one subject with SD (RECIST 1.1), tumor regression of 27% in a single target lesion was demonstrated.
  • the primary efficacy objective of this trial is to estimate the CBR at 24 weeks (defined as complete response [CR], partial response [PR], or SD) (by RECIST 1.1) of Formula IX 9 mg and of Formula IX 18 mg given PO daily in subjects with estrogen receptor positive and androgen receptor positive (ER+/AR+) BC who have centrally confirmed AR+ status.
  • the secondary efficacy objectives are to estimate the CBR at 24 weeks (by RECIST 1.1) of Formula IX 9 mg and 18 mg in all subjects randomized who receive at least one dose of study medication (the full analysis set [FAS]) regardless of AR status as determined by the central laboratory.
  • the additional secondary efficacy objectives apply to both centrally confirmed AR+ subjects (the evaluable subset of the FAS) as well as to all subjects in the FAS: (a) Estimate the objective response rate (ORR; defined as CR or PR) (by RECIST 1.1) of Formula IX 9 mg and 18 mg at 24 weeks; (b) Estimate the best overall response rate (BOR) of Formula IX 9 mg and 18 mg; (c) Estimate the progression free survival (PFS) of subjects receiving Formula IX 9 mg and 18 mg; (d) Estimate the TTP of subjects receiving Formula IX 9 mg and 18 mg; and (e) Estimate duration of response (time from documentation of tumor response to disease progression or death) of subjects receiving Formula IX 9 mg and 18 mg.
  • ORR objective response rate
  • BOR Best overall response rate
  • PFS progression free survival
  • TTP of subjects receiving Formula IX 9 mg and 18 mg
  • Estimate duration of response time from documentation of tumor response to disease progression or death
  • the tertiary objectives apply to both centrally confirmed AR+ subjects (the evaluable subset of the FAS) as well as to all subjects in the FAS (a) Assess the effect of Formula IX 9 mg and 18 mg on serum PSA; (b) Assess the effect of Formula IX 9 mg and 18 mg on Quality of Life (QoL) as measured by EQ-5D-5L; (c) Assess the effect of Formula IX 9 mg and 18 mg on circulating tumor cells (CTCs); (d) Assess the impact of duration of prior CB on outcome; (e) Assess the impact of time from diagnosis of metastases to randomization on outcome; (f) Describe the effect of Formula IX 9 mg and 18 mg on tumor volumetrics; (g) Assess the effect of plasma concentrations of Formula IX and Formula IX glucuronide on CBR at 24 weeks.
  • the safety objective is to describe the safety profile of Formula IX 9 mg and 18 mg PO daily in subjects with ER+/AR+ BC with centrally confirmed AR+ as well as in all subjects randomized and treated.
  • the pharmacokinetic objective To describe the plasma concentrations of Formula IX and Formula IX glucuronide at each of the assessed time points.
  • Formulation, Packaging, and Labelling Formula IX 3.0 mg Softgels will be supplied as opaque, white to off-white, size 5, oval Softgel capsules containing 3.0 mg of Formula IX.
  • the liquid Softgel fill is composed of Formula IX dissolved in polyethylene glycol 400.
  • Formula IX 3.0 mg Softgels will be packaged in blister packs.
  • Each blister pack will contain sufficient study drug for one (1) week of dosing.
  • subjects will be provided with a carton of study drug containing 7 blister packs, equivalent to 7 weeks of dosing.
  • Subjects will be requested to bring with them the carton box with all blister packs at every visit.
  • Each blister pack will be comprised of an appropriate number of blister strips (1 blister for the 9 mg treatment arm and 2 blisters for the 18 mg treatment arm) encased in a child-resistant heat-sealed card.
  • the blister strips are composed of a PVC/ACLAR base and an aluminum foil/PVC/PVAC copolymer and polymethacrylate (product contact) lidding. Perforations on the back of the heat-seal card overlay the foil lidding. To remove the study drug, subjects will release the appropriate perforation by depressing a release button on the inside of the card. Once released, the perforation can be removed and the study drug pushed through the foil.
  • Pharmacokinetic assessment [000519] Blood samples for pharmacokinetic assessment will be collected at baseline (pre-dose), Visit 3 (week 6), Visit 5 (week 18), and Visit 6 (week 24).
  • One blood sample will be collected in a 6 mL K2-ethylenediaminetetraacetic acid (EDTA) blood collection tube on each of these days. The exact time (hh:mm) and date that each blood sample is collected will be recorded on the electronic Case Repot Form.
  • the blood sample should be collected before the subject is given their first dose of Formula IX.
  • Visits 3 (week 6), 5 (week 18), and 6 (week 24) the date and approximate time of the last dose of Formula IX prior to the blood sample should be recorded; i.e., it should be documented whether the subject took the previous dose that morning or the evening before.
  • the tubes will be gently inverted several times to mix the anticoagulant with the blood sample.
  • Blood samples will be kept on wet ice (ice packs in a water bath is also acceptable) for up to 20 minutes until processed.
  • the plasma fraction will be separated by placing the collection tube into a centrifuge for 10 minutes at 1,500 x g.
  • the plasma fraction will be withdrawn by pipette and divided into two 2 mL polypropylene transfer vials (with each tube receiving approximately equal aliquots).
  • All sample collection and freezing tubes will be clearly labeled in a fashion which identifies the subject, the study number, the visit number, and freezing tube aliquot letter. Labels will be fixed to freezing tubes in a manner that will prevent the label from becoming detached after freezing. Samples will be stored in a freezer at –20°C or lower.
  • Protocol G100402 a Phase 1 multiple ascending dose study in 50 healthy, young, male volunteers, and 23 elderly male volunteers with truncal obesity; 3. Protocol G100503, a Phase 1 single dose pharmacokinetic study to assess the effect of a dosage regimen that simulates a sustained release formulation to an immediate release formulation in 18 healthy, young male volunteers and 18 postmenopausal women; 4. Protocol G100506, a Phase 1 single dose pharmacokinetic study to assess the relative bioavailability of a 3 mg hard shell capsule formulation to be used during continued clinical development and to assess the effect of food on the pharmacokinetics of the 3 mg softgel formulation in 27 healthy, young, male volunteers; 5.
