CN116981451A - Pharmaceutical composition for treating breast cancer and method of use thereof - Google Patents

Pharmaceutical composition for treating breast cancer and method of use thereof Download PDF

Info

Publication number
CN116981451A
CN116981451A CN202280021307.2A CN202280021307A CN116981451A CN 116981451 A CN116981451 A CN 116981451A CN 202280021307 A CN202280021307 A CN 202280021307A CN 116981451 A CN116981451 A CN 116981451A
Authority
CN
China
Prior art keywords
breast cancer
subject
positive
treating
inhibitor
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
CN202280021307.2A
Other languages
Chinese (zh)
Inventor
米切尔·施泰纳
拉梅什·纳拉亚南
安淳洲
詹姆斯·多尔顿
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 CN116981451A publication Critical patent/CN116981451A/en
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

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)

Abstract

The present invention relates to treating breast cancer in a subject. The method comprises the following steps: these methods comprise administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound and a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor by first determining 18F-16 beta-fluoro-5 alpha-dihydrotestosterone (18F-DHT) tumor uptake and identifying the subject as having AR-positive breast cancer based on F-DHT tumor uptake.

Description

Pharmaceutical composition for treating breast cancer and method of use thereof
Technical Field
The invention is thatTo the treatment of androgen receptor positive breast cancer in a subject (e.g., a male or female subject). Accordingly, the present invention provides the following method: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject having AR positive breast cancer with or without expression of Estrogen Receptor (ER), progesterone Receptor (PR) and/or human epidermal growth factor receptor 2 (HER 2); i) Treating a subject having Triple Negative Breast Cancer (TNBC); j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treatment, prevention, containment or inhibition of the disease Metastasis in a subject with breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the breast cancer in the subject; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound.
Background
Breast cancer is a disease that causes over 45000 women to die annually in the united states alone. More than 180000 new breast cancer cases are diagnosed each year, and it is estimated that one-eighth of women will develop breast cancer. These numbers indicate that breast cancer is one of the most dangerous diseases facing women today. Breast cancer also occurs in men, but with much lower incidence. Cancer research has not been able to identify the etiology of breast cancer and has not yet found a suitable treatment or prevention.
Genotyping has long been used to screen women who may be genetically predisposed to breast cancer. It is another diagnostic or prognostic tool that can be used to determine the effectiveness of a treatment. Some women are susceptible to breast cancer based on the presence of germline (i.e., genetic) mutations in the breast cancer susceptibility gene (BRCA) type 1 (BRCA 1) or BRCA 2. Both SERMs, tamoxifen 1999 and raloxifene 2007, were approved for primary prevention of breast cancer in high risk patient populations based on family history and/or genotypic considerations. However, hysterectomy or prophylactic mastectomy is generally considered a more definitive precaution in these patients. In 2018 and 2019, inhibitors of talazopania (Talzenna) and olapamide (lynparaza), poly ADP Ribose Polymerase (PARP) were approved for metastatic ER positive and HER2 negative breast cancer patients who had received chemotherapy and had some inherited BRCA mutations (hormone therapy if ER was positive). In 2019, an inhibitor of apicalist (Piqray), a phosphatidylinositol-3-kinase (PI 3K), has been approved for patients with certain functional gain mutations in the gene encoding the catalytic alpha-subunit of PI3K (PIK 3 CA), with inhibitory activity mainly against PI3K alpha. These mutations lead to activation of PI3kα and Akt signaling, cell transformation, and tumor production in vitro and in vivo models. PI3K inhibition by apicalist treatment has been shown to induce an increase in Estrogen Receptor (ER) transcription in breast cancer cells. In xenograft models derived from ER positive, PIK3CA mutated breast cancer cell lines, the combination of apilimbus and fulvestrant demonstrated enhanced antitumor activity compared to either treatment alone. PIK3CA mutations are present in about 30% -40% of breast cancer tumors, and are most prevalent in ER positive patients.
Current standards of care include screening for tumor expression levels of hormone receptor, estrogen Receptor (ER) and Progesterone Receptor (PR) and human epidermal growth factor receptor 2 (HER 2) kinase. Currently, women diagnosed with breast cancer may be initially treated by surgery, chemotherapy (and in some cases optional) and radiation therapy prior to initiation of targeted therapy. Hormone receptor positive breast cancers are sensitive to hormone therapy (also known as endocrine therapy) using selective estrogen receptor modulators or SERMs (e.g., tamoxifen, toremifene, raloxifene), aromatase inhibitors or AI (e.g., anastrozole, letrozole, exemestane), or selective estrogen receptor degradants or SERDs (e.g., fulvestrant). Hormonal therapies such as gonadotrophin releasing hormone (GnRH) agonists (commonly used in perimenopausal and perimenopausal women) and Aromatase Inhibitors (AI) (commonly used in postmenopausal women or in perimenopausal or perimenopausal women with GnRH agonists) block estrogen production in vivo, while SERMs and SERDs block the proliferative effects of estrogen on breast cancer cells. Although the prognosis is relatively good for most early ER-positive breast cancer patients compared to non-hormonal cancers, failure of adjuvant hormone therapy does lead to recurrence, including distant metastasis (i.e., advanced breast cancer). Metastatic or advanced breast cancers, whether hormone-naive or progressive despite endocrine therapy, are generally still ER positive and still rely on ER axis growth. Treatment of advanced breast cancer is rapidly evolving, from the use of endocrine monotherapy such as SERM or AI or fulvestrant, to endocrine therapy in combination with recently approved kinase inhibitors including cyclin dependent kinase 4/6 (CDK 4/6) inhibitors (pamphlet (2015 approval), rebaudinib (2017 approval) or abbe's (2017 approval), troraxili, len Luo Xili) or the mechanical targets of rapamycin (mTOR) inhibitors (everolimus (2012 approval)). These combination therapies delay progression of advanced breast cancer compared to endocrine therapy alone, and are replacing endocrine therapy alone in advanced breast cancer.
In this context, we propose the use of tumor genotyping (deep DNA sequencing) as a means of determining the status of estrogen receptor mutations in breast cancer patients as a basis for rational selection of therapies. Some common mutations in estrogen receptor α may occur during treatment and may be resistant to approved endocrine therapies, even when combined with various kinase therapies as described above. We have found that at least one of the mutations described herein (e.g., Y537S) is still sensitive to the androgen agonists of the invention despite the presence of SERM, AI and fulvestrant resistance. Thus, even advanced ER-positive AR-positive breast cancer patients who have been exposed to a complete endocrine and targeted therapy setting may still have further hormonal treatment options prior to being included in chemotherapy. If certain ER mutants are found by screening, they can be personalized, including the use of SARM to delay disease progression and/or to regress tumors.
HER2 positive breast cancers are sensitive to HER2 kinase inhibitors (e.g., trastuzumab, lapatinib, lenatinib, and icotinib) and are commonly used for metastatic disease. Anti-angiogenic therapy (bevacizumab) was also approved for metastatic disease, but FDA has abrogated this use of bevacizumab in 2011. Despite these multiple levels of targeted therapy, patients often suffer from or develop refractory forms of breast cancer. Examples of refractory breast cancers include primary tumors that are triple negative (lack ER, PR, HER 2), hormone resistant (SERM resistant, SERD resistant, or AI resistant), or kinase inhibitor resistant (e.g., inhibitors of CDK 4/6, mTor, and/or HER 2), or metastatic breast cancer tumors. Once all targeted therapies fail, e.g., the metastatic tumor is reactivated or the tumor metastasizes further, radiation therapy and high dose chemotherapy are required to ablate the refractory breast cancer tumor. Chemotherapeutic agents currently available for the treatment of refractory breast cancers include anthracyclines, taxanes and epothilones, which are toxic, dangerous, expensive, by intravenous injection and often ineffective, especially in the treatment of metastatic disease.
A great deal of clinical evidence suggests that androgens generally inhibit breast growth. For example, androgen deficient women have an increased risk of suffering from breast cancer. Androgen signalling plays a key role in mammary gland homeostasis, counteracting the proliferative effects of estrogen signalling in the mammary gland. However, when steroid androgens are biologically converted to estrogens (via the aromatase pathway), they increase the risk of cell proliferation and breast carcinogenesis. Historically, the steroid androgen receptor agonists testosterone, fluoxymesterone and carbosterone have been used to treat advanced breast cancer. These drugs have side effects such as excessive maleation, cross-reactivity with estrogen receptors, and aromatization of estrogens. The use of steroid androgens in advanced breast cancer precedes the screening of breast cancer hormone and kinase receptors. Recently, AR was found to be expressed in 50% -90% of breast tumors, providing a mechanism for targeted treatment of AR positive breast cancers using androgens.
Although most breast cancers are considered hormone receptor positive (ER, PR or HER 2), 15% -20% of women diagnosed with breast cancer will have Triple Negative Breast Cancer (TNBC) characterized by a lack of expression of ER, PR or HER 2. TNBC is more common in young patients (< 50 years) and generally exhibits more aggressive behavior. Until recently, those with advanced TNBC were limited to standard palliative treatment options, such as cytotoxic chemotherapy. The efficacy of chemotherapy is improved by delivering the active metabolite SN-38 of topoisomerase inhibitor I irinotecan to TNBC patients expressing Trop-2 (human trophoblast cell surface antigen 2) on their breast cancer cells using a conjugate therapy for antibodies with golimumab Sha Tuozhu (Trodelvy). Topoisomerase I and II are normal host enzymes found in mammalian nuclei and are required for normal DNA replication and cell division. These enzymes produce and subsequently repair single-stranded nicks in cellular DNA. The nicks allow the supercoiled double-stranded DNA to unwind and relax, allowing replication. Once the DNA torsional strain is relieved, the topoisomerase re-occludes the relaxed duplex. Topoisomerase activity is increased especially in rapidly dividing cells and cancer cells. If topoisomerase is inhibited, accumulation of DNA breaks can lead to inhibition of DNA replication and cell death. It represents a suitable but non-selective target for anticancer therapies.
Experimental microtubule disrupting agent saber Sha Bulin [ (2- (1H-indol-3-yl) imidazol-4-yl) (3, 4, 5-trimethoxyphenyl) methanone and the alternative resonating form (2- (1H-indol-3-yl) imidazol-5-yl) (3, 4, 5-trimethoxyphenyl) methanone ] has been demonstrated to bind to the colchicine binding site of tubulin and inhibit (CBSI) tubulin dynamics. These agents are orally active and, unlike colchicine, these CBSIs are not substrates for the outflow channel of anticancer agents pumped from cancer cells, and CBSIs can overcome taxane resistance. CBSI-based tubulin kinetics inhibit apoptotic cell death due to arrest in the G2/M phase of mitosis. Deng et al recently characterized sabelune in various in vitro and in vivo models of TNBC and demonstrated effective and efficient tumor growth inhibition in primary and metastatic tumors, as well as prevention of metastasis to distant organs including liver, lung, spleen and brain in taxane-sensitive (MDA-MB-231) and taxane-resistant TNBC models including xenografts derived from patients.
However, even after a preliminary response to chemotherapy, the duration of the response may be short and the likelihood of visceral metastasis, rapid disease progression and low survival is higher compared to hormone receptor positive breast cancer. Thus, research has focused on determining therapeutic targets in TNBC.
A new method of treating TNBC is approved, wherein antibodies blocking programmed death ligand 1 (PD-L1) are used in PD-LI positive patients. One such antibody is atilizumab (tecontriq; approved for TNBC in 2019), and the other is pembrolizumab (Keytruda; approved for TNBC in 2020). The median progression-free survival (PFS) was observed to be 7.4 months for patients with atino Li Zhushan antibody and albumin-bound paclitaxel, whereas the median progression-free survival was 4.8 months for patients with placebo and albumin-bound paclitaxel, which resulted in approval of atino Li Zhushan antibody. For pembrolizumab, median PFS was 9.7 months in the pembrolizumab plus chemotherapy group and 5.6 months in the placebo group.
One hormone receptor target that remains useful in ER positive, HER2 positive and triple negative breast cancers is the Androgen Receptor (AR). AR is the most highly expressed steroid receptor in breast cancer, with up to 95% of ER-positive breast cancers expressing AR (see example 9 below). In TNBC, up to 30% of cancers express AR. Historically, AR has been considered antiproliferative and beneficial in hormone receptor positive breast cancers. In TNBC, the data demonstrate that the presence of AR and androgen synthase correlates with lower proliferation, lower tumor grade, better overall survival and more favorable clinical outcome than those patients with TNBC that do not express AR. Evidence also suggests that the AR target gene Prostate Specific Antigen (PSA) is a favorable prognostic marker in breast cancer (not just TNBC). Based on these findings, research has focused on AR as a potential therapeutic target.
Long-term treatment of cancer with estrogen synthesis inhibitors (AI or GnRH agonists) or ER antagonists (SERMs or SERDs) leads to mutation of the target protein and activation of the resistance pathway. For example, continued treatment of ER-positive breast cancers with ER antagonists or Aromatase Inhibitors (AI) may develop resistance due to mutations in the ER Ligand Binding Domain (LBD). Clinical studies estimated that more than 30% of breast cancers treated with tamoxifen became refractory and relapsed as resistant cancers, and more than 40% of recurrent breast cancers expressed mutated ER. The mutant ER present in the treatment has escaped inhibition of the hormonal axis and is unable to respond to endocrine therapy, and therefore these patients will need to be treated with chemotherapeutic agents. Such cancers require new, non-toxic or less toxic effective endocrine therapies. One possibility is pharmacogenomic screening of tumor or circulating tumor cells to find mutant ERs that are resistant to existing endocrine therapies. This may be done when the molecular phenotype is ER positive (i.e. early stage disease) or in patients who fail endocrine therapy (i.e. late stage disease), such as SERM, AI, SERD and/or GnRH agonists, whether in combination with CDK 4/6 or mTor inhibitors.
Selective Androgen Receptor Modulators (SARMs) are compounds that exhibit AR-mediated tissue selective activity. Unlike their steroid precursors, SARMs are non-aromatic, generally exhibit no activity at other steroid receptors including ER and PR, and are non-masculinizing. Furthermore, SARMs may be beneficial to refractory breast cancer patients because they have high muscle anabolic effects, which would increase their tolerance to high doses of chemotherapy. In addition, SARM has beneficial osteoblast and anti-osteoclast effects in bone, which may reduce the risk of metastasis to bone or may reduce the risk of osteoporosis during endocrine and/or chemotherapy.
There is an urgent need at both the basic science and clinical level for new innovative approaches to develop compounds that can be used for the following purposes: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treatment of a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitors (ibuzol (Ibrance), rebabocili (Kisqali), abbe's cili (Vorzenio), trasturil, ly Luo Xili), apifil (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3kα)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olapamide (Lynparza) or talazopali (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitors (lapatinib, lenatinib (Nerlynx), dacatinib (Vizimpro) or tukartinib (Tukysa)), HER2 antibodies (trastuzumab (Herceptin), pertuzumab (perta), maceraxib (margenzab)), HER2 antibody drug conjugates (HER 2 ADC) (trastuzumab (enzab), kazadol, or anti-panaxomum (tzbead anti-panaxzepan) and anti-therapeutic anti-panaxoml (therapeutic anti-panaxoml) or anti-panaxoml (tzbezoma) (therapeutic anti-panaxmajub) (pantizob) or anti-panaxmajus (tzjus) (pantizob) or anti-panaxmajus (pantijus); l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; and/or p) extending progression free survival of a subject having breast cancer; q) treating, preventing, suppressing or inhibiting AR positive triple negative breast cancer; r) treating a subject having HER2 positive breast cancer; s) treating a subject having a breast cancer that expresses an ER mutant, t) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or u) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the subject's breast cancer.
Disclosure of Invention
In one aspect, the invention provides a pharmaceutical composition comprising a Selective Androgen Receptor Modulator (SARM) compound and a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor, wherein the SARM compound is represented by the structure of formula I:
wherein the method comprises the steps of
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 (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, CN, NO 2 、COR、COOH、CONHR、CF 3 、Sn(R) 3 Or R is 3 Together with the benzene ring to which it is attached, form a fused ring system represented by the structure:
z is NO 2 CN, COR, COOH or CONHR;
y is CF 3 F, br, cl, I, CN 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 A, B or C:
n is an integer from 1 to 4; and is also provided with
m is an integer of 1 to 3, or
Optical isomers, racemic mixtures, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides or crystals thereof.
In some embodiments, the SARM compounds of the compositions of the present invention are represented by the structure of formula VIII, IX, X, XI, XII, XIII or XIV:
in a further aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof and a CDK 4/6 inhibitor,
in some embodiments of the compositions of the invention, the CDK 4/6 inhibitor is palbociclib, rebabociclib, trazocili, la Luo Xili, or abbe cili. In certain embodiments, CDK 4/6 is palbociclib. In some embodiments, the CDK 4/6 inhibitor is arbeli.
In another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and palbociclib,
in another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and Rabociclib,
In another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and treamxili,
in another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and L Luo Xili,
in another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and Abeli,
in another aspect, the invention provides a method for treating a subject having breast cancer, the method comprising administering to the subject a pharmaceutical composition of the invention as described herein.
In some embodiments of the methods of the invention, the breast cancer is 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 failed with the following treatment: selective Estrogen Receptor Modulators (SERMs) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonists (goserelin), aromatase Inhibitors (AI) (letrozole, anastrozole, exemestane), fluvalirome, cyclin dependent kinase 4/6 (CDK 4/6) inhibitors (cabonine (Ibrance), rapaminib (Kisqali), abbe-zenio), trazoxiline, ly Luo Xili), axiliziram (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olpataline (Lynparza) or talazopanil (Talzenna)), human growth factor receptor 2 (CDK 4/6) inhibitors (lapatinib, letinib (nerlozenb), dasatib (vic), vicat or vicat), anti-bead anti-tuzumab (Herceptin) or anti-bead anti-tuzumab (Herceptin) anti-HER 2 (Herceptin), anti-bead anti-tuzumab (tuzuelan) (panaxb) or anti-tuzuelan) anti-tuzuelan (HER 2 (panaxb) or anti-tuzuelan) anti-tuzuelan (panaxb) Pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) treatment.
In some embodiments, breast cancer fails to be treated with cyclin dependent kinase 4/6 (CDK 4/6) inhibitors. In some embodiments, a subject in a method of the invention is resistant to or non-responsive to a CDK 4/6 inhibitor. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance), rebabociclib (Kisqali), traracili, len Luo Xili, or abbe cili (Vorzenio). In certain embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance). In some embodiments, the CDK 4/6 inhibitor is abbe-cili (Vorzenio).
In some embodiments, the compositions of the invention as described herein re-sensitize the breast cancer to treatment with a CDK 4/6 inhibitor. In some embodiments, the compositions of the invention as described herein overcome estrogen endocrine resistance. In some embodiments, the compositions of the invention as described herein overcome resistance to a combination therapy of estrogen endocrine and CDK 4/6 inhibitors. In some embodiments, the estrogenic endocrine therapy comprises at least one of tamoxifen, toremifene, raloxifene, exemestane, letrozole, anastrozole, and fulvestrant. In some embodiments of the methods of the invention, the CDK 4/6 inhibitor is at least one of palbociclib (Ibrance), rebabociclib (Kisqali), trazoxili, len Luo Xili, and abbe zenio.
Drawings
The subject matter regarded as the invention is particularly pointed out and distinctly claimed in the concluding portion of the specification. The invention, however, both as to organization and method of operation, together with objects, features, and advantages thereof, may best be understood by reference to the following detailed description when read with the accompanying drawings in which:
FIGS. 1A-1J show that DHT and the compound of formula IX inhibit MDA-MB-231 triple negative breast cancer cell growth. FIG. 1A shows MDA-MB-231 cell expression of AR after transfection. FIG. 1B shows IC in AR positive MDA-MB-231 cells 50 . Figures 1C to 1J show the effect of DHT, formula IX, bicalutamide and the (R) enantiomer of formula IX on the percent (%) cell viability. (the cells of FIGS. 1C, 1E, 1G and 1I were treated in activated charcoal-desorbed FBS. The cells of FIGS. 1D, 1F, 1H and 1J were treated in whole serum). ● MDA-MB-231 having lacZ; MDA-MB-231 with AR 200. Mu.L; has an AR of 500. Mu.L MDA-MB-231.
Figures 2A-2H show that DHT and formula IX inhibit HCC-38 triple negative breast cancer cell growth. FIG. 2A shows HCC-38 cell expression of AR after transfection. FIG. 2B showsIllustrating IC in AR-positive HCC-38 cells 50 . Figures 2A to 2H show the effect of DHT, formula IX and bicalutamide on the percent (%) cell viability. (FIG. 2C, FIG. 2E and FIG. 2G cells were treated in activated charcoal-desorbed FBS. The cells of FIG. 2D, FIG. 2F and FIG. 2H were treated in whole serum). ● HCC-38 with lacZ; HCC-38 with AR 200. Mu.L; has an AR of 500. Mu.L HCC-38.
Figures 3A to 3E show that the effect of DHT and formula IX on MDA-MB-231 cells was reversed by bicalutamide. Figures 3A to 3D show the effect of DHT or formula IX on the percent (%) cell viability in the presence or absence of bicalutamide. (FIG. 3A and FIG. 3C cells were treated in activated charcoal-desorbed FBS. The cells of FIG. 3B and FIG. 3D were treated in whole serum). ● lacZ and 10. Mu.M bicalutamide was used; o lacZ; is AR and uses 10 μm bicalutamide; ΔAR. FIG. 3E shows IC in AR positive cells in the presence or absence of pretreatment with bicalutamide 50 Values.
Fig. 4A-4Q show that AR agonists inhibit triple negative breast cancer cell growth. Fig. 4A, 4B, 4E, 4F, 4G, 4H, 4K, 4L, 4M, 4N, 4O and 4P show the effect of AR agonist on the percent (%) cell viability. Fig. 4C and 4D show the effect of AR antagonists on the percent (%) cell viability. Fig. 4I and 4J show the effect of AR non-binding agent on the percent (%) cell viability. Fig. 4A, 4C, 4E, 4G, 4I, 4M and 4O cells were treated in activated charcoal-desorbed FBS. The cells of fig. 4B, 4D, 4F, 4H, 4J, 4L, 4N and 4P were treated in whole serum. FIG. 4Q shows EC in AR positive cells 50 Value sum IC 50 Values.
FIG. 5 shows that the growth inhibitory ligand is an AR agonist in MDA-MB-231 cells.
FIGS. 6A-6E show that growth inhibition in MDA-MB-231 cells is selective for AR. FIGS. 6A and 6B show expression of ERα or ERβ, respectively, in MDA-MB-231 cells after transfection. Fig. 6C, 6D and 6E show the effect of estradiol (E2) or ICI 182,780 (ICI) on the percent (%) cell viability. (cells of FIG. 6C were treated in activated carbon desorbed serum. Cells of FIGS. 6D and 6E were treated in whole serum).
FIG. 7 shows that DHT alters the morphology of MDA-MB-231 cells.
Figure 8 shows the effect of formula VIII on steroid receptor transactivation (agonist mode).
Fig. 9 depicts dose response curves for PR activity (antagonist mode) for compounds of formula VIII, compounds of formula IX, R-enantiomer of formula IX, and RU 486. The filled circles (+) correspond to data points VIII (IC) 50 =17.05 nM); the open circle (. Smallcircle.) corresponds to formula IX (IC 50 =162.9 nM); the solid triangle (, t) corresponds to the R-enantiomer of formula IX (IC 50 =1689 nM); the hollow triangle (delta) corresponds to RU486 (IC 50 =0.048nM)。
FIGS. 10A-10B illustrate that SARM (formula VIII) inhibits MDA-MB-231-AR tumor growth. Body weight (10A) and tumor size (10B) were measured in intact female nude mice with 150mm3-200mm3 tumors from MDA-MB-231-AR triple negative breast cancer cells for 35 days, followed by oral administration of vehicle Or 30mg/kg of formula VIII.
FIG. 11 shows that SARM (formula VIII) inhibits MDA-MB-231-AR tumor growth. After 35 days, 150mm with triple negative breast cancer cells from MDA-MB-231-AR 3 -200mm 3 Tumors were then measured in mm in intact female nude mice that received vehicle or 30mg/kg of VIII orally administered 3 Tumor size (left pane) and change in tumor size (middle pane) and tumor weight (right pane) were counted.
FIG. 12 shows the morphology of MDA-MB-231 breast cancer cells stably transfected with AR (MDA-MB-231-AR cells). The results indicate that AR agonist, DHT, formula IX and formula VIII change morphology to a more anchored phenotype compared to vehicle, bicalutamide or inactive isomer of formula IX. This may be indicative of a less metastatic breast cancer phenotype.
FIGS. 13A-13C show binding and transactivation of the indicated ligand to HEK-293 (13A) or MDA-MB-231 (13B and 13C) cells. DHT, formula IX and formula VIII are agonists of AR in breast cancer cells. Example 16
FIG. 14 shows the antiproliferative activity of DHT and SARM in MDA-MB-231 breast cancer cells stably transfected with AR. MDA-MB-231 cells stably transfected with AR using lentiviruses were treated with the indicated ligands for 6 days and the cell numbers were counted using a Coulter counter. DHT and SARM (VIII and IX) instead of the AR antagonist bicalutamide inhibited proliferation of MDA-MB-231 triple negative breast cancer cells stably transfected with AR.
FIG. 15 presents microarray results showing that activated AR (AR activated by the compound of formula VIII) inhibits expression of more genes than their expression induced in MDA-MB-231-AR xenograft breast cancer cells.
Fig. 16 depicts verification of microarray results.
FIG. 17 shows that formula VIII inhibits the growth of MCF-7-AR triple positive xenografts.
Figure 18 presents inhibition of uterine weight gain in estrogen-supplemented animals treated with formula VIII demonstrating the ability of SARMs to counteract estrogen stimulation in vivo.
Fig. 19 shows that the AR expression pattern in response to AR agonist (formula VIII) is similar to that observed in prostate cancer cells.
Fig. 20 depicts verification of microarray results.
FIG. 21 shows upregulation of JNK phosphorylation in MCF7-AR tumors using formula VIII.
Figure 22 shows inhibition of Triple Negative Breast Cancer (TNBC) growth using formulas VIII and IX. In the TNBC model using MDA-MB-231-AR cells in nude mice, formulas VIII and IX demonstrated about 85% TGI at all doses tried (5 mg/kg, 10mg/kg for formula VIII; 5mg/kg, 10mg/kg, 30mg/kg for formula IX).
Figure 23 shows inhibition of triple negative breast cancer using formulas VIII and IX. The tumor weight was also reduced for all doses of formula VIII and formula IX. Spleen enlargement was observed only in vehicle treated mice (680 mg versus 200mg-300mg in normal mice), possibly indicating prevention of tumor metastasis to the spleen by SARM.
Figure 24 shows body weight gain by SARM at all doses of formula VIII and formula IX, indicating healthy growth and lack of toxicity. By comparison, vehicle treated animals did not grow robustly.
FIGS. 25A-25E depict antagonism of SARM's ability to activate ER target genes in MCF-7-AR cells. FIGS. 25B and 25D show that addition of AR to MCF-7-AR cells (as opposed to Green Fluorescent Protein (GFP) as seen in FIGS. 25A and 25C) increases the effect of estradiol (when not antagonized) on ER target genes PR and PS2, respectively. The addition of AR to MCF-7-AR cells in the presence of SARM or SARM+ estradiol (E2) alone inhibited activation of these ER targets compared to GFP transfected cells (i.e., no AR; FIGS. 25A and 25C). FIG. 25E shows that SARM enhances the AR target gene even in the presence of estradiol.
FIGS. 26A and 26B depict immunohistochemistry of two regions of the same BR-0001 tumor (triple negative breast cancer (TNBC)). They showed that AR expression was consistent throughout formalin fixed, paraffin embedded (FFPE) tissues stained with AR antibody (AR N20 from SCBT). Fig. 26C depicts immunohistochemical staining of AR negative TNBC FFPE tumors as negative control.
Fig. 27A-27C depict the inhibition of BR-0001 tumor xenograft growth over time of formula IX in terms of breast cancer tumor volume (fig. 27A and 27B) and weight (fig. 27C) compared to enzalutamide (Enza) or vehicle. Experiments 1 and 2 are repeated experiments with n=5 and n=10 animals, respectively, at different times. Fig. 27A provides the results of experiment 1, fig. 27B provides the results of experiment 2, and fig. 27C provides the results of experiment 2. Will be 1mm 3 NOD Scid Gamma (NSG) mice were subcutaneously implanted with (approximately) BR-0001TNBC fragments. Once the tumor reached 100mm 3 -200mm 3 Animals were randomized and orally treated with vehicle, 10 mg/kg/day of formula IX or enzalutamide. Tumor volumes were measured three times per week. Animals were sacrificed and tumors were weighed.
FIGS. 28A-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% decrease in Ki-67 staining after 2 weeks of treatment. Tumors from experiment 2 were fixed in formalin and paraffin embedded. Sections were cut and stained with Ki-67 antibody (fig. 28A), ki-67 staining was reduced in tumors of animals treated with formula IX. Ki-67 positive cells in each slide were counted (200 total cells per field) and expressed as stained cells (FIG. 28B). For reference, the illustrations in the figures are bars of 200 micrometers (μm) length.
FIG. 29 depicts Z scores for 50 genes (PAM 50) used to identify BR-0001. PAM50 is a set of 50 genes used to classify breast cancer. PAM50 gene expression data indicated that BR-0001 tumors belonged to the basal-like breast cancer (BLBC) subtype of TNBC. The expression of the 50 genes required to classify breast cancer is given here (Z score).
Fig. 30A and 30B depict gene expression data that can be used to classify basal-like breast cancers (BLBC) as sub-classifications as compared to published genes (Pietenpol set). Sub-classification suggests that BR-0001 belongs to the Luminal Androgen Receptor (LAR) and mesenchymal stem cell-like (MSL) subtypes. Six TNBC subtypes according to the Pietenpol group include two basal-like (BL 1 and BL 2) subtypes, an Immunomodulatory (IM) subtype, a mesenchymal (M) subtype, a mesenchymal stem cell-like (MSL) subtype, and a Luminal Androgen Receptor (LAR) subtype. GE-gene expression.
FIG. 31 depicts the change in gene expression in BR-0001 tumors treated with formula IX.
Figure 32 depicts the reduction of tumor growth for ER positive, PR positive, HER2 positive and AR positive tumors composed of HCI-007 cells with formula IX.
Fig. 33A and 33B depict the use of formula IX in xenografts composed of HCI-013 cells to effectively reduce tumor growth. The HCI-013 phenotype was tri-positive and also expressed AR. Fig. 33A: tumor volume change (%) and fig. 33B: tumor weight (g).
Fig. 34A-34E depict AR agonists inhibiting proliferation of ER-positive and AR-positive breast cancer cells. FIG. 34A depicts that formula IX inhibits proliferation of ZR-75-1 cells. ZR-75-1 breast cancer cells (n=4/treatment) inoculated in growth medium were treated with the indicated dose of formula IX was treated for 6 days, medium was changed and re-treated on day 3. After 6 days of treatment, cells were harvested and counted. FIG. 34B depicts that formula IX inhibits proliferation of MCF-7 cells expressing AR. MCF-7 cells stably transfected with GFP (MCF-7-GFP) or AR (MCF-7-AR) were seeded in 96-well plates (n=4/treatment) in growth medium and treated with the indicated doses of formula IX. After 3 days the medium was changed and reprocessed. Cells were fixed 6 days after treatment and cell viability was measured by SRB assay. FIG. 34C depicts the secretion of factors by breast cancer fibroblasts treated with AR agonists that inhibit MCF-7-GFP cells lacking supplemented AR. Primary fibroblasts obtained from breast cancer patients were cultured in growth medium and treated in triplicate with vehicle, 10nM DHT, 1 μm enzalutamide, or 1 μm formula IX. The medium was changed and the cells were reprocessed on days 4 and 7. The medium was collected, combined in triplicate and stored at-80 ℃. 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 inoculated in the growth medium. 24 hours after inoculation, the cells were fed with conditioned medium obtained from fibroblasts derived from the patient as described above. The cells were fed with conditioned medium for 10 days, with medium changes at day 4 and day 7. After 10 days of treatment, cells were fixed and viability was measured by SRB assay. FIG. 34D depicts that AR ligands do not inhibit the growth of ER-negative AR-positive HCI-9 PDX. As 1mm, AR-positive but ER-negative HCI-9PDX was used 3 Fragments were surgically implanted under the mammary fat pad of NSG mice (n=8-10/group). Once the tumor reached 100mm 3 -200mm 3 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 volumes were measured three times per week. Figure 34E depicts that HCI-13ER- α is resistant to the ER antagonists fulvestrant and tamoxifen (right pane) compared to wt-ER- α (left pane). ER-alpha from HCI-13 was cloned into pCR3.1 vector. Wild-type ER-alpha and HCI-13 ER-alpha, ERE-LUC and CMV-LUC were transfected into COS-1 cells using lipofectamine. 24 hours after transfection, cells were treated with vehicle, 0.1nM estradiol, 10nM fulvestrant or 1. Mu.M tamoxifen in combination with 0.1nM estradiol. Collecting after 24 hours of treatmentCells were obtained and luciferase assays were performed. ER antagonists in wt-ER-alpha are significantly different from vehicle-treated wt-ER-alpha, e.g. p<0.05. AR-androgen receptor; GFP-green fluorescent protein; DHT-5 alpha-dihydrotestosterone; e2-17 beta-estradiol; ER-estrogen receptor; SARM-selective androgen receptor modulators; SRB-sulforhodamine B; mpk-mg/kg body weight. Values are expressed as n=3-4/mean of data points ± s.e.
Fig. 35A-35D depict AR agonists inhibiting proliferation and growth of wild-type and mutant ER and AR positive xenografts. FIG. 35A depicts proteins from HCIPDX (HCI-7, 9 or 13) tumor fragments extracted and fractionated on SDS-PAGE and Western blotted against AR. AR was also quantified at mRNA level and expressed as fold change in AR from LNCaP prostate cancer cells (numbers provided under the blot). Fig. 35B (same as fig. 32) and fig. 35C depict the inhibition of HCI-7 tumor growth by formula IX. AR-positive HCI-7PDX expressing wild type ER was taken as 1mm 3 Fragments were surgically implanted under the mammary fat pad of NSG mice (n=8-10/group). Once the tumor reached 100mm 3 -200mm 3 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 volumes were measured weekly. At the time of sacrifice, tumors were removed, weighed (fig. 35C) and stored for further analysis. Fig. 35D depicts subcutaneous implantation of MCF-7 cells (MCF-7-AR) (300 tens of thousands of cells/mouse) stably expressing AR into ovariectomized mice (n=8/group) supplemented with 17β -estradiol. Once the tumor reached 100mm 3 -200mm 3 Mice were randomized and treated with vehicle or formula IX (10 mpk p.o.). Tumor volumes were measured twice weekly. * =p <0.05; HCI-hounsmei cancer institute; AR-androgen receptor; ER-estrogen receptor; NSG-NOD SCID gamma; PDX-patient-derived xenograft; OVX-ovariectomy; PR-progesterone receptor; mpk-mg/kg body weight.
Fig. 36A-36K depict AR agonists inhibiting proliferation and growth of mutant ER and AR positive xenografts. Figure 36A depicts that HCI-13PDX growth is independent of circulating estrogen. HCI-13PDX tumor fragment was used as 1mm 3 Fragment passageThe sham or ovariectomized NSG mice (n=6/group) were surgically implanted under the mammary fat pad. Tumor volumes were measured weekly. FIGS. 36B and 36C depict inhibition of growth of HCI-13PDX by an AR agonist (formula IX). AR positive HCI-13PDX expressing mutant ER was taken as 1mm 3 Fragments were surgically implanted under the mammary fat pad of NSG mice (n=8-10/group). Once the tumor reached 100mm 3 -200mm 3 Mice were randomized and treated with vehicle (DMSO: PEG-300 (15%: 85%)) or formula IX (10 mpk p.o.). Tumor volumes were measured weekly. At the time of sacrifice, tumors were removed, weighed (fig. 36C) and stored for further analysis. FIGS. 36D-36G depict inhibition of ER target genes in HCI-13 ex vivo sponge cultures by AR agonists, but not AR or ER antagonists. HCI-13 tumor (1 mm) 3 ) Cultured on gelatin sponge in growth medium (n=3/group; each n is obtained by combining 5 fragments). Tissues were treated with vehicle, 10nM DHT, 1. Mu.M formula IX, 1. Mu.M enzalutamide or 100nM fulvestrant for 3 days. RNA was extracted from the tissues, gene expression was measured by real-time PCR and normalized to GAPDH. Fig. 36H-36J depict the effect of formula IX on ER positive breast cancer patient samples. Breast cancer samples obtained from patients were cultured on gelatin sponge (n=1; each n was obtained from 5 tumor fragments). Tissues were treated with vehicle, 1. Mu.M formula IX or 100nM fulvestrant for 3 days. RNA was extracted from the tissues, gene expression was measured by real-time PCR and normalized to GAPDH. The table in FIG. 36K shows the fold difference in expression of AR and ER at the mRNA level compared to HCI-13 tumors. * =p<0.05; HCI-hounsmei cancer institute; AR-androgen receptor; ER-estrogen receptor; NSG-NOD SCID gamma; PDX-patient-derived xenograft; MKI67-mRNA of the Ki67 proliferation index protein; OVX-ovariectomy; PR-progesterone receptor; mpk-mg/kg body weight.
FIGS. 37A-37H depict gene expression studies in HCI-13PDX, which demonstrate inhibition of the ER pathway by AR agonists. RNA was isolated from HCI-13PDX xenografts treated with vehicle or formula IX (fig. 36B-36C) and subjected to microarray (n=4/group). The different genes in the formula IX treatment group (q < 0.05) are represented in the heat map (upper 1/5 of the left column of the heat map (vehicle treatment) was mainly up-regulated (initially red), while lower 4/5 of the heat map was mainly down-regulated (initially green), in contrast to the reverse column of the formula IX treatment (green top, red bottom)) (fig. 37A). The log fold change in expression of the top up-and down-regulated genes is shown in figure 37B. The classical pathway, upstream regulatory factors and diseases represented by the enriched genes obtained from the Inventive Pathway Analysis (IPA) are shown in fig. 37C. Representative ER and AR target genes and the uppermost and lowermost genes are shown in FIGS. 37D-37G. Figure 37H depicts that GSEA KEGG pathway analysis provides the ERBB2 (ERBB is an abbreviation for erythroblastic oncogene B; also often referred to as HER2 (from human epidermal growth factor receptor 2) or HER 2/neu) pathway, as one of the pathways highly correlated with formula IX treatment (bottom four rows in the left column (vehicle treatment) are down-regulated genes (original color blue) while most rows are up-regulated genes (original color red), in contrast to the column of formula IX treatment (right) which is the exact opposite. * =q <0.05; ER-estrogen receptor; AR-androgen receptor; PDX-patient-derived xenograft; GSEA-gene set enrichment analysis; KEGG-kyoto gene, genome encyclopedia.
FIGS. 38A-38H depict rearrangements of ChIP sequencing showing ER and AR binding to DNA. Fig. 38A depicts chromatin immunoprecipitation (ChIP) assays with ER in tumors treated with vehicle (n=4) or 10 mg/kg/day of formula IX (n=3) or AR (n=1) (tumors from animals shown in fig. 36B-36C). Next generation sequencing was performed to determine ER and AR binding to the whole genome of DNA. A heat map of significantly different peaks (ER peak and corresponding AR peak q < 0.05) is shown. The top enriched motif is shown in figure 38H. FIG. 38B shows representative peaks from KLK3 regulatory regions of ER and AR ChIP-Seq. FIG. 38C shows a Principal Component Analysis (PCA) plot of vehicle corresponding to the ER-ChIP peak and the samples treated with formula IX. FIG. 38D depicts the ChIP assay with AR or ER antibodies in HCI-13 samples treated with vehicle or formula IX and real-time PCR of specific regions with primers and Taqman probes. FIG. 38E depicts a pie chart showing ER enrichment distribution in a HCI-13 sample treated with formula IX. For the down-regulated locus (left panel), the "distal regulatory region" is 56%, the intron is 38%, the exon is 5% and the promoter is 2%. For the enrichment site (right cake), the "remote regulatory protein" accounted for 53%, the intron accounted for 36%, the exon accounted for 8% and the promoter accounted for 3%. FIG. 38F depicts a Venn diagram showing the overlap between depleted FOXA1RE and ERE regions and enriched ARE, GRE and FOXA1 RE. FIG. 38G depicts SRC-1 interacting 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 blotted with SRC-1. AR-androgen receptor; ER-estrogen receptor; chIP-chromatin immunoprecipitation; ARE-androgen response elements; ERE-estrogen responsive element; GRE-glucocorticoid response element; SRC-1-steroid receptor coactivator-1, FOXARRE, a response element of the fork A1. FIG. 38H depicts an up-regulated motif (ER).