  • Protocol 006 a Phase 1 single dose and multiple dose pharmacokinetic study in 24 postmenopausal, Japanese women; 6. Protocol G200501, a Phase 2 study in 60 postmenopausal women and 60 elderly men to assess lean body mass and physical function; 7. Protocol 003, a Phase 1b study in 44 postmenopausal women; 8. Protocol G200502, a Phase 2b study in 159 men and postmenopausal women with cancer to assess lean body mass and physical function; 9. Protocol G100511, a Phase 1 study to assess the effect of severe renal impairment on the pharmacokinetics of Formula IX; 10. Protocol G100508, a Phase 1 study to assess the effect of mild and moderate hepatic impairment on the pharmacokinetics of Formula IX; 11.
  • Protocol G100509 a Phase 1 mass balance study of Formula IX in healthy volunteers; 12. Protocol G100507, a Phase 1 study to assess the pharmacokinetics and absolute oral bioavailability of Formula IX in Caucasian and African American men and women; 13. Protocol G100510, a single-dose, randomized, double-blind, comparative, positive and placebo-controlled, four-period crossover Phase 1 study to define the electrocardiogram (ECG) effects of Formula IX, at therapeutic and supratherapeutic doses, in healthy male and female subjects: a thorough ECG trial; 14.
  • ECG electrocardiogram
  • Protocol G100512 a Phase 1 study to assess the effect of ketoconazole (Cytochrome P450, Family 3,Subfamily A [CYP3A4] inhibitor) on the pharmacokinetics of Formula IX; 15. Protocol G100513, a Phase 1 study to assess the effect of rifampin (CYP3A4 inducer) on the pharmacokinetics of Formula IX; 16. Protocol G100514, a Phase 1 study to assess the pharmacokinetic drug:drug interaction of Formula IX and celecoxib (CYP2C9); 17. Protocol G100515, a Phase 1 study to assess the pharmacokinetic drug:drug interaction of Formula IX and probenecid (UGT2B7); 18.
  • ketoconazole Cytochrome P450, Family 3,Subfamily A [CYP3A4] inhibitor
  • Protocol G100516, a Phase 1 study to assess the pharmacokinetic drug:drug interaction of Formula IX and rosuvastatin (breast cancer resistance protein [BCRP]); 19. Protocol G300504, a Phase 3 randomized, double-blind, placebo-controlled study of the effect of Formula IX on muscle wasting in 321 subjects with non-small cell lung cancer receiving first line platinum plus a taxane chemotherapy; 20. Protocol G300505, a Phase 3 randomized, double-blind, placebo-controlled study of the effect of Formula IX on muscle wasting in 320 subjects with non-small cell lung cancer receiving first line platinum plus a non-taxane chemotherapy; 21.
  • Protocol G200801 an ongoing, Phase 2, open label study to examine AR status and the activity of Formula IX hormonal therapy in 22 women with ER-positive metastatic breast cancer who have previously responded to hormone therapy.
  • the 18 mg dose Formula IX has been evaluated in 21 completed and ongoing clinical studies enrolling over 1,500 total subjects.
  • Formula IX has been generally well- tolerated, including single doses up to 100 mg and multiple doses up to 30 mg once daily for up to 14 days. In longer studies, Formula IX has also been generally well tolerated, including 1, 3, and 9 mg daily doses for up to 184 days.
  • Previous clinical studies demonstrated that daily doses up to 30 mg of Formula IX were well tolerated in healthy male volunteers.
  • Formula IX 3 mg was chosen for its anabolic activity in muscle for the completed Phase 3 program, a dose of 9 mg once daily was selected for hormonal therapy in the ongoing Phase 2 trial in ER+ and AR+ metastatic breast cancer in order to achieve a higher exposure that is both safe and more likely to be efficacious in women with advanced breast cancer.
  • CB clinical benefit
  • SD stable disease
  • Dosing Formula IX at 15–20 mg per day may provide therapeutic benefit in hormone receptor positive breast cancer by two separate mechanisms: activating AR and inhibiting progesterone receptor, thereby increasing potential efficacy.
  • Progesterone receptor expression in cancer stem cells has been shown to be involved in proliferation of cancer epithelial cells, and inhibiting progesterone receptor’s activity is now considered a novel approach to treating breast cancer.
  • Formula IX at higher doses might provide dual anti- proliferative effects in breast cancer.
  • doses of 15–20 mg per day should provide saturation of the AR potentially providing better efficacy as opposed to a lower dose with partial occupancy of the AR and absence of any progesterone receptor inhibitory effect.
  • the 18 mg dose is expected to be safe and generally well tolerated. However, in the event that a subject has a Grade 3 or greater toxicity, the 18 mg dose may be reduced to 9 mg until the AE resolves or for the remainder of treatment based on the Investigator’s discretion. The 9 mg dose has been previously studied in postmenopausal women with metastatic breast cancer and was safe and well tolerated. [000531] In TNBC patients, the 18 mg dose is preferred over a lower dose due to the aggressive phenotype of the disease and poor prognosis.
  • the 18 mg dose is more likely to saturate the AR and may lead to better clinical outcomes than a lower dose without receptor saturation or progesterone receptor inhibition.
  • the 18 mg dose may provide greater efficacy in TNBC without compromising subject safety. However, in the event that a subject has a Grade 3 or greater toxicity, the 18 mg dose may be reduced to 9 mg until the AE resolves or for the remainder of treatment based on the Investigator’s discretion. The 9 mg dose has been previously studied in postmenopausal women with metastatic breast cancer and was safe and well tolerated. [000533] In TNBC patients, the 18 mg dose is preferred over a lower dose due to the aggressive phenotype of the disease and poor prognosis.
  • Simon’s two-stage (optimal) design will be used to assess primary efficacy and will require up to 41 evaluable subjects; i.e., subjects with centrally confirmed AR+ who receive at least one dose of study drug.
  • 21 to 55 subjects including over-enrollees (see below) will be enrolled to receive a daily PO dose of Formula IX 18 mg.
  • Fourteen of the aforementioned subjects may be over-enrollees to allow for replacement of subjects to account for lack of centrally confirmed AR+ status, or for the rare subject who is enrolled but does not receive study drug.
  • the trial will test for an unacceptably low clinical benefit rate (CBR) of ⁇ 5% versus a CBR more consistent with ⁇ 20%.
  • CBR clinical benefit rate