FIG. 39 depicts co-localization of AR and ER- α in luminal B breast cancer samples.
FIG. 40 depicts representative ChIP Seq peaks in regulatory regions of genes. Peaks were color coded with top plot up (red) and plot 2 down (blue) from top plot, plot 3 from top plot (CERS 3) down with the exception of the last row (IX (AR)), and bottom plot (miR 4471) down with the exception of the last row (IX (AR)).
FIGS. 41A-41E depict phosphorylated proteomic analysis of HCI-13 PDX. Fig. 41A-41C depict lysates (as shown in fig. 36B-36C) from HCI-13 tumor samples (n=4) of PDX printed onto nitrocellulose-coated sections. The array was probed with a total of 174 antibodies targeting a broad range of protein kinases and activating them by phosphorylation. The array was stained with anti-rabbit or anti-mouse biotinylated secondary antibodies. The signal was amplified and streptavidin conjugated IRDye680 was used as a second signal detector. Images were acquired and quantified. Fig. 41D-41E depict that activation of PKC overcomes the inhibition of formula IX. HCI-13 tissue fragments were incubated on gelatin sponge and treated with 100nM phorbol 12-myristate 13-acetate (PMA) or 100ng/mL EGF for 30 min, followed by 1. Mu.M formula IX. EGF is treated twice daily due to its short stability. Tissues were harvested 3 days after treatment, RNA was isolated, and expression of various genes was measured by real-time PCR. * P <0.05 for vehicle treated samples; # p <0.05 from formula IX treated samples. n=3 per group (each sample obtained from 5 separate fragments. PMA-phorbol 12-myristate 13-acetate; EGF-epidermal growth factor; PDX-patient-derived xenograft; HCI-hensmal cancer institute).
Fig. 42 shows a model depicting modulation of ER function by AR agonists.
FIGS. 43A and 43B present a baseline [ 18 F]-relation of 16 beta-fluoro-5 alpha-dihydrotestosterone (FDHT) SUVmax (FDHT uptake) to AR. Fig. 43A depicts that AR positive (n=7) status tumors determined by biopsy had higher baseline FDHT uptake (baseline FDHT-SUVmax) compared to AR negative (n=2) status tumors. ND-AR state is undetermined; UNK-is an unknown AR state. Fig. 43B depicts the correlation between baseline SUVmax and AR levels determined by biopsy. The correlation was 0.41 (p-value=0.27). Excluding one outlier (as shown, ar= 64,1946, baseline suvmax=1), a higher trend in baseline FDHT SUVmax was observed, and the quantitative AR expression level was higher (r=0.71, p=0.046).
Fig. 44A and 44B present correlations of baseline FDHT uptake (baseline FDHT-SUVmax) with optimal relief (clinical benefit (CB) or no CB). Fig. 44A depicts a baseline FDHT-SUVmax median of 2.93 (range 1-4.38) for 7 patients with CB (defined as Complete Remission (CR), partial Remission (PR), or disease Stabilization (SD) according to RECIST criteria) at 12 weeks post-treatment, and a baseline FDHT-SUV max median of 2.15 (0.96-3.77) for 4 patients with Progressive Disease (PD). Fig. 44B depicts that patients with CB at 12 weeks had decreased FDHT uptake change from baseline to six weeks (SUVmax change from baseline to six weeks) while patients with PD did not. Disc (AE) -interrupted due to an adverse event; disc-interrupt.
FIGS. 45A and 45B present that formula IX (SARM) re-sensitizes a human breast cancer model to CDK 4/6 inhibition in a CDK 4/6 resistant model. Fig. 45A depicts growth curves for PDX GAR15-13 treated with vehicle (n=8), formula IX labeled SARM (n=8), palbo (n=7), or combination (Combo) sarm+palbo (n=11). Asterisks indicate significantly different tumor volumes at ethical endpoints as determined by two-tailed unpaired student t-testSpecific for Palbo versus Combo (t=3.7246, d.f. =14, p=0.0022), and SARM versus Combo (t=4.094, d.f. =15, p=0.0010). Data expressed as mean ± s.e.m. fig. 45B depicts Palb R Proliferation of cells in response to AR agonists (100 nM of formula IX) and Palb (125 nM), alone or in combination. Data represent mean ± s.e.m. of four replicate cell culture wells per condition. Data were analyzed using one-way ANOVA (f=79.71, d.f. =23, p<0.0001 A Tukey multiple comparison test is then performed. The P-value is indicated by grey asterisks, where P for all highlighted comparisons<0.0001。
Detailed Description
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject. Accordingly, the present invention provides the following method: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treatment of a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (pamtepanib (Ibrance), rapmactinib (Kisqali), abbe-cili (Vorzenio), trasturil, ly Luo Xili), abarism (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3kα), mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olapatinib (lynparazapal) or talzenia)), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nertinib (nertinib), dactylb (virlof) or fig. 2 (virlozenb) Cartinib (tukusa)), HER2 antibody (trastuzumab (Herceptin), pertuzumab (Perjeta), mactuximab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (Enhertu), enmtuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), atentizumab (tecentiq) (PD-L1 blocking antibody), pemab (Keytruda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating, preventing, suppressing or inhibiting AR positive triple negative breast cancer; r) treating a subject having HER2 positive breast cancer; s) treating a subject having a breast cancer that expresses an ER mutant, t) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or u) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer, and treats the breast cancer in the subject by administering to the subject a therapeutically effective amount of a compound of formula I-XIV of the invention as described herein and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof. In one embodiment, the subject is male. In one embodiment, the subject is female.
In one embodiment of the invention, there is provided a method for treating a subject having breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having metastatic breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the subject's metastatic breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having refractory breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the refractory breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the AR positive breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In one embodiment, the AR positive breast cancer is ER, PR and HER2 positive. In another embodiment, the AR positive breast cancer is ER, PR and HER2 negative. In one embodiment, the AR positive breast cancer is ER positive and is PR and HER2 negative. In another embodiment, the AR positive breast cancer is ER and PR positive and HER2 negative. In another embodiment, the AR positive breast cancer is ER and HER2 positive and is PR negative. In yet another embodiment, the AR positive breast cancer is ER negative and PR and HER2 positive. In another embodiment, the AR positive breast cancer is ER and PR negative and HER2 positive. In yet another 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.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive refractory breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the AR positive refractory breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive metastatic breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an analogue, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, in an amount effective to treat the AR positive metastatic breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive and ER positive breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the AR positive metastatic breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having ER positive breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the ER positive breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In one embodiment, ER positive breast cancer is AR positive. In another embodiment, ER positive breast cancer is AR negative. In one embodiment, the ER positive breast cancer is a triple positive (ER, PR, HER 2) breast cancer. In another embodiment, the ER positive breast cancer is not a triple positive breast cancer.
In another embodiment of the present invention, there is provided a method for treating a subject having triple negative breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the present invention and/or an 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 the triple negative breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive triple negative breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the AR positive triple negative breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for treating a subject having advanced breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the advanced breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, a method is provided for treating a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonist (goserelin), aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), rebaciclovir (Kisqali), abbe' zenio), trazoxiline, ly Luo Xili), axist (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3kα), mTOR inhibitor (ivermectin), polyribopolymerase (PARP) inhibitor (lypatadine (lyazane) or tazoril (tazernia)), human epidermal growth factor 2 (HER), tuyezotinib (tuyezob), anti-bead (Herceptin), anti-tuyered (panaxy) antibody (herception 2), anti-tuyered (panaxy 2) or anti-tuyered (panaxy 2) antibody (panaxy 2) An enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), altlizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) treatment failure, the method comprising the step of administering a compound of formula I-XIV of the invention and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof to a subject in an amount effective to treat breast cancer in the subject that failed with: selective Estrogen Receptor Modulators (SERMs) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonists (goserelin), aromatase Inhibitors (AI) (letrozole, anastrozole, exemestane), fluvalirome, cyclin dependent kinase 4/6 (CDK 4/6) inhibitors (cabonine (Ibrance), rapaminib (Kisqali), abbe-zenio), trazoxiline, ly Luo Xili), axiliziram (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olpataline (Lynparza) or talazopanil (Talzenna)), human growth factor receptor 2 (CDK 4/6) inhibitors (lapatinib, letinib (nerlozenb), dasatib (vic), vicat or vicat), anti-bead anti-tuzumab (Herceptin) or anti-bead anti-tuzumab (Herceptin) anti-HER 2 (Herceptin), anti-bead anti-tuzumab (tuzuelan) (panaxb) or anti-tuzuelan) anti-tuzuelan (HER 2 (panaxb) or anti-tuzuelan) anti-tuzuelan (panaxb) Pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) treatment. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment, the invention provides a method for treating a subject having HER2 positive breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the HER2 positive breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In one embodiment, the HER2 positive breast cancer is HER2 positive refractory breast cancer. In another embodiment, 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, HER2 positive breast cancer is AR positive. In another embodiment, HER2 positive breast cancer is AR negative.
In a certain embodiment, HER2 positive breast cancer is ER positive, PR positive and AR positive. In another embodiment, HER2 positive breast cancer is ER positive, PR negative, and AR positive. In another embodiment, HER2 positive breast cancer is ER positive, PR negative, and AR negative. In other embodiments, HER2 positive breast cancer is ER positive, PR positive, and AR negative. In another embodiment, HER2 positive breast cancer is ER negative, PR negative, and AR positive. In another embodiment, HER2 positive breast cancer is ER negative, PR positive, and AR positive. In another embodiment, HER2 positive breast cancer is ER negative, PR positive, and AR negative. In a certain embodiment, HER2 positive breast cancer is ER negative, PR negative and AR negative. In a certain embodiment, the HER2 positive breast cancer is a triple positive HER2 breast cancer.
In another embodiment, the present invention provides a method for treating a subject having breast cancer that expresses an ER mutant, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the subject's breast cancer that expresses an ER mutant. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a Y537S mutation.
In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the D351Y mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing an E380Q mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the V422del mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the S432L mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a G442A mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the S463P mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L469V mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536R mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536H mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536P mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536Q mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a Y537N mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a Y537C mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the Y537D mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a D538G mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the E542G mutation. In one embodiment, breast cancer expressing an ER mutant refers to a mutant of ER- α.
In a certain embodiment, the breast Cancer expressing the ER mutant is such as Cancer Cell,2018, volume 33, pages 173-186, or Nat Rev Cancer,2018, volume 18, phase 6: pages 377-388, which are incorporated herein by reference. In one embodiment, breast cancer expressing an ER mutant refers to a mutant of ER- α.
In one aspect, the invention provides a pharmaceutical composition comprising a Selective Androgen Receptor Modulator (SARM) compound and a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor, wherein the SARM compound is represented by the structure of formula I:
wherein the method comprises the steps of
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 (CF) 2 CF 3
R2 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, CN, NO 2 、COR、COOH、CONHR、CF 3 、Sn(R) 3 Or R is 3 Together with the benzene ring to which it is attached, form a fused ring system represented by the structure:
z is NO 2 CN, COR, COOH or CONHR;
y is CF 3 F, br, cl, I, CN 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 A, B or C:
n is an integer from 1 to 4; and is also provided with
m is an integer of 1 to 3, or
Optical isomers, racemic mixtures, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides or crystals thereof.
In some embodiments, the SARM compound in the compositions of the present invention is represented by the structure of formula II:
wherein the method comprises the steps of
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 A, B or C:
r is C 1 -C 4 Alkyl, aryl, phenyl, alkenyl, hydroxy, C 1 -C 4 Haloalkyl, halogen or haloalkenyl; and is also provided with
R 1 Is CH 3 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
In some embodiments, the SARM compounds of the compositions of the present invention are represented by the structure of formula VIII, IX, X, XI, XII, XIII or XIV:
/>
in another aspect, the invention provides a pharmaceutical composition comprising formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and a CDK 4/6 inhibitor,
In some embodiments of the compositions of the invention, the CDK 4/6 inhibitor is palbociclib, rebabociclib, trazocili, la Luo Xili, or abbe cili. In certain embodiments, CDK 4/6 is palbociclib. In some embodiments, the CDK 4/6 inhibitor is rebaudinib. In some embodiments, the CDK 4/6 inhibitor is trazoyside. In some embodiments, the CDK 4/6 inhibitor is from Luo Xili. In some embodiments, the CDK 4/6 inhibitor is arbeli.
In another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and palbociclib,
in another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and Rabociclib,
in another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and treamxili,
In another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and L Luo Xili,
in another aspect, the invention provides a pharmaceutical composition comprising compound IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and Abeli,
in some embodiments, the compositions of the present invention are in the form of pills, tablets, capsules, solutions, suspensions, emulsions, elixirs, gels, creams, suppositories, or parenteral formulations.
In another aspect of the invention, the pharmaceutical composition of the invention is useful for treating breast cancer.
In one aspect, the invention provides a method for treating a subject having breast cancer, the method comprising administering to the subject a pharmaceutical composition of the invention as described herein.
In some embodiments of the methods of the invention, the pharmaceutical composition comprises a Selective Androgen Receptor Modulator (SARM) compound and a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor, wherein the SARM compound is represented by the structure of formula I:
Wherein the method comprises the steps of
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 (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, CN, NO 2 、COR、COOH、CONHR、CF 3 、Sn(R) 3 Or R is 3 Together with the benzene ring to which it is attached, form a fused ring system represented by the structure:
z is NO 2 CN, COR, COOH or CONHR;
y is CF 3 F, br, cl, I, CN 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 A, B or C:
n is an integer from 1 to 4; and is also provided with
m is an integer of 1 to 3, or
Optical isomers, racemic mixtures, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides or crystals thereof.
In some embodiments of the methods of the invention, the SARM compound is represented by the structure of formula II:
wherein the method comprises the steps of
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 A, B or C:
r is C 1 -C 4 Alkyl, aryl, phenyl, alkenyl, hydroxy, C 1 -C 4 Haloalkyl, halogen or haloalkenyl; and is also provided with
R 1 Is CH 3 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
In some embodiments of the methods of the invention, the SARM compound is represented by the structure of formula VIII, IX, X, XI, XII, XIII or XIV:
/>
in some embodiments of the methods of the invention, the pharmaceutical composition comprises formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof, and a CDK 4/6 inhibitor,
in some embodiments of the methods of the invention, the CDK 4/6 inhibitor is palbociclib, rebabociclib, trazoxili, len Luo Xili, or abbe cilide. In certain embodiments, CDK 4/6 is palbociclib. In some embodiments, the CDK 4/6 inhibitor is rebaudinib. In some embodiments, the CDK 4/6 inhibitor is trazoyside. In some embodiments, the CDK 4/6 inhibitor is from Luo Xili. In some embodiments, the CDK 4/6 inhibitor is arbeli.
In some embodiments of the methods of the invention, the pharmaceutical composition comprises formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof and palbociclib,
In some embodiments of the methods of the invention, the pharmaceutical composition comprises formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof and Rabociclib,
in some embodiments of the methods of the invention, the pharmaceutical composition comprises formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal and fraxil thereof,
in some embodiments of the methods of the invention, the pharmaceutical composition comprises formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal sum thereof Luo Xili,
in some embodiments of the methods of the invention, the pharmaceutical composition comprises formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof and Abeli,
in some embodiments of the methods of the invention, the breast cancer is 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 an inhibitor employing a 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 (cabonine), rapazetinib (kiquali), abbe zenio, trazoprib, ly Luo Xili), alist (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3kα), mTOR inhibitor (ADP), polysaccharase (PARP) inhibitor (oxpanni (lyzantinib) or jenzolamide), anti-bead 2 (HER 2), anti-bead 2 (panzotinib), anti-bead 2 (panoramide), anti-bead 2 (HER 2) or anti-bead 2 (HER 2) antibody (HER 2) Enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), actlizumab (Tecentriq) (PD-L1 blocking antibody), pembrolizumab (Keystuda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) treat failed breast cancer.
In some embodiments, the breast cancer is AR positive metastatic breast cancer. In certain embodiments, the breast cancer is AR positive refractory breast cancer.
In some embodiments, the ER-positive breast cancer is AR-positive and ER-positive breast cancer, or AR-negative and ER-positive breast cancer.
In some embodiments, 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 positive and HER2 negative; ER positive, PR negative and HER2 positive; or ER positive, PR positive and HER2 positive.
In some embodiments, breast cancer fails treatment with a Selective Estrogen Receptor Modulator (SERM). In some embodiments, the SERM is tamoxifen, toremifene, or raloxifene.
In some embodiments, breast cancer fails to be treated with cyclin dependent kinase 4/6 (CDK 4/6) inhibitors. In some embodiments, the subject is resistant to or non-responsive to a CDK 4/6 inhibitor. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance), rebabociclib (Kisqali), traracili, len Luo Xili, or abbe cili (Vorzenio). In certain embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance). In some embodiments, the CDK 4/6 inhibitor is rebaudinib. In some embodiments, the CDK 4/6 inhibitor is trazoyside. In some embodiments, the CDK 4/6 inhibitor is from Luo Xili. In some embodiments, the CDK 4/6 inhibitor is arbeli.
In some embodiments of the methods of the invention, the compositions of the invention as described herein insensitize the breast cancer to treatment with a CDK 4/6 inhibitor. In some embodiments, the CDK 4/6 inhibitor is at least one of palbociclib (Ibrance), rebabociclib (Kisqali), traracili, ly Luo Xili, and abbe zenio. In certain embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance). In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali). In some embodiments, the CDK 4/6 inhibitor is trazoyside. In some embodiments, the CDK 4/6 inhibitor is from Luo Xili. In some embodiments, the CDK 4/6 inhibitor is arbeli.
In some embodiments of the methods of the invention, the compositions of the invention as described herein overcome estrogen endocrine resistance. In some embodiments, the estrogenic endocrine therapy comprises at least one of tamoxifen, toremifene, raloxifene, exemestane, letrozole, anastrozole, and fulvestrant. In some embodiments, the estrogenic endocrine therapy comprises tamoxifen. In some embodiments, the estrogenic endocrine therapy comprises toremifene. In some embodiments, the estrogenic endocrine therapy comprises raloxifene. In some embodiments, the estrogenic endocrine therapy comprises exemestane. In some embodiments, the estrogenic endocrine therapy comprises letrozole. In some embodiments, the estrogenic endocrine therapy comprises anastrozole. In some embodiments, the estrogenic endocrine therapy comprises fulvestrant.
Another aspect of the invention is that the compositions of the invention as described herein overcome resistance to a combination therapy of estrogenic endocrine and CDK 4/6 inhibitors. In some embodiments, the CDK 4/6 inhibitor is at least one of palbociclib (Ibrance), rebabociclib (Kisqali), traracili, ly Luo Xili, and abbe zenio. In certain embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance). In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali). In some embodiments, the CDK 4/6 inhibitor is trazoyside. In some embodiments, the CDK 4/6 inhibitor is from Luo Xili. In some embodiments, the CDK 4/6 inhibitor is arbeli. In some embodiments, the estrogenic endocrine therapy comprises at least one of tamoxifen, toremifene, raloxifene, exemestane, letrozole, anastrozole, and fulvestrant. In some embodiments, the estrogenic endocrine therapy comprises tamoxifen. In some embodiments, the estrogenic endocrine therapy comprises toremifene. In some embodiments, the estrogenic endocrine therapy comprises raloxifene. In some embodiments, the estrogenic endocrine therapy comprises exemestane. In some embodiments, the estrogenic endocrine therapy comprises letrozole. In some embodiments, the estrogenic endocrine therapy comprises anastrozole. In some embodiments, the estrogenic endocrine therapy comprises fulvestrant.
In some embodiments, when the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is palbociclib (Ibrance) and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is trazoxili and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is trazoysin and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is trazoysin and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is trazoyside and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is trazoysib and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is trazoyside and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is trazoyside and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is trazoxili and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is trazoysin and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is abbe-zenio (vorzeio) and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is abbe-cili (Vorzenio) and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is abbe-cili (Vorzenio) and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is abbe-cili (Vorzenio) and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is arbitide (Vorzenio) and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is abbe-cili (Vorzenio) and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is abbe-cili (Vorzenio) and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is rebaudinib (Kisqali) and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is trazoxili and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is trazoysin and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is trazoysin and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is trazoyside and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is trazoysib and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is trazoyside and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is trazoyside and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is Luo Xili and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments, where the compositions of the invention as described herein overcome resistance to a combination therapy of an estrogen endocrine and a CDK 4/6 inhibitor, the CDK 4/6 inhibitor is arbeli and the estrogen endocrine therapy comprises tamoxifen. In some embodiments, the CDK 4/6 inhibitor is arbelii and the estrogen endocrine therapy comprises toremifene. In some embodiments, the CDK 4/6 inhibitor is arbelii and the estrogen endocrine therapy comprises raloxifene. In some embodiments, the CDK 4/6 inhibitor is arbeli and the estrogen endocrine therapy comprises exemestane. In some embodiments, the CDK 4/6 inhibitor is arbeli and the estrogen endocrine therapy comprises letrozole. In some embodiments, the CDK 4/6 inhibitor is arbitraconazole and the estrogen endocrine therapy comprises anastrozole. In some embodiments, the CDK 4/6 inhibitor is arbeli and the estrogen endocrine therapy comprises fulvestrant.
In some embodiments of the methods of the invention, breast cancer fails to be treated with an mTOR inhibitor. In some embodiments, the mTOR inhibitor is everolimus, sirolimus, temsirolimus, or sirolimus.
In some embodiments, the methods of the invention further extend the survival of a subject having breast cancer or extend the progression free survival of a subject having breast cancer.
In some embodiments, the compositions of the invention are administered intravenously, intra-arterially, intramuscularly, subcutaneously, orally, or topically. In some embodiments, the compositions of the present invention are administered orally.
In some embodiments, the dosage of the selective androgen receptor modulator is from 1mg to 50mg per day. In some embodiments, the selective androgen receptor modulator dose is from about 1mg to about 5mg, or from about 5mg to about 50mg, or from about 5mg to about 10mg, or from about 5mg to about 15mg, or from about 5mg to about 20mg, or from about 5mg to about 30mg, or from about 10mg to about 50mg, or from about 10mg to about 40mg, or from about 10mg to about 30mg, or from about 10mg to about 20mg, or from about 15mg to about 50mg, or from about 20mg to about 50mg, or from about 25mg to about 50mg, or from about 30mg to about 40mg, per day. In some embodiments, the dosage of the selective androgen receptor modulator is about 9mg per day or 18mg per day.
As used herein, in one embodiment, the term "treatment" may refer to treating, delaying progression of, preventing relapse, or treating relapse. In one embodiment, the term "treating" refers to reducing the morbidity, mortality, or a combination thereof associated with breast cancer.
The term "preventing" may refer to preventing the initial occurrence of a disease, reducing risk factors, minimizing disability or potential health risks of the disease.
As used herein, the term "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 ER, PR and/or HER2 expression; triple positive breast cancer (ER, PR and HER2 positive), AR positive breast cancer with or without ER expression; ER positive breast cancer with or without AR expression; AR-positive and ER-positive breast cancers; refractory breast cancer; AR positive refractory breast cancer; ER positive refractory breast cancer; AR positive metastatic breast cancer; ER positive metastatic breast cancer; using a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotropin releasing hormone (GnRH) agonist (goserelin), aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (Pabosib (Ibrance), raboscalin (Kisqali), abeli (Vorzenio), trazoysin, lei Luo Xili), mTOR inhibitor (everolimus), trastuzumab (Herbeeptin, enmetrastuzumab), pertuzumab (Perjeta), apilimbus (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), lapatinib, lenatinib (Neynx), olaparza (ADP) (polynuclear) Inhibitors of carbohydrate polymerase (PARP), bevacizumab (Avastin) and/or fulvestrant treatment of failed breast cancer; triple negative breast cancer; or ingest 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) breast cancer; or any combination thereof.
In one embodiment, the term "breast cancer" refers to a disease characterized by abnormal rapid proliferation of abnormal cells in one or both breasts of a subject. Abnormal cells are commonly referred to as "tumor cells," which in some embodiments refer to transformed cells capable of forming solid tumors. In some embodiments, the term "tumor" refers to an abnormal cell mass or population (i.e., two or more cells) caused by excessive or abnormal cell division (whether malignant or benign) as well as pre-cancerous and cancerous cells. Malignant tumors differ from benign growths or tumors in that, in addition to uncontrolled cell proliferation, they can invade surrounding tissues and metastasize.
In breast cancer, tumor cells may be identified in only one or both breasts and not in the other tissue or organ, in one or both breasts and one or more adjacent tissues or organs (e.g., lymph nodes), or in one or more non-adjacent tissues or organs to which breast and breast cancer cells have metastasized.
In some embodiments, the term "metastasis" refers to a process in which cancer cells move from one organ or tissue to another non-adjacent organ or tissue. Cancer cells in the breast can spread to tissues and organs of the subject, and conversely, cancer cells from other organs or tissues can invade or metastasize to the breast. Cancer cells from the breast can 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. In some embodiments, the term "invasion" refers to the spread of cancer cells to adjacent surrounding tissues.
As used herein, the term "advanced breast cancer" refers to cancer that has spread to other parts of the body and that is generally not cured or controlled with current therapies.
As used herein, the term "AR positive breast cancer" may refer to breast cancer in which at least a portion of the cancer cells express at least the Androgen Receptor (AR).
As used herein, the term "ER positive breast cancer" may refer to breast cancer in which at least a portion of the cancer cells express at least an Estrogen Receptor (ER).
As used herein, the term "triple negative breast cancer" may refer to breast cancer cells that do not have Estrogen Receptor (ER), progesterone Receptor (PR), or substantial HER2/neu protein. "triple negative breast cancer" may also be referred to herein as "ER negative PR negative HER2/neu negative breast cancer".
As used herein, the term "triple positive breast cancer" may refer to breast cancer cells that express Estrogen Receptor (ER), progesterone Receptor (PR), and a number of HER2/neu (HER 2) proteins. "triple positive breast cancer" may also be referred to herein as "ER positive PR positive HER2/neu positive breast cancer" or "ER, PR and HER2 breast cancer.
As used herein, the term "refractory" may refer to breast cancer that is unresponsive to treatment. Breast cancer may be resistant at the beginning of treatment or it may become resistant during treatment. "refractory breast cancer" may also be referred to herein as "resistant cancer".
As used herein, the term "HER2 positive breast cancer" may refer to breast cancer in which at least a portion of the cancer cells express high levels of HER2 protein (HER 2 (from human epidermal growth factor receptor 2) or HER 2/neu) that promote rapid cell growth.
As used herein, the term "breast cancer expressing an ER mutant" may refer to breast cancer that expresses a mutated estrogen receptor α (ER- α) that confers resistance to treatment. Typically these mutations are located within the ligand binding domain of ER- α, are therapeutic in appearance, and/or confer resistance to some or all endocrine therapies (such as SERM, AI, SERD and/or GnRH agonists). As used herein, the term "breast cancer expressing a Y537S ER mutant" may refer to breast cancer that expresses estrogen receptor α (ER- α) with a point mutation Y537S.
In another embodiment of the invention, there is provided a method for treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for prolonging the survival of a subject suffering from breast cancer, comprising the step of administering to the subject a compound of formula I-XIV of the present invention and/or an 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 the subject suffering from breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for slowing the progression of breast cancer in a subject, comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for prolonging the progression free survival of a subject suffering from breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 progression free survival of the subject suffering from breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In one embodiment, the breast cancer of the invention: in one embodiment, ER positive metastatic breast cancer; in another embodiment, ER positive refractory breast cancer; in another embodiment, ER positive PR positive HER2 negative breast cancer; in another embodiment AR positive ER positive breast cancer; in another embodiment AR positive ER positive refractory breast cancer; in another embodiment AR positive ER positive metastatic breast cancer; in another embodiment, a triple positive breast cancer; in another embodiment refers to advanced ER-positive breast cancer; in another embodiment AR positive; in another embodiment, ER positive breast cancer; and in another embodiment refers to the use of a 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), raplocillin (kiquali), abbe-li (Vorzenio), trasturil, lya Luo Xili), an inhibitor of abaricline (Piqray) (phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha), an mTOR inhibitor (everolimus), a Poly ADP Ribose Polymerase (PARP) inhibitor (olazapanib (lynpzarzali) or talzenia), a human epidermal growth factor receptor 2 (2) kinase inhibitor (lapatinib, nepalenib (nepalen), a (fluzab), a-ziram (ziram), a (guanoxydol) or an anti-beads (HER) anti-tuzumab) or anti-tuzumab (HER) antibody (HER 2), or anti-tuzumab (tuzumab) antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) treat failed breast cancers.
In another embodiment of the present invention, there is provided a method for reducing biomarker levels in a subject having breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the present invention and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof in an amount effective to reduce biomarker levels in the subject. In another embodiment, the method comprises administering a compound of formula I-XIV of the invention. As used herein, the term "biomarker" may refer to a substance that serves as an indicator of a process, event, or condition. The biomarker may be a biological molecule such as a nucleic acid molecule (e.g., microrna, genomic DNA, etc.), protein, polysaccharide, etc. Biomarkers include tumor antigens and tumor markers. In one embodiment, the biomarker is indicative of the presence of cancer (e.g., breast cancer). In one embodiment, the biomarker may be used to determine the efficacy of a treatment. In one embodiment, the biomarker can be used to determine the progression of a disorder (e.g., breast cancer).
MUC-1 associated antigen or CA27.29 is a cancer antigen that is highly associated with breast cancer. As used herein, the term "CA27.29 biomarker" refers to a biomarker of breast cancer. In one embodiment, CA27.29 is a biomarker for advanced breast cancer.
"PSA (prostate specific antigen) biomarkers" are used as biomarkers for prostate cancer, however, higher levels of PSA are also found in the blood of women with breast cancer compared to women without breast cancer. PSA can also be used as a biomarker for breast cancer.
"CTX biomarker" and "NTX biomarker" are C-terminal peptide and N-terminal peptide of type I collagen, respectively, for use as biomarkers of bone turnover. NTX and CTX biomarkers can be sensitive indicators of the presence of bone metastasis in breast cancer patients.
In one embodiment, the method of the invention reduces a CA27.29 biomarker in a subject. In one embodiment, the methods of the invention reduce PSA in a subject. In one embodiment, the methods of the invention reduce CTX biomarkers in a subject. In one embodiment of the invention, the methods of the invention reduce an NTX biomarker in a subject. In another embodiment, the method of the invention maintains CA27.29 levels in a subject. In another embodiment, the methods of the invention maintain PSA levels in a subject. In another embodiment, the methods of the invention maintain the level of CTX biomarker in the subject. In another embodiment, the methods of the invention maintain the level of an NTX biomarker. In one embodiment, the subject has breast cancer. In one embodiment, the subject has advanced breast cancer. In another embodiment, the subject has refractory breast cancer. In another embodiment, the subject has AR positive breast cancer. In yet another embodiment, the subject has ER positive breast cancer.
In one embodiment, the invention relates to a method of treating breast cancer in a subject, the method comprising the steps of: determination of 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer, followed by administration of a Selective Androgen Receptor Modulator (SARM) compound to the AR positive breast cancer subject.
In another embodiment, the selective androgen receptor modulator compound is a compound of formula I-XIV.
In one embodiment, the tumor is a metastatic breast cancer tumor. In one embodiment, the tumor is an ER positive metastatic breast cancer tumor. In one embodiment, the tumor is an ER positive metastatic breast cancer tumor that failed treatment with FDA approved hormones and/or kinases.
In one embodiment, the AR positive breast cancer is ER positive. In another embodiment, the AR positive breast cancer is metastatic. In another embodiment, the breast cancer is any one of the following: refractory breast cancer; AR positive breast cancer; AR positive refractory breast cancer; AR positive metastatic breast cancer; AR-positive and ER-positive breast cancers; AR positive breast cancer with or without expression of Estrogen Receptor (ER), progesterone Receptor (PR) and/or human epidermal growth factor receptor 2 (HER 2); triple Negative Breast Cancer (TNBC); advanced breast cancer; use of Selective Estrogen Receptor Modulators (SERM) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonists (goserelin), aromatase Inhibitors (AI) (letrozole, anastrozole, exemestane), fluvalirome, cyclin dependent kinase 4/6 (CDK 4/6) inhibitors (Pabosib (Ibrance), raboscalid (Kisqali), abeli (Vorzonio), trazoxib, latif Luo Xili), arpelisis (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (Olaparib (Lynparaza) or Talazopanib (Talzenna)), human epidermal growth factor receptor 2 (CDK) inhibitors (Lapatinib, latinib (Nerlx), tavantinib (Vizib) or Tuzijia), anti-beads (HER) (anti-tuzumab) anti-bead anti-tuzumab (HER) or anti-tuzumab (HER) beads (HERBUGmbH) or anti-tuyetin (HER) anti-tuyered (HER) antibody (HER) anti-tuyered (HER) beads (HER) anti-tuyered (HER) antibody (HER) anti-tuyered (HER) anti-GYtuyered (HER) or (HERX) to HEYTAR) to HETAMER (YMER-MER), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed to treat breast cancer; ER positive breast cancer; HER2 positive breast cancer; breast cancer expressing ER mutant, or breast cancer expressing Y537S ER mutant.
In another embodiment, the breast cancer is estrogen receptor positive (er+) metastatic breast cancer.
In one embodiment, the compounds of the invention are antagonists. In another embodiment, the compounds of the invention are agonists. In another embodiment, the compounds of the invention are partial agonists/partial antagonists. In one embodiment, the compounds of the invention are AR agonists. In another embodiment, the compound is an AR antagonist. In another embodiment, the compounds are partial AR agonists and AR antagonists. In one embodiment, the compounds of the invention are PR agonists. In another embodiment, the compound is a PR antagonist. In another embodiment, the compounds are partial PR agonists and PR antagonists.
In one embodiment, the compounds of the invention are AR agonists and PR antagonists.
In some embodiments, the SARM compounds of the present invention are useful for: a) Treating, preventing, delaying the onset of, increasing the time to first appearance of, suppressing or inhibiting the occurrence of, or reducing the risk of a first bone-related event (SRE) in a subject, such as a pathological fracture, bone surgery, bone radiation, spinal cord compression, new bone metastasis and/or bone loss; b) Treating, preventing, suppressing or inhibiting or reducing the risk of developing a variety of hormone-related conditions in a subject, e.g., for increasing the force ratio; and/or c) improving the quality of life of the subject.
Osteoporosis is a systemic skeletal disease characterized by low bone mass and degeneration of bone tissue, resulting in increased bone fragility and susceptibility to fracture. In the united states, the condition affects more than 2500 thousands of people, resulting in more than 130 thousands of fractures each year, including 50 thousands of spinal fractures, 25 tens of thousands of hip fractures, and 24 tens of thousands of wrist fractures each year. Hip fractures are the most serious consequence of osteoporosis, with 5% -20% of patients dying within one year, and more than 50% of survivors losing work. The risk of osteoporosis for the elderly is greatest, and thus it is expected that this problem will increase significantly as the population ages. The global fracture incidence is expected to increase by three times in the next 60 years, and one study estimated that there will be 450 tens of thousands of hip fractures worldwide in 2050.
Women are more at risk of osteoporosis than men. Women experience dramatically accelerated bone loss within five years after menopause. Other risk-increasing factors include smoking, alcoholism, sedentary lifestyle and low calcium intake. However, osteoporosis also frequently occurs in males. It is well known that bone mineral density in males decreases with age. Reduced amounts of bone mineral content and density correlate with reduced bone strength and fracture is prone to occur. The molecular mechanisms of sex hormones under pleiotropic effects in non-reproductive tissues have just begun to be understood, but it is clear that physiological concentrations of androgens and estrogens play an important role in maintaining bone homeostasis throughout the life cycle. Thus, when androgen or estrogen deprivation occurs, the rate of bone remodeling increases, tilting the balance of resorption and formation toward favorable resorption, resulting in an overall loss of bone mass. In males, the natural decline in sex hormones at maturity (direct decline in androgens and decline in estrogen levels resulting from peripheral aromatization of androgens) is associated with bone frailty. This effect is also observed in castrated males.
In one embodiment, the invention provides the use of a compound as described herein or a prodrug, analog, isomer, metabolite, derivative, pharmaceutically acceptable salt, pharmaceutical product, polymorph, crystal, impurity, N-oxide, hydrate or any combination thereof, in the following: a) Treating bone related diseases; b) Preventing bone related diseases; c) Suppressing bone related diseases; d) Inhibiting bone related diseases; e) Increasing the strength of bone in the subject; f) Increasing bone mass in the subject; g) For inhibiting osteoclast production; and/or h) for stimulating osteoblast production.
In one embodiment, the invention provides the use of a compound as described herein or a prodrug, analog, isomer, metabolite, derivative, pharmaceutically acceptable salt, pharmaceutical product, polymorph, crystal, impurity, N-oxide, hydrate or any combination thereof, in the following: a) Accelerating bone repair; b) Treating bone diseases; c) Treating loss of bone density; d) Treating low Bone Mineral Density (BMD); e) Treating reduced bone mass; f) Treating metabolic bone diseases; g) Promote bone growth or regeneration; h) Promoting bone repair; i) Promoting fracture repair; j) Promoting bone remodeling; k) Treating bone injuries after reconstructive surgery, including bone injuries of the face, hip, or joint; l) enhancing bone strength and function; m) increasing cortical bone mass; n) increasing trabecular connectivity; o) preventing, inhibiting or delaying metastasis to bone; and/or p) preventing, inhibiting or delaying the growth of metastatic tumors of the bone.
In one embodiment, the bone-related disease is a genetic disease, or in another embodiment, is induced as a result of a treatment regimen for a given disease. For example, in one embodiment, the compounds as described herein may be used to treat a bone-related disease caused by cancer metastasis to bone, or in another embodiment, a bone-related disease caused by androgen deprivation therapy administered, for example, in response to prostate carcinogenesis in a subject.
As used herein, "estrogen deprivation therapy" may refer to a therapy administered in response to breast cancer in a subject. Known treatments include treatment with GnRH agonists, SERMs, SERDs or Aromatase Inhibitors (AI). For example, in one embodiment, the compounds as described herein may be used to treat a bone-related disease caused by cancer metastasis to bone, or in another embodiment, a bone-related disease caused by estrogen deprivation therapy, e.g., administered in response to breast cancer in a subject. Menopause may also be induced using GnRH agonists such as goserelin (Zoladex), which maintains endogenous estrogen at low levels by inhibiting the hypothalamic-pituitary-gonadal axis.
In one embodiment, the bone-related disorder is loss of Bone Mineral Density (BMD). In another embodiment, the bone-related disorder is osteoporosis. In another embodiment, the bone-related disorder is osteopenia. In another embodiment, the bone-related disorder is increased bone resorption. In another embodiment, the bone-related disorder is a bone fracture. In another embodiment, the bone-related disorder is bone frailty. In another embodiment, the bone-related disorder is any combination of osteoporosis, osteopenia, increased bone resorption, bone fracture, bone frailty, and loss of BMD. Each disease represents a separate embodiment of the present invention.