  • the first stage will be assessed among the first 21 evaluable subjects. If at least 2/21 subjects achieve clinical benefit (CB) (defined as complete response [CR], partial response [PR], or stable disease [SD], per Response Evaluation Criteria in Solid Tumors [RECIST], Version 1.12) at week 16, then the trial will proceed to the second stage of recruitment of up to a total of 41 subjects in the evaluable subset of the Full Analysis Set (FAS). Otherwise, the trial will be discontinued for lack of efficacy. [000535] Subjects who are not confirmed AR+ may remain on the trial, but will not be part of the primary efficacy analysis – these subjects will contribute to secondary and tertiary analyses.
  • Subjects who experience an adverse event (AE) with Grade ⁇ 3 intensity may have a dose reduction from 18 mg to 9 mg per day or a drug interruption based on the medical judgment of the Investigator and after confirmation by the study Medical Monitor.
  • AE adverse event
  • NCI-CTCAE National Cancer Institute Common Terminology Criteria for Adverse Events
  • the subjects who demonstrate clinical benefit (CB) will be treated for up to 12 months from the date of the first dose of study treatment (as long as they continue to demonstrate CB from the treatment during these 12 months).
  • Subjects who continue to demonstrate a beneficial response from the study treatment at 12 months will be offered to continue in a safety extension study under a separate protocol. All subjects will be followed- up for one month after the last dose of Formula IX is received, for safety purposes.
  • Tertiary objectives The following tertiary efficacy objectives apply to both centrally confirmed AR+ subjects (the evaluable subset of the FAS) as well as to all subjects in the FAS: • Assess the effect of Formula IX 18 mg on serum prostate specific antigen (PSA). • Assess the effect of Formula IX 18 mg on Quality of Life (QoL) as measured by EQ- 5D-5L. • Assess the effect of Formula IX 18 mg on circulating tumor cells (CTCs). • Assess the impact of duration of prior CB on outcome. • Assess the impact of time from diagnosis of metastases to study enrollment on outcome.
  • PSA serum prostate specific antigen
  • QoL Quality of Life
  • CTCs circulating tumor cells
  • Subjects eligible for inclusion in this study must meet all of the following criteria: • Able and willing to give voluntary, written and signed, informed consent; • Women ⁇ 18 years of age; • Women with TNBC who have received at least one but no more than two prior chemotherapy regimens for the treatment of advanced or metastatic TNBC; • Confirmation of AR+ (defined as ⁇ 10% nuclear AR staining by immunohistochemistry [IHC]) TNBC in either the primary or metastatic lesion, assessed during the screening period by a local laboratory or by medical history; • TNBC confirmed by medical history as: human epidermal growth factor receptor 2 [HER2]-negative (confirmed by IHC 0, 1+ regardless of fluorescence in situ hybridization [FISH] ratio; IHC 2+ with FISH ratio lower than 2.0 or HER2 gene copy less than 6.0; FISH ratio of 0, indicating gene deletion, when positive and negative in situ hybridization [ISH] controls are present); estrogen receptor (ER) negative (confirmed as ER expression less than or equal to
  • Metastatic tumor tissue is preferred when possible; • Subjects must have either measurable disease or bone-only non-measurable disease, evaluable according to RECIST 1.1; • Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 at the time of screening and enrollment; • Negative pregnancy test in women of childbearing potential (premenopausal or less than 12 months of amenorrhea post-menopause, and who have not undergone surgical sterilization), no more than 7 days before the first dose of study treatment; • For women of childbearing potential who are sexually active, agreement to use a highly effective, non-hormonal form of contraception during and for at least 6 months after completion of study treatment; OR, a fertile male partner willing and able to use effective non-hormonal means of contraception (barrier method of contraception in conjunction with spermicidal jelly, or surgical sterilization) during and for at least 6 months after completion of study treatment; • Adequate organ function as shown by: Absolute neutrophil count ⁇ 1,500 cells/mm 3 ;
  • Formulation, Packaging, and Labelling [000544] Formula IX 3.0 mg Softgels will be supplied as opaque, white to off-white, size 5, oval Softgels.
  • the liquid Softgel fill is composed of Formula IX dissolved in polyethylene glycol 400. Dosing instructions will be provided on the study drug label and in the subject information sheet.
  • EXAMPLE 28 FORMULA IX REDUCED THE GROWTH OF HER2-POSITIVE TUMORS Methods
  • HCI-007 tumor pieces (1 mm 3 ) were implanted surgically (one per mouse) under the skin on the flanks of NSG mice. Simultaneously, 17 ⁇ -estradiol pellet (Innovative Research of America) was implanted surgically under the skin of each mouse.
  • Tumors were allowed to grow and reach approximately 100 mm 3 volume (l*W*W*0.526). Mice were randomized and treated orally with vehicle (15% DMSO+85%PEG-300), Formula IX (10 mg/kg), or enzalutamide (20 mg/kg). Tumor volume was measured weekly and represented as % change in tumor volume ( Figure 32). Mice were sacrificed and tumors stored for further analysis. Results [000546] As described in Figure 32, ER-positive, PR-positive, HER2-positive and AR- positive tumors of animals treated with vehicle and enzalutamide grew comparably, while the tumors of mice treated with Formula IX grew slowly. Tumors of animals treated with Formula IX regressed during the first 7 days, before started to slowly increase.
  • mice were randomized and treated orally with vehicle (15% DMSO + 85% PEG-300) or Formula IX (10 mg/kg). Tumor volume was measured weekly and represented as % change in tumor volume. Mice were sacrificed, tumors weighed, and stored for further analysis. Results [000549] As described in Figures 33A and 33B, triple positive HER2 tumors of animals treated with vehicle grew robustly, while the tumors of mice treated with Formula IX grew very slowly. Tumors of Formula IX treated animals did not grow appreciably through the duration of the experiment suggesting that there is almost a 100% tumor growth inhibition (TGI) ( Figure 33A). The tumor volume results are reflected in tumor weights observed at the end of the experiment ( Figure 33B).
  • TGI tumor growth inhibition
  • Example 30 INHIBITION OF PROLIFERATION AND GROWTH OF PATIENT-DERIVED XENOGRAFTS (PDX) AND TISSUES THAT EXPRESS WILDTYPE AND MUTANT REFRACTORY ER
  • PDX patient-derived xenografts
  • SARMs tissue-selective AR modulators
  • TaqMan PCR primers and fluorescent probes, master mixes, and Cells-to-Ct reagents were obtained from Life Technologies (Carlsbad, CA).
  • Cell culture medium and charcoal-stripped fetal bovine serum (csFBS) were purchased from Fisher Scientific (Waltham, MA).
  • FBS was purchased from Hyclone (San Angelo, TX).
  • AR-N20 antibody was procured from Santa Cruz Biotechnology (Santa Cruz, CA).
  • Enzalutamide was purchased from MedKoo Biosciences (Chapel Hill, NC).
  • ER- ⁇ (D8H8) antibody was procured from Cell Signaling (Danvers, MA).
  • Actin antibody, DHT, tamoxifen, and fulvestrant were purchased from Sigma (St. Louis, MO). Vetspon dental cubes/sponges (Patterson Veterinary Supplies Inc., NC0654350) were obtained from Fisher Scientific (Waltham, MA). Epidermal growth factor (EGF) was purchased from R&D systems (Minneapolis, MN), phorbol 12-myristate 13- acetate (PMA) was obtained from Acros organics, and 17 ⁇ -estradiol was obtained from Tocris (Bristol, UK). All other reagents used were analytical grade.
  • EGF Epidermal growth factor
  • PMA phorbol 12-myristate 13- acetate
  • Tocris was obtained from Tocris (Bristol, UK). All other reagents used were analytical grade.
  • Cell culture [000553] MCF-7 and ZR-75-1 cells were obtained from American Type Culture Collection (ATCC, Manassas, VA).
  • MCF-7 stable cells were generated by lentiviral infection of green-fluorescent protein (GFP) or the AR cloned into pLenti U6 Pgk-puro vector as described earlier (Narayanan et al. (2014) PLoS One 9, e103202; Yang et al. (2010) Canc Res 70, 8108-8116; Yepuru et al.
  • GFP green-fluorescent protein
  • HCI-7, HCI-9, and HCI-13 PDXs were gifts kindly donated by Dr. Alana Welm (Huntsman Cancer Institute, Salt Lake City, Utah).
  • HCI PDX tumor fragments (1 mm 3 ) were surgically implanted under the mammary fat pad in female NOD SCID Gamma (NSG) mice. Tumor volume was measured twice weekly for MCF-7 xenograft and once or twice weekly for HCI PDXs.
  • NSG NOD SCID Gamma mice.
  • Tumor volume was measured twice weekly for MCF-7 xenograft and once or twice weekly for HCI PDXs.
  • animals were sacrificed and tumors were excised, weighed, and stored for various analyses.
  • Patient specimen collection [000557] Specimens from breast cancer patients were collected with patient consent under a protocol approved by the UTHSC Institutional Review Board (IRB).
  • Specimens were collected immediately after surgery in RPMI medium containing penicillin:streptomycin and Fungizone and transported to the laboratory on ice. The tissues were finely minced and treated with collagenase for 2 hours. The digested tissues were washed with serum-free medium and frozen in liquid nitrogen in freezing medium (5% DMSO+95% FBS) or implanted under the mammary fat pad in female NSG mice.
  • Sponge culture [000558] HCI-13 tumors grown in female mice were allowed to reach 500-1000 mm 3 before the animals were sacrificed and the tumors were excised to be used for sponge culture. Patient specimens frozen in liquid nitrogen in freezing medium were used for sponge culture.
  • Sponge cultures were performed in accordance to the protocol published earlier (Dean et al. (2012) Cell Cycle 11, 2756-2761; Hu et al. (2016) Cancer Res 76, 5881-5893; Ochnik et al. (2014) Menopause 21, 79-88).
  • Tumors were sliced into small pieces ( ⁇ 1 mm 3 ) and incubated on pre- soaked gelatin sponges (5 fragments/sponge) in 12 well plates containing 1.5 mL medium (MEM+10% FBS+2 mM L-glutamine+10 ⁇ g/mL insulin+10 ⁇ g/mL hydrocortisone + penicillin:streptomycin).
  • the cultures were performed in triplicates for HCI-13 and singly for patient specimens.
  • RNA from tumors was extracted and verified qualitatively and quantitatively.
  • the arrays Human ST2.0, Affymetrix, Santa Clara, CA
  • Affymetrix Fluidics station 450 washed and stained on Affymetrix Fluidics station 450 and scanned on an Affymetrix GCS 3000 scanner.
  • Data from microarrays were normalized using Affymetrix Expression Console. Mean, Standard Deviation, and Variance were calculated across the groups. Fold Change from vehicle-treated samples was calculated, and a fold change of 1.5 was used as cutoff.
  • Arrays were probed with a total of 174 antibodies targeting a wide range of protein kinases and their activation via phosphorylation. Antibodies specificity was tested using standard immunoblotting on a panel of cell lysates. Selected arrays were stained with an anti-rabbit or anti-mouse biotinylated secondary antibody alone (Vector Laboratories Inc., Burlingame, CA and Dako Cytomation, Carpinteria, CA, respectively) and used as negative controls for nonspecific binding/background subtraction.
  • ChIP-Seq Chromatin immunoprecipitation assay
  • HCI-13 xenograft specimens were snap frozen and stored for ChIP-sequencing analysis.
  • ChIP-Seq study was performed in vehicle or Formula IX -treated HCI-13 PDX grown in NSG mice.
  • a whole cell lysate was made from tissues.
  • the lysate was sonicated using a Covaris E210 machine (Covaris Inc., Woburn, MA), for 30 min per sample (settings: duty cycle 20%, intensity 8 at 200 cycles per burst).
  • ER or AR was immunoprecipitated, washed, and the complex eluted.
  • the DNA-protein complex was reverse cross-linked by incubating at 65°C for 6 hours to overnight. After reverse cross-linking, precipitated and input DNA was purified using QIAquick PCR purification columns (Qiagen). [000565]
  • ThruPLEX-FD Prep Kit Rubicon Genomics, Ann Arbor, MI
  • Blocks of tissue were retrieved and sectioned at a thickness of 3 ⁇ M and mounted on glass slides. In order to assess co-localisation mirror image sectioning was used. The slides were then stained for ER ⁇ and AR (ER ⁇ , 1:50 dilution, Clone 6F11, Leica; AR, 1:50 dilution, Clone AR441, Dako) using immunohistochemistry as previously described (McNamara et al. (2013) Cancer Sci 104, 639-646; Niikawa et al. (2008) Clin Cancer Res 14, 4417-4426). Statistics [000567] Statistical analysis was performed using GraphPad prism software (La Jolla, CA).
  • SARM Formula IX is an AR agonist that binds to and activates the AR at less than 10 nM (Narayanan et al. (2014) PLoS One 9, e103202; Ponnusamy et al. (2017) Hum Mol Genet. 26(13), 2526-2540).
  • Formula IX has been evaluated in over 1000 patients in multiple clinical trials (see Example 27 for a partial list) and was shown to increase lean mass and physical function without having significant virilizing side effects (Dobs et al. (2013) Lancet Oncol 14, 335-345).