In one embodiment, "osteoporosis" refers to the thinning of bone and reduction of bone mass due to depletion of calcium and bone proteins. In another embodiment, osteoporosis is a systemic skeletal disease characterized by low bone mass and degeneration of bone tissue, resulting in increased bone fragility and susceptibility to fracture. In one embodiment, the patient with osteoporosis has abnormal bone strength, resulting in an increased risk of fracture. In another embodiment, osteoporosis depletes calcium and collagen normally found in bone, in one embodiment resulting in abnormal bone mass or reduced bone density. In another embodiment, bone affected by osteoporosis may fracture only by a slight fall or injury that does not normally cause the fracture. In one embodiment, the fracture may be in the form of a fracture (as in a hip fracture) or collapse (as in a spinal compression fracture). The spine, hip and wrist are common areas of osteoporosis-induced fractures, although fractures may occur in other skeletal areas as well. In another embodiment, uninhibited osteoporosis may result in altered posture, physical abnormalities, and reduced mobility.
In one embodiment, the osteoporosis is caused by androgen deprivation. In another embodiment, osteoporosis occurs after androgen deprivation. In another embodiment, the osteoporosis is caused by estrogen deprivation therapy. In another embodiment, osteoporosis occurs after estrogen deprivation therapy. In another embodiment, 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 may predispose a breast cancer patient to bone metastasis and/or predispose the patient to a bone related event.
In another embodiment, 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.
According to this aspect of the invention and in one embodiment, the bone-related disorder is treated with a compound as described herein or a combination thereof. In another embodiment, additional bone stimulating compounds may be provided to the subject prior to, concurrently with, or after administration of one or more compounds as described herein. In one embodiment, such bone stimulating compounds may comprise natural or synthetic materials.
In one embodiment, the bone stimulating compound may include Bone Morphogenic Proteins (BMP), growth factors such as Epidermal Growth Factor (EGF), fibroblast Growth Factor (FGF), transforming growth factor (TGF, insulin Growth Factor (IGF)), platelet-derived growth factor (PDGF) hedgehog proteins (such as sonic, indian and desert hedgehog), hormones (such as follicle stimulating hormone, parathyroid hormone-related peptide, activin, inhibin, follistatin, frizzled-related protein, BMP binding proteins (such as chordin and fetuin), cytokines (such as IL-3, IL-7, GM-CSF), chemokines (such as eosinophil-activating chemokines), collagen, osteocalcin, osteonectin, and the like, as will be appreciated by those skilled in the art.
In another embodiment, the compositions of the present invention for treating bone disorders may comprise one or more compounds described herein, one or more additional bone stimulating compounds, and osteoblasts. In one embodiment, the osteoblast cells may be stem cells or progenitor cells that can be induced to differentiate into osteoblast cells. In another embodiment, the cell may be an osteoblast. In another embodiment, a nucleic acid encoding a bone stimulating compound may be administered to a subject, which is considered part of the present invention.
In one embodiment, the invention provides a method of treating, preventing, suppressing, or inhibiting or reducing the risk of occurrence of a bone related event (SRE), such as a fracture, bone surgery, bone radiation, spinal cord compression, new bone metastasis, bone loss, or a combination thereof, in a subject having cancer, the method comprising administering a compound as described herein and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, or any combination thereof. The invention is particularly directed to the treatment of SRE with the compounds of formula I-XIV of the invention in the following subjects: (a) A subject with prostate cancer who is receiving or has received Androgen Deprivation Therapy (ADT); or (b) a subject having breast cancer who is receiving or has received estrogen deprivation therapy.
In one embodiment, the bone-related event treated using the methods provided herein and/or with the compositions provided herein is a fracture, which in one embodiment is a pathologic fracture, a non-traumatic fracture, a vertebral fracture, a non-vertebral fracture, a morphometric fracture, or a combination thereof. In some embodiments, the fracture may be a simple fracture, a compound fracture, a transverse fracture, a green branch fracture, or a comminuted fracture. In one embodiment, the fracture may be against any bone in the body, in one example, the fracture is a fracture in any one or more bones of an arm, a wrist, a hand, a finger, a leg, an ankle, a foot, a toe, a hip, a collarbone, or a combination thereof. In breast cancer, metastasis most commonly occurs in the hips and vertebrae. In one embodiment, the bone-related fracture is a fracture of the hip and/or vertebra.
In another embodiment, the methods and/or compositions provided herein are effective in treating, preventing, suppressing, inhibiting, or reducing the risk of a bone-related event, such as a pathological fracture, spinal cord compression, hypercalcemia, bone-related pain, or a combination thereof.
In another embodiment, the bone-related event sought to be treated using the methods provided herein and/or with the compositions provided herein includes the necessity of bone surgery and/or bone radiation, which in some embodiments is used to treat pain caused in one embodiment by bone injury or nerve compression. In another embodiment, the bone-related event sought to be treated using the methods provided herein and/or with the compositions provided herein comprises spinal cord compression of the subject, or the necessity of an anti-tumor therapy change (including hormonal therapy change). In some embodiments, a bone-related event that is sought to be treated using the methods provided herein and/or with the compositions provided herein includes treating, suppressing, preventing, reducing the incidence of, or slowing the progression or severity of bone metastasis or bone loss. In one embodiment, bone loss may include osteoporosis, osteopenia, or a combination thereof. In one embodiment, the bone related event may comprise any combination of the embodiments listed herein.
In one embodiment, the methods provided herein and/or utilizing the compositions provided herein are effective to reduce metastasis to bone, such as in terms of number of lesions, lesion size, or a combination thereof. According to this aspect of the invention and in one embodiment, provided herein is a method of preventing or inhibiting metastasis of cancer to bone in a subject, the method comprising the step of administering to the subject a composition comprising toremifene, raloxifene, tamoxifen, or an analog, functional derivative, metabolite, or combination thereof, or a pharmaceutically acceptable salt thereof. In one embodiment, such metabolites may include ospemifene, non-ospemifene, or a combination thereof. In one embodiment, the cancer is prostate cancer. In one embodiment, the cancer is breast cancer.
In one embodiment, the bone-related event is the result of a cancer treatment. In one embodiment, the bone related events are the result of hormone deprivation therapy, while in another embodiment they are the products of Androgen Deprivation Therapy (ADT), and in another embodiment they are the products of estrogen deprivation therapy.
As used herein, the term "force ratio" may refer to libido, or as defined in example 9.
As used herein, the term "quality of life" may refer to the health and life concerns of a subject suffering from a disorder or disease (e.g., suffering from breast cancer) after treatment until the end of life. It covers physical, psychological and economic problems faced by subjects outside of the diagnostic and therapeutic stages. The term "quality of life" may also be referred to herein as "survival". In one embodiment, survival includes problems associated with the ability to obtain health care and follow-up treatment, the post-treatment effects, secondary cancer, and quality of life. Family members, friends, and caregivers are also considered to be part of the survival experience.
In one embodiment, the methods of the invention are useful in a subject, which is a human. In one embodiment, the subject is male. In another embodiment, the subject is female. In some embodiments, while the methods as described herein may be used to treat males or females, females may have a more favorable response to the administration of certain compounds for certain methods. In other embodiments, although the methods as described herein may be used to treat males or females, for certain methods, males may respond more favorably to the administration of certain compounds.
Selective Androgen Receptor Modulator (SARM) compounds
In one embodiment, the compounds of the present invention are effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treatment of a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase4/6 (CDK 4/6) inhibitors (ibuzol (Ibrance), rebabocili (Kisqali), abbe's cili (Vorzenio), trasturil, ly Luo Xili), apifil (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3kα)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olapamide (Lynparza) or talazopali (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitors (lapatinib, lenatinib (Nerlynx), dacatinib (Vizimpro) or tukartinib (Tukysa)), HER2 antibodies (trastuzumab (Herceptin), pertuzumab (perta), maceraxib (margenzab)), HER2 antibody drug conjugates (HER 2 ADC) (trastuzumab (enzab), kazadol, or anti-panaxomum (tzbead anti-panaxzepan) and anti-therapeutic anti-panaxoml (therapeutic anti-panaxoml) or anti-panaxoml (tzbezoma) (therapeutic anti-panaxmajub) (pantizob) or anti-panaxmajus (tzjus) (pantizob) or anti-panaxmajus (pantijus); l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; and/or p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the structure of formula I and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof:
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 (CF) 2 CF 3
R2 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, CN, NO 2 、COR、COOH、CONHR、CF 3 、Sn(R) 3 Or R is 3 Together with the benzene ring to which it is attached, form a fused ring system represented by the structure:
z is NO 2 CN, COR, COOH or CONHR;
y is CF 3 F, br, cl, I, CN 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 A, B or C:
n is an integer from 1 to 4; and is also provided with
m is an integer of 1 to 3.
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject (e.g., a female subject). Accordingly, the present invention provides a method 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 having breast cancer; e) Extending progression free survival of a subject having breast cancer; f) Treating a subject having ER positive breast cancer; g) Treating a subject having metastatic ER-positive breast cancer; h) Treating a subject having refractory ER-positive breast cancer; i) Treating a subject having AR positive ER positive breast cancer; j) Treating a subject having AR positive ER positive refractory breast cancer; k) Treating a subject having AR positive ER positive metastatic breast cancer; l) treating a subject with AR-positive and ER-positive breast cancer; m) treating a subject with AR positive ER positive breast cancer with or without PR and/or HER2 expression; n) treating a subject with advanced ER positive breast cancer; o) treating a subject having ER-positive breast cancer with a 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 (Paboscalid (Ibrance), rebaboscalid (Kisqali), abeli (Vorzinio), trastumide, lassa Luo Xili), apicalicheast (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitor (Evidin), polysaccharase polymerase (PARP) inhibitor (Olaparnib (Lynpza) or Talazolparli (Talrina)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (Papanib, nepaltinib (Takaratinib), albizib (Takara) or anti-bead anti-tuzumab (Heraclidinb) (HERBARP), or anti-bead (HERBARY) ) Trastuzumab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (Enhertu), entrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), atizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) failed treatment; p) treating, preventing, suppressing or inhibiting metastasis in a subject having ER positive breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; t) treating a subject with AR positive HER2 positive breast cancer; u) treating a subject having a breast cancer that expresses an ER mutant, v) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or w) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifying the subject as having AR positive breast cancer to treat breast cancer in the subject, comprising administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by the compound 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 (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, CN, NO 2 、COR、COOH、CONHR、CF 3 、Sn(R) 3 Or R is 3 Together with the benzene ring to which it is attached, form a fused ring system represented by the structure:
z is NO 2 CN, COR, COOH or CONHR;
y is CF 3 F, br, cl, I, CN 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 A, B or C:
n is an integer from 1 to 4; and is also provided with
m is an integer of 1 to 3;
and/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In another embodiment, the invention provides a method for the following uses: a) Treating a subject having HER2 positive breast cancer; b) Treating a subject having HER2 positive refractory breast cancer; c) Treating a subject having HER2 positive metastatic breast cancer; d) Treating a subject having HER2 positive and ER negative breast cancer; e) Treating a subject having HER 2-positive and ER-positive breast cancer; f) Treating a subject having HER2 positive and PR positive breast cancer; g) Treating a subject having HER2 positive and PR negative breast cancer; h) Treating a subject having HER 2-positive and AR-positive breast cancer; i) Treating a subject having HER2 positive and AR negative breast cancer; j) Treating a subject having HER2 positive, ER positive, PR positive, and AR positive breast cancer; k) Treating a subject having HER2 positive, ER positive, PR negative, and AR positive breast cancer; l) treating a subject having HER2 positive, ER positive, PR negative and AR negative breast cancer; m) treating a subject having HER2 positive, ER positive, PR positive and AR negative breast cancer; n) treating a subject having HER2 positive, ER negative, PR negative and AR positive breast cancer; o) treating a subject with HER2 positive, ER negative, PR positive and AR positive breast cancer; p) treating a subject having HER2 positive, ER negative, PR positive and AR negative breast cancer; and/or q) treating a subject having HER2 positive, ER negative, PR negative, and AR negative breast cancer; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound 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 (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, CN, NO 2 、COR、COOH、CONHR、CF 3 、Sn(R) 3 Or R is 3 Together with the benzene ring to which it is attached, form a fused ring system represented by the structure:
z is NO 2 CN, COR, COOH or CONHR;
y is CF 3 F, br, cl, I, CN 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 A, B or C:
n is an integer from 1 to 4; and is also provided with
m is an integer of 1 to 3;
and/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In one embodiment, 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 in formula I 1 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 . In another embodiment, 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.
Substituent Z, Y and R 3 Can be at any position of a ring bearing these substituents (hereinafter referred to as "A ring"). In one embodiment, substituent Z is in the para position of the A ring. In another embodiment, substituent Y is in the meta position of the A ring. In another embodiment, 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 may be in any position of a ring bearing these substituents (hereinafter referred to as "B ring"). In one embodiment, substituent Q is in the para position of the B ring. In another embodiment, the substituent R 2 In the meta position of the B ring. In another embodiment, substituent Q is CN and is in the para position of the B ring.
As contemplated herein, when the integers m and n are greater than 1, the substituent R 2 And R is 3 Is not limited to one particular substituent and may be any combination of the substituents listed above.
In another embodiment, the compounds of the present invention are effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the mammary glands of the subject Progression of cancer; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the compound of formula II:
wherein the method comprises the steps of
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 A, B or C:
r is C 1 -C 4 Alkyl, aryl, and,Phenyl, alkenyl, hydroxy, C 1 -C 4 Haloalkyl, halogen or haloalkenyl; and is also provided with
R 1 Is CH 3 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject (e.g., a female subject). Accordingly, the present invention provides a method 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 having breast cancer; e) Extending progression free survival of a subject having breast cancer; f) Treating a subject having ER positive breast cancer; g) Treating a subject having metastatic ER-positive breast cancer; h) Treating a subject having refractory ER-positive breast cancer; i) Treating a subject having AR positive ER positive breast cancer; j) Treating a subject having AR positive ER positive refractory breast cancer; k) Treating a subject having AR positive ER positive metastatic breast cancer; l) treating a subject with AR-positive and ER-positive breast cancer; m) treating a subject with AR positive ER positive breast cancer with or without PR and/or HER2 expression; n) treating a subject with advanced ER positive breast cancer; o) treating a subject having ER-positive breast cancer with a 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 (Pabosib (Ibrance), raboscalid (Kisqali), abeli (Vorzinio), fraxiril, leylactide, le Luo Xili), apilimus (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitor (Evidin), polysaccharase polymerase (PARP) inhibitor (Olaparnib (Lynpza) or Talafazii (Talrna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (Alapatinib, nepal, latinib (Tuyn), or Vigortinib (Vigordona)), or Viqrap (Viqra) ER2 antibody (trastuzumab (Herceptin), pertuzumab (Perjeta), migratuximab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (Enhertu), enmtuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzzxf (Phesgo)), atentizumab (tecentiq) (PD-L1 blocking antibody), pemetuzumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) failed treatment; p) treating, preventing, suppressing or inhibiting metastasis in a subject having ER positive breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; t) treating a subject with AR positive HER2 positive breast cancer; u) treating a subject having a breast cancer that expresses an ER mutant, v) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or w) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifying the subject as having AR positive breast cancer to treat breast cancer in the subject, comprising administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by the compound of formula II:
Wherein the method comprises the steps of
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 A, B or C:
r is C 1 -C 4 Alkyl, aryl, phenyl, alkenyl, hydroxy, C 1 -C 4 Haloalkyl, halogen or haloalkenyl; and is also provided with
R 1 Is CH 3 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
And/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In another embodiment, the invention provides a method for the following uses: a) Treating a subject having HER2 positive breast cancer; b) Treating a subject having HER2 positive refractory breast cancer; c) Treating a subject having HER2 positive metastatic breast cancer; d) Treating a subject having HER2 positive and ER negative breast cancer; e) Treating a subject having HER 2-positive and ER-positive breast cancer; f) Treating a subject having HER2 positive and PR positive breast cancer; g) Treating a subject having HER2 positive and PR negative breast cancer; h) Treating a subject having HER 2-positive and AR-positive breast cancer; i) Treating a subject having HER2 positive and AR negative breast cancer; j) Treating a subject having HER2 positive, ER positive, PR positive, and AR positive breast cancer; k) Treating a subject having HER2 positive, ER positive, PR negative, and AR positive breast cancer; l) treating a subject having HER2 positive, ER positive, PR negative and AR negative breast cancer; m) treating a subject having HER2 positive, ER positive, PR positive and AR negative breast cancer; n) treating a subject having HER2 positive, ER negative, PR negative and AR positive breast cancer; o) treating a subject with HER2 positive, ER negative, PR positive and AR positive breast cancer; p) treating a subject having HER2 positive, ER negative, PR positive and AR negative breast cancer; and/or q) treating a subject having HER2 positive, ER negative, PR negative, and AR negative breast cancer; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound of formula II:
Wherein the method comprises the steps of
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 A, B or C:
r is C 1 -C 4 Alkyl, aryl, phenyl, alkenyl, hydroxy, C 1 -C 4 Haloalkyl, halogen or haloalkenyl; and is also provided with
R 1 Is CH 3 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
And/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In one embodiment, 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 in formula II 1 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. In another embodiment, Q in formula II is NHCOCH 3 . In another embodiment, Q in formula II is in the para position. In another embodiment, Z in formula II is in the para position. In another embodiment, Y in formula II is in the meta position. In another embodiment, 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. In another embodiment, 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 may be in any position of the ring bearing these substituents (hereinafter referred to as "A ring"). In one embodiment, substituent Z is in the para position of the A ring. In another embodiment, substituent Y is in the meta position of the A ring. In another embodiment, substituent Z is in the para position of the A ring and substituent Y is in the meta position of the A ring.
The substituent Q may be at any position of a ring having the substituent (hereinafter referred to as "B ring"). In one embodiment, substituent Q is in the para position of the B ring. In another embodiment, substituent Q is CN and is in the para position of the B ring. [000209]In another embodiment, the compounds of the present invention are effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Abilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), ago Sha Tuo Beadmab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) treatment failed; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or u) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the breast cancer in the subject, the compound being a compound represented by the structure of formula III:
wherein the method comprises the steps of
Z is NO 2 CN, COOH, COR, NHCOR or CONHR;
y is CF 3 、F、I、Br、Cl、CN、C(R) 3 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 A, B or C:
r is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F、CHF 2 、CF 3 、CF 2 CF 3 An aryl group,
Phenyl, halogen, alkenyl or OH.
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject (e.g., a female subject). Accordingly, the present invention provides a method 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 having breast cancer; e) Extending progression free survival of a subject having breast cancer; f) Treating a subject having ER positive breast cancer; g) Treating a subject having metastatic ER-positive breast cancer; h) Treating a subject having refractory ER-positive breast cancer; i) Treating a subject having AR positive ER positive breast cancer; j) Treating a subject having AR positive ER positive refractory breast cancer; k) Treating a subject having AR positive ER positive metastatic breast cancer; l) treating a subject with AR-positive and ER-positive breast cancer; m) treating a subject with AR positive ER positive breast cancer with or without PR and/or HER2 expression; n) treating a subject with advanced ER positive breast cancer; o) treating a subject having ER-positive breast cancer with a 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 (Pabosinib (Ibrance), rabosinib (Kisqali), abelib (Vorzinio), fraxib, leonum Luo Xili), apicalichease (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitor (Evidin), polysaccharase polymerase (PARP) inhibitor (Olaparnib (Lynpza) or Talazolpali (Talrna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (Lapannib, netinib), nepalinib (Tacrolimus) Treatment failed with either tinib (Vizimpro) or tukatinib (Tukysa)), HER2 antibody (trastuzumab (Herceptin), pertuzumab (Perjeta), mactuximab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (Enhertu), enmtuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), atentizumab (tecantrioq) (PD-L1 blocking antibody), pemuzumab (Keytruda) (PD-L1 blocking antibody), go Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin); p) treating, preventing, suppressing or inhibiting metastasis in a subject having ER positive breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; t) treating a subject with AR positive HER2 positive breast cancer; and/or u) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the breast cancer in the subject; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound of formula III:
Wherein the method comprises the steps of
Z is NO 2 CN, COOH, COR, NHCOR or CONHR;
y is CF 3 、F、I、Br、Cl、CN、C(R) 3 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 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 analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In another embodiment, the invention provides a method for the following uses: a) Treating a subject having HER2 positive breast cancer; b) Treating a subject having HER2 positive refractory breast cancer; c) Treating a subject having HER2 positive metastatic breast cancer; d) Treating a subject having HER2 positive and ER negative breast cancer; e) Treating a subject having HER 2-positive and ER-positive breast cancer; f) Treating a subject having HER2 positive and PR positive breast cancer; g) Treating a subject having HER2 positive and PR negative breast cancer; h) Treating a subject having HER 2-positive and AR-positive breast cancer; i) Treating a subject having HER2 positive and AR negative breast cancer; j) Treating a subject having HER2 positive, ER positive, PR positive, and AR positive breast cancer; k) Treating a subject having HER2 positive, ER positive, PR negative, and AR positive breast cancer; l) treating a subject having HER2 positive, ER positive, PR negative and AR negative breast cancer; m) treating a subject having HER2 positive, ER positive, PR positive and AR negative breast cancer; n) treating a subject having HER2 positive, ER negative, PR negative and AR positive breast cancer; o) treating a subject with HER2 positive, ER negative, PR positive and AR positive breast cancer; p) treating a subject having HER2 positive, ER negative, PR positive and AR negative breast cancer; and/or q) treating a subject having HER2 positive, ER negative, PR negative, and AR negative breast cancer; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound of formula III:
Wherein the method comprises the steps of
Z is NO 2 CN, COOH, COR, NHCOR or CONHR;
y is CF 3 、F、I、Br、Cl、CN、C(R) 3 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 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 analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In one embodiment, 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 embodimentIn formula III, Y 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 and Y is CF 3 Or halogen, and Q is CN or F. In another embodiment, Z is NO 2 Y is CF 3 And Q is NHCOCH 3 F or Cl.
In another embodiment, the compounds of the present invention are effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (Kisqali), abbe-cili (Vorzenio), trazoxilinide, ly Luo Xili), axist (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3K α), mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olpatanib (lynpara) or talzenia), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepatib (nepafenib), trazotinib (tujingku), moxib (virtuzumab (zhengag), or anti-beads (Herceptin 2) HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (Enhertu), emtrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), atilizumab (tecantriq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the structure of formula IV and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof:
Wherein the method comprises the steps of
X is a bond, O, CH 2 NH, S, se, PR, NO or NR;
g is O or S;
R 1 is CH 3 、CH 2 F、CHF 2 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
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;
a is a ring selected from:
b is a ring selected from the group consisting of:
wherein A and B cannot be both benzene rings;
z is NO 2 CN, COOH, COR, NHCOR or CONHR;
y is CF 3 、F、I、Br、Cl、CN、C(R) 3 Or Sn (R) 3
Q 1 And Q 2 Independently hydrogen, alkyl, halogen, CF 3 、CN、C(R) 3 、Sn(R) 3 、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, SR, or
Q 3 And Q 4 Are independently of each other hydrogen, alkyl, halogen, CF 3 、CN、C(R) 3 、Sn(R) 3 、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;
W 1 o, NH, NR, NO or S; and is also provided with
W 2 Is N or NO.
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject (e.g., a female subject). Accordingly, the present invention provides a method 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 having breast cancer; e) Extending progression free survival of a subject having breast cancer; f) Treating a subject having ER positive breast cancer; g) Treating a subject having metastatic ER-positive breast cancer; h) Treating a subject having refractory ER-positive breast cancer; i) Treating a subject having AR positive ER positive breast cancer; j) Treating a subject having AR positive ER positive refractory breast cancer; k) Treating a subject having AR positive ER positive metastatic breast cancer; l) treating a subject with AR-positive and ER-positive breast cancer; m) treating a subject with AR positive ER positive breast cancer with or without PR and/or HER2 expression; n) treating a subject with advanced ER positive breast cancer; o) treating a subject having ER positive breast cancer with a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonist (goserelin), aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclane), rapuqali (kiquali), abbe-cilia (Vorzenio), trazoxiline, ly Luo Xili), apifil (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha), mTOR inhibitor (everin), polysaccharase polymerase (PARP) inhibitor (lynpza) or talazapril (talpanna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (rapalotinib, nepafenib (tuyene), trazoxan (tuyene), anti-bead anti-tuzumab (Herceptin), or anti-tuzumab (trazotinib) antibody (rufin 2) or (trazotinib) bead anti-tuzumab (Herceptin) antibody (Herceptin 2) Anti/hyaluronidase-zzxf (Phesgo)), atenolizumab (tecantrioq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; p) treating, preventing, suppressing or inhibiting metastasis in a subject having ER positive breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; t) treating a subject with AR positive HER2 positive breast cancer; u) treating a subject having a breast cancer that expresses an ER mutant, v) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or w) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifying the subject as having AR positive breast cancer to treat breast cancer in the subject, comprising administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound of formula IV:
wherein the method comprises the steps of
X is a bond, O, CH 2 NH, S, se, PR, NO or NR;
G is O or S;
R 1 is CH 3 、CH 2 F、CHF 2 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
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;
a is a ring selected from:
b is a ring selected from the group consisting of:
wherein A and B cannot be both benzene rings;
z is NO 2 CN, COOH, COR, NHCOR or CONHR;
y is CF 3 、F、I、Br、Cl、CN、C(R) 3 Or Sn (R) 3
Q 1 And Q 2 Independently hydrogen, alkyl, halogen, CF 3 、CN、C(R) 3 、Sn(R) 3 、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, SR, or
Q 3 And Q 4 Are independently of each other hydrogen, alkyl, halogen, CF 3 、CN、C(R) 3 、Sn(R) 3 、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;
W 1 o, NH, NR, NO or S; and is also provided with
W 2 Is N or NO;
and/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In another embodiment, the invention provides a method for the following uses: a) Treating a subject having HER2 positive breast cancer; b) Treating a subject having HER2 positive refractory breast cancer; c) Treating a subject having HER2 positive metastatic breast cancer; d) Treating a subject having HER2 positive and ER negative breast cancer; e) Treating a subject having HER 2-positive and ER-positive breast cancer; f) Treating a subject having HER2 positive and PR positive breast cancer; g) Treating a subject having HER2 positive and PR negative breast cancer; h) Treating a subject having HER 2-positive and AR-positive breast cancer; i) Treating a subject having HER2 positive and AR negative breast cancer; j) Treating a subject having HER2 positive, ER positive, PR positive, and AR positive breast cancer; k) Treating a subject having HER2 positive, ER positive, PR negative, and AR positive breast cancer; l) treating a subject having HER2 positive, ER positive, PR negative and AR negative breast cancer; m) treating a subject having HER2 positive, ER positive, PR positive and AR negative breast cancer; n) treating a subject having HER2 positive, ER negative, PR negative and AR positive breast cancer; o) treating a subject with HER2 positive, ER negative, PR positive and AR positive breast cancer; p) treating a subject having HER2 positive, ER negative, PR positive and AR negative breast cancer; and/or q) treating a subject having HER2 positive, ER negative, PR negative, and AR negative breast cancer; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound of formula IV:
Wherein the method comprises the steps of
X is a bond, O, CH 2 NH, S, se, PR, NO or NR;
g is O or S;
R 1 is CH 3 、CH 2 F、CHF 2 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
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;
a is a ring selected from:
b is a ring selected from the group consisting of:
wherein A and B cannot be both benzene rings;
z is NO 2 CN, COOH, COR, NHCOR or CONHR;
y is CF 3 、F、I、Br、Cl、CN、C(R) 3 Or Sn (R) 3
Q 1 And Q 2 Independently hydrogen, alkyl, halogen, CF 3 、CN、C(R) 3 、Sn(R) 3 、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, SR, or
Q3 and Q4 are independently of each other hydrogen, alkyl, halogen, CF 3 、CN、C(R) 3 、Sn(R) 3 、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;
W 1 o, NH, NR, NO or S; and is also provided with
W 2 Is N or NO;
and/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In one embodiment, G in formula IV is O. In another embodiment, X in formula II is O. In another embodiment, T in formula IV is OH. In another embodiment, R in formula IV 1 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 in formula II 1 Is CN. In another embodiment, Q in formula IV 1 Is F. In another embodiment, Q in formula IV 1 Is Cl. In another embodiment, Q in formula II 1 Is NHCOCH 3 . In another embodiment, Q in formula IV 1 Is positioned at the para-position. In another embodiment, Z in formula IV is in the para position. In another embodiment, Y in formula IV is in the meta position. In another embodiment, 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 may be in any position of the ring bearing these substituents (hereinafter referred to as "A ring"). In one embodiment, substituent Z is in the para position of the A ring. In another embodiment, substituent Y is in the meta position of the A ring. In another embodiment, substituent Z is in the para position of the A ring and substituent Y is in the meta position of the A ring.
Substituent Q 1 And Q 2 Can be located at any position of a ring bearing these substituents (hereinafter referred to as "B ring"). In one embodiment, substituent Q 1 Is positioned at the para position of the B ring. In another embodiment, substituent Q 2 Is H. In another embodiment, substituent Q 1 In para position of ring B and substituent Q 2 Is H. In another embodiment, substituent Q 1 Is CN and is in the para position of the B ring, and the substituent Q 2 Is H.
As contemplated herein, other specific embodiments of the compounds are included within the scope of the present invention and are useful for: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treatment of a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (pamoxmide (Ibrance), rapazetinib (Kisqali), abbe-cili (Vorzenio), trasturil, ly Luo Xili), apiril (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3kα), mTOR inhibitor (everolimus), a Poly ADP Ribose Polymerase (PARP) inhibitor (olaparnib (lynparazapal) or talzenia)), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, lenatinib (Nerlynx), dactyltinib (Vizimpro) or tukutinib (Tukysa)), HER2 antibody (trastuzumab (Herceptin), pertuzumab (Perjeta), magtuximab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (Enhertu), enmtuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), atizumab (tecentiq) (PD-L1 blocking antibody), pemumab (Keytruda) (PD-L1 blocking antibody), golimumab (trodulvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the breast cancer in the subject, these compounds are of formula V or VI. It is understood that included within the scope of the present invention are analogs, derivatives, metabolites, isomers, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, polymorphs, crystals, prodrugs, or combinations thereof of these compounds:
wherein the method comprises the steps of
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.
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject (e.g., a female subject). Accordingly, the present invention provides a method 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 having breast cancer; e) Extending progression free survival of a subject having breast cancer; f) Treating a subject having ER positive breast cancer; g) Treating a subject having metastatic ER-positive breast cancer; h) Treating a subject having refractory ER-positive breast cancer; i) Treating a subject having AR positive ER positive breast cancer; j) Treating a subject having AR positive ER positive refractory breast cancer; k) Treating a subject having AR positive ER positive metastatic breast cancer; l) treating a subject with AR-positive and ER-positive breast cancer; m) treating a subject with AR positive ER positive breast cancer with or without PR and/or HER2 expression; n) treating a subject with advanced ER positive breast cancer; o) treating a subject having ER-positive breast cancer with a 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 (Pabosinib (Ibrance), rabosinib (Kisqali), abeli (Vorzenio), fraxib, laziprasidi, lavender Luo Xili), apicalis (Piqray) (phosphatidylinositol-3-kinase subunit alpha) PI3kα), mTOR inhibitor (everolimus), poly ADP Ribose Polymerase (PARP) inhibitor (olnparza) or talazolepali (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, lenatinib (Nerlynx), dacatinib (Vizimpro) or tucattinib (Tukysa)), HER2 antibody (trastuzumab (Herceptin), pertuzumab (Perjeta), mactututuximab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (Enhertu), enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzzxf (phesgocriptin), actlizumab (tentriq) (PD-L1 blocking antibody), petuzumab (keytrub) (bc-binding antibody), HER2 antibody (Sha Tuozhu) or anti-prasugrel antibody (tnvalbumb) (therapeutic failure; p) treating, preventing, suppressing or inhibiting metastasis in a subject having ER positive breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; t) treating a subject with AR positive HER2 positive breast cancer; u) treating a subject having a breast cancer that expresses an ER mutant, v) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or w) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifying the subject as having AR positive breast cancer to treat breast cancer in the subject, comprising administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by the structure of formula V or VI:
wherein the method comprises the steps of
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 analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In another embodiment, the invention provides a method for the following uses: a) Treating a subject having HER2 positive breast cancer; b) Treating a subject having HER2 positive refractory breast cancer; c) Treating a subject having HER2 positive metastatic breast cancer; d) Treating a subject having HER2 positive and ER negative breast cancer; e) Treating a subject having HER 2-positive and ER-positive breast cancer; f) Treating a subject having HER2 positive and PR positive breast cancer; g) Treating a subject having HER2 positive and PR negative breast cancer; h) Treating a subject having HER 2-positive and AR-positive breast cancer; i) Treating a subject having HER2 positive and AR negative breast cancer; j) Treating a subject having HER2 positive, ER positive, PR positive, and AR positive breast cancer; k) Treating a subject having HER2 positive, ER positive, PR negative, and AR positive breast cancer; l) treating a subject having HER2 positive, ER positive, PR negative and AR negative breast cancer; m) treating a subject having HER2 positive, ER positive, PR positive and AR negative breast cancer; n) treating a subject having HER2 positive, ER negative, PR negative and AR positive breast cancer; o) treating a subject with HER2 positive, ER negative, PR positive and AR positive breast cancer; p) treating a subject having HER2 positive, ER negative, PR positive and AR negative breast cancer; and/or q) treating a subject having HER2 positive, ER negative, PR negative, and AR negative breast cancer; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound of formula V or VI:
Wherein the method comprises the steps of
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 analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In one embodiment, 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
In another embodiment, the compounds of the present invention are effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) prolonged suffering from milk Progression free survival of the subject of the adenocarcinoma; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the structure of formula VII and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof:
wherein Z is Cl or CF 3
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject (e.g., a female subject). Accordingly, the present invention provides a method 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 having breast cancer; e) Extending progression free survival of a subject having breast cancer; f) Treating a subject having ER positive breast cancer; g) Treating a subject having metastatic ER-positive breast cancer; h) Treating a subject having refractory ER-positive breast cancer; i) Treating a subject having AR positive ER positive breast cancer; j) Treating a subject having AR positive ER positive refractory breast cancer; k) Treating a subject having AR positive ER positive metastatic breast cancer; l) treating a subject with AR-positive and ER-positive breast cancer; m) treating a subject with AR positive ER positive breast cancer with or without PR and/or HER2 expression; n) treating a subject with advanced ER positive breast cancer; o) treating a subject having ER-positive breast cancer with a Selective Estrogen Receptor Modulator (SERM) ) (tamoxifen, toremifene, raloxifene), gonadotropin releasing hormone (GnRH) agonists (goserelin), aromatase Inhibitors (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitors (pamoxnib (Ibrance), rebamipril (Kisqali), abbe-cili (Vorzenio), fraxil, L Luo Xili), apifil (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (lyarza) or talazolepali (Talzenna)); human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, lenatinib (Nerlynx), dactyltinib (Vizimpro) or tucarttinib (Tukysa)), HER2 antibody (trastuzumab (Herceptin), pertuzumab (Perjeta), migratuzumab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (detrastuzumab (enherty), enmettrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), atizumab (tecentiq) (PD-L1 blocking antibody), a, pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; p) treating, preventing, suppressing or inhibiting metastasis in a subject having ER positive breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; t) treating a subject with AR positive HER2 positive breast cancer; and/or u) treating a subject having a breast cancer that expresses an ER mutant, v) treating a subject having a breast cancer that expresses a Y537SER mutant; and/or w) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifying the subject as having AR positive breast cancer to treat breast cancer in the subject, comprising administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by the structure of formula VII:
wherein Z is Cl or CF 3
And/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject
In another embodiment, the invention provides a method for the following uses: a) Treating a subject having HER2 positive breast cancer; b) Treating a subject having HER2 positive refractory breast cancer; c) Treating a subject having HER2 positive metastatic breast cancer; d) Treating a subject having HER2 positive and ER negative breast cancer; e) Treating a subject having HER 2-positive and ER-positive breast cancer; f) Treating a subject having HER2 positive and PR positive breast cancer; g) Treating a subject having HER2 positive and PR negative breast cancer; h) Treating a subject having HER 2-positive and AR-positive breast cancer; i) Treating a subject having HER2 positive and AR negative breast cancer; j) Treating a subject having HER2 positive, ER positive, PR positive, and AR positive breast cancer; k) Treating a subject having HER2 positive, ER positive, PR negative, and AR positive breast cancer; l) treating a subject having HER2 positive, ER positive, PR negative and AR negative breast cancer; m) treating a subject having HER2 positive, ER positive, PR positive and AR negative breast cancer; n) treating a subject having HER2 positive, ER negative, PR negative and AR positive breast cancer; o) treating a subject with HER2 positive, ER negative, PR positive and AR positive breast cancer; p) treating a subject having HER2 positive, ER negative, PR positive and AR negative breast cancer; and/or q) treating a subject having HER2 positive, ER negative, PR negative, and AR negative breast cancer; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by a compound of formula VII:
Wherein Z is Cl or CF 3
And/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In another embodiment, the compound is effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having breast cancer with a 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), rapbosil (kiquali), abbe-cili (Vorzenio), trazoxiline, ly Luo Xili), apifil (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha), mTOR inhibitor (everolimus), poly (ADP) inhibitor (oxipamil) or tazizopanil (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepatib (neomycin), trazotinib (tujingku), anti-bead 2 (herception) or anti-HER 2 (herception), anti-bead 2 (HER 2) antibody (perzhuzafiomb) or anti-panitude (HER 2) Treatment with either zumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), atilizumab (tecontriq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the structure of formula VIII and/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combination thereof:
In another embodiment, the compound is effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having breast cancer with selectivityAn estrogen receptor modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonist (goserelin), aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib), rapaminib (Kisqali), abbe-li (Vorzenio), trazoxiline, ly Luo Xili), apifil (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3 alpha)), mTOR inhibitor (everolimus), poly ADP-ribose polymerase (PARP) inhibitor (lynpazazab) or talazopani (Talzenna)), human growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, letinib (nerox), dactinotinib (vic) or tuzizane (valcanib), anti-bead anti-tuzumab (Herceptin) antibody (herception) or anti-bead anti-tuzuelapsib) (anti-tuzuelapsib) anti-beads (Herceptin 2) or anti-tuzuelapsib (Herceptin) (Herceptin 2) anti-beads (Herceptin) anti-tuzuelapsib) anti-tuzuelas (Herceptin) or anti-tuzuelas (herqua) and anti-tuzuelas (tujatizuelas) and (kajab) may be made to be released to a, pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifying the subject as having AR-positive breast cancer to treat the subject's breast cancer, the compound being a compound represented by the structure of formula IX and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug thereofOr any combination thereof:
in another embodiment, the compound is effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Treatment with atenolizumab (tecantroq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) Failure of therapy; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having breast cancer expressing an ER mutant; s) treating a subject having breast cancer expressing a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the structure of formula X and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof:
in another embodiment, the compound is effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treatment of a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (pamoate) West (Ibrance), rebaudinib (Kisqali), abbe west (Vorzenio), trasturil, ly Luo Xili), apicalist (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3kα)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olaparib (lynparaza) or talazopania (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitors (lapatinib, lenatinib (Nerlynx), dacomitinib (Vizimpro) or fig. calitinib (Tukysa)), HER2 antibodies (trastuzumab (Herceptin), pertuzumab (Perjeta), maceraxib (margenzab)), HER2 antibody drug conjugates (HER 2 ADC) (entuzumab), enmtuzumab (kadbc) or other anti-panaxatrix (tntuzumab) or anti-panaxzebra) antibodies (apbook), or anti-panaxzebra antibody (apbook) and anti-panaxmzbezob (tzqq) (therapeutic antibody) (therapeutic anti-panaxmzvalb) or anti-panaxs1-panaxb (tzpanaxb); l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the structure of formula XI and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof:
in another embodimentIn (2), the compound is effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) prolonging progression free survival of a subject having breast cancer A period; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the breast cancer in the subject, the compound being a compound represented by the structure of formula XII and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof:
in another embodiment, the compound is effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treatment of a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (pamoxmide (Ibrance), rapazetinib (Kisqali), abbe-cili (Vorzenio), trasturil, lenaline Luo Xili), axist (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3kα), mTOR inhibitor (everolimus), a Poly ADP Ribose Polymerase (PARP) inhibitor (olapatinib (lynparaza) or tala) Zolpidil (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, lenatinib (Nerlynx), dactyltinib (Vizimpro) or tukyra), HER2 antibody (trastuzumab (Herceptin), pertuzumab (Perjeta), migrituximab (Margenza)), HER2 antibody drug conjugate (HER 2 ADC) (Enhertu), enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzzxf (Phesgo)), altlizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golizumab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) failed therapy; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject with breast cancer expressing an ER mutant, S) treating a subject with breast cancer expressing a Y537S ER mutant, and/or t) determining the presence of a target in the subject by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the breast cancer in the subject, the compound being a compound represented by the compound of formula XIII and/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combination thereof:
in another embodiment, the compound is effective: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating patientsA subject with AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat breast cancer in the subject, the compound being a compound represented by the structure of formula XIV and/or an analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof:
in one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject (e.g., a female subject). Accordingly, the present invention provides a method 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 having breast cancer; e) Extending progression free survival of a subject having breast cancer; f) Treating a subject having ER positive breast cancer; g) Treating a subject having metastatic ER-positive breast cancer; h) Treating a subject having refractory ER-positive breast cancer; i) Treating a subject having AR positive ER positive breast cancer; j) Treating a subject having AR positive ER positive refractory breast cancer; k) Treating a subject having AR positive ER positive metastatic breast cancer; l) treating a subject with AR-positive and ER-positive breast cancer; m) treating a subject with AR positive ER positive breast cancer with or without PR and/or HER2 expression; n) treating a subject with advanced ER positive breast cancer; o) treating a subject having ER-positive breast cancer with a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor (Pabosini (Ibrance), rabosini (Kisqali), abeli (Vorzinio), fraxiril, laziprairie, lassa Luo Xili), april (Piqray) (phosphorus) Inhibitors of acyl inositol-3-kinase subunit α (PI 3kα), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olrapanib (Lynparza) or talazopania), human epidermal growth factor receptor 2 (HER 2) kinase inhibitors (lapatinib, lenatinib (Nerlynx), dacatinib (Vizimpro) or tukyra), HER2 antibodies (trastuzumab (Herceptin), pertuzumab (Perjeta), megtutuximab (Margenza)), HER2 antibody drug conjugates (HER 2 ADC) (detrituximab (Enhertu), enmetuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), actigb (tecentiq) (PD-L1 blocking antibodies), gautuzumab (Avastin-62) and anti-therapeutic antibodies (trastum) or anti-therapeutic antibodies (trastum 1/trvam); p) treating, preventing, suppressing or inhibiting metastasis in a subject having ER positive breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; and/or t) treating a subject with AR positive HER2 positive breast cancer; u) treating a subject having a breast cancer that expresses an ER mutant, v) treating a subject having a breast cancer that expresses a Y537SER mutant; and/or w) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifying the subject as having AR positive breast cancer to treat breast cancer in the subject, comprising administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by the structure of formula VIII, IX, X, XI, XII, XIII or XIV:
/>
and/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In another embodiment, the invention provides a method for the following uses: a) Treating a subject having HER2 positive breast cancer; b) Treating a subject having HER2 positive refractory breast cancer; c) Treating a subject having HER2 positive metastatic breast cancer; d) Treating a subject having HER2 positive and ER negative breast cancer; e) Treating a subject having HER 2-positive and ER-positive breast cancer; f) Treating a subject having HER2 positive and PR positive breast cancer; g) Treating a subject having HER2 positive and PR negative breast cancer; h) Treating a subject having HER 2-positive and AR-positive breast cancer; i) Treating a subject having HER2 positive and AR negative breast cancer; j) Treating a subject having HER2 positive, ER positive, PR positive, and AR positive breast cancer; k) Treating a subject having HER2 positive, ER positive, PR negative, and AR positive breast cancer; l) treating a subject having HER2 positive, ER positive, PR negative and AR negative breast cancer; m) treating a subject having HER2 positive, ER positive, PR positive and AR negative breast cancer; n) treating a subject having HER2 positive, ER negative, PR negative and AR positive breast cancer; o) treating a subject with HER2 positive, ER negative, PR positive and AR positive breast cancer; p) treating a subject having HER2 positive, ER negative, PR positive and AR negative breast cancer; and/or q) treating a subject having HER2 positive, ER negative, PR negative, and AR negative breast cancer; the method comprises administering to the subject a therapeutically effective amount of a Selective Androgen Receptor Modulator (SARM) compound represented by the following formula VIII, IX, X, XI, XII, XIII or XIV structure:
/>
And/or analogs, derivatives, isomers, metabolites, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, crystals, polymorphs, prodrugs, or any combinations thereof, as described herein. In one embodiment, the subject is a female subject. In one embodiment, the subject is a male subject.
In one embodiment, the methods of the present invention utilize a compound of formula VIII. In one embodiment, the methods of the present invention utilize a compound of formula IX. In one embodiment, the methods of the present invention utilize a compound of formula X. In one embodiment, the methods of the present invention utilize compounds of formula XI. In one embodiment, the process of the present invention utilizes a compound of formula XII. In one embodiment, the methods of the present invention utilize compounds of formula XIII. In one embodiment, the methods of the present invention utilize compounds of formula XIV.
In one embodiment, the methods of the invention comprise administering an analog of a compound of formula I-XIV. In another embodiment, the method of the invention comprises administering a derivative of a compound of formula I-XIV. In another embodiment, the methods of the invention comprise administering an isomer of a compound of formula I-XIV. In another embodiment, the method of the invention comprises administering a metabolite of the compounds of formula I-XIV. In another embodiment, the methods of the invention comprise administering a pharmaceutically acceptable salt of a compound of formula I-XIV. In another embodiment, the method of the invention comprises administering a pharmaceutical product of a compound of formula I-XIV. In another embodiment, the method of the invention comprises administering a hydrate of a compound of formula I-XIV. In another embodiment, the method of the invention comprises administering an N-oxide of a compound of formula I-XIV. In another embodiment, the methods of the invention comprise administering a polymorph of a compound of formula I-XIV. In another embodiment, the method of the invention comprises administering crystals of the compound of formula I-XIV. In another embodiment, the methods of the invention comprise administering a prodrug of a compound of formula I-XIV. In another embodiment, the methods of the invention comprise administering any combination of analogs, derivatives, metabolites, isomers, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides, polymorphs, crystals, or prodrugs of the compounds of formulas I-XIV.
In one embodiment, the method of the invention comprises administering a compound of formula I-XIV. In another embodiment, the methods of the invention comprise administering a compound of formula I. In another embodiment, the methods of the invention comprise administering a compound of formula II. In another embodiment, the methods of the invention comprise administering a compound of formula III. In another embodiment, the methods of the invention comprise administering a compound of formula IV. In another embodiment, the method of the invention comprises administering a compound of formula V. In another embodiment, the methods of the invention comprise administering a compound of formula VI. In another embodiment, the method of the invention comprises administering a compound of formula VII. In another embodiment, the method of the invention comprises administering a compound of formula VIII. In another embodiment, the methods of the invention comprise administering a compound of formula IX. In another embodiment, the method of the invention comprises administering a compound of formula X. In another embodiment, the methods of the invention comprise administering a compound of formula XI. In another embodiment, the method of the invention comprises administering a compound of formula XII. In another embodiment, the method of the invention comprises administering a compound of formula XIII. In another embodiment, the method of the invention comprises administering a compound of formula XIV.
The compounds of the invention, alone or as pharmaceutical compositions, are useful for: a) Treating a subject having breast cancer: b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression;i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) extending the survival of a subject with ER positive breast cancer; r) slowing the progression of ER positive breast cancer in the subject; s) extending progression-free survival of a subject having ER-positive breast cancer; t) treating a subject having AR positive HER2 positive breast cancer, u) treating a subject having breast cancer expressing an ER mutant, v) treating a subject having breast cancer expressing a Y537S ER mutant; and/or w) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) swellingTumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer to treat the subject's breast cancer.
The compounds of the present invention provide significant advances over steroid androgen therapy because 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, non-cross-reactivity with other steroid receptors, non-aromatizability and long biological half-life.
In one embodiment, the invention relates to treating androgen receptor positive breast cancer in a subject. Accordingly, the present invention provides the following method: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treatment of subjects with breast cancer employing a 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 (pamtezenib (ibrence), rapmaceniib (kiquali), abbe-cili (Vorzenio), trazoxiline, ly Luo Xili), apirilsey (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitor (everolimus), poly ADP-ribose polymerase (PARP) inhibitor (oxipam (lynpara) or tazepali (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, neptinib (nepafex), dacliziram (tuyene (zhengliziram), or anti-beads (panaxomuta 2), anti-beads (panaxomuta 2) or anti-beads (perpanaxat) (Margeneza)), HER2 antibody drug conjugate (HER 2 ADC) (Detrastuzumab (Enheretu), enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), actiglizumab (Tecentriq) (PD-L1 blocking antibody), pembrolizumab (Kertuda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; and/or p) extending progression free survival of a subject having breast cancer; q) treating a subject having HER2 positive breast cancer; r) treating a subject having a breast cancer that expresses an ER mutant, S) treating a subject having a breast cancer that expresses a Y537S ER mutant; and/or t) by first determining 18 F-16 beta-fluoro-5 alpha-dihydrotestosterone 18 F-DHT) tumor uptake and basis 18 F-DHT tumor uptake identifies the subject as having AR positive breast cancer, and treats the breast cancer in the subject by administering to the subject a therapeutically effective amount of a selective androgen receptor modulator of formula I-XIV of the invention and/or its analog, derivative, isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide, crystal, polymorph, prodrug or any combination thereof, as described herein.
As used herein, 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 the "isomer".
In one embodiment, the invention encompasses the use of various optical isomers of selective androgen receptor modulators. Those skilled in the art will appreciate that the selective androgen receptor modulators of the present invention contain at least one chiral center. Thus, the selective androgen receptor modulators used in the methods of the present invention may exist in optically active or racemic forms and be isolated in these 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 forms possess properties suitable for use in the treatment of androgen-related conditions described herein. In one embodiment, the selective androgen receptor modulator is the pure (R) -isomer. In another embodiment, the selective androgen receptor modulator is the pure (S) -isomer. In another embodiment, the selective androgen receptor modulator is a mixture of the (R) isomer and the (S) isomer. In another embodiment, the selective androgen receptor modulator is a racemic mixture comprising equal amounts of the (R) isomer and the (S) isomer. It is well known in the art how to prepare optically active forms (e.g. by resolution of the racemic form using recrystallization techniques, by synthesis from optically active starting materials, by chiral synthesis or by chromatographic separation using chiral stationary phases).
The present invention encompasses "pharmaceutically acceptable salts" of the compounds of the present invention, which salts may be produced by the reaction of a compound of the present invention with an acid or base.
The invention includes pharmaceutically acceptable salts of amino-substituted compounds with organic and inorganic acids, such as citric acid and hydrochloric acid. The invention also includes N-oxides of amino substituents of the compounds described herein. Pharmaceutically acceptable salts can also be prepared from phenolic compounds by treatment with an inorganic base such as sodium hydroxide. In addition, esters of phenolic compounds can be prepared with aliphatic and aromatic carboxylic acids, such as acetates and benzoates.
Suitable pharmaceutically acceptable salts of the compounds of formula I-XIV may be prepared from inorganic acids or from organic acids. In one embodiment, examples of inorganic salts of the compounds of the present invention are bisulfate, borate, bromide, chloride, hemisulfate, hydrobromide, hydrochloride, 2-isethionate (hydroxyethane sulfonate), iodate, iodide, isoparaffinate, nitrate, homosulfate, phosphate, sulfate, sulfamate, sulfanilate, sulfonic acid (alkyl sulfonate, aryl sulfonate, halogen-substituted alkyl sulfonate, halogen-substituted aryl sulfonate), sulfonate, and thiocyanate.
In one embodiment, examples of inorganic salts of the compounds of the present invention may be selected from the group consisting of organic acids of aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic acids, examples thereof are acetic acid, arginine, aspartic acid, ascorbate, adipate, anthranilate, alginate, alkane carboxylate, substituted alkane carboxylate, alginate, benzenesulfonate, benzoate, bisulfate, butyrate, bicarbonate, bitartrate, citrate, camphorate, camphorsulfonate, cyclohexylsulfamate, cyclopentanepropionate, calcium ethylenediamine tetraacetate, dextromethorsulfonate, carbonate, clavulanate (clavulanates), cinnamate, dicarboxylate, digluconate, dodecylsulfonate, dihydrochloride, decanoate, heptanoate, ethane sulfonate, ethylenediamine tetraacetate, ethane disulfonate, propionate dodecylsulfate, ethane sulfonate, fumarate, formate, fluoride, galacturonate, gluconate, glutamate, glycolate, glucarate, glucoheptonate, glycerophosphate, glucoheptonate, paracetamolate, glutarate, glutamate, heptanoate, caproate, hydroxymaleate, hydroxycarboxylic acid, hexylresorcinol, hydroxybenzoate, hydroxynaphthoate, hydrofluoric acid, lactate, lactobionate, laurate, malate, maleate, methylenebis (beta-oxynaphthoate), malonate, mandelate, methanesulfonate, methyl bromide, methyl nitrate, methylsulfonate, monopotassium maleate, mucinate, monocarboxylate, nitrate, naphthalene sulfonate, 2-naphthalene sulfonate, nicotinate, naphthalene sulfonate, N-methyl glucamine, oxalate, caprylate, oleate, pamoate, phenylacetate, picrate, phenylbenzoate, trimethylacetate, propionate, phthalate, phenylacetate, pectate, phenylpropionate, palmitate, pantothenate, polygalacturonate, pyruvate, quinic acid, salicylate, succinate, stearate, sulfanilate, basic acetate, tartrate, theophyllinate, p-toluenesulfonate (tosylate), trifluoroacetate, terephthalate, tannate, theate, trihaloacetate, triethyliodide, tricarboxylate, undecanoate, and valerate.
In one embodiment, the salt may be formed in a conventional manner, for example by reacting the free base or product in the free acid form with one or more suitable acids or equivalents of the 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 ion exchange of the existing salt for another ion or suitable ion exchange resin.
The invention also includes derivatives of selective androgen receptor modulators. The term "derivative" includes, but is not limited to, ether derivatives, acid derivatives, amide derivatives, ester derivatives, and the like. In addition, the present invention also includes hydrates of selective androgen receptor modulators. The term "hydrate" includes, but is not limited to, hemihydrate, monohydrate, dihydrate, trihydrate, and the like.
The invention also includes metabolites of selective androgen receptor modulators. The term "metabolite" means any substance produced by another substance through metabolism or metabolic processes.
The invention also includes pharmaceutical products of selective androgen receptor modulators. The term "pharmaceutical product" refers to a composition suitable for pharmaceutical use (pharmaceutical composition), as defined herein.
The invention also includes prodrugs of selective androgen receptor modulators. The term "prodrug" refers to a substance that can be converted to a biologically active agent in vivo by a reaction such as hydrolysis, esterification, de-esterification, activation, salification, and the like.
The invention also includes crystals of the selective androgen receptor modulator. Furthermore, the present invention provides polymorphs of a selective androgen receptor modulator. The term "crystal" refers to a substance in a crystalline state. The term "polymorph" refers to a particular crystalline state of a substance having particular physical properties such as X-ray diffraction, IR spectrum, melting point, and the like.
In one embodiment of the present invention, there is provided a method for treating a subject having breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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 the breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having metastatic breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the subject's metastatic breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having refractory breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the refractory breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the AR positive breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In one embodiment, the AR positive breast cancer is ER, PR and HER2 positive. In another embodiment, the AR positive breast cancer is ER, PR and HER2 negative. In one embodiment, the AR positive breast cancer is ER positive and is PR and HER2 negative. In another embodiment, the AR positive breast cancer is ER and PR positive and HER2 negative. In another embodiment, the AR positive breast cancer is ER and HER2 positive and is PR negative. In yet another embodiment, the AR positive breast cancer is ER negative and PR and HER2 positive. In another embodiment, the AR positive breast cancer is ER and PR negative and HER2 positive. In yet another 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.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive refractory breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the subject's AR positive refractory breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive metastatic breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the subject's AR positive metastatic breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having ER positive breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the ER positive breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR positive and ER positive breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the subject's AR positive metastatic breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having ER-positive refractory breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the subject's ER-positive refractory breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having ER-positive metastatic breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the subject's ER-positive metastatic breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In one embodiment, ER positive breast cancer is AR positive. In another embodiment, ER positive breast cancer is AR negative.
In another embodiment of the present invention, there is provided a method for treating a subject having advanced breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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 the advanced breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having AR-positive and ER-positive breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the subject's AR-positive and ER-positive refractory breast cancer. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for treating a subject having AR positive and ER negative breast cancer, comprising administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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 the subject's AR positive and ER negative metastatic breast cancer
Is carried out by a method comprising the steps of. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having triple negative breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the 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 the triple negative breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, a method is provided for treating a subject with breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonist (goserelin), aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), rebaciclovir (Kisqali), abbe' zenio), trazoxiline, ly Luo Xili), axist (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3kα), mTOR inhibitor (ivermectin), polyribopolymerase (PARP) inhibitor (lypatadine (lyazane) or tazoril (tazernia)), human epidermal growth factor 2 (HER), tuyezotinib (tuyezob), anti-bead (Herceptin), anti-tuyered (panaxy) antibody (herception 2), anti-tuyered (panaxy 2) or anti-tuyered (panaxy 2) antibody (panaxy 2) An enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), actlizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC) and/or bevacizumab (Avastin) treatment failure, the method comprising the step of administering to the subject a selective androgen modulator of formula I-XIV of the 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 that failed to treat: selective Estrogen Receptor Modulators (SERMs) (tamoxifen, toremifene, raloxifene), gonadotrophin releasing hormone (GnRH) agonists (goserelin), aromatase Inhibitors (AI) (letrozole, anastrozole, exemestane), fluvalirome, cyclin dependent kinase 4/6 (CDK 4/6) inhibitors (cabonine (Ibrance), rapaminib (Kisqali), abbe-zenio), trazoxiline, ly Luo Xili), axiliziram (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olpataline (Lynparza) or talazopanil (Talzenna)), human growth factor receptor 2 (CDK 4/6) inhibitors (lapatinib, letinib (nerlozenb), dasatib (vic), vicat or vicat), anti-bead anti-tuzumab (Herceptin) or anti-bead anti-tuzumab (Herceptin) anti-HER 2 (Herceptin), anti-bead anti-tuzumab (tuzuelan) (panaxb) or anti-tuzuelan) anti-tuzuelan (HER 2 (panaxb) or anti-tuzuelan) anti-tuzuelan (panaxb) Pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) treatment. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for treating and/or preventing a bone-related event in a subject, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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 a bone-related event in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for increasing the force ratio in a subject, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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 increase the force ratio in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for improving the quality of life of a subject, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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 the quality of life of the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the present invention, there is provided a method for treating, preventing, suppressing or inhibiting metastasis in a subject suffering from breast cancer, the method comprising the step of administering to the subject a selective androgen receptor modulator of formula I-XIV of the present 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. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In another embodiment of the invention, there is provided a method for treating a subject having HER2 positive breast cancer, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the HER2 positive breast cancer in the subject. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In one embodiment, the HER2 positive breast cancer is HER2 positive refractory breast cancer. In another embodiment, 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, HER2 positive breast cancer is AR positive. In another embodiment, HER2 positive breast cancer is AR negative.
In a certain embodiment, HER2 positive breast cancer is ER positive, PR positive and AR positive. In another embodiment, HER2 positive breast cancer is ER positive, PR negative, and AR positive. In another embodiment, HER2 positive breast cancer is ER positive, PR negative, and AR negative. In another embodiment, HER2 positive breast cancer is ER positive, PR positive, and AR negative. In another embodiment, HER2 positive breast cancer is ER negative, PR negative, and AR positive. In another embodiment, HER2 positive breast cancer is ER negative, PR positive, and AR positive. In other embodiments, HER2 positive breast cancer is ER negative, PR positive, and AR negative. In a certain embodiment, HER2 positive breast cancer is ER negative, PR negative and AR negative.
In another embodiment of the invention, there is provided a method for treating a subject having breast cancer that expresses an ER mutant, the method comprising the step of administering to the subject a compound of formula I-XIV of the invention and/or an 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 the subject's breast cancer that expresses an ER mutant. In one embodiment, the subject is a female subject. In another embodiment, the subject is a male subject.
In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a Y537S mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the D351Y mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing an E380Q mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the V422del mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the S432L mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a G442A mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the S463P mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L469V mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536R mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536H mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536P mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the L536Q mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a Y537N mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a Y537C mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the Y537D mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing a D538G mutation. In a certain embodiment, the breast cancer expressing the ER mutant is a breast cancer expressing the E542G mutation. In one embodiment, breast cancer expressing an ER mutant refers to a mutant of ER- α.
In a certain embodiment, the breast Cancer expressing the ER mutant is such as Cancer Cell,2018, volume 33, pages 173-186, or Nat Rev Cancer, month 6 of 2018, volume 18, phase 6: pages 377-388, which are incorporated herein by reference. In one embodiment, breast cancer expressing an ER mutant refers to a mutant of ER- α.
Substituent R is defined herein as alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CH 2 F、CHF 2 、CF 3 、CF 2 CF 3 The method comprises the steps of carrying out a first treatment on the surface of the Aryl, phenyl, halogen, alkenyl Or Hydroxy (OH).
"alkyl" refers to saturated aliphatic hydrocarbons, including straight, branched, and cyclic alkyl groups. In one embodiment, the alkyl group has 1 to 12 carbons. In another embodiment, the alkyl group has 1 to 7 carbons. In another embodiment, the alkyl group has 1 to 6 carbons. In another embodiment, the alkyl group has 1 to 4 carbons. The alkyl group may be unsubstituted or substituted with one or more groups selected from: halogen, hydroxy, alkoxycarbonyl, amido, alkylamido, dialkylamido, nitro, amino, alkylamino, dialkylamino, carboxyl, thio or thioalkyl.
"haloalkyl" refers to an alkyl group as defined herein substituted with one or more halogen atoms, such as F, cl, br or I.
"aryl" refers to an aromatic group having at least one carbocyclic aromatic group or heterocyclic aromatic group, which aryl group may be unsubstituted or substituted with one or more groups selected from the group consisting of: halogen, haloalkyl, hydroxy, alkoxycarbonyl, amido, alkylamido, dialkylamido, nitro, amino, alkylamino, dialkylamino, carboxy or thio or thioalkyl. Non-limiting examples of aromatic rings are phenyl, naphthyl, pyranyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyrazolyl, pyridinyl, furanyl, thienyl, thiazolyl, imidazolyl, isoxazolyl, and the like.
"hydroxy" refers to an OH group. "alkenyl" refers to a group having at least one carbon-carbon double bond. Halogen groups refer to F, cl, br or I.
"arylalkyl" refers to an alkyl group bonded to an aryl group, wherein alkyl and aryl are as defined above. An example of an aralkyl group is benzyl.
Bioactivity of selective androgen receptor modulators
Selective androgen receptor modulators provided herein are a novel class of compounds that inhibit the growth of AR positive breast cancer. The compounds of the invention have tissue-selective myoanabolic activity characteristics of non-steroidal ligands for the androgen receptor. Furthermore, the compounds of the present invention are non-aromatic, non-maleated, and generally do not cross-react with ER and PR. Furthermore, in one embodiment, the Selective Androgen Receptor Modulators (SARMs) of the present invention are beneficial to refractory breast cancer patients undergoing chemotherapy due to anabolism.
As contemplated herein, appropriately substituted selective androgen receptor modulators of the present invention may be used: a) Treating a subject having breast cancer; b) Treating a subject having metastatic breast cancer; c) Treating a subject having refractory breast cancer; d) Treating a subject having AR positive breast cancer; e) Treating a subject having AR positive refractory breast cancer; f) Treating a subject having AR positive metastatic breast cancer; g) Treating a subject having AR-positive and ER-positive breast cancer; h) Treating a subject with AR positive breast cancer with or without ER, PR and/or HER2 expression; i) Treating a subject having triple negative breast cancer; j) Treating a subject having advanced breast cancer; k) Treating a subject having a breast cancer employing a Selective Estrogen Receptor Modulator (SERM) (tamoxifen, toremifene, raloxifene), a gonadotropin releasing hormone (GnRH) agonist (goserelin), an Aromatase Inhibitor (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitor (palbociclib (Ibrance), raplocillin (kiquali), abbe-li (Vorzenio), trazoxiline, lycra Luo Xili), abapelisine (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), an mTOR inhibitor (everolimus), a poly ADP-ribose polymerase (PARP) inhibitor (olaparnib (lynparar) or taziram), a human epidermal growth factor receptor 2 (HER 2) kinase inhibitor (lapatinib, nepafenib (neomycin), a (guanoxydant) or an anti-bead anti-tuzumab (Herceptin), an anti-bead anti-tuzumab (panaxomb) or an anti-tuzumab (HER 2) antibody (HER) or an anti-tuzuelan antibody (HER) Atilizumab (tecentiq) (PD-L1 blocking antibody), pembrolizumab (Keytruda) (PD-L1 blocking antibody), golian Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) failed treatment; l) treating a subject with ER positive breast cancer; m) treating, preventing, suppressing or inhibiting metastasis in a subject having breast cancer; n) extending the survival of a subject suffering from breast cancer; o) slowing the progression of breast cancer in the subject; p) extending progression free survival of a subject having breast cancer; q) treating HER2 positive breast cancer; r) treating breast cancer expressing an ER mutant, and/or s) treating a subject with breast cancer expressing a Y537SER mutant.
In one embodiment, a "refractory breast cancer" is a breast cancer that does not respond to treatment. In another embodiment, a "refractory breast cancer" is a breast cancer that is resistant to treatment. In one embodiment, the refractory breast cancer is refractory metastatic breast cancer. In one embodiment, refractory breast cancer pairs employ anthracyclines, taxanes, capecitabine, ixabepilone, selective Estrogen Receptor Modulators (SERM) (inhibitors of tamoxifen, toremifene, raloxifene), gonadotropin releasing hormone (GnRH) agonists (goserelin), aromatase Inhibitors (AI) (letrozole, anastrozole, exemestane), fulvestrant, cyclin dependent kinase 4/6 (CDK 4/6) inhibitors (Paboscalid), rapaminib (Kisqali), abeli (Vorzeno), trazoxiline, lei Luo Xili), apilimus (Piqray) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitors (Evidin), poly ribose polymerase (PARP) inhibitors (Olapazamide (Lynpzazamide) or Talazol (Talrzenna) human epidermal growth factor 2), HER (HER 4/6) inhibitors (Paenib), anti-tuzandrazotinib (HER), anti-bead (HER), anti-tuzandrazotinib (HER) or anti-tussa antibody (HER) (HER 2), anti-bead (HER 2) or anti-bead (HER 2) Enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), actlizumab (Tecentriq) (PD-L1 blocking antibody), pembrolizumab (Kettuda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) treatment, or any combination thereof, is non-reactive.
In one embodiment, a "triple negative breast cancer" is defined as a lack of expression of estrogen, progesterone, and ErbB2 (also known as human epidermal growth factor receptor 2 (HER 2)) receptors. This subtype accounts for 15% of all types of breast cancer. This subtype of breast cancer is clinically characterized as more aggressive, less responsive to standard treatment, and is associated with a poor overall prognosis for the patient.
In one embodiment, the methods of the invention relate to treating a subject with AR positive breast cancer, regardless of grade, stage, or previous treatment.
In one embodiment, the methods of the invention relate to treating a subject having HER2 positive breast cancer, regardless of grade, stage, or previous treatment.
In one embodiment, the method of the invention is a first-line, second-line, third-line or fourth-line treatment of breast cancer. First line treatment refers to medical treatment recommended for the initial treatment of a disease, sign or symptom. The second line treatment is administered when the initial treatment (first line treatment) is inactive or ceases to be active. Three-wire therapy is administered when neither the initial therapy (first-line therapy) nor the subsequent therapy (second-line therapy) is active or ceases to be active, or the like.
As used herein, a "kinase" is a group of enzymes that catalyze the transfer of phosphate groups from a donor (such as ADP or ATP) to an acceptor. In one embodiment, phosphorylation results in functional changes in the target protein (substrate) by altering enzyme activity, cellular localization, or binding to other protein kinases. Kinases regulate most cellular pathways, particularly those involving signal transduction. In one embodiment, deregulated kinase activity is a common cause of diseases, particularly cancer, in which kinase regulation controls many aspects of cell growth, movement and death. In one embodiment, the drug that inhibits a particular kinase is used to treat kinase related diseases, including cancer. In one embodiment, HER2 positive breast cancer is sensitive to HER2 kinase inhibitors (e.g., trastuzumab and lapatinib), and is commonly used for metastatic disease. However, some breast cancers are refractory to HER2 kinase inhibitor treatment.
As used herein, the receptors for extracellular signaling molecules are collectively referred to as "cell signaling receptors". Many cell signaling receptors are transmembrane proteins on the cell surface; when they bind to extracellular signaling molecules (i.e., ligands), they are activated, thereby producing intracellular signaling cascades that alter cellular behavior. In contrast, in some cases, the receptors are intracellular and the signaling ligands must enter the cell to activate them; thus, these signaling molecules must be small enough 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 invention relates to androgen receptors. In one embodiment, the invention relates to androgen receptor agonists. In one embodiment, the invention relates to progesterone receptors. In one embodiment, the invention relates to progesterone receptor antagonists.
In addition to ligand binding to the receptor, the receptor may be blocked to prevent ligand binding. When a substance binds to a recipient, the three-dimensional structure of the substance fits into the space created by the three-dimensional structure of the recipient in a ball-and-socket configuration. The better the ball fits the socket, the tighter the ball is held. This phenomenon is called affinity. If one substance has a greater affinity than the original hormone, it competes with the hormone and binds to the binding site more frequently. Once bound, a signal may be sent into the cell through the receptor, causing the cell to react in some way. This is called activation. Upon activation, the activated receptor directly modulates transcription of a particular gene. But the substance and receptor may have certain properties other than affinity in order to activate the cell. Chemical bonds may be formed between atoms of the substance and atoms of the acceptor. In some cases, this results in a change in receptor configuration that is sufficient to initiate the activation process (known as signal transduction).
In one embodiment, the compounds of the invention inhibit intratumoral expression of genes and pathways that promote breast cancer progression through their effect on AR. In one embodiment, the compounds of the invention inhibit intratumoral expression of Muc1, SLUG, VCAM1, SPARC or MMP2 or any combination thereof. In another embodiment, formula VIII inhibits gene expression that promotes breast cancer.
In one embodiment, the receptor antagonists are substances that bind to receptors and inactivate them. In one embodiment, a selective androgen receptor modulator is a molecule that exhibits in vivo tissue selectivity that activates signaling activity of the Androgen Receptor (AR) to a greater extent in anabolic (muscle, bone, etc.) tissue than in androgenic tissue. Thus, in one embodiment, the selective androgen receptor modulators of the present invention may be used to bind to and activate steroid hormone receptors. In one embodiment, the SARM compounds of the present invention are agonists that bind to the androgen receptor. In another embodiment, the compound has a high affinity for the androgen receptor.
Assays to determine whether a compound of the invention is an AR agonist or antagonist are well known to those of skill in the art. For example, AR agonistic activity can be determined by monitoring the ability of a selective androgen receptor modulator to maintain and/or stimulate growth of AR-containing androgen tissues (such as the prostate and seminal vesicles) in a castrated animal, as measured by weight. The AR antagonistic activity can be determined by monitoring the ability of a selective androgen receptor modulator to inhibit the growth of AR-containing tissue in whole animals or to combat testosterone effects in castrated animals.
Androgen Receptor (AR) is an androgen receptor of any species (e.g., mammalian). In one embodiment, the androgen receptor is a human androgen receptor. Thus, in another embodiment, the selective androgen receptor modulator binds reversibly to a human androgen receptor. In another embodiment, the selective androgen receptor modulator reversibly binds to an androgen receptor of a mammal.
As contemplated herein, in one embodiment, the term "selective androgen receptor modulator" (SARM) is a molecule that exhibits in vivo tissue selectivity that activates signaling activity of an androgen receptor to a greater extent in anabolic (muscle, bone, etc.) tissue than in androgen tissue. In another embodiment, the selective androgen receptor modulator selectively binds to an androgen receptor. In another embodiment, the selective androgen receptor modulator selectively affects signaling through an androgen receptor. In one embodiment, the SARM is a partial agonist. In one embodiment, the SARM is a tissue-selective agonist, or in some embodiments, a tissue-selective antagonist.
In one embodiment, the SARM of the present invention exerts its effect on the androgen receptor in a tissue-dependent manner. In one embodiment, the SARM of the present invention will have an IC for AR 50 Or EC50, as determined using AR transactivation assays known in the art, or in other embodiments as described herein.
In some embodiments, the term "IC 50 "refers to SARM concentration that reduces the activity of a target (e.g., AR) to a half maximum level.
In some embodiments, the term "EC 50 "refers to the concentration of SARM that produces half maximal effect.
For example, using a transactivation assay, FIG. 5 shows that compounds of the invention exhibit AR agonist activity in MDA-MB-231 cells transfected with AR.
As defined herein, "contacting" refers to introducing a selective androgen receptor modulator of the present invention into a sample containing the receptor in a tube, flask, tissue culture, chip, array, plate, microplate, capillary, etc., and incubating at a temperature and for a time sufficient to allow the selective androgen receptor modulator to bind to the receptor. Methods of contacting a sample with a selective androgen receptor modulator or other specific binding component are known to those skilled in the art and can be selected according to the type of assay protocol to be performed. The incubation method is also standard and known to those skilled in the art.
In another embodiment, the term "contacting" refers to introducing a selective androgen receptor modulator of the present invention into a subject undergoing treatment and contacting the selective androgen receptor modulator with the androgen receptor in vivo.
As used herein, the term "treatment" includes disease modifying treatment. As used herein, the terms "reduce," "inhibit," and "inhibit" have their commonly understood meaning of alleviation or reduction. As used herein, the term "progression" refers to an increase in scope or severity, development, growth, or worsening. As used herein, the term "recurrence" refers to the recurrence of a disease after remission. As used herein, the term "delay" refers to stopping, impeding, slowing, deferring, blocking, or reversing. As used herein, the term "metastasis" refers to the transfer of a disease from one organ or portion thereof to another organ or portion thereof that is not directly connected thereto. Metastasis can occur, for example, due to malignant cells being transferred from one organ (e.g., breast) to another organ.
In one embodiment, "treatment" refers to reducing tumor growth by 75%, as demonstrated, for example, in example 8. In another embodiment, treatment refers to reducing tumor growth by at least 75%. In another embodiment, treatment refers to reducing tumor growth by at least 50%. In another embodiment, treatment refers to reducing tumor growth by at least 25%. In another embodiment, treatment refers to a 50% -100% reduction in tumor growth. In another embodiment, treatment refers to a reduction in tumor growth by 70% -80%. In another embodiment, treatment refers to a reduction in tumor growth by 25% -125%.
In another embodiment, "treatment" refers to a 50% reduction in tumor weight, as demonstrated, for example, in example 8. In another embodiment, treatment refers to reducing tumor weight by at least 50%. In another embodiment, treatment refers to reducing tumor weight by at least 40%. In another embodiment, treatment refers to reducing tumor weight by at least 30%. In another embodiment, treatment refers to reducing tumor weight by at least 20%. In another embodiment, treatment refers to a 25% -75% reduction in tumor weight. In another embodiment, treatment refers to a 25% -100% reduction in tumor weight.
As used herein, the term "administering" refers to contacting an individual with a compound of the present invention. As used herein, administration can be accomplished in vitro, i.e., in a test tube, or in vivo, i.e., in cells or tissues of a living organism (e.g., a human). In one embodiment, the invention encompasses the administration of a compound of the invention to a subject.
In one embodiment, the compounds of the invention are administered to a subject weekly. In another embodiment, the compounds of the invention are administered to a subject twice weekly. In another embodiment, the compounds of the invention are administered to a subject three times per week. In another embodiment, the compounds of the invention are administered to a subject four times per week. In another embodiment, the compounds of the invention are administered to a subject five times per week. In another embodiment, the compounds of the invention are administered to a subject daily. In another embodiment, the compounds of the invention are administered to a subject weekly. In another embodiment, the compounds of the invention are administered to a subject once every two weeks. In another embodiment, the compounds of the invention are administered to a subject monthly.
In one embodiment, the methods of the invention comprise administering a selective androgen receptor modulator as the sole active ingredient. However, methods for hormone therapy, for treating breast cancer, for delaying progression of breast cancer, and for preventing and treating breast cancer recurrence and/or breast cancer metastasis are also contemplated within the scope of the invention, including administration of a selective androgen receptor modulator in combination with one or more therapeutic agents. These agents include, but are not limited to: 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 (palbocicle), rebacicenib (Kisqali), arbelii (Vorzenio), trazoyside, len Luo Xili), abaricilast (Piqray)) (inhibitors of phosphatidylinositol-3-kinase subunit alpha (PI 3K alpha)), mTOR inhibitors (everolimus), poly ADP Ribose Polymerase (PARP) inhibitors (olaparib (Lynparza) or talazopania (Talzenna)), human epidermal growth factor receptor 2 (HER 2) kinase inhibitors (lapatinib, lenatinib (Nerlynx), dacatinib (Vizimpro) or tukatinib (Tukysa)), HER2 antibodies (trastuzumab (Herceptin), pertuzumab (Perjeta), megnetuzumab (Margenza)), HER2 antibody drug conjugates (HER 2 ADC) (detrastuzumab (Enhertu), enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zxf (pheztgo)), attitum (teconiq) (PD-L blocking antibodies), prasugrel-L1 (tuzotinib) (tukuda), anti-HDAC (HDAC), anti-HDAC (prasugrel) and anti-tumor drugs (HDAC) or anti-prasugrel antibodies (prasugrel) and (prasugrel) drugs (HDAC) or (HDAC) and (visliquamide inhibitors (HDAC) or (visuquamide inhibitors (3-visuja) or their vaccine or anti-toxin inhibitors (HDAC) CTLA-4 (cytotoxic T lymphocyte-associated protein-4) inhibitors (trimemumab), PD-1 inhibitors (pembrolizumab), PD-L1 inhibitors (atenolizumab, avilamab, dulvalli You Shan antibody), chemotherapeutic agents, taxanes, anthracyclines, epothilones, LHRH analogs, reversible anti-androgens, antiestrogens, anticancer agents, 5-alpha reductase inhibitors, progestins, agents that act through other nuclear hormone receptors such as progesterone and estrogen receptors, estrogens, progestins, PDE5 inhibitors, apomorphine, bisphosphonates, growth factor inhibitors (such as those that inhibit VEGF, IGF, etc.), or one or more additional Selective Androgen Receptor Modulators (SARMs).