  • SARMs are non-metabolizable SARMs to weaker androgen or estrogen metabolites which confound results in breast cancer, which is in contrast with steroidal androgens such as DHT.
  • Formula IX inhibited (ER, PR, and AR positive) breast cancer cell proliferation
  • ZR-75-1 breast cancer cells that endogenously express AR, ER, and PR were treated with vehicle or a dose response regimen of Formula IX and the number of cells were counted after 6 days of treatment.
  • Proliferation of ZR-75-1 cells was significantly reduced dose dependently by Formula IX ( Figure 34A).
  • the results were reproduced in MCF-7 cells stably transfected with AR, but not with GFP ( Figure 34B).
  • Tumor microenvironment contains tumor epithelial cells, stromal cells, cancer- associated fibroblasts (CAFs), and endothelial cells. The collective function of these cells promotes the aggressive growth of tumors due to secretion of paracrine factors.
  • the CAFs are important for the sustained growth of cancers, and they differ from normal fibroblasts in their capacity to secrete factors that promote proliferation of cancer cells.
  • CAFs were isolated from an ER, PR, and AR-positive breast cancer tissue obtained from a 59-year-old African American patient (Sample ID 1005).
  • the CAFs were treated with vehicle, 10 nM DHT, or 1 ⁇ M Formula IX or 1 ⁇ M of an AR antagonist, enzalutamide.
  • Medium was collected over a period of 10 days and pooled.
  • CAFs were stained with SRB to evaluate the effect of AR ligands on proliferation.
  • MCF-7 cells stably transfected with GFP that lack AR were plated in 96 well plates and fed with conditioned medium obtained from CAFs treated with vehicle, DHT, Formula IX, or enzalutamide. Conditioned medium was replaced on days 4 and 7 and the cells were stained with SRB to measure viability. Both DHT and Formula IX -treated conditioned medium, but not enzalutamide-treated medium, inhibited the proliferation of MCF-7-GFP cells ( Figure 34C, right side).
  • Formula IX inhibited wild-type ER-positive breast cancer PDX (HCI-7) growth [000573] To determine if the growth inhibitory properties of Formula IX in vitro could be observed in vivo, Formula IX was tested in a PDX expressing wildtype AR. From the several PDXs available, three AR-positive PDXs were identified, based on gene expression profile.
  • HCI-13 was obtained from a patient who was treated with and relapsed from drugs ranging from ER-targeted therapeutics to chemotherapy (Table 12). To determine whether this ER mutant expressing PDX is dependent on estrogen for growth, HCI-13 tumors were implanted under the mammary fat pad in sham-operated and ovariectomized mice. Tumor growth was monitored over a period of 4 weeks. The growth rate in both sham-operated and in ovariectomized mice was comparable, indicating that the ER in HCI-13 is constitutively active and does not require estrogen to grow (Figure 36A).
  • breast cancer is also heterogeneous in its genomic profile as well as in its response to treatments.
  • To determine the effect of Formula IX and fulvestrant on growth inhibition breast cancer specimens obtained from patients were cultured, on dental sponges, as indicated above. The specimens were treated with vehicle, 1 ⁇ M Formula IX, or 100 nM fulvestrant. Three days after treatment, RNA was isolated from the tissues and expression of ER- and AR-target genes was measured.
  • HCI-13 expresses AR at levels comparable to that of LNCaP prostate cancer cells and the other specimens ranged from 0.2-20%, with the triple-negative specimen, 2005, having the least expression. Fulvestrant inhibited the ER function in 4 of 8 specimens, while Formula IX inhibited the ER function in 3 of 8 specimens ( Figure 36H- Figure 36I). Interestingly, Formula IX inhibited the ER function in specimen 1005, where fulvestrant functioned as an agonist. Specimen 1005 could be comparable to that of HCI-13 in its response to ER antagonists.
  • RNA from HCI-13 tumors obtained from animals shown in Figure 36B- Figure 36C were subjected to Affymetrix microarray.
  • 3029 genes were differentially regulated by Formula IX in HCI-13 tumors compared to vehicle-treated tumors.
  • Formula IX up-regulated 1792 genes and down-regulated 1237 genes.
  • Heatmap of the differentially regulated genes clearly indicates a shift in the expression pattern of genes due to Formula IX treatment ( Figure 37A).
  • Some of the most up-regulated genes include Cyp4F8, MYBPC1, RAB3B, LRRC26, AQP4, and CST4 ( Figure 37B).
  • ER-target genes such as TFF1, PGR, NRIP1 were down-regulated by Formula IX (not shown), other ER-target genes such as CTSD and CCND1 were not inhibited by Formula IX.
  • Formula IX functions in breast cancer by at least partially inhibiting the ER-signaling pathway to reduce the growth of cancer.
  • Some direct and indirect regulation of ER-targets were observed in Formula IX - treated samples. ER increases PDZK1 expression, which in-turn inhibits the expression of SLC26A3, a tumor suppressor gene .
  • Formula IX significantly inhibited the expression of PDZK1, which restored the expression of the tumor-suppressor gene, SLC26A3.
  • anti-apoptotic gene BCL-2 and genes present in its network such as PARP and WT1 were significantly down-regulated by Formula IX. Although these genes do not belong to the list of ER-direct target genes, cross-talk between ER and the BCL-2 pathway has been reported previously.
  • Expression of another class of oncogenic proteins, histone class was inhibited by Formula IX. About 17 members of histone group were inhibited significantly by Formula IX. The histone class has been implicated in aggressive cancers and endocrine-resistance (Nayak et al. (2015) Horm Cancer 6, 214-224).
  • IPA analysis did not provide any evidence for regulation of ERBB2 (human epidermal growth factor receptor 2 or HER2/neu) pathway by Formula IX
  • GSEA enrichment analysis revealed that Formula IX affected the genes regulated by ERBB2 ( Figure 37H). It is not clear at this point whether the regulation of ERBB2 is a result of growth inhibition or inhibition of the ER pathway. Irrespective of the mechanism, a downregulation of ERBB2 pathway, which is an oncogenic and tumor-promoting pathway, may be an added advantage of using Formula IX or an AR agonist in ER-positive breast cancers.
  • ChIP-Seq analysis demonstrates that Formula IX reprogrammed ER and AR cistromes [000585] Previous studies have demonstrated that the interaction of Y537S mutant ERwith DNA has been reprogrammed and might share limited similarity to the wildtype ER genome interaction. To determine if the effect of Formula IX on ER function is due to direct effect on ER binding to DNA, ChIP-sequencing was performed in the tumor samples obtained from animals shown in Figure 36B- Figure 36C. ER binding to 1248 regions (q ⁇ 0.05) on the DNA was reprogrammed by Formula IX, with 792 regions enriched with ER and 456 regions depleted of ER ( Figure 38A and Figure 38H). AR showed a similar pattern of DNA binding i.e.
  • ER regions enriched with ER were also enriched for AR, and the regions that were depleted of ER were also depleted of AR ( Figure 38A and Figure 38H).
  • the motifs that were enriched by the ER represent androgen response element (ARE; SEQ ID NO: 1), glucocorticoid response element (GRE; SEQ ID NO: 2), and Forkhead box protein A1 or FOXA1 response elements (FOXA1RE; SEQ ID NO: 3), while the regions that were depleted of ER represent estrogen response element (ERE; SEQ ID NO: 4) and FOXA1RE (SEQ ID NO: 5) ( Figure 38A and Figure 38H).
  • ARE androgen response element
  • GRE glucocorticoid response element
  • FOXA1RE Forkhead box protein A1 or FOXA1 response elements
  • the enriched cistrome motifs represent ARE, GRE, and FOXA1RE, while the depleted cistrome motifs represent ERE and FOXA1RE.
  • FOXA1 pioneering transcription factor is important for the function of both AR and ER and has overlapping binding sites with ARE and ERE, it is highly possible that the activated AR might sequester FOXA1 from the FOXA1REs adjacent to the EREs to open up the nucleosome and facilitate its binding to ARE and GRE.
  • ER is functioning as a complex with AR, it is also sequestered from EREs and FOXA1REs towards AREs, GREs, and FOXA1REs.
  • AR and ER colocalized in luminal B breast cancers
  • immunohistochemistry was performed in several luminal B breast cancer specimens. Nuclear immunoreactivity of both AR and ER and expression at high levels was observed in all the breast cancer specimens examined ( Figure 39). Additionally, several samples had moderate levels of cytoplasmic immunoreactivity for both markers.
  • Phospho-proteomic analysis showed the inhibition of oncogenic and induction of tumor- suppressor protein phosphorylation by AR agonist [000590]
  • phospho-proteomics was performed in HCI-13 tumors treated with vehicle or Formula IX.
  • Formula IX inhibited the phosphorylation of various oncogenic proteins such as pERK, PKC ⁇ , RSK3, Ezrin, BCL2, ELF4G, and ER ( Figures 41A- Figure 41C).
  • Formula IX also inhibited the expression of proliferation marker Ki67.
  • Formula IX increased the phosphorylation of tumor suppressor proteins such as p53, p27, ACC, and the AR.
  • Formula IX also increased the phosphorylation of STAT5, which could be a tumor suppressor or an oncogene depending on the context ( Figures 41A- Figure 41C). These results demonstrate that activating the AR with an agonist promotes the alteration of appropriate pathways that facilitate tumor growth inhibition. [000591] To understand the consequences of these protein alterations, cell signaling activators and inhibitors in HCI-13 sponge cultures were used to determine their effects on AR and ER target gene expression, FKBP5 and pS2, respectively. Because ERK and PKC phosphorylation were down-regulated by Formula IX, HCI-13 sponge cultures were treated with activators of the two pathways, EGF and PMA ( Figure 41D- Figure 41E).
  • the FOXA1 is sequestered away from EREs towards AREs to open the nucleosomes and facilitate the binding of the complex.
  • SARMs such as Formula IX
  • heterogeneity is seen the response of ER-positive clinical specimens and hence it might be optimal to pharmacogenomically screen for Y537S ER mutant expressing breast cancers in order to enriched the clinical benefit rate of Formula IX.
  • Tissue-selective AR agonism might offer an alternative hormonal approach for hormone receptor-positive breast cancers.
  • a prospective imaging sub-study was designed to demonstrate the proof-of-principle that FDHT-PET can be used to non-invasively image the presence of AR expression in breast cancer and to explore the potential of FDHT-PET as an imaging biomarker for evaluating response to SARM therapy.
  • Controlled Clinical Trials 1989; 10: 1-10 Subjects were randomized in a 1:1 fashion to one of the two dose arms. [000601] Randomization was stratified by subjects presenting with bone only metastases and all other subjects, and further by setting of immediately preceding therapy (adjuvant setting or metastatic setting) in order to balance the proportion of subjects with these presenting features in each dose arm. There was no intent to statistically compare the two dose arms, but to determine whether either or both doses resulted in an acceptable clinical benefit response (CBR), defined as the proportion of evaluable subjects (i.e., subjects with centrally confirmed AR+ and who received at least one dose of study drug) with either CR, PR, or SD by RECIST 1.1 at week 24 while maintaining an acceptable safety profile.
  • CBR clinical benefit response
  • Subjects on the 18 mg treatment arm who experienced an adverse event (AE) with Grade ⁇ 3 intensity may have had a dose reduction from 18 mg to 9 mg per day or a drug interruption based on the medical judgment of the Investigator and after confirmation by the study Medical Monitor.
  • the drug interruption may have lasted for a period of up to 5 days after which the subject must be rechallenged with study drug (18 mg or 9 mg) or discontinued from the study.
  • a dose reduction once the AE had resolved or reduced in intensity to Grade 1, the subject may have been rechallenged with 18 mg or maintained at 9 mg at the discretion of the Investigator.
  • Subjects on the 9 mg treatment arm who experienced an AE with Grade ⁇ 3 intensity (NCI-CTCAE 4.0) and/or intolerance may have had a drug interruption based on the medical judgment of the Investigator and after confirmation by the study Medical Monitor. The drug interruption may last for a period of up to 5 days after which the subject must be rechallenged with study drug (9 mg) or discontinued from the study.
  • For safety analysis subjects were analyzed in the treatment arm in which they were initially dosed.
  • For efficacy analysis subjects were analyzed according to the treatment arm to which they were randomized.
  • the subjects who demonstrated CB were treated for up to 24 months from the date of randomization (as long as they continued to demonstrate CB from the treatment during these 24 months).