Additional therapeutic agents that may be administered in combination with the selective androgen receptor modulator compounds of the present invention include, but are not limited to: abeli, a process for preparing,(methotrexate), ->(paclitaxel albumin stabilized nanoparticle formulation), enmetrastuzumab, adriamycin PFS (doxorubicin hydrochloride), adriamycin RDF (doxorubicin hydrochloride),>(fluorouracil),>(everolimus), apilimus, anastrozole, and +>(anastrozole),>(exemestane), velumab, atilizumab, bicalutamide, bupirimate,/i >(pegylated liposomal doxorubicin), capecitabine, carboplatin, cisplatin,/-and->(cyclophosphamide), cyclophosphamide,(cyclophosphamide), docetaxel, doxorubicin hydrochloride, dulcitol You Shan, and->(fluorouracil),(epirubicin hydrochloride), entinostat, enzalutamide, epirubicin hydrochloride, eribulin, ethinyl estradiol, everolimus,>(raloxifene), exemestane, < >>(toremifene),>(fulvestrant),(letrozole),>(5-fluorouracil), fluorouracil, fluoxytestosterone, < >>(methotrexate), ->(methotrexate), fulvestrant, gemcitabine hydrochloride,/o>(gemcitabine hydrochloride), ->(eribulin mesylate), a->(trastuzumab), ixabepilone, < ->(ixabepilone), lapatinib ditosylate, letrozole, megestrol acetate, methotrexate LPF (methotrexate), a,(methotrexate), ->(methotrexate), ->(cyclophosphamide), ->(nelipepimut-S)、/>(citric acid tamoxifen), paclitaxel albumin stable nanoparticle formulations, palbociclib, pembrolizumab,/->(pertuzumab), pertuzumab,>(apirism), raloxifene, rebaudinib, tamoxifen citrate, tasselisib, < > >(paclitaxel),>(docetaxel), trastuzumab, tiuximab, toremifene, and/or->(lapatinib ditosylate), vinorelbine and(capecitabine).
Thus, in one embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and a selective estrogen receptor modulator. Thus, in one embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and selective estrogen receptor reductionA lytic agent (fulvestrant). Thus, in one embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and a CDK4/6 inhibitor (palbociclib, rebabociclib, abbe-cilib, treacli, lei Luo Xili). Thus, in one embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and a PIK3A inhibitor (apicalist, bupirimate, tapeelinib). Thus, in one embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and a HER2 inhibitor (lapatinib, trastuzumab, lenatinib). Thus, in one embodiment, the methods of the invention comprise the combined administration of Sub>A selective androgen receptor modulator and Sub>A VEGF-Sub>A inhibitor (bevacizumab). Thus, in one embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and a chemotherapeutic agent. In one embodiment, the chemotherapeutic agent is a taxane. In another embodiment, the chemotherapeutic agent is an anthracycline. In one embodiment, the chemotherapeutic agent is epothilone (ixabepilone). Thus, in one embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and an LHRH analog. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and a reversible anti-androgen. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and an antiestrogen. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and an anticancer drug. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and a 5-alpha reductase inhibitor. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and an aromatase inhibitor. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and a progestogen. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and an agent that acts through other nuclear hormone receptors. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and a Selective Estrogen Receptor Modulator (SERM). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with a progestogen or an antiprogestin. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator and an estrogen. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and a PDE5 inhibitor. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and apomorphine. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and a bisphosphonate (pamidronate, zoledronic acid). In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and denomabIn another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and a growth factor inhibitor. In another embodiment, the methods of the invention comprise administering a selective androgen receptor modulator of the invention in combination with one or more additional Selective Androgen Receptor Modulators (SARMs).
In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and(methotrexate). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(paclitaxel albumin stabilized nanoparticle formulation). In another embodiment, the present inventionThe method comprises administering a selective androgen receptor modulator of the present invention in combination with enmetrastuzumab. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(doxorubicin hydrochloride). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and Adriamycin>(doxorubicin hydrochloride). In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and(fluorouracil). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(everolimus). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with anastrozole. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +. >(anastrozole). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(exemestane). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with capecitabine. In another embodiment, the methods of the invention comprise administering the present invention in combinationInventive selective androgen receptor modulators and +.>(cyclophosphamide). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with cyclophosphamide. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(cyclophosphamide). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with docetaxel. In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with doxorubicin hydrochloride. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +. >(fluorouracil). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(epirubicin hydrochloride). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with epirubicin hydrochloride. In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with everolimus. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and exemestane. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and(support)Remiprofen). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(raloxifene). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(fulvestrant). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +. >(letrozole).
In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and(5-fluorouracil). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with fluorouracil. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(methotrexate). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and Folex->(methotrexate). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with fulvestrant. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and gemcitabine hydrochloride.In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(gemcitabine hydrochloride). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +. >(trastuzumab). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and Ibrance (palbociclib). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with Kisquari (Rabociclib). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and Verenzio (abenzli). In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with trazoyside. In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with l Luo Xili. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and ixabepilone. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(ixabepilone). In another embodiment, the method of the invention comprises administering the selective androgen receptor modulator of the invention in combination with lapatinib xylene sulfonate. In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with letrozole. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and methotrexate. In another embodiment, the present The inventive method comprises the combined administration of a selective androgen receptor modulator of the invention and methotrexate LPF (methotrexate). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(methotrexate). In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and(methotrexate). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(cyclophosphamide). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(tamoxifen citrate). In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and paclitaxel. In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and a paclitaxel albumin stabilized nanoparticle formulation. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +. >(pertuzumab). In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and pertuzumab. In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with tamoxifen citrate. In a further embodiment of the present invention,the method of the invention comprises the combined administration of the selective androgen receptor modulator of the invention and +.>(paclitaxel). In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(docetaxel). In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and trastuzumab. In another embodiment, the method of the invention comprises administering a selective androgen receptor modulator of the invention in combination with romifene. In another embodiment, the method of the invention comprises the combined administration of a selective androgen receptor modulator of the invention and +.>(lapatinib ditosylate). In another embodiment, the methods of the invention comprise the combined administration of a selective androgen receptor modulator of the invention and (capecitabine).
In one embodiment, the methods of the invention comprise administering a pharmaceutical composition (or pharmaceutical formulation, used interchangeably herein) comprising a selective androgen receptor modulator of the 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.
Pharmaceutical composition
As used herein, "pharmaceutical composition" refers to a therapeutically effective amount of a selective androgen receptor modulator, as well as suitable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers. As used herein, "therapeutically effective amount" means that the amount is givenThe condition and the amount of therapeutic effect provided by the administration regimen. Such compositions are liquid or lyophilized or otherwise dried formulations and include various buffer contents (e.g., tris-HCl, acetate, phosphate), pH and ionic strength diluents, additives for preventing adsorption to surfaces such as albumin or gelatin, detergents (e.g., tweenBile acid salts), solubilizing agents (e.g., glycerol, polyglycol glycerol), antioxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., +. >Benzyl alcohol, parabens), bulking substances or tonicity modifiers (e.g., lactose, mannitol), covalent attachment of polymers such as polyethylene glycol to proteins, complexation with metal ions, or incorporation of the material into or onto particulate formulations of polymeric compounds such as polylactic acid, polyglycolic acid, hydrogels, etc., or onto liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroplasts. Such compositions will affect physical state, solubility, stability, in vivo release rate, and in vivo clearance rate. Controlled or sustained release compositions include formulations in lipophilic depots (e.g., fatty acids, waxes, oils).
In one embodiment, a pharmaceutical composition comprising a compound of the invention is used in a method of the invention at a dose of 1mg to 50mg of the compound of the invention. In another embodiment, the dose is 1mg, 3mg, 9mg, 10mg, 18mg or 30mg. In another embodiment, a pharmaceutical composition comprising a compound of the invention uses a 1mg dose of a compound of the invention in a method of the invention. In another embodiment, a pharmaceutical composition comprising a compound of the invention is used in a method of the invention at a dose of 3mg of the compound of the invention. In another embodiment, a pharmaceutical composition comprising a compound of the invention uses a 9mg dose of a compound of the invention in a method of the invention. In another embodiment, a pharmaceutical composition comprising a compound of the invention uses a 10mg dose of a compound of the invention in a method of the invention. In another embodiment, a pharmaceutical composition comprising a compound of the invention uses a dose of 18mg of a compound of the invention in a method of the invention. In another embodiment, a pharmaceutical composition comprising a compound of the invention is used in a method of the invention at a dose of 30mg of the compound of the invention.
The invention also includes particulate compositions coated with a polymer (e.g., poloxamer or poloxamer). Other embodiments of the compositions of the present invention include protective coatings in particulate form, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral. In one embodiment, the pharmaceutical composition is administered parenterally, paracancerous, transmucosally, transdermally, intramuscularly, intravenously, intradermally, subcutaneously, intraperitoneally, intraventricularly, intravaginally, intracranially, and intratumorally.
Furthermore, as used herein, "pharmaceutically acceptable carriers" are well known to those skilled in the art and include, but are not limited to, 0.01M-0.1M and preferably 0.05M phosphate buffer or about 0.8% saline. In addition, such pharmaceutically acceptable carriers can 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, alcohol/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 nutritional supplements, electrolyte supplements (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, finishing agents, inert gases and the like.
Controlled or sustained release compositions include formulations in lipophilic depots (e.g., fatty acids, waxes, oils). The invention also includes particulate compositions coated with a polymer (e.g., poloxamer or poloxamer) and particulate compositions of compounds coupled to antibodies directed against or to tissue specific receptors, ligands or antigens.
Other embodiments of the compositions of the present invention include protective coatings in particulate form, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral.
Compounds modified by covalent attachment of water-soluble polymers such as polyethylene glycol, copolymers of polyethylene glycol and polypropylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinylpyrrolidone or polyproline are known to exhibit significantly longer half-lives in blood after intravenous injection than the corresponding unmodified compounds (Abuchowski et al, 1981; newmark et al, 1982; and Katre et al, 1987). Such modifications can also increase the solubility of the compound in aqueous solutions, eliminate aggregation, enhance the physical and chemical stability of the compound, and greatly reduce the immunogenicity and reactivity of the compound. Thus, by administering such polymer-compound adducts less frequently or in lower doses than unmodified compounds, the desired in vivo biological activity can be achieved.
In another embodiment, the pharmaceutical composition may be delivered in a controlled release system. For example, the agent may be administered using intravenous infusion, implantable osmotic pumps, transdermal patches, liposomes, or other modes of administration. In one embodiment, a pump (see Langer, supra; sefton, CRC Crit. Ref. Biomed. Eng., volume 14: page 201 (1987)), buchwald et al, surgery, volume 88: page 507 (1980), saudek et al, N.Engl. J. Med., volume 321: page 574 (1989)) may be used, in another embodiment, a polymeric material may be used, in another embodiment, the controlled release system may be placed in proximity to the therapeutic target (i.e., brain) and thus requires only a portion of the systemic dose (see, e.g., goodson, medical Applications of Controlled Release, supra, volume 2, pages 115-138 (1984)), other controlled release systems are discussed in Langer's review (Science, volume 249: pages 1527-1533).
The pharmaceutical formulations may contain the selective androgen receptor modulator alone or may further contain a pharmaceutically acceptable carrier, and may be in solid or liquid form such as tablets, powders, capsules, pills, 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. Pharmaceutical formulations containing selective androgen receptor modulators may be administered to a subject by means of, for example, subcutaneously implanted pills; in another embodiment, the bolus provides controlled release of the selective androgen receptor modulator over a period of time. The formulations may also be administered by intravenous, intra-arterial or intramuscular injection of liquid formulations, oral administration of liquid or solid formulations, or by topical administration. Administration may also be accomplished through the use of rectal suppositories or urethral suppositories.
The pharmaceutical formulations of the present invention may be prepared by known dissolution, mixing, granulation or tablet formation methods. For the administration by oral administration,
the selective androgen receptor modulator or its physiologically tolerable derivative such as a salt, ester, N-oxide, etc. is admixed with additives conventionally used for this purpose such as vehicles, stabilizers or inert diluents and converted by conventional means into a suitable administration form such as tablets, coated tablets, hard or soft gelatin capsules, aqueous, alcoholic or oily solutions. Examples of suitable inert vehicles are conventional tablet bases such as lactose, sucrose or corn starch; with binders such as acacia, corn starch, gelatin; with disintegrating agents, such as corn starch, potato starch, alginic acid; or in combination with a lubricant such as stearic acid or magnesium stearate.
Examples of suitable oily vehicles or solvents are vegetable or animal oils, such as sunflower oil or cod liver oil. The formulation may be in the form of dry granules and wet granules. For parenteral administration (subcutaneous, intravenous, intraarterial or intramuscular injection), the selective androgen receptor modulators or their physiologically tolerable derivatives such as salts, esters, N-oxides, etc. are converted into solutions, suspensions or emulsions, if desired, with substances customary and suitable for this purpose, for example solubilizers or other auxiliaries. Examples are sterile liquids such as water and oils, with or without the addition of surfactants and other pharmaceutically acceptable adjuvants. Illustrative oils are those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil or mineral oil. In general, water, physiological saline, dextrose and related aqueous sugar solutions, and glycols such as propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions.
The preparation of pharmaceutical compositions containing active ingredients is well known in the art. Such compositions may be prepared as aerosols of the active ingredient for delivery to the nasopharynx, or as injections, as liquid solutions or suspensions; however, solid forms suitable for dissolution or suspension in a liquid prior to injection may also be prepared. The preparation can also be emulsified. The active therapeutic ingredient is typically admixed with excipients that 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.
In addition, the composition may contain minor amounts of auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, which enhance the effectiveness of the active ingredient.
The active ingredient may be formulated into the composition in the form of a neutralized pharmaceutically acceptable salt. Pharmaceutically acceptable salts include acid addition salts (formed with free amino groups of the polypeptide or antibody molecule) with inorganic acids such as hydrochloric or phosphoric acids or organic acids such as acetic, oxalic, tartaric, mandelic, and the like. Salts formed from the free carboxyl groups may also be derived from inorganic bases such as sodium, potassium, ammonium, calcium or ferric hydroxides, and organic bases such as isopropylamine, trimethylamine, 2-ethylaminoethanol, histidine, procaine and the like.
For topical application to body surfaces using, for example, creams, gels, drops, and the like, selective androgen receptor modulators or their physiologically tolerable derivatives such as salts, esters, N-oxides, and the like are prepared and administered as solutions, suspensions, or emulsions in physiologically acceptable diluents, with or without a pharmaceutical carrier.
In another embodiment, the active compounds can be delivered in vesicles, in particular liposomes (see Langer, science, volume 249: pages 1527-1533 (1990); treat et al, "Liposomes in the Therapy of Infectious Disease and Cancer", lopez-Berestein and Fidler, eds., lists, new York, pages 353-365 (1989); lopez-Berestein, supra, pages 317-327; see generally supra).
For pharmaceutical use, the salt of the selective androgen receptor modulator will be a pharmaceutically acceptable salt. However, other salts may be used to prepare the compounds of the invention or pharmaceutically acceptable salts thereof. Suitable pharmaceutically acceptable salts of the compounds of the invention include acid addition salts, which may be formed, for example, by mixing a solution of the compound of the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, methanesulfonic acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
In one embodiment, the term "about" refers to a deviation from the indicated number or range of numbers between 0.0001% -5%. In one embodiment, the term "about" refers to a deviation from the indicated number or range of numbers of between 1% -10%. In one embodiment, the term "about" refers to a deviation of up to 25% from the indicated number or range of numbers.
In some embodiments, the term "comprising" or grammatical forms thereof is meant to include a specified active agent, such as a compound of the present invention, as well as including other active agents, and pharmaceutically acceptable carriers, excipients, emollients, stabilizers, and the like, as known in the pharmaceutical industry. In some embodiments, the term "consisting essentially of … …" means that the only active ingredient of the composition is the indicated active ingredient, however, other compounds may be included for stabilizing, preserving, etc., the formulation, but not directly related to the therapeutic effect of the indicated active ingredient. In some embodiments, the term "consisting essentially of … …" may refer to a component that exerts a therapeutic effect via a different mechanism than the indicated active ingredient. In some embodiments, the term "consisting essentially of … …" may refer to a component that exerts a therapeutic effect and that belongs to a class of compounds different from the indicated active ingredient. In some embodiments, the term "consisting essentially of … …" may relate to components of a class of compounds that exert a therapeutic effect and that belong to a different class of compounds than the indicated active ingredient, which components function by, for example, different mechanisms of action and represent one embodiment of the invention, the polypeptide comprising a T cell epitope present in the composition may be specifically combined with the polypeptide comprising a B cell epitope. In some embodiments, the term "consisting essentially of … …" may refer to a component that facilitates the release of an active ingredient. In some embodiments, the term "consisting of … …" refers to a composition containing an active ingredient and a pharmaceutically acceptable carrier or excipient.
Furthermore, as used herein, the term "comprising" is intended to mean that the system comprises the listed elements, but does not exclude other elements that may be optional. The phrase "consisting essentially of … …" refers to a method that includes the listed elements but excludes other elements that may have a significant impact on the performance of the method. Thus, "consisting of … …" shall mean excluding more than trace amounts of other elements. Embodiments defined by each of these transitional terms are within the scope of the invention.
In one embodiment, the invention provides a combined preparation. In one embodiment, the term "combination preparation" is defined in particular as "kit of parts" in the sense that the combination partners as defined above can be administered independently or by using different fixed combinations with different amounts of the combination partners, i.e. simultaneously, concurrently, separately or sequentially. In some embodiments, the parts of the kit of parts may then be administered, for example, 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. In some embodiments, the ratio of the total amounts of the combination partners may be administered in a combined formulation. In one embodiment, the combined preparation may be varied, for example in order to meet the needs of a patient sub-population to be treated or the needs of a single patient, these different needs may be due to a specific disease, severity of the disease, age, sex or weight, which can be readily made by a person skilled in the art.
In one embodiment, the term "a" means at least one. In one embodiment, the phrase "two or more" may be any naming suitable for a particular purpose. In one embodiment, "about" may include a deviation 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% from the indicated term.
The following examples are presented to more fully illustrate the preferred embodiments of the invention. However, the examples should in no way be construed as limiting the broad scope of the invention.
Experimental details
General experimental method
Cell growth conditions
HCC 1937, HCC 1954, HCC 38, T47D-KBlu, MDA-MB-453 and MDA-MB-231 cells were grown in RPMI-1640 medium containing 2mM L-glutamine supplemented with 10% Fetal Bovine Serum (FBS). 5% CO to maintain cells at 37℃ 2 95% air humidified atmosphere. MCF-7 cells were grown in minimal essential medium supplemented with 10% FBS.
Breast cancer tumors typically express AR 70% -90% of the time, whereas breast cancer cell lines typically do not express AR. This makes the development of preclinical models for studying the effect of androgens on breast cancer very difficult. Thus, AR has been introduced into some breast cancer cell lines used in the following studies by adenovirus infection (stably integrated into the genome).
Sulfonyl Rhodamine B (SRB) assay
SRB assay was used to determine cell numbers during cytotoxicity experiments. The following scheme was used:
1. cells were isolated with 0.25% trypsin.
2. The experimental cultures were grown in 96-well microtiter plates (200. Mu.L of growth medium per well; 1,000-200,000 cells per well).
3. Cultures were fixed with 50. Mu.L of 50% TCA (4 ℃). (see cell immobilization protocol for details).
4. The fixed cells were stained with 50 μl of 0.4% (w/v) SRB in 1% acetic acid for 10 min.
5. SRB was removed and cultures were rinsed 5 times with 1% acetic acid rapidly to remove unbound dye. * *
6. The cultures were air dried overnight until no moisture was visible.
7. The cellular protein bound SRB was lysed with 200. Mu.L of unbuffered Tris base (10 mM, pH 10.5) on a rocking platform for 30 min.
8. Absorbance was read at 540 nm.
* The rinsing process was performed rapidly to prevent desorption of protein-bound SRB by vigorously flicking the plate over the sink to completely remove residual wash solution.
Immobilization of cells attached to a Plastic matrix
The following protocol was used to immobilize cells:
a. mu.L of 50% TCA (4 ℃) was gently spread on top of the growth medium in each well to give a final TCA concentration of 10%.
b. Cultures were incubated at 4℃for 1 hour.
c. The culture was washed 5 times with tap water to remove TCA, growth medium, low molecular weight metabolites and serum proteins.
d. The plates were air dried until no moisture was visible.
Example 1
Effect of formula IX on growth in different breast cancer cell lines expressing androgen receptor
Materials and methodsMethod of
MDA-MB-231 and HCC-38 triple negative breast cancer cells were used to analyze the growth effects of various compounds.
MDA-MB-231 and HCC-38 triple negative breast cancer cells were infected with 200. Mu.L or 500. Mu.L adenovirus containing lacZ (negative control) or AR and treated with various AR ligands (agonists: DHT and formula IX, and antagonists: bicalutamide) or non-AR binders of similar structure to formula IX (R-enantiomer of formula IX). Treatment of cells in activated charcoal-desorbed FBS (FIG. 1C, FIG. 1E, FIG. 1G and FIG. 1I; FIG. 2C, FIG. 2E and FIG. 2G or whole serum (FIG. 1D, FIG. 1F, FIG. 1H and FIG. 1J; FIG. 2D, FIG. 2F and FIG. 2H) for 3 days, fixation and staining with Sulfonyl Rhodamine Blue (SRB) to measure cell viability. IC was calculated 50 Values.
Results
Expression of AR in AR or lacZ infected cells was assessed using western blotting (fig. 1A and 2A).
Only the AR agonist DHT and formula IX inhibited MDA-MB-231 and HCC-38 triple negative breast cancer cell growth (FIG. 1C, FIG. 1D, FIG. 1E, FIG. 1F and FIG. 2C, FIG. 2D, FIG. 2E and FIG. 2F). This inhibition was only observed in the presence of AR (comparison of lacZ-containing and AR-containing). IC of DHT and formula IX in AR-positive cells 50 The values are shown in fig. 1B and 2B.
Example 2
Reversal of the Effect of formula IX on growth
Materials and methods
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 treated with the AR agonist DHT or formula IX in the presence or absence of the AR antagonist bicalutamide. Cell viability was measured by staining cells with activated charcoal-desorbed FBS (figures 3A and 3C or whole serum (figures 3B and 3D for 3 days, fixed and stained with sulforhodamine blue (SRB) & calculate IC 50 Values.
Results
AR is required for both DHT and formula IXInhibiting MDA-MB-231 cell growth, as demonstrated by the attenuated growth inhibition in the presence of bicalutamide (FIGS. 3A-3D). DHT and IC of formula IX in AR positive cells pretreated with or without bicalutamide 50 The values are shown in fig. 3E.
Example 3
Effect of AR ligands on breast cancer cell growth
Materials and methods
To determine if all AR ligands inhibited growth of triple negative breast cancer cells, MDA-MB-231 cells were infected with adenovirus containing lacZ or AR, and with various AR ligands (agonists: DHT, formula VIII, formula IX, formula X, formula XIII, formula XIV; antagonists: bicalutamide) and non-AR binders: treatment of the R-enantiomer of formula IX. Cells were treated in activated charcoal-desorbed FBS (fig. 4A, 4C, 4E, 4G, 4I, 4K, 4M and 4O) or whole serum (fig. 4B, 4D, 4F, 4H, 4J, 4L, 4N and 4P) for 3 days, fixed and stained with Sulfonyl Rhodamine Blue (SRB) to measure cell viability. Calculation of antiproliferative IC in breast cancer cells 50 Values and transactivation values generated in HEK-293 cells, i.e.EC 50 (agonist) values and IC 50 (antagonist) value comparison. The growth regulating properties of these molecules in breast cancer cells were comparable to the transactivation values obtained in HEK-293 cells.
Results
Only AR agonists inhibited the growth of MDA-MB-231 cells (fig. 4A, 4B, 4E-4H and 4K-4P), and the growth inhibitory potential of these ligands was ranked in order of their agonist activity observed in HEK-293 cells (fig. 4Q).
Example 4 also demonstrates that AR agonists inhibit proliferation of MDA-MB-231 cells stably transfected with AR.
Example 4
AR transactivation assay in breast cancer cells
Materials and methods
To ensure that the ligand responsible for the growth inhibitory properties is an agonist in MDA-MB-231 cells, an AR transactivation assay was performed in MDA-MB-231 cells. Although AR transactivation assays are performed in HEK-293 cells, the ability of the ligand to function as an agonist or antagonist depends on the cellular microenvironment. Thus, MDA-MB-231 cells were transfected with lipofectamine and AR, GRE-LUC and CMV-LUC were used as normalization controls. Cells were treated 24 hours after transfection and luciferase assay was performed 48 hours after transfection.
Results
FIG. 5 shows that all AR ligands that elicit antiproliferative activity are agonists and that their agonist and growth inhibitory properties are reasonably good in MDA-MB-231 cells transfected with AR. In other words, in MDA-MB-231 cells transfected with AR, the growth inhibitory ligand is an AR agonist.
Example 5
Analysis of growth inhibition in breast cancer cells expressing estrogen receptors
Materials and methods
To ensure that growth inhibition in MDA-MB-231 cells is selective for AR, and to determine whether ligand-dependent growth inhibition is directed against AR alone, and to ensure that the effect is not an artefact of adenovirus infection, MDA-MB-231 triple negative breast cancer cells are infected with ER- α or ER- β adenovirus constructs, and with ER agonists: 17 beta-estradiol (E2) or ER antagonist: ICI 182,780 (ICI) serum desorbed in activated carbon (FIG. 6C)
Or whole serum (fig. 6D and 6E) for 3 days. Cells were fixed and stained with sulfonylrhodamine blue (SRB) to measure cell viability. Expression of ER in infected cells was assessed using Western blotting.
Results
FIGS. 6A-6B show the presence or absence of ERα or ERβ in MDA-MB-231 cells after transfection. These results indicate that the antiproliferative effect observed with androgens is specific to ligand-activated AR, rather than adenovirus artifacts. Figures 6C-6E show that overexpression of ER- α or ER- β in MDA-MB-231 cells in the presence of ER agonists or antagonists fails to promote growth inhibition. Thus, the growth inhibition observed in MDA-MB-231 cells is selective for the presence of AR and AR agonists.
Example 6
Influence of AR agonists on breast cancer cell morphology
Materials and methods
MDA-MB-231 cells were stably transfected with AR using lentiviruses. Following transfection, cells were treated with DHT or bicalutamide at the indicated concentrations for 3 days. Live cells were observed using an optical microscope and photographed. Cells were imaged under the same magnification and under the same microscopic conditions.
Results
Fig. 7 shows that DHT changes the morphology of MDA-MB-231 cells to more anchorage dependent and differentiated cells, indicating that AR agonist-bound AR-expressing breast cancer cells will be less invasive and migratory (e.g., less likely to metastasize).
DHT and SARM alter the morphology of AR positive MDA-MB-231 cells. MDA-MB-231 cells were stably transfected with AR using lentiviruses and treated with either vehicle or AR agonist at the indicated concentrations. At the end of the 3 days of incubation, the cells were imaged under a microscope (40X).
DHT and SARM, rather than the AR antagonist bicalutamide (data not shown) or inactive isomer of formula IX, altered the morphology of the cells to a more anchorage dependent phenotype (fig. 12).
Example 7
Cross-reactivity of formula VIII with other nuclear hormone receptors
To determine whether the compounds of the invention affect other nuclear hormone receptor signaling, compounds of formula VIII are analyzed for their ability to stimulate (agonists) or inhibit (antagonists) erα, erβ, GR, PR or MR mediated transcriptional activation.
Materials and methods
Instantaneous timeTransfection
Rats GR, MR, PR, ER-alpha and ER-beta were cloned into pCR3.1 vector backbones, respectively. Sequencing was performed to verify that no mutations were present. HEK-293 cells were seeded at 90,000 cells/well in Dulbecco's minimal essential medium supplemented with 5% charcoal-desorbed FBS in 24-well plates. Cells were transfected using Lipofectamine (Invitrogen, carlsbad, calif.) using 0.25 μg GRE-LUC for GR, MR and PR, ERE-LUC for ER- α and ER- β, 0.5ng CMV-LUC (Renilla luciferase) for each receptor and 12.5ng-25ng of the corresponding expression vector. 24 hours after transfection, cells were treated with formula VIII in the absence (agonist mode) and presence (antagonist mode) of known agonists (17β -estradiol for ER; dexamethasone for GR; aldosterone for MR; progesterone for PR) as controls. Luciferase assays were performed 48 hours after transfection. The transcriptional activation values are expressed as firefly luciferases normalized to Renilla luciferases.
Results
The agonist effect of formula VIII on ER-beta, ER-alpha, GR, PR and MR was tested and compared to the activity of known ligands (FIG. 8). The compounds of formula VIII failed to activate either ER- β or ER- α even at the highest tested concentration (1 μm), whereas 1nm 17 β -estradiol induced ER α and ER β mediated transactivation 3-fold and 5-fold, respectively. Compounds of formula VIII failed to activate GR or MR mediated transactivation. The compound of formula VIII does not induce GR or MR mediated transactivation at all concentrations tested, whereas the known ligands (dexamethasone and aldosterone) induce GR or MR activity up to 70-fold and 60-fold, respectively, at concentrations of 1 nM. However, compounds of formula VIII increased trans-activation of PR by a factor of 3 and 8 at 1 μm and 10 μm, respectively. Progesterone activates PR up to 23-fold at 1nM concentration, indicating that the compound of formula VIII is more than 10,000-fold weaker than the endogenous agonist of PR.
Compounds of formula VIII were also tested for their ability to inhibit the effect of known agonists on each of the receptors described above.
Co-incubation of HEK 293 cells with indicated concentrations of formula VIII failed to alter 17 β -estradiol induced ER- β or ER- α activity, dexamethasone induced GR-mediated transactivation or aldosterone induced MR-mediated transactivation.
The dose response curve for the compounds of formula VIII in antagonist mode demonstrated a significant partial inhibition of PR activity (fig. 9). Formula VIII is 10-fold and 100-fold more potent than the R-enantiomer of formula IX, as compared to formula IX. In contrast to RU486, formula VIII is about 1/1,000 of RU486 as a PR antagonist.
The compounds of formulas VIII and IX are specific for AR and do not stimulate or inhibit receptor-mediated transactivation of erα, erβ, GR or MR. Unexpectedly, formula VIII exhibited moderate potent partial agonist activity and potent partial antagonism of PR (see fig. 9). The combined AR agonism and PR antagonism would be beneficial for certain breast cancers (e.g., PR positive breast cancers).
Example 8
Formula VIII and formula IX inhibit triple negative breast cancer cell tumor growth in mice
Materials and methods
MDA-MB-231-AR triple negative breast cancer cells (2 million cells/mouse; MDA-MB-231 cells stably transfected with AR using lentiviruses) were mixed with matrigel (1:1) and subcutaneously injected into the flanks of intact female nude mice (n=5/group). When the tumor reaches 150mm 3 -200mm 3 At this time, the animals were divided into two groups, one group receiving vehicle and the other group receiving 30mg/kg of formula VIII orally. Tumor volumes were measured three times per week and% Tumor Growth Inhibition (TGI) was calculated. At the end of 35 days of treatment, animals were sacrificed, tumors were resected, weighed and collected for various analyses. Blood was collected and serum was isolated for drug concentration measurement.
Results
Formula VIII significantly reduced tumor growth with a TGI of about 75% (fig. 10B). By treatment with formula VIII, tumor weight was also reduced by more than 50% (fig. 11, right panel), tumor size was also reduced by more than 50% (fig. 11, left panel (mm) 3 ) And center graph (% change)). Formula VIII caused these results without any associated toxicity or weight change (fig. 10A). Uterine weight also increased in response to treatment of formula VIII (not shown), indicatingIn vivo androgen response.
The results, as shown in figure 24, demonstrate the weight gain produced by SARM at all doses of formula VIII and formula IX, indicating healthy growth and lack of toxicity. By comparison, vehicle treated animals did not grow robustly.
In summary, as described above and below, the formula VIII SARM is extremely effective in causing regression of growth of triple negative breast cancer xenografts expressing AR in mice, and may be effective against a variety of AR positive breast cancers in humans.
Example 9
Effect of formula IX on women with metastatic or ER and/or AR positive refractory breast cancer
This clinical trial assessed the safety and efficacy of 9mg of the compound represented by the structure of formula IX (formula IX) in 22 postmenopausal women suffering from Estrogen Receptor (ER) positive metastatic breast cancer and previously responsive to adjuvant and/or rescue endocrine therapy. The aim of this study was to determine the importance of AR status as a therapeutic target in women with ER positive Metastatic Breast Cancer (MBC) who have previously responded to hormone therapy. Treatment continues until disease Progression (PD).
The primary endpoint is Clinical Benefit Remission (CBR) at 6 months (m), defined as patients with Complete Remission (CR), partial Remission (PR), or disease Stabilization (SD). CBR will be related to AR status of metastatic tumor biopsies.
Serum Prostate Specific Antigen (PSA) was evaluated as a biomarker for AR activity. [000380] results: formula IX is well tolerated, has no drug-related serious adverse events and does not exceed grade 3. Conclusion: formula IX demonstrates promise as a novel targeted therapy for AR positive MBC. The primary endpoint has been reached, with 6/17 of the ar+ patients reaching a statistical threshold of success, as summarized in tables 1-5 below. Serum PSA appears to be a surrogate marker for AR activity and disease response.
Materials and methods
Population of subjects
Female subjects with ER positive metastatic breast cancer have previously been treated with up to 3 prior hormone therapies for the treatment of breast cancer. The subject must receive and respond to adjuvant therapy for 3 years or more or metastatic disease hormone therapy for 6 months or more before the disease progresses. The details of the subject selection criteria are as follows:
in order to qualify for participation in the present study, the subject must meet all of the following criteria, including: voluntarily signing an informed consent form according to the agency policy; is a female diagnosed with ER-positive metastatic breast cancer; and is clinically confirmed to be postmenopausal. Prior to the start of the study, subjects must undergo spontaneous menopause, drug menopause, or surgical menopause. (spontaneous menopause is defined as the natural cessation of ovarian function, manifested as amenorrhea for at least 12 months. If the subject has amenorrhea for > 6 months but <12 months, they must have a serum FSH concentration of > 50mIU/mL and a 17 beta-estradiol concentration of < 25 pg/mL; medical menopause is defined as treatment with a luteinizing hormone receptor hormone agonist; and operative menopause is defined as bilateral ovariectomy).
Additional requirements that a subject must meet include: they must receive and respond to prior adjuvant hormone therapy for 3 years or more or prior hormone therapy for metastatic disease for 6 months or more before the disease progresses; they did not undergo radiation therapy for breast cancer within 2 weeks of randomization in this study, and did not plan to undergo radiation therapy during participation in this study. The subject must be willing to provide a tissue sample of a biopsy of the metastatic tumor lesion to determine AR and ER status. Tissue samples from biopsies of primary tumor lesions will also be provided, if possible. Additional subjects must have an ECOG score of 2 or less and an age of 18 or more.
Subjects with any of the following exclusion criteria will not qualify for the group study: has triple negative breast cancer; at the discretion of the researcher, suffering from clinically significant concomitant diseases or psychological, familial, social, geographic or other concomitant disorders, adequate follow-up and compliance with the research protocol is not allowed; suffering from uncontrolled hypertension, congestive heart failure or angina; suffering from stage 4 Chronic Obstructive Pulmonary Disease (COPD); from HBsAg (liver B)Inflammatory surface antigen) is positive unless the subject is prior to entry into the group >Is diagnosed for 10 years and has no evidence of active liver disease; ALT/SGOT or AST/SGPT with 1.5 times higher than the Upper Limit of Normal (ULN); positive screening of hepatitis A antibody IgM or HIV; receiving metastatic breast cancer chemotherapy within 3 months prior to the study of the group or anticipated to receive metastatic breast cancer chemotherapy during the study; testosterone, methyltestosterone and androstane oxide are currently being takenHydroxymethyl longan, danazol, fluoxymesterone ++>Testosterone-like drugs (such as Dehydroepiandrosterone (DHEA), androstenedione, and other androgenic compounds, including herbal medicines) or antiandrogens; past therapies with testosterone and testosterone-like drugs are acceptable and have a 30 day washout period (if the previous testosterone therapy was long term storage therapy over the past 6 months, the study center should contact the medical monitor of the study to determine the appropriate washout period); with untreated or uncontrolled brain metastases; has been diagnosed with cancer or has been treated for cancer in the past two years, with the exception of breast cancer or non-melanoma skin cancer
After group entry, all primary and/or metastatic focus subjects were evaluated for Androgen Receptor (AR) status. It was observed that most (17/19) subjects with ER-positive breast cancer also expressed AR in their primary tumor samples, which correlates well with the previous literature predicting that 70% -95% will be AR positive (Niemeier LA et al, "Androgen receptor in breast cancer: expression in estrogen receptor-positive tumors and in estrogen-negative tumors with apocrine differentiation.", modern Pathology, vol. 23:205-212, 2010; narita D et al, "Immunohistochemical expression of androgen receptor and prostate-specific antigen in breast cancer.", folia Histochemica Et Cytobiologica, vol. 44:165-172, 2006).
A high percentage (72% -84%) of metastatic lesions obtained from women with advanced breast Cancer have also been found to be AR positive (Lea OA et al, "Improved measurement of androgen receptors in human breast Cancer.", cancer Research, volume 49: pages 7162-7167, 1989).
Since 70% or more of ER-positive breast cancer females are expected to have AR-positive tumors, the study was designed to recruit about 27 AR-positive breast cancer subjects (40 subjects initially scheduled for recruitment) in each dose group, enabling the assessment of primary endpoints for AR-positive subjects, as well as secondary and tertiary endpoints based on a subset of AR status (i.e., all subjects, AR-positive subjects, and AR-negative subjects).
At the time of writing this document, patient demographics are: average age 63.7 years, average diagnostic time 11.0 years, 72.7% of previous chemotherapy, 89% (17/19) AR+,41% detectable baseline PSA and 86.4% of previous radiation.
Table 1: baseline characteristics of the reaction are as follows
/>
/>
/>
Treatment of
Subjects received a 9mg daily dose of formula IX and were assessed for baseline and periodic studies of safety and efficacy.
During this study, measurable and unmeasurable lesions (primary and/or metastatic) were identified and evaluated by a modified solid tumor remission evaluation criteria (RECIST 1.1) classification (described in detail below).
Duration of the study
Each subject participating in the study received intervention until a Progression Free Survival (PFS) endpoint (tumor progression or death) was reached. Subjects were only followed after discontinuation of treatment due to life status.
End of efficacy
The primary efficacy analysis was the clinical benefit in subjects with AR positive breast cancer at 6 months as measured by the improved solid tumor remission assessment criteria (RECIST 1.1) classification. Critical secondary endpoints of clinical benefit in all subjects and AR negative subjects were also assessed, as well as objective remission rates, progression free survival, progression time, remission duration, SRE incidence, and first SRE time in subgroups based on AR status (i.e., all subjects, AR positive subjects, and AR negative subjects). The effect on CA 27-29, PSA, bone turnover markers, QOL and force ratios were evaluated as tertiary endpoints.
Primary endpoint
Clinical benefit in a subject is defined as complete remission [ CR ], partial remission [ PR ] or disease stabilization [ 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, volume 45:pages 228-247, 2009).
For subjects with non-measurable (non-target) disease only at baseline, SD is defined as those subjects with non-CR/non-PD combined remission. The primary endpoint of this study was to evaluate the proportion of subjects (PCB) who had clinical benefit at 6 months (cr+pr+sd) in subjects with AR positive breast cancer.
Secondary endpoint
Secondary efficacy endpoints included:
evaluate the clinical benefit of all breast cancer subjects receiving treatment of formula IX. Clinical benefit is defined as the proportion of subjects who completely remit [ CR ] + partially remit [ PR ] + disease stable [ 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, volume 45: pages 228-247, 2009).
For subjects with non-measurable (non-target) disease only at baseline, SD is defined as those subjects with non-CR/non-PD combined remission.
Evaluate the Objective Remission Rate (ORR) of breast cancer subjects receiving treatment of formula IX. Objective remission rate is defined as the proportion of subjects who reached CR or PR at 6 months, as measured by improved RECIST 1.1. For subjects with non-measurable (non-target) disease only at baseline, ORR is defined as the proportion of subjects that reached CR at 6 months, as measured by the modified RECIST 1.1.
Assessing Progression Free Survival (PFS) of a breast cancer subject receiving treatment of formula IX. PFS is defined as the time that passes between the onset of treatment and tumor progression, as measured by modified RECIST 1.1 or death.
Time To Progression (TTP) of breast cancer subjects receiving treatment of formula IX was assessed. Tumor progression time is defined as the time elapsed between the onset of treatment and tumor progression, as measured by the modified RECIST 1.1.
Assessing the duration of remission in a breast cancer subject receiving treatment of formula IX.
Assessing the incidence of bone related events (SREs) in subjects receiving the treatment of formula IX.
Assessing the time to first appearance of a bone related event (SRE) in a subject receiving treatment of formula IX.
Three-stage endpoint
Serum CA 27-29 changes in breast cancer subjects receiving treatment of formula IX were assessed.
Serum PSA changes were assessed in breast cancer subjects receiving treatment of formula IX.
Evaluation of the changes in bone turnover markers (serum osteocalcin, serum type I collagen-crosslinked C-terminal peptide [ CTX ], serum type I collagen-crosslinked N-terminal peptide [ NTX ], serum bone-specific alkaline phosphatase, and urinary NTX) in subjects receiving treatment of formula IX.
Assessing the effect of formula IX on quality of life (QOL), as measured by the facility-F questionnaire in subjects receiving treatment of formula IX.
The effect of formula IX on the force ratio was evaluated as measured by the female sexual index (FSFI) questionnaire in subjects receiving treatment of formula IX.
Exploring the relationship between different levels of AR expression as determined by immunohistochemistry and primary, secondary and tertiary targets.
Results
After a median follow-up of 81 days (d) (range 7d-304 d), the preliminary results for 22 patients were as follows: 9 SD with a median duration of 212d was observed for optimal relief. Current status of all patients: 15 PDs after the median 80d (range 15d-304 d), 4 SDs, 3 early interrupts (d 7, 28, 255). Of the patients reaching 6m, 6 were AR positive with SD and increased PSA.1 had not reached 6m, and CR or PR was not yet observed. Formula IX is well tolerated, has no drug-related serious adverse events and does not exceed grade 3.
No useful trend was observed with the following biomarkers of bone turnover: bone-specific alkaline phosphatase, C-terminal peptide, N-terminal peptide and osteocalcin. Likewise, breast cancer biomarkers CA 27-29 did not show any useful trend.
As observed in 20 of the 22 patients measured, PSA levels appeared to increase in response to treatment of formula IX, but the correlation with clinical benefit or disease progression was not yet apparent.
The following non-serious adverse events were observed:
atrial fibrillation (1); anxiety/mood changes (5), joint pain (6), bloating (2), bruise (1), cellulitis (1), chills (1), constipation (2), cough (1), dehydration (1), diarrhea (3), dizziness (2), dysgeusia (1), dyspepsia (1), dyspnea (3), edema (2), fatigue (14), fever (1), flatulence (1), glaucoma (1), headache (4), hot and night sweats (7), hypertension (2), infections (1), insomnia (2), myalgia (5), nail discoloration (1), nausea (11), pain (22), paresthesia (1), pleural effusion (1), polyuria (1), post-menopausal bleeding (3), rash/acne (3), stiffness (1), tendinitis (1), vision changes (3), vomiting (2), weight gain (2), and weight loss (2).
Liver enzymes (ALT, AST and bilirubin) return to baseline without interrupting the treatment and without an increase in total bilirubin.
Conclusion: formula IX demonstrates promise as a novel targeted therapy for AR positive MBC. The primary endpoint was reached, with 6/17AR positive patients reaching a statistical threshold of success. Serum PSA appears to be a surrogate marker for AR activity and disease response.
/>
Improved RECIST 1.1
The following modified RECIST 1.1 definition applies:
Measurable lesions
A measurable lesion is defined as one lesion with a Longest Diameter (LD) that can be precisely measured as ≡10mm by CT or MRI techniques using a 5mm continuous reconstruction algorithm.
The measurable lesions must be at least 2 times the thickness of the slice or at least 2 times the size of the CT scan interval cut.
Lesions found in chest X-ray but not confirmed by CT or MRI scans may not be measurable lesions for this study.
When assessed by CT scanning (CT scan slice thickness is recommended to be no greater than 5 mm), the lymph nodes must be >15mm in the short axis to be considered pathologically enlarged and measurable. In baseline and follow-up, only the short axis will be measured and tracked.
Measurable disease is defined as the presence of at least one measurable lesion.
All measurements were made and recorded in millimeters using an electronic measurement method.
Non-measurable lesions
An unmeasurable lesion is defined as any lesion or truly unmeasurable lesion (or disease site) that is smaller than the measurable lesion criteria described above (non-nodular lesions <10mm in longest diameter or pathological lymph nodes with short axes ≡10mm to <15 mm). Lesions that are considered truly unmeasurable are bone lesions (no soluble lesions or mixed soluble-proliferative lesions that can identify soft tissue components, and proliferative lesions), leptomeningeal diseases, ascites, pleural/pericardial effusions, cutaneous/pulmonary lymphangitis, inflammatory breast diseases, abdominal masses and cystic lesions that are not confirmed by imaging techniques.
Target focus
The target lesion must be a measurable lesion.
All target lesions representing all relevant organs (up to two lesions per organ, five lesions total) will be selected/identified as target lesions and recorded and measured at baseline.
The target lesions should be selected based on their size (lesions with longest diameter) and their suitability for accurate repeated measurement by CT/MRI imaging techniques, and most representative of the tumor burden of the subject.
The target lesion will be measured in one dimension by a size estimate of its diameter. The sum of the diameters of all target lesions at each time point (the diameter of the non-nodular lesion is the longest and the diameter of the nodular lesion is the shortest) will be calculated and reported. The baseline sum of diameters will be used as a reference to further characterize objective tumor remission in the measurable dimension of the disease.
Non-target lesions
All other lesions (or disease sites) and any measurable lesions not selected as target lesions should be identified as non-target lesions and indicated as present at baseline.
Measurements of non-target lesions may be made, however the continued presence or absence and status of disappearance or progression of these lesions will be recorded throughout the follow-up assessment.
New lesions
Whether the new lesion occurs in the anatomical region of the routine follow-up, or in the region that is free of disease at baseline and is subsequently scanned for clinical suspicion of the new disease, the follow-up will be considered a new lesion. The new lymph node needs to have a minimum size of 10mm on its shortest axis. The new non-nodular lesions need not be measurable or of minimal size. Measurements of new lesions may be made.
Mitigation criteria definition
The following remission criteria will apply to both target and non-target lesions:
target lesion remission criteria
Complete Remission (CR): all target lesions disappeared. Target lymph node lesions with shortest diameters <10mm will be considered normal (non-pathological) and their actual measurements will be recorded. Thus, if all target lymph node lesions become <10mm and all other non-lymph node lesions have disappeared (whether of the target or non-target type), then overall remission will be considered CR.
Partial Relief (PR): taking the baseline sum of diameters as a reference, the sum of diameters of target lesions is reduced by at least 30%.
Disease Stabilization (SD): taking the minimum sum of diameters (nadir) as a reference, there is neither enough shrinkage to conform to PR nor enough increase to conform to PD.
Progressive Disease (PD): taking as a reference the sum of the smallest diameters recorded since the start of the treatment (nadir), the sum of the diameters of the target lesions increases by at least 20%. In addition to a relative increase of 20%, the sum of the diameters must also show an absolute increase of at least 5 mm.
Inestimable (NE): NE may be applied if repeated measurements cannot be evaluated due to insufficient or missing imaging, etc.
Non-target lesion remission criteria
Complete Remission (CR): disappearance of all non-target lesions. All lymph nodes must be non-pathological in size (< 10mm minor axis). The bone lesions determined by bone scintigraphy disappeared.
non-CR/non-PD: the persistence of one or more non-target lesions. Stabilization, reduction or slight increase in bone focus uptake upon bone scintigraphy.
Progressive Disease (PD): there is a clear progression of non-target lesions. The increase in perceived bone disease in pre-existing areas will not be considered progression.
For bone scintigraphy, at least two new lesions are required to infer the clear presence of new lesions unless one or more of these lesions are confirmed by radiography, CT or MRI.
Inestimable (NE): NE may be applied if duplicate evaluations cannot be evaluated due to insufficient imaging or lack thereof, etc.
Definition of combined mitigation for each time point
The overall remission at each time point was determined based on a combination of target remission, non-target remission, and the presence or absence of new lesions using the algorithm outlined in tables C1 and C2 below.
Table C1: summary of definitions of remission for patients with measurable (target) disease at baseline
Table C2: summary of definitions of remission for patients with non-measurable (non-target) disease at baseline only
/>
Example 10
Synthesis of the (S) enantiomer of formula VIII
(2R) -1-methacryloylpyrrolidine-2-carboxylic acid. D-proline, 14.93g,0.13 mol) was dissolved in 71mL 2N NaOH and cooled in an ice bath; the resulting alkaline solution was diluted with acetone (71 mL). Methacryloyl chloride (13.56 g,0.13 mol) in acetone (71 mL) and 2N NaOH solution (71 mL) were added simultaneously to an aqueous solution of D-proline in an ice bath over 40 minutes. The pH of the mixture was maintained at 10℃to 11℃during the addition of methacryloyl chloride. After stirring (3 hours, room temperature), the mixture was evaporated in vacuo at a temperature of 35 ℃ to 45 ℃ to remove acetone. The resulting solution was washed with diethyl ether and acidified to pH 2 with concentrated HCl. The acidic mixture was saturated with NaCl and extracted with EtOAc (100 mL. Times.3). The combined extracts were dried over Na2SO4, and purified by Filtered and evaporated in vacuo to give the crude product as a colourless oil. Recrystallisation of the oil from diethyl ether and hexane gives 16.2g (68%) of the desired compound as colourless crystals: mp 102-103 ℃; NMR spectra of this compound confirm the presence of two rotamers of the title compound. 1 H NMR (300 MHz, DMSO-d 6) delta 5.28(s) and 5.15(s) are both for the first rotamer, 5.15(s) and 5.03(s) are both for the second rotamer (2H total of both rotamers, vinyl CH) 2 ) 4.48-4.44 for the first rotamer, 4.24-4.20 (m) for the second rotamer (total of 1H for both rotamers, CH at chiral center), 3.57-3.38 (m, 2H, CH) 2 ),2.27-2.12(1H,CH),1.97-1.72(m,6H,CH 2 ,CH,Me); 13 C NMR (75 MHz, DMSO-d 6) delta for the major rotamer 173.3,169.1,140.9,116.4,58.3,48.7,28.9,24.7,19.5: for minor rotamer 174.0,170.0,141.6,115.2,60.3,45.9,31.0,22.3,19.7; IR (KBr) 3437 (OH), 1737 (c=o), 1647 (CO, COOH), 1584,1508,1459,1369,1348,1178cm -1 ;[α]D 26 +80.8 ° (c=1, meoh); analytical calculations of C9H13NO 3: and C59.00,H 7.15,N 7.65. Experimental values: and C59.13,H7.19,N 7.61.
(3R, 8 aR) -3-bromomethyl-3-methyl-tetrahydro-pyrrolo [2,1-c][1,4]Oxazine-1, 4-dione. A solution of NBS (23.5 g,0.132 mol) in 100mL of DMF was added dropwise to a stirred solution of (meth-acryl) -pyrrolidine (16.1 g,88 mmol) in 70mL of DMF under argon at room temperature, and the resulting mixture was stirred for 3 days. The solvent was removed in vacuo, and a yellow solid precipitated. The solid was suspended in water, stirred overnight at room temperature, filtered and dried to give 18.6g (81%) (weight reduced about 34% when dried) of the title compound as a yellow solid: mp 152-154 ℃; 1 H NMR (300 mhz, dmso-d 6) delta 4.69 (dd, j=9.6 hz, j=6.7 hz,1H, CH at chiral center), 4.02 (d, j=11.4 hz,1H, chha), 3.86 (d, j=11.4 hz,1H, chhb), 3.53-3.24 (m, 4H, CH) 2 ),2.30-2.20(m,1H,CH),2.04-1.72(m,3H,CH 2 And CH), 1.56 (s, 2H, me); 13 C NMR(75MHz,DMSO-d 6 )δ167.3,163.1,83.9,57.2,45.4,37.8,29.0,22.9,21.6;IR(KBr)3474,1745(C=O),1687(C=O),1448,1377,1360,1308,1227,1159,1062cm -1 ;[α] D 26 +124.5 ° (c=1.3, chloroform); c (C) 9 H 12 BrNO 3 Is calculated by analysis of: and C41.24,H 4.61,N 5.34. Experimental values: and C41.46,H 4.64,N 5.32.
(2R) -3-bromo-2-hydroxy-2-methylpropanoic acid. Bromide (18.5 g)71 mmol) in 300ml24% HBr for 1 hour. The resulting solution was diluted with brine (200 mL) and extracted with ethyl acetate (100 ml×4). The combined extracts were treated with saturated NaHCO 3 (100 mL. Times.4) washing. The aqueous solution was acidified to ph=1 with concentrated HCl and the acidified aqueous solution was then extracted with ethyl acetate (100 ml×4). The combined organic solutions were treated with Na 2 SO 4 Dried, filtered through Celite, and evaporated to dryness in vacuo. Recrystallisation from toluene gives 10.2g (86%) of the desired compound as colourless crystals: mp 107-109 ℃; 1 H NMR(300MHz,DMSO-d 6 )δ3.63(d,J=10.1Hz,1H,CHH a ),3.52(d,J=10.1Hz,1H,CHH b ),1.35(s,3H,Me);IR(KBr)3434(OH),3300-2500(COOH),1730(C=O),1449,1421,1380,1292,1193,1085cm- 1 ;[α] D 26 +10.5°(c=2.6,MeOH);C 4 H 7 BrO 3 is calculated by analysis of: c26.25, H3.86. Experimental values: c26.28, h 3.75.
Synthesis of (2R) -3-bromo-N- (3-chloro-4-cyanophenyl) -2-hydroxy-2-methylpropanamide. Thionyl chloride (7.8 g,65.5 mmol) was added dropwise to a cooled (below 4 ℃) solution of (R) -3-bromo-2-hydroxy-2-methylpropanoic acid (9.0 g,49.2 mmol) in 50mL THF under argon atmosphere. The resulting mixture was stirred under the same conditions for 3 hours. Et3N (6.6 g,65.5 mol) was added thereto and stirred under the same conditions for 20 minutes. After 20 min, 4-amino-2-chlorobenzonitrile (5.0 g,32.8 mmol) and 100mL THF were added and the mixture was then allowed to stir at room temperature overnight. The solvent was removed under reduced pressure to give a solid which was taken up in 100mL of H 2 O was treated and extracted with EtOAc (2X 150 mL). The combined organic extracts were washed with saturated NaHCO 3 The solution (2X 100 mL) and brine (300 mL) were washed. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give a solid which was purified from column chromatography using EtOAc/hexanes (50:50) to give 7.7g (49.4%) of the target compound as a brown solid.
1 H NMR(CDCl 3 /TMS)δ1.7(s,3H,CH 3 ),3.0(s,1H,OH),3.7(d,1H,CH),4.0(d,1H,CH),7.5(d,1H,ArH),7.7(d,1H,ArH),8.0(s,1H,ArH),8.8(s,1H,NH)。MS:342.1(M+23)。Mp 129℃。
Synthesis of (S) -N- (3-chloro-4-cyanophenyl) -3- (4-cyanophenoxy) -2-hydroxy-2-methylpropanamide (formula VIII). A mixture of bromoamide (2.0 g,6.3 mmol), anhydrous K2CO3 (2.6 g,18.9 mmol) in 50mL of acetone was heated at reflux for 2 hours and then concentrated under reduced pressure to give a solid. The resulting solid was treated with 4-cyanophenol (1.1 g,9.5 mmol) and anhydrous K2CO3 (1.7 g,12.6 mmol) in 50mL 2-propanol, heated under reflux for 3 hours, and then concentrated under reduced pressure to give a solid. The residue was taken up with 100mL H 2 O was treated and then extracted with EtOAc (2X 100 mL). The combined EtOAc extracts were washed sequentially with 10% naoh (4×100 mL) and brine. The organic layer was dried over MgSO4 and then concentrated under reduced pressure to give an oil which was purified by column chromatography using EtOAc/hexane (50:50) to give a solid. Solid from CH 2 Cl 2/hexane was recrystallized to give 1.4g (61.6%) of (S) -N- (3-chloro-4-cyanophenyl) -3- (4-cyanophenoxy) -2-hydroxy-2-methylpropanamide as a colorless solid.
1 H NMR(CDCl 3 /TMS)δ1.61(s,3H,CH 3 ) 3.25 (s, 1h, oh), 4.06 (d, j=9.15 hz,1h, ch), 4.50 (d, j=9.15 hz,1h, ch), 6.97-6.99 (m, 2h, arh), 7.53-7.59 (m, 4h, arh), 7.97 (d, j=2.01 hz,1h, arh), 8.96 (s, 1h, nh). Calculated mass: 355.1, [ M+Na ]] + 378.0。Mp:103℃-105℃。
Example 11
Synthesis of the (S) enantiomer of formula IX
(2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl]-2-hydroxy-2-methylpropanamideIs a synthesis of (a). Thionyl chloride (46.02 g,0.39 mol) was added dropwise to a cooled (below 4 ℃) solution of (R) -3-bromo-2-hydroxy-2-methylpropanoic acid (51.13 g,0.28 mol) in 300mL THF under argon atmosphere. (R) -3-bromo-2-hydroxy-2-methylpropanoic acid was prepared as described in example 10. The resulting mixture was stirred under the same conditions for 3 hours. Et is added thereto 3 N (39.14 g,0.39 mol) and stirred under the same conditions for 20 minutes. After 20 minutes, 5-amino-2-cyano benzotrifluoride (40.0 g,0.21 mol), 400mL THF was added, and the mixture was then stirred at room temperature overnight. The solvent was removed under reduced pressure to give a solid, which was quenched with 300mL of H 2 O was treated and extracted with EtOAc (2X 400 mL). The combined organic extracts were washed with saturated NaHCO3 solution (2X 300 mL) and brine (300 mL). The organic layer was dried over MgSO 4 Dried and concentrated under reduced pressure to give a solid, which was concentrated using CH 2 Cl2/EtOAc (80:20) was purified by column chromatography to give a solid. The solid is taken from CH 2 Cl 2/hexane was recrystallized to give 55.8g (73.9%) of (2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl as a pale yellow solid]-2-hydroxy-2-methylpropanamide.
1 H NMR(CDCl 3 /TMS)δ1.66(s,3H,CH 3 ) 3.11 (s, 1h, oh), 3.63 (d, j=10.8 hz,1h, ch 2), 4.05 (d, j=10.8 hz,1h, ch 2), 7.85 (d, j=8.4 hz,1h, arh), 7.99 (dd, j=2.1, 8.4hz,1h, arh), 8.12 (d, j=2.1 hz,1h, arh), 9.04 (bs, 1h, nh). Calculated mass: 349.99, [ M-H ]]-349.0。M.p.:124℃-126℃。
Synthesis of (S) -N- (4-cyano-3- (trifluoromethyl) phenyl) -3- (4-cyanophenoxy) -2-hydroxy-2-methylpropanamide (formula IX). Bromo amide ((2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl)]A mixture of 2-hydroxy-2-methylpropanamide, 50g,0.14 mol), anhydrous K2CO3 (59.04 g,0.43 mol), 4-cyanophenol (25.44 g,0.21 mol) in 500mL of 2-propanol was heated under reflux for 3 hours and then concentrated under reduced pressure to give a solid. The residue obtained was taken up in 500mL H 2 O was treated and then extracted with EtOAc (2X 300 mL). Will be combinedWith 10% NaOH (4X 200 mL) and
washing with brine. The organic layer was dried over MgSO 4 Dried and then concentrated under reduced pressure to give an oil which was treated with 300mL of ethanol and activated carbon. The reaction mixture was heated to reflux for 1 hour, and the hot mixture was then passed through And (5) filtering. The filtrate was concentrated under reduced pressure to give an oil. The oil was prepared using CH 2 Cl2/EtOAc (80:20) was purified by column chromatography to give an oil which was taken from CH 2 Cl 2 Crystallization in hexane gives 33.2g (59.9%) of (S) -N- (4-cyano-3- (trifluoromethyl) phenyl) -3- (4-cyanophenoxy) -2-hydroxy-2-methylpropanamide as a colourless solid (cotton).
1 H NMR (CDCl 3/TMS) δ1.63 (s, 3H, ch 3), 3.35 (s, 1H, oh), 4.07 (d, j=9.04 hz,1H, ch), 4.51 (d, j=9.04 hz,1H, ch), 6.97-6.99 (m, 2H, arh), 7.57-7.60 (m, 2H, arh), 7.81 (d, j=8.55 hz,1H, arh), 7.97 (dd, j=1.95, 8.55hz,1H, arh), 8.12 (d, j=1.95 hz,1H, arh), 9.13 (bs, 1H, nh). Calculated mass: 389.10, [ M-H ]]-388.1。Mp:92℃-94℃。
Example 12
Synthesis of the (R) enantiomer of formula IX
(2S) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl]-synthesis of 2-hydroxy-2-methylpropanamide (precursor of the R-enantiomer of formula IX). Thionyl chloride (46.02 g,0.39 mol) was added dropwise to a cooled (below 4 ℃) solution of (S) -3-bromo-2-hydroxy-2-methylpropanoic acid (51.13 g,0.28 mol) in 300mL THF under argon atmosphere. The resulting mixture was stirred under the same conditions for 3 hours. Et3N (39.14 g,0.39 mol) was added thereto and stirred under the same conditions for 20 minutes. After 20 minutes, 5-amino-2-cyano benzotrifluoride (40.0 g,0.21 mol), 400mL of THF was added, and the mixture was then stirred at room temperature Overnight. The solvent was removed under reduced pressure to give a solid, which was quenched with 300mL of H 2 O was treated and extracted with EtOAc (2X 400 mL). The combined organic extracts were washed with saturated NaHCO 3 The solution (2X 300 mL) and brine (300 mL) were washed. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give a solid, which was concentrated using CH 2 Cl2/EtOAc (80:20) was purified by column chromatography to give a solid. The solid was recrystallized from EtOAc/hexanes to give 55.8g (73.9%) of the target compound as a pale yellow solid.
1 H NMR(CDCl3/TMS)δ1.66(s,3H,CH3),3.11(s,1H,OH),3.63(d,J=10.8Hz,1H,CH2),4.05(d,J=10.8Hz,1H,CH2),7.85(d,J=8.4Hz,1H,ArH),7.99(dd,J=2.1,8.4Hz,1H,ArH),8.12(d,J=2.1Hz,1H,ArH),9.04(bs,1H,NH)。
Calculated mass: 349.99, [ M-H ] -349.0.Mp: 124-126 ℃.
Synthesis of (R) -N- (4-cyano-3- (trifluoromethyl) phenyl) -3- (4-cyanophenoxy) -2-hydroxy-2-methylpropanamide (R-enantiomer of formula IX). Bromoamide (50.0 g,0.14 mol), anhydrous K 2 CO 3 A mixture of (59.04 g,0.43 mol), 4-cyanophenol (25.44 g,0.21 mol) in 500mL of 2-propanol was heated under reflux for 3 hours, then concentrated under reduced pressure to give a solid. The residue obtained was taken up in 500mL H 2 O was treated and then extracted with EtOAc (2X 300 mL). The combined EtOAc extracts were washed with 10% naoh (4×200 mL) and brine. The organic layer was dried over MgSO4 and then concentrated under reduced pressure to give an oil which was treated with 300mL of ethanol and activated carbon. The reaction mixture was heated to reflux for 1 hour, and the hot mixture was then passed through And (5) filtering. The filtrate was concentrated under reduced pressure to give an oil. The oil was purified by column chromatography using hexane/EtOAc (20:80) to give an oil which was crystallized from EtOAc/hexane to give 33.2g (59.9%) of (R) -N- (4-cyano-3- (trifluoromethyl) as a colorless solidPhenyl) -3- (4-cyanophenoxy) -2-hydroxy-2-methylpropanamide (R-isomer of formula IX).
1 H NMR(CDCl 3 /TMS)δ1.63(s,3H,CH 3 ) 3.44 (s, 1h, oh), 4.07 (d, j=9.16 hz,1h, ch), 4.51 (d, j=9.16 hz,1h, ch), 6.97-6.99 (m, 2h, arh), 7.57-7.59 (m, 2h, arh), 7.81 (d, j=8.54 hz,1h, arh), 7.97 (dd, j=2.07, 8.54hz,1h, arh), 8.12 (d, j=2.07 hz,1h, arh), 9.15 (bs, 1h, nh). Calculated mass: 389.10, [ M-H ]]-388.1。Mp:92℃-94℃。
Example 13
Synthesis of the (S) enantiomer of formula X
(2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl]-synthesis of 2-hydroxy-2-methylpropanamide. Thionyl chloride (46.02 g,0.39 mmol) was added dropwise to a cooled (below 4 ℃) solution of (R) -3-bromo-2-hydroxy-2-methylpropanoic acid (51.13 g,0.28 mmol) in 300mL THF under argon atmosphere. The resulting mixture was stirred under the same conditions for 3 hours. Et3N (39.14 g,0.39 mol) was added thereto and stirred under the same conditions for 20 minutes. After 20 minutes, 5-amino-2-cyano benzotrifluoride (40.0 g,0.21 mol), 400mL THF was added, and the mixture was then stirred at room temperature overnight. The solvent was removed under reduced pressure to give a solid, which was quenched with 300mL of H 2 O was treated and extracted with EtOAc (2X 400 mL). The combined organic extracts were washed with saturated NaHCO 3 The solution (2X 300 mL) and brine (300 mL) were washed. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give a solid, which was concentrated using CH 2 Cl 2 EtOAc (80:20) was purified by column chromatography to give a solid. The solid is taken from CH 2 The Cl 2/hexane was recrystallized to give the target compound (55.8 g, 73.9%) as a pale yellow solid.
1 H NMR(CDCl3/TMS)δ1.66(s,3H,CH3),3.11(s,1H,OH),3.63(d,J=10.8Hz,1H,CH 2 ),4.05(d,J=10.8Hz,1H,CH 2 ),7.85(d,J=8.4Hz,1H,ArH),7.99(dd, j=2.1, 8.4hz,1h, arh), 8.12 (d, j=2.1 hz,1h, arh), 9.04 (bs, 1h, nh). Calculated mass: 349.99, [ M-H ]]-349.0。Mp:124℃-126℃。
Synthesis of (S) -N- (4-cyano-3- (trifluoromethyl) phenyl) -3- (4-fluorophenoxy) -2-hydroxy-2-methylpropanamide (formula X). A mixture of bromoamide (10.0 g,28.5 mmol), anhydrous K2CO3 (11.8 g,85.4 mmol) in 150mL of acetone was heated at reflux for 1h and then concentrated under reduced pressure to give a solid. The residue obtained was treated with 4-fluorophenol (4.8 g,42.7 mmol), anhydrous K 2 CO 3 (7.9 g,57.0 mmol), 150mL of 2-propanol, and then heated under reflux for 2 hours. The resulting mixture was concentrated under reduced pressure to give a solid. The solid was treated with 300mL of H 2 O was treated and extracted with EtOAc (2X 250 mL). The combined EtOAc extracts were washed with saturated NaHCO3 solution (2×250 mL) and brine. The organic layer was dried over MgSO4 and then concentrated under reduced pressure to give an oil which was purified using CH 2 Cl2/EtOAc (80:20) was purified by column chromatography to give a solid. The solid is taken from CH 2 Cl 2 Hexane was recrystallized to give (S) -N- (4-cyano-3- (trifluoromethyl) phenyl) -3- (4-fluorophenoxy) -2-hydroxy-2-methylpropanamide (formula X,10.04g, 92.2%) as a colorless solid.
1 H NMR (CDCl 3/TMS) δ1.59 (s, 3H, ch 3), 3.36 (s, 1H, oh), 3.95 (d, j=9.00 hz,1H, ch), 4.43 (d, j=9.00 hz,1H, ch), 6.87-6.88 (m, 2H, arh), 6.96-7.02 (m, 2H, arh), 7.81 (d, j=8.45 hz,1H, arh), 7.94-7.98 (m, 1H, arh), 8.10 (d, j=1.79 hz,1H, arh), 9.11 (s, 1H, nh). Calculated mass: 382.31, [ M-H ]]-380.9。Mp:139℃-141℃。
Example 14
Synthesis of the (S) enantiomer of formula XIII
(2R)-3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl]-synthesis of 2-hydroxy-2-methylpropanamide. Thionyl chloride (46.02 g,0.39 mol) was added dropwise to a cooled (below 4 ℃) solution of R-18 (51.13 g,0.28 mol) in 300mL THF under argon. R-18 is (R) -3-bromo-2-hydroxy-2-methylpropanoic acid prepared as described in example 10. The resulting mixture was stirred under the same conditions for 3 hours. Et3N (39.14 g,0.39 mol) was added thereto and stirred under the same conditions for 20 minutes. After 20 minutes, 5-amino-2-cyano benzotrifluoride (40.0 g,0.21 mol), 400mL THF was added, and the mixture was then stirred at room temperature overnight. The solvent was removed under reduced pressure to give a solid, which was quenched with 300mL of H 2 O was treated and extracted with EtOAc (2X 400 mL). The combined organic extracts were washed with saturated NaHCO 3 The solution (2X 300 mL) and brine (300 mL) were washed. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give a solid which was purified from column chromatography using CH2Cl2/EtOAc (80:20) to give a solid. The solid is taken from CH 2 Cl 2 Recrystallisation of hexane gives 55.8g (73.9%) of (2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl as a pale yellow solid]-2-hydroxy-2-methylpropanamide (R-19).
1 H NMR(CDCl3/TMS)δ1.66(s,3H,CH3),3.11(s,1H,OH),3.63(d,J=10.8Hz,1H,CH 2 ),4.05(d,J=10.8Hz,1H,CH 2 ) 7.85 (d, j=8.4 hz,1h, arh), 7.99 (dd, j=2.1, 8.4hz,1h, arh), 8.12 (d, j=2.1 hz,1h, arh), 9.04 (bs, 1h, nh). Calculated mass: 349.99, [ M-H ]]-349.0。M.p.:124℃-126℃。
Synthesis of (S) -N- (4-cyano-3- (trifluoromethyl) phenyl) -3- (4-cyano-3-fluorophenoxy) -2-hydroxy-2-methylpropanamide (formula XIII). Bromo amide ((2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl)]A mixture of 2-hydroxy-2-methylpropanamide R-19 (2.0 g,5.70 mmol)), anhydrous K2CO3 (2.4 g,17.1 mmol) in 50mL of acetone was heated at reflux for 2 hours and then concentrated under reduced pressure to give a solid. The resulting solid was treated with 2-fluoro-4-hydroxybenzonitrile (1.2 g,8.5 mmol) and anhydrous K 2 CO 3 (1.6 g,11.4 mmol) of a 50mL 2-propanol solution was treated, heated under reflux for 3 hours, and then concentrated under reduced pressure to give a solid. The residue was taken up with 100mL H 2 O was treated and then extracted with EtOAc (2X 100 mL). The combined EtOAc extracts were washed sequentially with 10% naoh (4×100 mL) and brine. The organic layer was dried over MgSO 4 Dried and then concentrated under reduced pressure to give an oil which is taken from CH 2 Crystallization of Cl 2/hexane afforded 0.5g (23%) of (S) -N- (4-cyano-3- (trifluoromethyl) phenyl) -3- (4-cyano-3-fluorophenoxy) -2-hydroxy-2-methylpropanamide as a colorless solid.
1 H NMR(CDCl 3 /TMS)δ1.63(s,3H,CH 3 ) 3.34 (bs, 1h, oh), 4.08 (d, j=9.17 hz,1h, ch), 4.50 (d, j=9.17 hz,1h, ch), 6.74-6.82 (m, 2h, arh), 7.50-7.55 (m, 1h, arh), 7.81 (d, j=8.50 hz,1h, arh), 7.97 (q, j=2.03, 8.50hz,1h, arh), 8.11 (d, j=2.03 hz,1h, arh), 9.12 (s, 1h, nh). Calculated mass: 407.1, [ M+Na ]] + 430.0。Mp:124℃-125℃。
Example 15
Synthesis of the (S) enantiomer of formula XIV
(2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl]-synthesis of 2-hydroxy-2-methylpropanamide. Thionyl chloride (46.02 g,0.39 mol) was added dropwise to a cooled (below 4 ℃) solution of R-18 (51.13 g,0.28 mol) in 300mL THF under argon. R-18 is (R) -3-bromo-2-hydroxy-2-methylpropanoic acid prepared as described in example 10. The resulting mixture was stirred under the same conditions for 3 hours. Et3N (39.14 g,0.39 mol) was added thereto and stirred under the same conditions for 20 minutes. After 20 minutes, 5-amino-2-cyano benzotrifluoride (40.0 g,0.21 mol), 400mL THF was added, and the mixture was then stirred at room temperature overnight. The solvent was removed under reduced pressure to give a solid, which was quenched with 300mL of H 2 O was treated and extracted with EtOAc (2X 400 mL). The combined organic extracts were washed with saturated NaHCO3 solution (2X 300 mL) and saltsWashed with water (300 mL). The organic layer was dried over MgSO 4 Dried and concentrated under reduced pressure to give a solid, which was concentrated using CH 2 Cl2/EtOAc (80:20) was purified by column chromatography to give a solid. The solid is taken from CH 2 Cl 2 Recrystallisation of hexane gives 55.8g (73.9%) of (2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl as a pale yellow solid]-2-hydroxy-2-methylpropanamide (R-19).
1 H NMR(CDCl 3 /TMS)δ1.66(s,3H,CH 3 ),3.11(s,1H,OH),3.63(d,J=10.8Hz,1H,CH 2 ),4.05(d,J=10.8Hz,1H,CH 2 ) 7.85 (d, j=8.4 hz,1h, arh), 7.99 (dd, j=2.1, 8.4hz,1h, arh), 8.12 (d, j=2.1 hz,1h, arh), 9.04 (bs, 1h, nh). Calculated mass: 349.99, [ M-H ]]-349.0。M.p.:124℃-126℃。
Synthesis of (S) -3- (4-chloro-3-fluorophenoxy) -N- (4-cyano-3- (trifluoromethyl) phenyl) -2-hydroxy-2-methylpropanamide (formula XIV). Bromo amide ((2R) -3-bromo-N- [ 4-cyano-3- (trifluoromethyl) phenyl)]A mixture of-2-hydroxy-2-methylpropanamide (R-19, 2.0g,5.70 mmol)), anhydrous K2CO3 (2.4 g,17.1 mmol) was heated under reflux for 2 hours and then concentrated under reduced pressure to give a solid. The resulting solid was treated with 4-chloro-3-fluorophenol (1.3 g,8.5 mmol) and anhydrous K 2 CO 3 (1.6 g,11.4 mmol) of a 50mL 2-propanol solution was treated, heated under reflux for 3 hours, and then concentrated under reduced pressure to give a solid. The residue was taken up with 100mL H 2 O was treated and then extracted with EtOAc (2X 100 mL). The combined EtOAc extracts were washed sequentially with 10% naoh (4×100 mL) and brine. The organic layer was dried over MgSO4 and then concentrated under reduced pressure to give an oil which was purified by column chromatography using EtOAc/hexanes (50:50) to give a solid which was purified from CH 2 Cl 2/hexane was recrystallized to give 1.7g (70.5%) of (S) -3- (4-chloro-3-fluorophenoxy) -N- (4-cyano-3- (trifluoromethyl) phenyl) -2-hydroxy-2-methylpropanamide as a colorless solid.
1 H NMR(CDCl 3 /TMS)δ1.60(s,3H,CH 3 ),3.28(s,1H,OH),3.98(d,J=9.05hz,1h, ch), 6.64-6.76 (m, 2h, arh), 7.30 (d, j=8.67 hz,1h, arh), 7.81 (d, j=8.52 hz,1h, arh), 7.96 (q, j=2.07, 8.52hz,1h, arh), 8.10 (d, j=2.07 hz,1h, arh), 9.10 (s, 1h, nh). Calculated mass: [ M-H ]]-414.9。Mp:132℃-134℃。
Example 16
Binding and transactivation of SARM in breast cancer cells
To determine whether the compounds of the invention are agonists in breast cancer cells, HEK-293 or MDA-MB-231 cells were transfected with 0.25 μg GRE-LUC, 10ng CMV-Renilla LUC and 25ng CMV-hAR using lipofectamine. 24 hours after transfection, cells were treated with DHT, a compound of formula VIII or a compound of formula IX and luciferase assays were performed 48 hours after transfection. Using known steroids and high affinity AR ligands [17α -methyl ] 3 H]Mibolone ([ solution ]) 3 H]MIB) and purified AR-LBD protein to measure competitive binding of DHT, the compound of formula VIII and the compound of formula IX.
Results
The compounds of formula DHT, VIII and IX are agonists of AR in breast cancer cells (HEK-293 cells in fig. 13A and MDA-MB-231 cells in fig. 13B-13C) as shown in fig. 13A-13C. The Relative Binding Affinities (RBA) of DHT, formula IX, formula VIII and bicalutamide to AR were 1.0, 0.330, 0.314 and 0.016, respectively, confirming the high affinity AR binding of the SARM compounds of the present invention (data not shown).
Example 17
Inhibition of intratumoral gene expression
AR agonists differentially modulate genes in ER positive and ER negative breast cancer cells. MDA-MB-231 and MCF-7 cells infected with adenovirus containing AR or GFP were maintained in medium containing activated carbon desorbed serum for 3 days and treated with DHT or formula VIII. After overnight treatment, cells were harvested, RNA isolated and real-time PCR was performed on the indicated genes. The measurements were normalized to GAPDH in response to expression of various genes of DHT or formula VIII and are given as composite data in table 6 (same effect for DHT and formula VIII).
Table 6: differential modulation of gene expression by AR ligands in ER-positive (MCF 7) and ER-negative (MDA-MB-231) breast cancers Node
VCAM 1-vascular cell adhesion protein-1-is important for cell anchorage-dependent growth and is also a chemoattractant.
SPARC-a secreted acidic and cysteine-rich protein (also known as osteonectin) -an extracellular glycoprotein important for angiogenesis.
MUC 1-mucin 1-an extracellular glycoprotein associated with cancer-has a strong ARE in its promoter.
SLUG-zinc finger transcription factor-its promoter has a strong ARE.
MMP 2-matrix metalloproteinase-2-a gene activated by intercellular aggregation.
Example 18
Gene expression array of MDA-MB-231-AR xenograft
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 microarrays were performed to determine changes in gene signature expression.
Results
The results shown in FIG. 15 demonstrate that activation of AR in MDA-MB-231-AR xenografts inhibits expression of more genes than they induced in these tumors. This pattern is unique in breast cancer cells and, unlike the gene expression results observed in prostate cancer cells, more genes are induced in prostate cancer cells than are suppressed (data not shown).
The results shown in FIG. 16 verify the microarray results shown in FIG. 15 by analyzing selected genes using real-time PCR TaqMan primers and probes in ABI 7900.
Example 19
Inhibiting growth of MCF-7-AR xenografts of formula VIII
MCF-7 cells stably transfected with AR using lentiviruses were implanted (2 million cells/mouse; n=5) into ovariectomized nude mice supplemented with 17β -estradiol (50 μg/day). Once the tumor reached 100mm 3 -200mm 3 Animals were randomized and treated with vehicle or 30 mg/kg/day of formula VIII. Tumor volumes and body weights were measured three times per week. At the end of the 5 week treatment, animals were sacrificed, tumors were weighed and stored for RNA and protein isolation and histological analysis. * Significant p<0.05。
In addition, uterine weights were measured in these xenograft studies and western blots of AR were probed from MCF-7 tumor xenografts.
Results
The graph shown in fig. 17 demonstrates the inhibition of triple positive breast cancers (ER, PR and HER 2) using formula VIII. The results show that formula VIII inhibits the growth of MCF-7 breast cancer cell xenografts by more than 50%.
The results shown in fig. 18 demonstrate that formula VIII inhibits uterine weight in these estrogen-supplemented animals.
The results shown in fig. 19 demonstrate that AR expression patterns in response to agonists (formula VIII) are similar to those observed in prostate cancer cells (data not shown).
Example 20
Up-regulating JNK phosphorylation in MCF7-AR tumors of formula VIII
Proteins from MCF-7-AR tumors treated with vehicle or compound of formula VIII were extracted and incubated with phosphorylated MAPK arrays to determine the effect of the compound of formula VIII on phosphorylation of various kinases.