  • Subjects who continued to demonstrate a CB from the study treatment at 24 months would have been offered to continue in a safety extension study under a separate protocol. For safety purposes, all subjects would have been followed-up for one month after the last dose of Formula IX was received. [000608] For safety purposes, all subjects were followed-up for one month after the last dose of Formula IX was received. [000609] Target Population: Adult postmenopausal women with metastatic or recurrent locally advanced ER+/AR+ BC. [000610] Study Duration: The study duration was estimated at 3 years.
  • TTP time to progression
  • the results demonstrate that, after a median duration on study of 81 days, 41 percent of all subjects (9/22) achieved CB as best response and also had increased PSA, which appeared to be an indicator of AR activity.
  • the study was still ongoing with one subject whose disease remained stable beyond 336 days.
  • Preclinical data with Formula IX suggested that it was also anabolic in bone and decreases bone turn over markers. Treatment with Formula IX may decrease bone turn over as compared with other hormonal therapies for the treatment of hormone receptor positive BC. Stronger bone microenvironment may decrease metastases to bone or delay time to skeletal related events.
  • the primary efficacy objective of this trial was to estimate the CBR at 24 weeks (defined as complete response [CR], partial response [PR], or SD) (by RECIST 1.1) of Formula IX 9 mg and of Formula IX 18 mg given PO daily in subjects with estrogen receptor positive and androgen receptor positive (ER+/AR+) BC who had centrally confirmed AR+ status.
  • the secondary efficacy objectives were to estimate the CBR at 24 weeks (by RECIST 1.1) of Formula IX 9 mg and 18 mg in all subjects randomized who received at least one dose of study medication (the full analysis set [FAS]) regardless of AR status as determined by the central laboratory.
  • the safety objective was to describe the safety profile of Formula IX 9 mg and 18 mg PO daily in subjects with ER+/AR+ BC with centrally confirmed AR+ as well as in all subjects randomized and treated.
  • the pharmacokinetic objective To describe the plasma concentrations of Formula IX and Formula IX glucuronide at each of the assessed time points.
  • Formulation, Packaging, and Labelling Formula IX 3.0 mg Softgels were supplied as opaque, white to off-white, size 5, oval Softgel capsules containing 3.0 mg of Formula IX.
  • the liquid Softgel fill was composed of Formula IX dissolved in polyethylene glycol 400.
  • Formula IX 3.0 mg Softgels were packaged in blister packs. Each blister pack contained sufficient study drug for one (1) week of dosing.
  • each blister pack was comprised of an appropriate number of blister strips (1 blister for the 9 mg treatment arm and 2 blisters for the 18 mg treatment arm) encased in a child- resistant heat-sealed card.
  • the blister strips were composed of a PVC/ACLAR base and an aluminum foil/PVC/PVAC copolymer and polymethacrylate (product contact) lidding. Perforations on the back of the heat-seal card overlay the foil lidding. To remove the study drug, subjects released the appropriate perforation by depressing a release button on the inside of the card. Once released, the perforation can be removed and the study drug pushed through the foil.
  • Pharmacokinetic assessment [000623] Blood samples for pharmacokinetic assessment were collected at baseline (pre-dose), Visit 3 (week 6), Visit 5 (week 18), and Visit 6 (week 24).
  • Table 13 describes a subset of the total number of patients. These were patients that were evaluable (AR+) with measurable disease at study entry and received palbociclib as a previous therapy. Table 13 [000628] According to Table 13, in the 9 mg Formula IX arm, there were 3 patients that previously were treated with palbociclib and failed. Among these 3 patients, one patient (33%) had a complete response (CR). In the 18 mg arm, there were 6 patients that were previously treated with palbociclib and failed.
  • Example 32 demonstrated that Formula IX maintained activity in patients that were resistant/non-responsive to the CDK 4/6 inhibitor (CDKi) palbociclib.
  • Inhibitors of cyclin-dependent kinase 4/6 in combination with estrogen endocrine therapy have become the standard of care for women with advanced breast cancer and these treatment failures represent a growing unmet need.
  • Tissue samples from an ER-positive, PR-negative, and HER2-negative (ER + , PR – , HER2 – ) patient that progressed on (i.e., was resistant to) palbociclib were implanted into animal (NSG mice) models and grown as a patient-derived xenograft (PDX).
  • Formula IX (SARM) is active alone or in combination and further Formula IX re-sensitized tumors to palbociclib. This is also observed in other models tested in vitro including a palbociclib resistant MCF-7 (MCF7 palbo R ) line ( Figure 45B). MCF7 palbo R cells (Palb R ) had a compensatory increase in CDK2 expression, indicative of resistance to CDK 4/6 inhibitors but maintained expression of AR and ER comparable to the parental line (data not shown). Lundberg, A. et al. Breast Cancer Res.21, 34 (2019).
  • Figure 45B demonstrated modest activity with either Formula IX alone (SARM) or palbociclib alone (Palbo) in Palb R cells but combination therapy (SARM + Palbo) demonstrated synergistic activity in MCF7 palbo R cells; providing another model where CDKi resistance can be overcome by Formula IX and palbociclib co-therapy.
  • this data supports the use of Formula IX alone or in combination with CDKi, and even after the patient has acquired resistance to CDKi and estrogen targeted endocrine co-therapies.
  • CDKi are approved for use with SERMs, aromatase inhibitors, and SERDs such as fulvestrant but eventual treatment failure produces a CDKi and estrogen endocrine therapy resistant population for which few treatment options are available.
  • This data supports use of Formula IX combined with a CDKi in this population, extending the time which ER-positive advanced breast cancers can be treated with hormonal therapies and kinase therapies rather than chemotherapies.
  • Formula IX can be more effective than existing estrogen endocrine therapies (for example, tamoxifen) or new CDKi (for example, palbociclib) standard-of-care treatments and, in the case of the latter, can be combined to unexpectedly enhance growth inhibition.
  • existing estrogen endocrine therapies for example, tamoxifen
  • new CDKi for example, palbociclib