Results
The results shown in FIG. 21 show that JNK phosphorylation in MCF-7-AR tumors is upregulated by treatment with compounds of formula VIII. JNK plays a key role in death receptor-mediated intrinsic and extrinsic apoptotic pathways. JNK activates apoptosis signaling by upregulating pro-apoptotic genes. The observed phosphorylation of the pro-apoptotic kinase JNK might suggest a possible mechanism explanation for anti-proliferation.
Example 21
Gene expression analysis of MDA-MB-231-AR and MCF-7-AR xenografts treated with formulas VIII and IX
RNA from MDA-MB-231-AR and MCF-7-AR tumors was subjected to microarray analysis to identify and compare changes in gene expression in ER-negative (MDA-MB-231-AR; triple negative) ER-positive (MCF-7-AR; triple positive) breast cancer tumors treated with the compound of formula VIII (30 mg/kg/day p.o., for 4 weeks). Affymetrix analysis of xenografts was performed on pooled samples of xenografts. The analysis included about 70,000 sequences and about 30,000 genes and their indicated variants, as well as micrornas. RNA was isolated and gene expression was assessed using a microarray (Affymetrix Human Gene ST 2.0 array). The expression of the gene in the sample treated with the compound of formula VIII was compared to the expression in the vehicle treated sample. Genes that are up-regulated or down-regulated by more than a factor of 2 are considered to be differentially regulated by compounds of formula VIII.
Results
Table 7 below shows the sum of up-and down-regulated genes in MDA-MB-231-AR and MCF-7-AR tumors.
TABLE 7
Type(s) Upregulation of Down-regulation of Sum total
MCF-7-AR 566 981 1547
MDA-MB-231-AR 720 816 1536
Of particular interest, of the 1547 regulatory genes identified in MCF-7-AR tumors and the 1536 regulatory genes identified in MDA-MB-231-AR tumors, the subset of overlapping genes is only 245 genes. The results indicate that formula VIII regulates different sets of genes in MCF-7-AR (ER positive; triple positive) and MDA-MB-231-AR (ER negative; triple negative) breast cancer cells.
Table 8 and Table 9 below show genes involved in breast tumorigenesis regulated by differences of formula VIII (at least 2-fold) in MDA-MB-231-AR tumors (Table 8) and MCF-7-AR tumors (Table 9). An indication of up or down is displayed in the rightmost column.
Table 8: breast cancer related genes regulated in MDA-MB-231-AR tumors
Table 9: in MCF-7-AR tumorsRegulated breast cancer-associated genes
The results shown in tables 7 and 8 demonstrate that SARM treatment (formula VIII) results in net down-regulation of genes in MDA-MB-231-AR tumors (N=1042 is inhibited; N=640 is induced; 2-2.5 fold-plus or-minus thresholds (note: curve is log fold change; follow-up RT-PCR shows 10-20 fold change). Well known androgen-dependent genes (e.g., FKP5 and MUC) 1 The method comprises the steps of carrying out a first treatment on the surface of the See table 10 below) increases, indicating penetration of SARMs into tumors. 29/36 known breast cancer-associated genes also showed a reduction, which provides a reasonable basis for the antiproliferative activity of formula VIII in ER-negative breast cancers.
Further analysis of MDA-MB-231-AR tumor results showed that formula VIII induced a known androgen response gene (Table 10 below). Thus, breast cancer-related genes such as β2-adrenergic receptor and PARP1 are inhibited by formula VIII; and the ARE-dependent gene is induced by the treatment of formula VIII.
Table 10
The results shown in tables 7 and 9 indicate that formula VIII does not have as strong gene inhibition in MCF-7-AR triple positive (ER positive) tumors as in triple negative (ER negative) tumors. Interestingly, however, MCF-7-AR analysis showed that the androgen-dependent gene was upregulated and the estrogen-dependent gene was inhibited (table 11 below), as verified by RT-PCR.
TABLE 11
The results shown in FIG. 20 verify the microarray results shown in the above analysis by analyzing the selected genes using real-time PCR TaqMan primers and probes in ABI 7900.
The results shown in figure 22 demonstrate the inhibition of triple negative breast cancer growth using formulas VIII and IX. In a triple negative breast cancer model using MDA-MB-231-AR cells in nude mice, formulas VIII and IX demonstrated about 85% TGI at all doses tried (5 mg/kg, 10mg/kg for formula VIII; 5mg/kg, 10mg/kg, 30mg/kg for formula IX).
The results shown in figure 23 demonstrate the inhibition of triple negative breast cancer using formulas VIII and IX. The tumor weight was also reduced for all doses of formula VIII and formula IX. Spleen enlargement was observed only in vehicle treated mice (680 mg versus 200mg-300mg in normal mice), possibly indicating prevention of tumor metastasis to the spleen by SARM.
In vitro data shown in MCF-7 cells with and without AR (fig. 25A-25E) support cofactors used by SARM-activated AR-sequesterable ERs. Addition of AR to MCF-7 cells increased the effect of 17β -estradiol (when not antagonized) on ER target genes PR and pS2, but antagonism was enhanced in this case (FIGS. 25B and 25D) by SARM or SARM+17β -estradiol (E2) alone compared to GFP (i.e., no AR; FIGS. 25A and 25C). FIG. 25E shows that SARM enhances the AR target gene even in the presence of 17β -estradiol.
Example 22
Xenograft experiments with IX
Xenograft experiments. NSG mice obtained from JAX laboratories were housed with 5 animals per cage and allowed free access to tap water and commercial rat feed (Harlan Teklad 22/5 rodent diet-8640). During the study, animals were maintained in the 12 hour light to dark cycle. Animals were anesthetized and 1mm was used 3 The BR-0001TNBC fragment was (approximately) subcutaneously implanted into NSG mice. Once the tumor size reached 100mm 3 -200mm 3 Animals were randomized and orally treated with vehicle control (polyethylene glycol: DMSO ratio of 9:1) or 10 mg/kg/day of formula IX (n=12) or enzalutamide. Weekly measurementTumor volumes were measured three times. Tumor volumes were calculated using the formula length x width x 0.5236. Once the tumor reaches more than 1500mm 3 -2000mm 3 Animals were sacrificed, tumors were weighed and stored for various analyses. Two regions of the same BR-0001 tumor (AR positive TNBC xenograft) were immunohistochemically stained with AR antibody (AR N20 from SCBT) (fig. 26A and 26B) and compared with AR negative (fig. 26C) TNBC as a negative control. Fig. 26A and 26B show that AR expression was consistent throughout formalin fixed, paraffin embedded (FFPE) tissue, while similar FFPEs showed no staining (no AR expression) in AR negative TNBC. The results of tumor xenograft efficacy experiments are provided in fig. 27A-27C, with fig. 27A and 27B being repeat experiments. In each experiment, formula IX (lower curve) produced some tumor growth inhibition for this AR positive TNBC tumor, while enzalutamide was indistinguishable from vehicle treatment (fig. 26A and 26B). Formula IX reduces tumor weight in experiment 2 by about 40%.
Example 23
Reduced Ki-67 staining in AR positive TNBC tumors in animals treated with formula IX
FIGS. 28A-28B show that Ki-67 staining was reduced by approximately 50% after 2 weeks of treatment. Tumors from repeat experiment 2 (fig. 27B) were fixed in formalin and paraffin embedded. Sections were cut and stained with Ki-67 antibody. Ki-67 positive cells in each section (total 200 cells were counted in each section) were counted and expressed as% stained cells, as shown in FIG. 28B. Ki-67 staining was reduced in tumors of animals treated with formula IX.
Example 24
Gene expression research and ChIP-SEQ research method in AR positive TNBC tumor xenograft
Chromatin immunoprecipitation assay (ChIP). Proteins were crosslinked by incubation with 1% formaldehyde (final concentration) for 10 min at 37 ℃. Tumors were homogenized using a probe-hand-held homogenizer. Cells were washed twice with 1 XPBS, scraped 1mL of protease inhibitor-containing PBS ([ aprotinin, leupeptin, anti-protease, benzamidine hydrochloride, and pepstatin each 1mg/mL ], 0.2mM phenylmethylsulfonyl fluoride, and 1mM sodium vanadate), precipitated and resuspended in SDS lysis buffer (1%SDS,10mM EDTA,50mM Tris-HCl [ pH 8.1 ]).
After lysis on ice for 10 minutes, the cell extract was sonicated in a cold chamber (Branson sonifier 250) at a constant duty cycle for 8 times, 10 seconds each, with an output of 3, and incubated on ice after each sonication. Fragments were pelleted at 13,000rpm for 10 min at 4℃and the supernatant diluted 10-fold with ChIP dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2mM EDTA, 16.7mM Tris Hcl[pH 8.1, 167mM NaCl). Protein was pre-clarified with 50. Mu.L of 1:1 protein A-agarose beads in TE. Aliquots (300. Mu.L) were retained as inputs while the remaining solution was incubated with 5. Mu.g of AR antibody (AR N20 SCBT) and 2. Mu.g of sheared salmon sperm DNA (Stratagene, la Jolla, calif.) at 4℃overnight with rotation.
protein-DNA-antibody complexes were precipitated by incubation with 100. Mu.L of 1:1 protein A-agarose gel beads and 2. Mu.g salmon sperm DNA for 2 hours at 4 ℃. The beads were precipitated and washed with low salt buffer (0.1% sodium dodecyl sulfate [ SDS ]]、1%Triton X-100、2mM EDTA、20mM Tris HCl[pH 8.1]Washed three times with 0.15M NaCl and 1 XDE (10 mM Tris HCl, 1mM EDTA); pH 8.0) was washed twice. By using 50. Mu.L of freshly prepared extraction buffer (1% SDS,0.1M NaHCO) 3 ) The beads were extracted three times to obtain a DNA-protein complex. Crosslinking of the DNA protein complex was reversed by incubation at 65 ℃ for 6 hours. DNA was extracted in 25. Mu.L final volume TE using QIAquick PCR purification kit (QIAGEN, varenchia (Valencia, calif.). Purified DNA was provided to tenaci university health science center molecular resource center (University of Tennessee Health Science Center Molecular Resource Center, UTHSC MRC) for next generation sequencing using an ionic proton sequencer.
RNA analysis and microarray. Tumors were homogenized, RNA isolated, purified and submitted to UTHSC MRC core facilities for microarray analysis (ST 2.0 array from Affymetrix).
Results
In the above gene expression studies, RNA was isolated from BR-0001TNBC tumors and the expression of the gene throughout the genome was measured by microarray (Affymetrix, ST2.0 array). In the ChIP-Seq study, chromatin immunoprecipitation was performed in untreated BR-0001 samples, and DNA immunoprecipitated with AR antibodies was sequenced using an ion torrent next generation sequencer. Promoters (-5 kb to +1 kb) showing about 20% AR occupancy were activated by androgens (mRNA increased >1.5 fold) (data not shown). Based on genomic enrichment analysis (GSEA), androgen therapy primarily affects cell cycle and metabolic processes (fig. 31). The expression of the TNBC subtype markers in fig. 30A and 30B suggests that the gene markers of LAR and MSL subtypes are highly expressed in SARM-treated tumors.
The gene expression data was compared with PAM50 to determine the tumor type to which BR-0001 belongs. The expression of the 50 genes required to classify breast cancer (Z scores) is given in fig. 29, where PAM50 indicates that the tumor belongs to basal-like breast cancer (BLBC) TNBC.
Triple Negative Breast Cancer (TNBC) is a heterogeneous group of breast cancers, the identification of its subtypes is critical for understanding the biological characteristics and clinical behavior of TNBC, and for developing personalized therapies. Based on 3,247 gene expression profiles from 21 breast cancer datasets, six TNBC subtypes were found, including 2 basal-like (BL 1 and BL 2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem cell-like (MSL) and Luminal Androgen Receptor (LAR) subtypes from 587 TNBC samples with unique gene expression patterns and bodies (Brian d.lehman et al, j.clin.invest.2011, volume 121, 7, pages 2750-2767). Cell line models representing each TNBC subtype also showed different sensitivities to targeted therapeutic agents. The gene expression data were compared to published genes (Pietenpol group) and BLBC was classified into sub-classifications. FIGS. 30A-30B depict the Pietenpol classification of TNBC indicating that BR-0001 tumors are LAR and MSL subtypes.
Example 25
Gene expression changes in AR positive TNBC xenograft tumors
FIG. 31 demonstrates that in BR-0001 tumors, formula IX upregulates gene expression. Approximately 4200 genes were up-regulated by formula IX compared to vehicle, and approximately 1170 genes were down-regulated by formula IX compared to vehicle. Formula IX recruits AR to 176 promoters (-5 kb to +1 kb). 20% of the promoters occupied by AR in response to formula IX also have genes up-regulated by formula IX. This suggests that these genes are direct targets of AR, rather than indirect effects. The inventive pathway analysis (http:// www.ingenuity.com/; QIAGEN, redwood City, calif.) indicated that genes involved in the cell cycle were altered by formula IX.
Example 26
Efficacy and safety of formula IX on metastatic or locally advanced ER+/AR+ Breast Cancer (BC) in postmenopausal women
Study design: this is an open-label, multicentric, multinational, randomized, parallel design phase 2 study and was used to evaluate the efficacy and safety of formula IX in postmenopausal subjects with er+/ar+bc. Subjects were randomized to receive 9mg or 18mg of formula IX administered orally (PO) per day for up to 24 months. Each dose group will be treated independently and the efficacy of each dose group is assessed using Simon r., "Optimal two-stage designs for Phase 2clinical trials", "Controlled Clinical Trials, 1989; volume 10: pages 1-10). Subjects will be randomized to one of the two dose groups in a 1:1 fashion.
Randomization will be stratified by subjects exhibiting only bone metastases and all other subjects, and further stratified by setting immediately prior to treatment (adjuvant setting or metastasis setting) in order to balance the proportion of subjects with these manifestation characteristics in each dose group. A statistical comparison of the two dose groups was not intended, but rather a determination was made as to whether one or both doses would produce acceptable Clinical Benefit Remission (CBR), which was defined as 30% agreement with acceptable safety profiles in the 24 th week according to the proportion of RECIST 1.1 to CR, PR or SD in an evaluable subject (i.e., a subject with centrally confirmed ar+ and receiving at least one study drug). Considering such results, future exploration of formula IX in ER+/AR+BC will be warranted at this dose level.
Thirty-six to eighty-eight (36-88) subjects with centrally confirmed ar+ who received at least one dose of study drug (evaluable subjects) would be required for primary efficacy analysis purposes and would be a subset of the Full Analysis Set (FAS). Thirty-six to one hundred-eighteen (36-118) subjects (including surrogate subjects) were randomized in a 1:1 fashion to receive daily oral doses of 9mg or 18mg of formula IX. 30 of the above subjects can be considered as surrogate subjects to address the lack of concentrated confirmation of ar+ status, or randomized but not study drug-receiving rare subjects (assuming 25% of enrolled subjects are not rated for primary efficacy analysis). Other statistical parameters as part of the sample size calculation were α=0.025 (one-sided) and test efficacy=90%. The first phase in each study group will be evaluated in the first 18 evaluable subjects. If at least 3/18 of the subjects reached CB (defined as CR, PR or SD) at week 24, the group will proceed to the second phase of recruitment until a total of 44 evaluable subjects per group. Otherwise, the group will be interrupted due to lack of efficacy. If at least 9/44 of the subjects in this group reached CB at week 24, then a statistical significance would be declared, i.e., it is more likely to reject the zero hypothesis of unacceptably low CBR+.10%, while supporting an alternative hypothesis indicating a higher ratio+.30%.
Subjects not centrally confirming ar+ can continue to participate in the trial but will not participate in the primary efficacy analysis-these subjects will contribute to the secondary and tertiary analysis.
Subjects in the 18mg treatment group who experienced Adverse Events (AEs) of intensity no less than grade 3 (national cancer institute-adverse events common terminology standard [ NCI-CTCAE ], version 4.0) and/or intolerance, based on the medical judgment of the researcher and confirmed by the research medical inspector, can either reduce the dose from 18mg daily to 9mg or undergo drug discontinuation. Drug discontinuation can last for up to 5 days after which the subject must re-prime or discontinue the study with study drug (18 mg or 9 mg). In the case of dose reduction, once AE has resolved or intensity reduced to grade 1, the subject may be re-challenged with 18mg or maintained at 9mg at the discretion of the investigator.
Subjects experiencing an AE of intensity > 3 grade (NCI-CTCAE 4.0) and/or intolerance in the 9mg treatment group may be subjected to drug disruption based on the medical judgment of the researcher and confirmed by the research medical inspector. Drug discontinuation may last for up to 5 days after which the subject must re-prime or discontinue the study with study drug (9 mg).
For safety analysis, subjects will be analyzed in the treatment group where dosing was initiated. For efficacy analysis, subjects will be analyzed according to their randomized treatment group profile.
Subjects exhibiting CB will receive treatment for up to 24 months from the randomized date (as long as they continue to exhibit CB during treatment during these 24 months). Subjects who continue to exhibit CB in study treatment at 24 months will be proposed to continue safety extension studies according to a separate regimen. For safety purposes, all subjects will receive a follow-up for one month after receiving the last dose of formula IX.
For safety purposes, all subjects will receive a follow-up for one month after receiving the last dose of formula IX.
Target population: adult postmenopausal women with metastatic or recurrent locally advanced er+/ar+bc.
Duration of study: the duration of the study was estimated to be 3 years.
Description of the agent or intervention: three (3) capsules of formula IX 3.0mg were orally administered with or without food at about the same time per day at a daily dose of 9mg, or six (6) capsules of formula IX 3.0mg at a daily dose of 18mg.
Potential benefits: based on the trial of example 9, 9mg of formula IX was studied once daily in 22 postmenopausal women with metastatic er+bc and previously responsive to hormone therapy. Primary endpoints of 17 AR positive subjects were assessed. 6 of these 17 subjects exhibited CB (SD) at 6 months. In a subject exhibiting SD (RECIST 1.1), 27% tumor regression was confirmed in a single target lesion. A total of 7 subjects (1 subject with unknown AR status) reached CB at 6 months. Of the 7 subjects who reached CB at 6 months, the Time To Progress (TTP) was estimated to be 10.2 months. The results also show that after a median duration of 81 days of study, 41% (9/22) of all subjects reached CB as optimal relief, and also had increased PSA, which appears to be an indicator of AR activity. By the time the protocol was finalized, the study was still in progress, with the disease in one subject remaining stable after 336 days.
Preclinical data of formula IX indicate that it is also anabolic in bone and reduces bone turnover markers. Treatment with formula IX may reduce bone turnover compared to other hormone therapies used to treat hormone receptor positive BC. A stronger bone microenvironment may reduce metastasis to bone or delay the time of a bone related event.
Therapeutic efficacy targets
The main therapeutic objective of this test was to estimate CBR (defined as complete relief [ CR ], partial relief [ PR ] or SD) (according to RECIST 1.1) at week 24 of daily oral administration of 9mg of formula IX and 18mg of formula IX in subjects with estrogen receptor positive and androgen receptor positive (er+/ar+) BC and with a centrally confirmed ar+ status.
The secondary efficacy objective was to estimate CBR (according to RECIST 1.1) at week 24 given 9mg and 18mg of formula IX in all randomized subjects (full analysis set [ FAS ]) receiving at least one dose of study drug, regardless of AR status as determined by the central laboratory.
Additional secondary efficacy targets apply to both centrally confirmed ar+ subjects (an evaluable subset of FAS) and to all subjects in FAS: (a) Objective remission rates (ORR; defined as CR or PR) at week 24 (according to RECIST 1.1) were estimated at 9mg and 18mg of formula IX; (b) Estimating the optimal overall remission rate (BOR) of 9mg and 18mg of formula IX; (c) Estimating Progression Free Survival (PFS) of subjects receiving 9mg and 18mg of formula IX; (d) estimating TTP in subjects receiving 9mg and 18mg of formula IX; (e) The duration of remission (time from recording tumor remission to disease progression or death) was estimated for subjects receiving 9mg and 18mg of formula IX.
Tertiary targets apply to both centrally confirmed ar+ subjects (an evaluable subset of FAS) and all subjects in FAS: (a) assessing the effect of 9mg and 18mg of formula IX on serum PSA; (b) Assessing the effect of 9mg and 18mg of formula IX on quality of life (QoL) as measured by EQ-5D-5L; (c) Assessing the effect of 9mg and 18mg of formula IX on Circulating Tumor Cells (CTCs); (d) evaluating the effect of the duration of the previous CB on the result; (e) Evaluating the effect of time to transition from diagnosis to randomization on the result; (f) describes the effect of 9mg and 18mg of formula IX on tumor volume; (g) The effect of plasma concentrations of glucuronide of formula IX and formula IX on CBR was assessed at 24 weeks.
The safety objective is to describe the safety profile of 9mg and 18mg per day of formula IX orally in subjects with er+/ar+bc and with centrally confirmed ar+ and in all randomized and treated subjects.
Pharmacokinetic targets: the plasma concentrations of the glucuronides of formula IX and formula IX at each evaluation time point are described.
Preparing, packaging and labeling: the 3.0mg soft capsule of formula IX will be provided in the form of an opaque, white to off-white, oval-shaped No. 5 soft capsule containing 3.0mg of formula IX. The liquid soft capsule fill consists of formula IX dissolved in polyethylene glycol 400. 3.0mg of formula IX soft capsules are packaged in blister packs. Each blister pack will contain enough study drug to be administered for one (1) week. At randomization (visit 2) and visit 3, visit 4 and visit 5, subjects will be provided with a carton containing 7 blister packs of study medication, corresponding to a 7 week dose. At visit 6, 8, 9, 10, 11, 12 and 13, to accommodate the visit schedule every 12 weeks (+ -7 days), subjects will receive two cartons of study medication (7 blister packs per carton) to cover 14 weeks of study treatment. At each visit, the subject was asked to carry a carton containing all blister packs.
Each blister pack will consist of an appropriate number of blister strips (1 blister for the 9mg treatment group and 2 blisters for the 18mg treatment group) enclosed in a child resistant heat seal card. The blister strip consists of a PVC/ACLAR substrate and an aluminum foil/PVC/PVAC copolymer and a polymethacrylate (product contact) cover. The perforations on the back of the heat seal card overlie the foil cover. To remove the study drug, the subject will release the appropriate perforation by pressing a release button on the inside of the card. Once released, the perforations may be removed and the study drug pushed through the foil.
Pharmacokinetic assessment
Blood samples for pharmacokinetic assessment were collected at baseline (pre-dose), at visit 3 (week 6), at visit 5 (week 18) and at visit 6 (week 24). On each of these days, one blood sample was collected in 6mL of K2-ethylenediamine tetraacetic acid (EDTA) blood collection tube. The exact time (hh: mm) and date of each blood sample collection will be recorded on an electronic case report form. At baseline visit, blood samples should be taken before subjects receive their first dose of formula IX. At visit 3 (week 6), visit 5 (week 18) and visit 6 (week 24), the date and approximate time of administration of the last dose of formula IX prior to blood sample collection should be recorded; that is, it should be noted whether the subject took the previous dose in the morning or in the evening of the previous day. Immediately after collection, the tube was gently inverted several times to mix the anticoagulant with the blood sample.
The blood sample is kept on wet ice (ice bags in a water bath are also acceptable) for up to 20 minutes until processing. The plasma fraction was isolated by placing the collection tube in a centrifuge and centrifuging at 1,500xg for 10 minutes. The plasma fraction was removed by pipette and split into two 2mL polypropylene transfer bottles (each tube receiving approximately equal aliquots).
All sample collection tubes and freezer tubes will be clearly labeled in a manner that identifies the subject, study number, visit number, and freezer tube sample letters. The label is secured to the freezer pipe in a manner that prevents the label from falling off after freezing. The sample will be stored in a refrigerator at-20 ℃ or less. The samples will be transported in insulated containers with enough dry ice to ensure that they remain frozen.
Any plasma sample remaining after completion of the pharmacokinetic analysis outlined in the protocol can be used to identify and quantify the metabolites of formula IX.
Example 27
Curative effect and safety of formula IX on advanced androgen receptor positive triple negative breast cancer (AR+TNBC)
Ongoing and completed clinical trials using formula IX: twenty-one clinical trials using stages 1, 2 and 3 of formula IX have been completed or are ongoing. These include:
1. regimen G100401, phase 1 single escalation dose study in 96 healthy young male volunteers; 2. scheme G100402, a multiple increasing dose study at stage 1 in 50 healthy young male volunteers and 23 older male volunteers with obese trunk; 3. regimen G100503, phase 1 single dose pharmacokinetic study, the effect of dosing regimen simulating a slow release formulation as an immediate release formulation was evaluated in 18 healthy young male volunteers and 18 postmenopausal women; 4. scheme G100506, phase 1 single dose pharmacokinetic study, assessing relative bioavailability of 3mg hard shell capsule formulation to be used during continuous clinical development, and assessing the effect of food on the pharmacokinetics of 3mg soft capsule formulation in 27 healthy young male volunteers; 5. scheme 006, phase 1 single and multi-dose pharmacokinetic studies in 24 postmenopausal japanese females; 6. regimen G200501, phase 2 study to assess lean body mass and body function in 60 postmenopausal women and 60 elderly men; 7. scheme 003, phase 1b study in 44 postmenopausal women; 8. regimen G200502, phase 2b study assessing lean body mass and body function in 159 men with cancer and postmenopausal women; 9. scheme G100511, phase 1 study to evaluate the effect of severe kidney injury on the pharmacokinetics of formula IX; 10. scheme G100508, phase 1 study to evaluate the effect of mild and moderate liver injury on formula IX pharmacokinetics; 11. scheme G100509, phase 1 mass balance study of formula IX in healthy volunteers; 12. regimen G100507 phase 1 study to evaluate the pharmacokinetics and absolute oral bioavailability of IX in caucasian and african american men and women; 13. scheme G100510, single dose, randomized, double blind, comparative, positive and placebo controlled, four-phase crossover phase 1 study of the Electrocardiographic (ECG) effect of formula IX on healthy male and female subjects for therapeutic and super-therapeutic doses: comprehensive ECG testing; 14. scheme G100512, phase 1 study to evaluate the effect of ketoconazole (cytochrome P450, family 3, subfamily a [ CYP3A4] inhibitor) on the pharmacokinetics of formula IX; 15. scheme G100513, phase 1 study to evaluate the effect of rifampicin (CYP 3A4 inducer) on the pharmacokinetics of formula IX; 16. scheme G100514, evaluation of pharmacokinetic drugs of formula IX and celecoxib (CYP 2C 9) phase 1 study of drug interactions; 17. scheme G100515, evaluation of pharmacokinetic drugs of formula IX and probenecid (UGT 2B 7) phase 1 study of drug interactions; 18. scheme G100516, phase 1 study of drug interactions to evaluate pharmacokinetic drugs of formula IX and rosuvastatin (breast cancer resistance protein [ BCRP ]); 19. regimen G300504, a phase 3 randomized, double-blind, placebo-controlled study in 321 non-small cell lung cancer subjects receiving first-line platinum plus taxane chemotherapy with respect to the effect of formula IX on muscle atrophy; 20. regimen G300505, a phase 3 randomized, double-blind, placebo-controlled study on the effect of formula IX on muscle atrophy in 320 non-small cell lung cancer subjects receiving first-line platinum plus non-taxane chemotherapy; 21. scheme G200801, an ongoing phase 2 open label study, examined the AR status and the activity of hormone therapy of formula IX in 22 women with ER positive metastatic breast cancer and who previously responded to hormone therapy.
Dosage of 18 mg: formula IX has been evaluated in 21 completed and ongoing clinical studies, which recruited a total of more than 1500 subjects. Formula IX is generally well tolerated, including single doses up to 100mg and multiple doses up to 30mg, once daily for up to 14 days. In longer studies, formula IX was also generally well tolerated, including doses of 1mg, 3mg and 9mg per day for up to 184 days.
Previous clinical studies have shown that healthy male volunteers have good tolerance to daily doses of up to 30mg of formula IX. Daily doses of 10mg and 30mg were evaluated for up to 14 days in regimen G100402. Elevated alanine Aminotransferase (ALT), any elevation beyond the upper limit of normal [ ULN ], is the most common Adverse Event (AE) experienced. In the 10mg dose group, no subjects discontinued the study due to ALT elevation. In the 30mg dose group, 6 subjects experienced an ALT increase of more than twice ULN.
The prevention and treatment of muscle atrophy (cachexia) in subjects with advanced non-small cell lung cancer and undergoing chemotherapy by 3mg of formula IX administered daily was evaluated in two completed phase 3 trials in more than 600 subjects. In both studies 3mg of formula IX increased lean body mass and was safe and well tolerated when administered for up to 168 days. Subjects in the formula IX and placebo groups experienced similar AEs, and these AEs were consistent with the background chemotherapy regimen.
While 3mg of formula IX was selected for the completed phase 3 project due to its anabolic activity in the muscle, in the ongoing phase 2 trial of er+ and ar+ metastatic breast cancer, a dose of 9mg once daily was selected for hormone therapy to achieve a higher exposure that was safe and more likely to be effective in women with advanced breast cancer. 7 of 22 subjects with advanced, largely pretreated (hormone therapy, radiation therapy and chemotherapy) breast cancer showed Clinical Benefit (CB) at 6 months (disease stabilization [ SD ]). In 1 subject who reached SD (according to solid tumor remission evaluation criteria [ RECIST ], version 1.12), 27% tumor regression was confirmed. In agreement with previous studies, formula IX remained safe and well tolerated (see example 9).
The reduction of Sex Hormone Binding Globulin (SHBG) has been identified as one of the most sensitive serum biomarkers for AR signaling in healthy volunteers and patients. In the G100402 regimen (listed as trial #2 above), SHBG was reduced by 15.1%, 15.6%, 18.2% and 18.4%, respectively, in young healthy volunteers receiving 1mg, 3mg, 10mg and 30mg of formula IX orally daily for 14 days, indicating that doses of 10mg and above maximally stimulated AR activity.
Dosing of formula IX at 15mg-20mg daily can provide therapeutic benefit in hormone receptor positive breast cancer by two separate mechanisms: activating AR and inhibiting progesterone receptor, thereby enhancing potential therapeutic effects. Expression of the progesterone receptor in cancer stem cells has been shown to be associated with proliferation of cancer epithelial cells, and inhibition of progesterone receptor activity is now considered a novel approach to the treatment of breast cancer. Thus, higher doses of formula IX may provide dual antiproliferative effects in breast cancer. In TNBC, a dose of 15mg-20mg per day should be able to saturate the AR, potentially providing better efficacy than lower doses that partially occupy the AR and do not have any progesterone receptor inhibitory effect.
Based on safety data collected so far in preclinical and clinical settings, 18mg doses are expected to be safe and generally well tolerated. However, in the event that the subject has grade 3 or higher toxicity, the 18mg dose may be reduced to 9mg until AE regresses or the remaining treatment is performed at the discretion of the researcher. The 9mg dose has been previously studied in postmenopausal women with metastatic breast cancer and is safe and well tolerated.
In TNBC patients, the 18mg dose is superior to the lower dose due to the aggressive phenotype and poor prognosis of the disease. Based on preclinical data, the 18mg dose is more likely to saturate the AR and may lead to better clinical outcome than lower doses without receptor saturation or progesterone receptor inhibition.
The 18mg dose may provide better efficacy in TNBC without compromising the safety of the subject. However, in the event that the subject has grade 3 or higher toxicity, the 18mg dose may be reduced to 9mg until AE regresses or the remaining treatment is performed at the discretion of the researcher. The 9mg dose has been previously studied in postmenopausal women with metastatic breast cancer and is safe and well tolerated.
In TNBC patients, the 18mg dose is superior to the lower dose due to the aggressive phenotype and poor prognosis of the disease. Based on preclinical data, the 18mg dose is more likely to saturate the AR and may lead to better clinical outcome than lower doses without receptor saturation or progesterone receptor inhibition. The 18mg dose may provide better efficacy in TNBC without compromising the safety of the subject.
Study design: this is an open-label, multicentric, multinational phase 2 study that evaluates the efficacy and safety of formula IX in female subjects with androgen receptor positive triple negative breast cancer (ar+tnbc). The subject will take (PO) 18mg of formula IX orally daily for up to 12 months. The two-stage (optimal) design of simon will be used to evaluate the primary efficacy and will require up to 41 evaluable subjects; i.e., subjects with centrally confirmed ar+ and receiving at least one dose of study drug. To obtain these numbers of evaluable subjects, 21 to 55 subjects, including excess recruiters (see below), were enrolled to receive a daily oral dose of 18mg of formula IX. 14 of the foregoing subjects may be overrated recruiters to allow replacement subjects to address cases of lack of centrally confirmed ar+ status, or rare subjects that have been recruited but not received study medication. The test compares unacceptably low Clinical Benefit Rates (CBR) of 5% or less to CBR more consistent with 20% or more. The first stage will be evaluated in the first 21 evaluable subjects. If at week 16, at least 2/21 of the subjects achieved Clinical Benefit (CB) (defined as complete remission [ CR ], partial remission [ PR ] or disease stabilization [ SD ] according to the solid tumor remission evaluation criteria [ RECIST ], version 1.12), the trial would proceed to a second phase of recruiting up to a total of 41 subjects in the evaluable subset of the Full Analysis Set (FAS). Otherwise, the trial will be discontinued due to lack of efficacy.
Subjects not confirming ar+ may continue to participate in the trial but will not participate in the primary efficacy analysis-these subjects will contribute to the secondary and tertiary analysis. Subjects experiencing grade 3 Adverse Events (AEs) (national cancer institute-adverse events common term standard [ NCI-CTCAE ], version 4.0) and/or intolerance, based on the medical judgment of the researcher and confirmed by the research medical inspector, may either reduce the dose from 18mg to 9mg daily or discontinue the medication. Subjects exhibiting Clinical Benefit (CB) will receive up to 12 months of treatment from the day of receiving the first dose of study treatment (as long as they continue to exhibit CB during these 12 months of treatment). Subjects who continue to demonstrate benefit relief in study treatment at 12 months will be proposed to continue safety extension studies according to a separate regimen. For safety purposes, all subjects will receive a follow-up for one month after receiving the last dose of formula IX.
The main therapeutic objective of this trial was to estimate the Clinical Benefit Rate (CBR) (defined as Complete Remission (CR), partial Remission (PR) or disease Stabilization (SD)) at week 16 of 18mg of formula IX given orally (PO) per day in subjects with TNBC and with centrally confirmed ar+ status (according to RECIST 1.1).
Secondary efficacy targets: CBR at week 16 was estimated at 18mg of formula IX given in all enrolled subjects receiving at least one dose of study drug (i.e., full Analysis Set (FAS)) regardless of the AR status determined by the central laboratory.
The following secondary efficacy targets apply to both centrally validated ar+ subjects (an evaluable subset of FAS) and to all subjects in FAS:
the objective remission rate (ORR; defined as CR or PR) at week 16 (according to RECIST 1.1) was estimated at 18mg of formula IX.
Estimated 18mg CBR of formula IX at 24 weeks.
Estimated 18mg ORR of formula IX at 24 weeks (defined as CR or PR).
The best overall remission rate (BOR) of 18mg formula IX was estimated.
Estimated Progression Free Survival (PFS) of subjects receiving 18mg of formula IX.
Estimated Time To Progression (TTP) for subjects receiving 18mg of formula IX.
The duration of remission (time from recording tumor remission to disease progression or death) was estimated for subjects receiving 18mg of formula IX.
Three-stage target: the following tertiary efficacy targets apply to both centrally validated ar+ subjects (an evaluable subset of FAS) and to all subjects in FAS:
the effect of 18mg of formula IX on serum Prostate Specific Antigen (PSA) was evaluated.
Evaluate the effect of 18mg of formula IX on quality of life (QoL) measured by EQ-5D-5L.
Evaluate the effect of 18mg of formula IX on Circulating Tumor Cells (CTCs).
Evaluate the effect of the duration of the previous CB on the result.
The effect of the time to transition from diagnosis to study entry on the results was evaluated.
Describe the effect of 18mg of formula IX on tumor volume.
The effect of plasma concentrations of glucuronide of formula IX and formula IX on CBR at 16 weeks and 24 weeks was assessed.
Security objective: safety profiles of 18mg per day of formula IX in subjects with TNBC and with centrally confirmed ar+ and in all enrolled and treated subjects are described.
Pharmacokinetic targets: the plasma concentrations of the glucuronides of formula IX and formula IX at each evaluation time point are described.
Target population: adult females with advanced TNBC and with centrally confirmed ar+.
Subject inclusion criteria: qualifying for inclusion in the study
Must meet all of the following criteria:
capable and willing to provide voluntary, written and signed informed consent;
women aged 18 or more;
women with TNBC who have received at least one but not more than two previous chemotherapy regimens for treating advanced or metastatic TNBC;
confirm ar+ (defined as ≡10% nuclear AR staining by immunohistochemistry [ IHC ]) in primary or metastatic lesions TNBC, assessed by local laboratory or by medical history during screening;
TNBC was confirmed by medical history as: human epidermal growth factor receptor 2[ HER2] -negative (confirmed by IHC 0, 1+ regardless of fluorescent in situ hybridization [ FISH ] ratio; IHC2+ with a FISH ratio below 2.0 or a HER2 gene copy below 6.0; FISH ratio of 0 when positive and negative in situ hybridization [ ISH ] controls are present, indicating gene deletion); estrogen Receptor (ER) negative (confirmed as positive tumor nuclei with ER expression less than or equal to 1%); progesterone receptor negative (confirmed that progesterone receptor expression is less than or equal to 1% of positive tumor nuclei);
availability of paraffin embedded or formalin fixed tumor tissue; alternatively, a minimum of 10 and a maximum of 20 sections of preserved tumor tissue were used for central laboratory confirmation of AR status and molecular typing. Metastatic tumor tissue is preferred if possible;
the subject must have a measurable disease or a disease where only bone is not measurable, which can be assessed according to RECIST 1.1;
at screening and group entry, the expression status of eastern tumor cooperative group (ECOG) was 0 or 1;
women with fertility potential (premenopausal or postmenopausal amenorrhea for less than 12 months and without sterilization surgery) were negative in pregnancy tests no more than 7 days prior to the first dose of study treatment;
For women with fertility potential and active sexual life, agreeing to use efficient, non-hormonal forms of contraception during and at least 6 months after completion of the study treatment; alternatively, a viable male partner would like and be able to use effective, non-hormonal forms of contraceptive (barrier contraception in combination with spermicidal ointment, or surgical sterilization) during the study treatment and for at least 6 months after completion;
organ function is sufficient, as follows: absolute neutrophil count greater than or equal to 1,500 cells/mm 3 The method comprises the steps of carrying out a first treatment on the surface of the Platelet count is greater than or equal to 100,000 cells/mm 3 The method comprises the steps of carrying out a first treatment on the surface of the Hemoglobin is more than or equal to 9g/dL; serum aspartate Aminotransferase (AST) and alanine Aminotransferase (ALT) are 2.5 times the upper limit of the normal range (ULN) (or 5 times if liver metastasis is present); total serum bilirubin is less than or equal to 2.0 XULN (unless the subject has been documented to have Gilbert syndrome); alkaline phosphatase level +.ltoreq.2.5XULN (+.5XULN in subjects with liver metastases); serum creatinine<2.0mg/dL or 177. Mu. Mol/L; international Normalized Ratio (INR) or activated partial thromboplastin time (aPTT)<1.5 XULN (unless anticoagulated at the time of screening);
capable of swallowing capsules;
any toxicity caused by previous chemotherapy has been eliminated or grade 1 (NCI-CTCAE, version 4.0).
Formulation, packaging and labelling
The 3.0mg formula IX soft capsule will be provided in the form of an opaque, white to off-white, oval shaped soft capsule No. 5. The liquid soft capsule fill consists of formula IX dissolved in polyethylene glycol 400. Dosage instructions will be provided on the study drug label and in the subject information table.