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Pain & Pain Management (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
EP22740195.7A 2021-01-15 2022-01-14 Pharmazeutische zusammensetzungen zur behandlung von brustkrebs und verfahren zur verwendung davon Pending EP4277613A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US17/151,108 US20210228529A1 (en) 2012-07-13 2021-01-15 Pharmaceutical compositions for treating breast cancers and methods of uses thereof
PCT/US2022/012631 WO2022155543A1 (en) 2021-01-15 2022-01-14 Pharmaceutical compositions for treating breast cancers and methods of uses thereof

Publications (1)

Publication Number Publication Date
EP4277613A1 true EP4277613A1 (de) 2023-11-22

Family

ID=82448697

Family Applications (1)

Application Number Title Priority Date Filing Date
EP22740195.7A Pending EP4277613A1 (de) 2021-01-15 2022-01-14 Pharmazeutische zusammensetzungen zur behandlung von brustkrebs und verfahren zur verwendung davon

Country Status (9)

Country Link
EP (1) EP4277613A1 (de)
JP (1) JP2024503718A (de)
KR (1) KR20230163362A (de)
CN (1) CN116981451A (de)
AU (1) AU2022208712A1 (de)
BR (1) BR112023014247A2 (de)
CA (1) CA3205212A1 (de)
IL (1) IL304467A (de)
WO (1) WO2022155543A1 (de)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115369089A (zh) * 2022-08-11 2022-11-22 中山大学孙逸仙纪念医院 两种er阳性her-2阴性乳腺癌cdk4/6抑制剂耐药株及其构建方法和用途
CN117982655A (zh) * 2023-11-21 2024-05-07 北京大学人民医院 芳香化酶抑制剂治疗相关的疼痛动物模型构建方法及应用

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007027582A2 (en) * 2005-08-31 2007-03-08 University Of Tennessee Research Foundation Treating renal disease, burns, wounds and spinal cord injury with selective androgen receptor modulators
US10987334B2 (en) * 2012-07-13 2021-04-27 University Of Tennessee Research Foundation Method of treating ER mutant expressing breast cancers with selective androgen receptor modulators (SARMs)
RU2022108295A (ru) * 2016-06-22 2022-04-06 Эллипсес Фарма Лтд Способы лечения ar+ рака молочной железы

Also Published As

Publication number Publication date
AU2022208712A1 (en) 2023-08-10
CN116981451A (zh) 2023-10-31
CA3205212A1 (en) 2022-07-21
JP2024503718A (ja) 2024-01-26
IL304467A (en) 2023-09-01
BR112023014247A2 (pt) 2023-10-03
WO2022155543A1 (en) 2022-07-21
KR20230163362A (ko) 2023-11-30

Similar Documents

Publication Publication Date Title
AU2018201450B2 (en) A method of treating androgen receptor (AR)-positive breast cancers with selective androgen receptor modulator (SARMS)
US10849873B2 (en) Non-invasive method of evaluating breast cancers for selective androgen receptor modulator (SARM) therapy
US9969683B2 (en) Method of treating estrogen receptor (ER)-positive breast cancers with selective androgen receptor modulator (SARMS)
AU2013203600B9 (en) A method of treating androgen receptor (AR)-positive breast cancers with selective androgen receptor modulator (SARMS)
US9622992B2 (en) Method of treating androgen receptor (AR)-positive breast cancers with selective androgen receptor modulator (SARMs)
US10258596B2 (en) Method of treating HER2-positive breast cancers with selective androgen receptor modulators (SARMS)
US9744149B2 (en) Method of treating androgen receptor (AR)-positive breast cancers with selective androgen receptor modulator (SARMs)
AU2022208712A1 (en) Pharmaceutical compositions for treating breast cancers and methods of uses thereof
US10314807B2 (en) Method of treating HER2-positive breast cancers with selective androgen receptor modulators (SARMS)
US20220257552A1 (en) Methods of prescreening and treating breast cancers with selective androgen receptor modulators
WO2020028593A9 (en) A method of treating er mutant expressing breast cancers with selective androgen receptor modulators (sarms)
US20210228529A1 (en) Pharmaceutical compositions for treating breast cancers and methods of uses thereof
US20230172897A1 (en) Methods of treating breast cancers with selective androgen receptor modulators and additional therapeutic agents

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230714

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)