Example 28
Formula IX reduces the growth of HER2 positive tumors
Method
HCI-007 tumor mass (1 mm) 3 ) The mice were surgically implanted (one for each) subcutaneously on the flank of NSG mice. At the same time, 17 β -estradiol pellets (Innovative Research of America) were implanted subcutaneously in each mouse. Grow the tumor and reach about 100mm 3 Volume (i.w.w.0.526). Mice were randomized and orally treated with vehicle (15% DMSO+85% PEG-300), formula IX (10 mg/kg) or enzalutamide (20 mg/kg). Tumor volume was measured weekly and expressed as change in tumor volume (fig. 32). Mice were sacrificed and tumors were stored for further analysis.
Results
As shown in figure 32, ER-positive, PR-positive, HER 2-positive and AR-positive tumors grew comparably in animals treated with vehicle and enzalutamide, while tumors in mice treated with formula IX grew slowly. Tumors of animals treated with formula IX regress within the first 7 days and then begin to slowly increase. (see also example 30 and fig. 35A to 35C).
Conclusion(s)
These results support previous results observed in MCF-7 cell xenografts, indicating that formula IX reduced the growth of HER2 positive tumors. (see example 30.)
Example 29
Formula IX inhibits triple positive (ER, PR, HER 2) from HCI-013 patient source and also expresses xenografts of AR Growth
Method
HCI-013 tumor mass (1 mm) 3 ) The mice were surgically implanted (one for each) subcutaneously on the flank of NSG mice. Grow the tumor and reach about 100mm 3 Volume (i.w.w.0.526). Mice were randomized and orally treated with vehicle (15% DMSO+85% PEG-300) or formula IX (10 mg/kg). Tumor volume was measured weekly and expressed as% change in tumor volume. Mice were sacrificed and tumors were weighed and stored for further analysis.
Results
As depicted in fig. 33A and 33B, the triple positive HER2 tumors of animals treated with vehicle grew robustly, whereas the tumors of mice treated with formula IX grew very slowly. Tumors of animals treated with formula IX did not grow significantly throughout the experiment, indicating the presence of almost 100% Tumor Growth Inhibition (TGI) (fig. 33A). Tumor volume results are reflected in the tumor weights observed at the end of the experiment (fig. 33B).
Conclusion(s)
These results indicate that formula IX is extremely effective in tumors that are triple positive (express ER, PR and HER 2) and also express AR. See also example 30, wherein HCI-13 is further characterized as including genotyping of ER in tumors, which genotyping shows that Y537S mutant ER is present in HCI-13 tumors.
Example 30
Inhibition of proliferation and tissue of patient-derived xenografts (PDX) and tissues expressing wild-type and mutant refractory ER Growth
In this study, it was found that proliferation and growth of patient-derived xenografts (PDX) and tissues expressing wild-type and mutant refractory ERs were inhibited by AR agonists and tissue-selective AR modulators (SARMs), but not by antagonists. AR agonists inhibit the growth of these tumors by reprogramming the ER cistron group and subsequently inhibiting ER function and by altering the phosphokinase group (phosphokinase) characteristics.
Materials and methods
Reagent(s)
TaqMan PCR primers and fluorescent probes, master mix, and Cells-to-Ct reagents were all purchased from Life Technologies (Carlsbad, calif.). Cell culture medium and activated charcoal desorbed fetal bovine serum (csFBS) was purchased from Fisher Scientific (Waltham, MA). FBS is available from Hyclone (San Angelo, TX) of Saint An Jiluo, texas. The AR-N20 antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, calif.). Enzalutamide was purchased from MedKoo Biosciences (Chapel Hill, N.C.) in Church mountain, N.Kaolina. ER-alpha (D8H 8) antibodies were obtained from Cell Signaling (Danvers, mass.). Actin antibodies, DHT, tamoxifen, and fulvestrant were purchased from Sigma (st louis, MO). Vetspon dental cubes/sponges (Patterson Veterinary Supplies inc., NC 0654350) are available from Fisher Scientific (waltherm, ma). Epidermal Growth Factor (EGF) was purchased from R & D systems (Minneapolis, MN)), phorbol 12-myristate 13-acetate (PMA) was purchased from Acros organics, and 17β -estradiol was purchased from Tocris (Bristol, UK). All other reagents used were analytical grade.
Cell culture
MCF-7 and ZR-75-1 cells were obtained from the American type culture Collection (ATCC, manassas, va.). Cells were cultured according to ATCC recommendations.
Growth assay
Cells were seeded at different densities in 96-well plate growth medium. Cells were treated as shown and viability was measured using a sulfonylrhodamine B (SRB) or cell numbers were counted using a coulter counter.
Transfection
MCF-7 stable cells were generated by lentiviral infection with Green Fluorescent Protein (GFP) or AR cloning into the pLenti U6 Pgk-puro vector k-puro vector, as previously described (Narayanan et al, (2014), PLoS One 9, page e 103202; yang et al, (2010), cancRes, volume 70, pages 8108-8116; yepsu et al, (2013), clin Cancer Res, volume 19, 20 th, pages 5613-5625).
Tumor xenograft experiments
All animal protocols were approved by the institutional animal care and use research committee of the University of Tennessee Health Science Center (UTHSC). Xenograft experiments were performed as previously disclosed (Narayanan et al, (2014), PLoS One 9, page e 103202). Simple and easyIn other words, 3 million MCF-7 cells were resuspended in 0.05ml MEM+10%FBS and 0.05ml Matrigel/nude mice and subcutaneously injected. Once the tumor size reached 100mm 3 -200mm 3 Animals were randomized and orally treated with the indicated drugs formulated in DMSO: PEG-300 (15:85). HCI-7, HCI-9 and HCI-13PDX are gifts donated by Alana Welm doctor (Henschel cancer institute, salt lake City, utah). HCIPDX tumor fragment (1 mm) 3 ) Female NOD SCID Gamma (NSG) mice were surgically implanted under the mammary fat pad. Tumor volumes were measured twice a week for MCF-7 xenografts and once or twice a week for HCIPDX. At the end of the study, animals were sacrificed, tumors excised, weighed and stored for various analyses.
Patient sample collection
Samples from breast cancer patients were collected under patient consent according to UTHSC Institutional Review Board (IRB) approved protocols. Samples were collected immediately after surgery in RPMI medium containing penicillin, streptomycin and amphotericin B and transported on ice to the laboratory. The tissues were minced and treated with collagenase for 2 hours. Digested tissue was washed with serum-free medium and frozen in liquid nitrogen in a freezing medium (5% dmso+95% fbs) or implanted under mammary fat pad of female NSG mice.
Sponge culture
To achieve 500mm HCI-13 tumor growth in female mice 3 -1000mm 3 Animals were then sacrificed and tumors excised for sponge culture. Patient samples frozen in a freezing medium in liquid nitrogen were used for sponge culture. Sponge culture was performed according to the protocol disclosed earlier (Dean et al, (2012), cell Cycle, volume 11, pages 2756-2761; hu et al, (2016), cancer Res, volume 76, pages 5881-5893; ochnik et al, (2014), menopause, volume 21, pages 79-88). Tumors were cut into small pieces (about 1mm 3 ) And incubated on presoaked gelatin sponge (5 fragments/sponge) in 12-well plates containing 1.5mL of medium (MEM+10% FBS+2mM L-glutamine+10. Mu.g/mL insulin+10. Mu.g/mL hydrocortisone+penicillin: streptomycin). For HCI-13Cultures were performed in triplicate and patient samples were cultured in duplicate. The pooled samples from each sponge (n=5/sponge) constitute one sample. The next day the medium was changed and treated as indicated. Tissues were harvested 3 days after treatment, RNA was extracted, and the expression of various genes was measured. Although the same procedure was used for samples obtained from breast cancer patients, samples were incubated in duplicate (n=5/sponge=one) instead of in triplicate as was done for HCI-13. The characteristics of the patient samples for PDX and sponge culture are provided in table 12.
Table 12
Microarray array
RNA was extracted from tumors and verified qualitatively and quantitatively. Total RNA from each sample (200 ng/sample; n=4/group) was amplified and labeled using the WT Plus kit from Affymetrix and processed according to the Affymetrix protocol. The array (human ST2.0, affymetrix, santa Clara, CA) was washed and stained on Affymetrix Fluidics station 450 and scanned on an Affymetrix GCS 3000 scanner.
Data from the microarray was normalized using the Affymetrix expression console (Affymetrix Expression Console). The mean, standard deviation and variance of each group were calculated. Fold changes in vehicle-treated samples were calculated and fold changes of 1.5 were used as cut-off values. Student's t-test was used to determine significance and a cutoff value of p-value <0.05 was used for significance discovery. The false discovery rate was calculated using the Benjamini & Hochberg method and a significant differential expression list was created using the cutoff value of FDR < 0.05. The gene candidate list was loaded into an original pathway analysis and genome enrichment analysis (GSEA) for further discovery. Microarray experiments were performed at the UTHSC Molecular Resource Center (MRC), and data analysis was performed by the UTHSC molecular bioinformatics (mhio) core facility.
Phosphorylated proteome
Frozen samples from HCI-13PDX treated with vehicle or formula IX were cut into 8 μm frozen sections and mounted on uncharged slides. Whole tissue lysates were prepared directly from tissue sections using a 1:1 mixture of T-PER (tissue protein extraction reagent; pierce, rockford, ill.) and 2 XTris-glycine SDS sample buffer (Invitrogen, calif.) supplemented with 5% 2-mercaptoethanol. The samples were boiled for 8 minutes and stored at-80 ℃ until aligned.
Samples and standard curves for internal quality assurance were printed onto nitrocellulose-coated slides (Grace Bio-labs, bend, OR) using an Ausson 2470 dot matrix instrument (Ausson BioSystems, billerica, mass.). The amount of protein in each sample was estimated using the selected array and using a Sypro Ruby Western blot stain (Molecular Probes, eugene, oreg.) protocol according to the manufacturer's instructions (Pin et al, (2014) Curr Protoc Prtein Sci, unit 2727). The remaining array was tested with a single primary antibody using an automated system (Dako Cytomation, carpinteria, CA) as described previously (baldeli et al, (2015), oncotarget, volume 6, pages 32368-32379). The array was first incubated with a Reblot antibody stripping solution (Chemicon, termekula, CA) and then washed 2 times in PBS and I-blocking solution (Tropix, bedford, MA) for a total of 4 hours. The array was probed with a total of 174 antibodies targeting a broad range of protein kinases and activating them by phosphorylation. Antibody specificity was tested for a set of cell lysates using standard immunoblotting. The selected arrays were stained with separate anti-rabbit or anti-mouse biotinylated secondary antibodies (Vector Laboratories inc., burlingame, CA) and Dako Cytomation, california (Carpinteria, CA), and used as non-specific binding/background subtraction negative controls.
Commercially available Signal Amplification System (CSA; dako Cytomation) and streptavidin conjugated IRDye 680 secondary antibody (LI-COR Biosciences, lincoln, NE) were used as signal detection methods. In the case of a laser-based PowerScanner (TECAN,switzerland) and analyzed for data as previously described (baldeli et al, (2015), oncotarget, volume 6, pages 32368-32379) using MicroVigene software version 5.1 (vipinge Tech, carlisle, MA). Reproducibility within and between assays has been previously reported (Pieopon et al, (2014), J-Proteome Res, vol.13, pp.2846-2855; rapkiewicz et al, (2007), cancer, vol.111, pp.173-184).
Chromatin immunoprecipitation assay (ChIP) -sequencing (ChIP-Seq)
HCI-13 xenograft samples were flash frozen and stored for ChIP sequencing analysis. ChIP-Seq studies were performed in either vehicle grown in NSG mice or HCI-13PDX treated with formula IX. ChIP was performed with ER (n=4/group) or AR (n=/group) antibodies and whole genome sequencing was performed on a NextSeq 500 sequencer. For ChIP, a standard SDS-based protocol was used, as already described (Carroll et al, (2005), cell, volume 122, pages 33-43). Briefly, whole cell lysates were prepared from tissues. Lysates were sonicated using a Covaris E210 machine (Covaris inc., woburn, MA) for 30 minutes per sample (set: 20% duty cycle, intensity 8, 200 cycles per pulse). Immunoprecipitation, washing and eluting the complex were performed on ER or AR. The DNA-protein complex was reverse cross-linked by incubation at 65 ℃ for 6 hours to overnight. After reverse cross-linking, the precipitated and imported DNA was purified using a QIAquick PCR purification column (Qiagen).
For library preparation, the ThruPLEX-FD preparation kit (Rubicon Genomics, ann Arbor, michigan) was used. For each library, 2ng-10ng DNA was used. After amplification, fragments of 200bp-600bp were selected using a Pippin Prep machine and using a cassette containing 2% agarose ethidium bromide (Sage Science, beverly, mass.). After size selection, the DNA was cleaned using Ampure beads and analyzed on a fragment analyzer (Advanced Analytical, ames, IA) in state of iowa. Sequencing was performed using the NextSeq 500 sequencing platform (Illumina, san Diego, CA). Human genome construction 19 (hg 19) was used as a reference genome. Sequencing data from ChIP experiments were aligned with human genomes using Bowtie. For peak detection MACS2 was used.
Immunohistochemistry
14 cases of invasive breast cancer of the luminal subtype B were randomly selected from formalin-fixed, paraffin-embedded samples available from the hospital pathology department tissue library at the northeast university. The luminal B classification of these samples is based on having greater than 1% erα expression and a Ki-67 marker index of greater than 20%. Samples had variable levels of PR expression (marker index, average 48.9, range 0-100) and other clinical pathology (Ki 67, average 38%, range 20-48%; norbukan grading, 1n=1, 2n=10, 3n=3). The use of these samples was approved by the university of northeast research institute medical ethics review board (Tohoku University School of Graduate Medicine Ethic review board) (2014-1-107). The tissue pieces were retrieved, sectioned at a thickness of 3 μm and fixed on a slide. To evaluate co-localization, mirror image slices were used. Slides were then subjected to ERα and AR staining (ERα,1:50 dilution, clone 6F11, leica; AR,1:50 dilution, clone AR441, dako) as previously described (McNamara et al, (2013), cancer Sci, volume 104, pages 639-646; niikawa et al, (2008), clin Cancer Res, volume 14, pages 4417-4426) using immunohistochemistry.
Statistics
Statistical analysis was performed using GraphPad prism software (La Jolla, CA). Experiments containing two groups were analyzed by a simple t-test, while experiments containing more than two groups were analyzed by one-way analysis of variance (ANOVA) and Tukey post-hoc test. Microarrays, phosphorylated proteomes, and ChIP-Seq statistical analysis are described under the respective methods.
All in vitro experiments were performed in at least triplicate. Data are presented as mean ± s.e.
Results
SARM of formula IX is an AR agonist that binds to and activates AR at less than 10nM (Narayanan et al, (2014), PLoS One 9, page e 103202; ponnusamy et al, (2017) Hum Mol Genet. 26, vol. 13:2526-2540). Clinically, formula IX (see example 27 for a partial list) has been evaluated in more than 1000 patients in multiple clinical trials and has been shown to increase lean body mass and body function without significant masculinizing side effects (Dobs et al, (2013) Lancet Oncol, volume 14, pages 335-345). One of the motivations for exploring SARM in preclinical studies of breast cancer is that SARM is a non-metabolizable SARM for weaker androgens or estrogen metabolites, confounding the results of breast cancer, in contrast to steroid androgens such as DHT.
Formula IX inhibits (ER, PR and AR positive) breast cancer cell proliferation
To determine the effect of AR agonists on proliferation of ER positive breast cancer cells, ZR-75-1 breast cancer cells endogenously expressing AR, ER and PR were treated with vehicle or dose response protocol of formula IX and the cell numbers were counted after 6 days of treatment. Proliferation of ZR-75-1 cells was significantly reduced by the dose dependence of formula IX (fig. 34A). The results were reproduced in MCF-7 cells stably transfected with AR, but not GFP (FIG. 34B). Although some previous reports indicate that MCF-7 cells express AR and respond to AR ligands (Buchanan et al, (2005) Cancer Res, page 65, pages 8487-8496), MCF-7 cell line clones under study lack or minimally express AR, consistent with other reports (De Amisis et al, (2010), breast Cancer Res Treat, volume 121, pages 1-11).
The tumor microenvironment comprises tumor epithelial cells, stromal cells, cancer-associated fibroblasts (CAF) and endothelial cells. The collective function of these cells promotes the invasive growth of tumors due to the secretion of paracrine factors. CAF is important for the sustained growth of cancer, and they differ from normal fibroblasts in their ability to secrete factors that promote proliferation of cancer cells. To determine how AR agonists affect paracrine factors secreted by CAF and subsequently affect epithelial cell proliferation, CAF (sample ID 1005) was isolated from ER, PR and AR positive breast cancer tissues obtained from 59 year old african american patients. CAF was treated with vehicle, 10nM DHT or 1 μm IX or 1 μm AR antagonist enzalutamide. Media was collected and pooled over 10 days. CAF was stained with SRB to evaluate the effect of AR ligands on proliferation. In the tested materials, neither AR agonist, DHT and formula IX nor the AR antagonist enzalutamide affected proliferation of breast cancer CAF (fig. 34C, left).
MCF-7 cells stably transfected with AR-deficient GFP (MCF-7-GFP) were seeded in 96-well plates and fed with conditioned medium obtained from CAF treated with vehicle, DHT, formula IX or enzalutamide. Conditioned medium was changed on days 4 and 7 and cells were stained with SRB to measure viability. DHT and formula IX treated conditioned medium, but not enzalutamide treated medium, inhibited proliferation of MCF-7-GFP cells (fig. 34C, right). The antiproliferative effect provided by DHT and formula IX will develop from the inhibition of any paracrine that occurs in CAF and cannot be a direct effect on AR negative MCF-7 clones.
Formula IX inhibits growth of wild type ER-positive breast cancer PDX (HCI-7)
To determine whether the in vitro growth inhibitory properties of formula IX can be observed in vivo, formula IX was tested in PDX expressing wild-type AR. Based on the gene expression profile, three AR positive PDXs were identified from several PDXs available. These PDX, HCI-7, HCI-9 and HCI-13 (Table 12) expressed high levels of AR comparable to that found in LNCaP cells (FIG. 35A). To determine the effect of formula IX on the growth of wild-type ER positive breast cancer PDX, HCI-7 (wtER positive, PR positive, AR positive) luminal a tumor fragments were implanted under the mammary fat pad of female NSG mice. Once the tumor reached 100mm 3 -200mm 3 The mice were randomized and treated with vehicle, 10mg/kg of Enzalutamide of formula IX or 30mg/kgOral treatment. The doses of enzalutamide were selected based on previously published experiments (Park et al, (2016), cancer Invest, volume 34, pages 517-520; poll et al, (2016), nat Chem Biol, volume 12, pages 795-801) and based on internal experiments performed in prostate Cancer xenografts and Hershberger studies. The growth of the slowly growing tumor HCI-7 was significantly inhibited by formula IX but not by enzalutamide (fig. 35B (see also fig. 32)). Tumor weights measured at the end of the study were also significantly smaller in the formula IX treatment group (fig. 35C).
To demonstrate the results obtained in HCI-7, xenografts were formed with MCF-7 cells (MCF-7-AR) (wtER, PR and HER2 positive) stably transfected with AR expressing wild type ER. Tumor volume measured three times per week was significantly reduced by formula IX, with a calculated tumor growth inhibition of greater than 60% (fig. 35D), which supports the use of SARM in ER-positive and AR-positive breast cancers.
To determine if the same effect was observed in AR positive but ER negative breast cancers, HCI-9 tumor fragments were implanted under the mammary fat pad of NSG mice. Once the tumor grows to 100mm 3 -200mm 3 Animals were randomized and treated with vehicle, formula IX or enzalutamide. Neither formula IX nor enzalutamide altered the tumor growth trace, indicating that AR agonists were ineffective in HCI-9PDX that did not express ER (FIG. 34D). Taken together, these results suggest that AR may require ER to inhibit cell proliferation and tumor growth.
AR agonists inhibit the growth of estrogen-independent mutant ER-positive PDX (HCI-13)
By internal sequencing and found in the literature, HCI-13PDX expressed ER mutated at Y537 in LBD (Sikora et al, (2014), cancer Res, volume 74, pages 1463-1474). Such mutations often occur in refractory ER-positive breast cancers that have been treated with ER antagonists (e.g., tamoxifen or fulvestrant) or aromatase inhibitors (e.g., letrozole, anastrozole, exemestane) (Jeselsohn et al, (2018), cancer Cell, volume 33, pages 173-186; toy et al, (2017), cancer Disc, volume 7, pages 277-287). Full genome ChIP-seq studies with cells expressing the mutant showed that the DNA binding characteristics of the mutant ER were different from those of the wild-type ER and that the mutation reprogrammed the ER cistron group. HCI-13 was obtained from patients treated with drugs and relapsed, ranging from therapies targeting ER to chemotherapy (table 12). To determine if this ER mutant expressing PDX was estrogen dependent, HCI-13 tumors were implanted under the mammary fat pad of sham operated and ovariectomized mice. Tumor growth was monitored over a period of 4 weeks. The growth rates of sham mice and ovariectomized mice were comparable, indicating that ER in HCI-13 was constitutively active and did not require estrogen growth (FIG. 36A).
To determine whether formula IX has the ability to inhibit the growth of constitutively active mutant ER-driven breast cancer, HCI-13 tumor fragments were implanted under the mammary fat pad of NSG mice. Once the tumor reached 100mm 3 -200mm 3 Animals were randomized and treated with vehicle or formula IX. Formula IX inhibited the growth of HCI-13 by almost 95% (FIGS. 36B and 36C). Tumors weighed at the time of sacrifice also reflected almost complete tumor inhibition (fig. 36C).
Since the Y537S mutation in ER-LBD results in resistance to ER antagonists, degradants and aromatase inhibitors, it is hypothesized that the inhibition of ER antagonists by mutant ER in HCI-13 may be refractory. To demonstrate this hypothesis, isolated sponge cultures were used to culture HCI-13. HCI-13 tumor fragments were cultured on gelatin sponge as described in methods and treated with vehicle, DHT, formula IX, enzalutamide, and fulvestrant. After 3 days of culture, tumors were harvested, RNA was isolated, and expression of ER and AR target genes was measured by real-time PCR (fig. 36D to 36G).
The results clearly show that DHT and formula IX are effective in inhibiting constitutive active ER-induced expression of pS2 and PR genes, although fulvestrant (a clinically useful effective ER-degrading agent) is ineffective (fig. 36D-36E). Both DHT and formula IX induced the AR-target gene FKBP5 (fig. 36F), indicating that AR is functional. Measurement of the proliferation marker MKI67 (i.e., ki 67) showed that, similar to the expression of pS2 and PR, the expression of MKI67 was inhibited by formulas IX and DHT, but not by fulvestrant or enzalutamide (fig. 36G). These results were reproduced in an ER transactivation assay using ER cDNA cloned from HCI-13 (FIG. 34E). Although fulvestrant and tamoxifen inhibited the activity of wild type ER (fig. 34E, left), HCI-13ER was not inhibited by either of these two compounds (fig. 34E, right). These results demonstrate that when ER inhibitors and degradants develop resistance, AR agonists may provide a mechanically distinct approach to inhibit resistant ER function.
In vitro culture with ER-positive (except 2005 and HCI-9) tumor samples indicated a response to ER and AR ligands Heterogeneity of
As with other cancers, breast cancer is also heterogeneous in its genomic profile and response to treatment. To determine the effect of formula IX and fulvestrant on growth inhibition, breast cancer samples obtained from patients were cultured on dental sponges as described above. Samples were treated with vehicle, 1 μm formula IX or 100nM fulvestrant. Three days after treatment, RNA was isolated from the tissues and expression of ER and AR target genes was measured. AR and ER expression mapped relative to HCI-13 expression indicated that these two targets were expressed at only a small fraction of the levels observed in HCI-13 (FIG. 36K). HCI-13 expressed AR at levels comparable to LNCaP prostate cancer cells, with other samples in the range of 0.2% -20%, with triple negative samples 2005 having minimal expression. Fulvestrant inhibits ER function in 4 of 8 samples, whereas formula IX inhibits ER function in 3 of 8 samples (fig. 36H-36I). Interestingly, formula IX inhibited ER function in sample 1005, where fulvestrant acted as an agonist. Sample 1005 may be comparable to HCI-13 in its response to ER antagonists. These results are consistent with the observation of HCI-13 that AR agonists may inhibit ER function even in cases where ER antagonists would not be able to inhibit. The ability of formula IX to act as an AR agonist was observed in 4 of 8 samples (fig. 36J), including all 3 samples for which formula IX was able to inhibit ER function. Furthermore, since most of these patients did not receive much treatment before tissue was obtained (table 12), no mutation in ER was expected.
Formula IX inhibits HCI-13 breast carcinogenesis by inhibiting function of constitutively active ERLong length
Gene expression studies in sponge cultures demonstrated that AR agonists inhibit ER target genes. To determine the mechanism of antiproliferative effect of formula IX in HCI-13, RNA from HCI-13 tumors obtained from animals shown in FIGS. 36B through 36C was subjected to Affymetrix microarray treatment. A total of 3029 genes were differentially regulated by formula IX in HCI-13 tumors compared to vehicle-treated tumors. Formula IX upregulates 1792 genes and downregulates 1237 genes. The heat map of the differentially regulated genes clearly shows the change in gene expression pattern due to the treatment of formula IX (fig. 37A). Some of the most upregulated genes included Cyp4F8, MYBPC1, RAB3B, LRRC26, AQP4 and CST4 (fig. 37B). Although the role of up-regulated genes such as Cyp4F8 and Mybpc1 in breast cancer is unclear and needs to be determined, down-regulated genes such as MUC-2 and IL10RA have been shown to play an important role in cancer.
The Inventive Pathway Analysis (IPA) showed that in the samples treated with formula IX, the ER target gene was even more highly enriched than the AR target gene (p-value 6.66 -11 Pair 2.83 -7 The method comprises the steps of carrying out a first treatment on the surface of the Fig. 37C). A subset of ER target genes were down-regulated by formula IX, while all AR target genes were up-regulated by formula IX (fig. 37D-37G). Although ER target genes such as TFF1, PGR, NRIP1 are down-regulated by formula IX (not shown), other ER target genes such as CTSD and CCND1 are not inhibited by formula IX. These results provide evidence that formula IX plays a role in breast cancer by at least partially inhibiting ER signaling pathways to reduce cancer growth.
Some direct and indirect modulation of ER targets was observed in samples treated with formula IX. ER increases PDZK1 expression, and thus inhibits the expression of the tumor suppressor SLC26A 3. Interestingly, formula IX significantly inhibited the expression of PDZK1, which restored the expression of the tumor suppressor gene SLC26 A3. Similarly, the anti-apoptotic gene BCL-2 and genes present in its network such as PARP and WT1 are significantly down-regulated by formula IX. Although these genes do not belong to the ER direct target gene list, crosstalk between ER and BCL-2 pathways has been previously reported. Expression of another class of oncoproteins, histones, is inhibited by formula IX. Approximately 17 members of the histone group are significantly inhibited by formula IX. Histones have been associated with invasive cancers and endocrine resistance (Nayak et al, (2015), horm Cancer, volume 6, pages 214-224).
Although IPA analysis did not provide any evidence of modulation of ERBB2 (human epidermal growth factor receptor 2 or HER 2/neu) pathway by formula IX, GSEA enrichment analysis revealed that formula IX affected genes regulated by ERBB2 (fig. 37H). It is currently unclear whether the modulation of ERBB2 is a result of growth inhibition or ER pathway inhibition. Regardless of the mechanism, downregulation of the ERBB2 pathway, which is an oncogenic and tumor promoting pathway, may be an additional advantage of using formula IX or AR agonists in ER-positive breast cancers.
ChIP-Seq analysis indicated that IX reprogrammed ER and AR cistron groups
Previous studies have shown that the interactions of the Y537S mutant ER with DNA have been reprogrammed and may share limited similarities with wild-type ER genomic interactions. To determine if the effect of formula IX on ER function was due to direct effect on ER binding to DNA, chIP sequencing was performed in tumor samples obtained from animals, as shown in fig. 36B-36C. Binding of ER to 1248 regions (q < 0.05) on DNA was reprogrammed by formula IX, with 792 regions enriched for ER and 456 regions depleted of ER (fig. 38A and 38H). AR shows a similar DNA binding pattern, i.e. the ER-rich region is also rich in AR, and the ER-depleted region is also depleted of AR (fig. 38A and 38H). This suggests that ER and AR may shuttle as a complex. The motifs enriched by ER represent the androgen response element (ARE; SEQ ID NO: 1), the glucocorticoid response element (GRE; SEQ ID NO: 2) and the fork box protein A1 or FOXA1 response element (FOXA 1RE; SEQ ID NO: 3), while the ER-depleted regions represent the estrogen response element (ERE; SEQ ID NO: 4) and the FOXA1RE (SEQ ID NO: 5) (FIGS. 38A and 38H). While the ER depleted region favors gene expression patterns, enrichment of ER at ARE and GRE is surprising and has not been previously reported. Principal Component Analysis (PCA) plots showed a clear demarcation in the clusters of vehicle and formula IX treated samples (fig. 38C). FIGS. 38B and 40 show representative regions of enrichment and depletion of ER and AR. Binding of AR and ER to pS2 ERE, PSA (KLK 3) promoter ARE and PSA enhancer ARE was verified by ChIP real-time PCR (fig. 38D). It is important to recognize that formula IX neither binds to nor alters ER activity (Kearbey et al, (2007), pharmaceutical Res, volume 24, pages 328-335; narayanan et al, (2008), molecular Endocrinology, volume 22, pages 2448-2465), the effect of which on ER cis-trans groups is mediated by activation of AR.
Since this was the first study to evaluate the effect of AR agonists such as DHT and formula IX on ER cistron groups in ER positive breast cancer, the region of ER binding was located in response to formula IX. 50% -60% of the ER enrichment and depletion sites are located in the distal regulatory region, while only about 2% -3% of the sites are located in the promoter region (FIG. 38E). Interestingly, while the percentage of intron and exon binding matched previous reports, the proportion of ER bound to the promoter and distal regulatory elements was different from that observed in response to estrogen or from that of constitutively active ER. Other studies have shown that ER cistron groups account for about 30% -40% of the distal regulatory region, 7% -22% of the proximal promoter region, and AR regulated ER cistron groups account for 50% -60% and 2% -3% of these regions, respectively.
Reprogramming FOXA1RE site of formula IX
Interestingly, FOXA1RE motifs were observed to be present in both the enriched and depleted ER cistron groups. Enriched cistron motifs represent ARE, GRE and FOXA1RE, whereas depleted cistron motifs represent ERE and FOXA1RE. Since FOXA1 precursor transcription factor is important for both AR and ER functions and has overlapping binding sites with ARE and ERE, activated AR is likely to sequester FOXA1 from FOXA1RE adjacent ERE, thereby opening nucleosomes and promoting their binding to ARE and GRE. Since ER acts as a complex with AR, it is also sequestered from ERE and FOXA1RE and faces ARE, GRE and FOXA1RE. To determine the validity of this hypothesis, motifs common to ERE and FOXA1RE were located in the down-regulated cistron group. Most ERE and FOXA1RE overlap in the down-regulation motif (fig. 38F). On the other hand, most GREs and ars in the up-regulation motif overlap with FOXA1RE. The results of this analysis confirm the following assumptions: ER. Foxa1 complex shuttles from ER binding site to AR binding site, promoting conversion of nucleosomes to open chromatin and AR binding.
To confirm that AR and ER are localized as complexes and that they migrate together between cistron groups, immunoprecipitation was performed with ER antibody and AR antibody, and western blotting was performed on SRC-1. It is hypothesized that if AR and ER exist as distinct, separate complexes, the treatment of formula IX will increase the interaction of SRC-1 with AR and decrease the interaction with ER. If AR and ER are present together as a complex, the treatment of formula IX will increase the interaction of both AR and ER with SRC-1. Treatment of HCI-13PDX with formula IX increases the interaction between AR and SRC-1 and between ER and SRC-1 (FIG. 38G). Although this is not direct evidence of the AR: ER complex, this evidence, combined with the ChIP-Seq data, suggests that the two proteins exist in complex form and that the main difference is cis-trans group binding leading to gene activation or inactivation.
AR and ER co-localize in luminal B breast cancer
To determine nuclear reactivity and potential co-localization of AR and ER in breast cancer samples, immunohistochemistry was performed in several luminal B breast cancer samples. Nuclear immunoreactivity and high levels of expression of both AR and ER were observed in all breast cancer samples examined (FIG. 39). In addition, several samples had moderate levels of cytoplasmic immunoreactivity for both markers. Since the expression level of both markers exceeded 60% in all samples, a high percentage of cells were positive for both ER and AR. The staining pattern between markers is also similar. In summary, the number of cells immunoreactive with AR in any one sample exceeded the number of cells immunoreactive with ER. However, the semi-quantitative nature of immunohistochemistry makes us unable to conclude that AR is expressed at higher levels than era. It is also possible to observe samples in which immunoreactivity for AR is weak or absent and ER immunoreactivity is present; however, these samples are not always present.
Analysis of phosphorylated protein fractions showed that AR agonists inhibit carcinogenicity and induce tumor suppressor protein phosphorylation
To determine the effect of formula IX on the function of various proteins, phosphorylated proteomes were performed in HCI-13 tumors treated with vehicle or formula IX.Formula IX inhibits various oncogenic proteins such as pERK,Phosphorylation of RSK3, ezrin, BCL2, ELF4G and ER (fig. 41A to 41C). Formula IX also inhibits the expression of the proliferation marker Ki 67. Alternatively, formula IX increases phosphorylation of tumor suppressor proteins such as p53, p27, ACC, and AR. Formula IX also increases phosphorylation of STAT5, STAT5 may be a tumor suppressor or oncogene, depending on the context (fig. 41A-41C). These results indicate that activation of AR with agonists promotes alterations in the appropriate pathways favoring tumor growth inhibition.
To understand the results of these protein changes, 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 was down-regulated by formula IX, HCI-13 sponge cultures were treated with two pathways of activators EGF and PMA (fig. 41D-41E). Treatment of the HCI-13 tumor fragment with PMA completely reversed the expression of the pS2 gene inhibited by formula IX, while EGF only slightly reversed the inhibition observed with formula IX. PMA reversed the effect of formula IX on pS2 gene expression without affecting the ability of formula IX to increase expression of the AR target gene FKBP5 (fig. 41E). This suggests that PMA is likely to act downstream of AR to regulate ER.
Discussion of the invention
Almost all tumor therapeutic agents are inhibitors or antagonists of their therapeutic targets. Prolonged use of these agents will lead to selective stress and ultimately resistance mutations. These resistance mutations may attenuate or prevent the antitumor activity of the drug or, in the worst case, convert the drug to an agonist and cause invasive tumor growth. In the presence of an agonist, the AR will exist in an agonistic conformation as observed in nature, rather than in an unstable antagonistic conformation. This conformational property may be less prone to AR mutations, thereby preventing AR binding or agonist action on the cistron group.
The results obtained in HCI-13 are very encouraging. Formula IX (AR agonist and nonsteroidal SARM) inhibited tumors that recurred and continued to grow in the presence of a range of therapeutic agents, tumor growth inhibition by more than 90%. This result, and results from ex vivo studies, support the use of SARM even after tumor recurrence due to other treatment options. Although HCI-13 is only one example of a mutant ER, this mutation Y537S is one of the common mutants found in the clinic and can be used as a proxy (Katzenellenbogen et al, (2018), nat Rev Cancer, vol.18, 6 th edition, pages 377-388).
The unique property of inhibiting ER function by activating AR demonstrates the complex interactions between various nuclear receptors and their related proteins. Microarray results indicate that inhibition of the ER and HER2 (human epidermal growth factor receptor 2) pathway by formula IX may provide greater benefit to patients in whom both oncogenic pathways are activated. This benefit is further enhanced by increasing the phosphorylation of various tumor suppressors and suppressing the phosphorylation of oncogenes.
The ChIP-Seq results indicate that AR and ER exist as complexes in the presence of formula IX and are transferred from ER cistron to AR cistron together with the precursor transcription factor FOXA 1. Based on these results, a model was proposed (fig. 42). In the absence of activated AR, constitutively active ERs bind to ERE by utilizing FOXA1RE to produce open chromatin and promote tumor growth. In the presence of AR agonists, AR interacts with ER and the complex migrates from ERE and adjacent FOXA1RE to ARE and adjacent FOXA1RE. In this case FOXA1 is sequestered away from ERE and towards ARE to open nucleosomes and promote binding of complexes.
Taken together, these mechanism-based preclinical and transformation studies support the use of SARMs such as formula IX for the treatment of refractory hormone receptor positive breast cancers. Furthermore, heterogeneity was observed in the response of ER positive clinical samples, so screening for breast cancer expressing Y537S ER mutant by pharmacogenomics may be optimal to promote clinical benefit rate of formula IX. Tissue selective AR agonism may provide an alternative hormonal approach for hormone receptor positive breast cancer.
Example 31
18 By F-16Imaging of breast cancer androgen receptor by beta-fluoro-5 alpha-dihydrotestosterone Positron Emission Tomography (PET)
The object is: [ 18 F]-16β -fluoro-5α -dihydrotestosterone (FDHT) is a novel radiotracer for imaging Androgen Receptors (AR) with PET. Most primary and metastatic breast cancer tumors express AR, and modulation of AR signaling has been considered a potentially important therapeutic target for metastatic breast cancer. As part of a phase II clinical trial to study SARMs (i.e., structures of formula IX) for estrogen receptor positive (ER (+)) metastatic breast cancer, prospective imaging sub-studies were designed to demonstrate the principle evidence that FDHT-PET can be used to non-invasively image the presence of AR expression in breast cancer, and explore FDHT-PET as an imaging biomarker for assessing the potential of response to SARM therapies.
The method comprises the following steps: 11 postmenopausal women with ER (+) metastatic breast cancer were recruited into the imaging sub-study and underwent FDHT-PET/CT at baseline and 6 and 12 weeks after initiation of SARM therapy (n=10, 9mg per day; n=1, 18mg per day). PET/CT scans were performed 45 minutes after intravenous administration of 333MBq (9 mCi) FDHT from the vertex of the skull to the middle of the thigh. All FDHT-PET/CT scans of the individual participants were obtained on the same scanner. Abnormal FDHT uptake in tumors is qualitatively defined as uptake greater than background in a pattern consistent with metastatic breast cancer and is quantified using SUVmax at baseline, 6 weeks and 12 weeks after initiation of SARM therapy. Concentrated examination of AR status (qualitative: positive/negative; quantitative: positive nuclei%) was performed on preserved or fresh tumor biopsy samples. The sum of the percent change in FDHT SUVmax between baseline and week 6 and week 12 scans was calculated. Tumor remission was assessed every 12 weeks according to RECIST 1.1. Patients are grouped according to their optimal overall remission as having clinical benefit (CB: complete/partial remission and disease stabilization) or Progressive Disease (PD). Statistical analysis is mainly descriptive due to the trial-and-error nature of the study.
Results: 9 patients completed all 3 FDHT-PET/CT scans; 2 patients were withdrawn from the study prior to week 12. For 9 patients who could obtain a tumor AR status, the baseline FDHT SUVmax was higher for AR positive (n=7) compared to AR negative (n=2) tumors (fig. 43A). A higher trend was observed for baseline FDHT SUVmax, excluding one outlier, and higher levels of quantitative AR expression (r=0.71, p=0.05) (fig. 43B). At 12 weeks post-treatment, 7 patients had clinical benefit and 4 patients had progressive disease. Median baseline FDHT SUVmax was 2.93 (range 1-4.38) for 7 CB patients and 2.15 (0.96-3.77) for 4 PD patients 12 weeks after treatment (fig. 44A). FDHT uptake was decreased in patients with CB (PR and SD) at 12 weeks, whereas FDHT uptake was not decreased in patients with progressive disease (PD or interruption (Disc (AE) or Disc)) (fig. 44B).
Conclusion: this hypothesis generated data supporting the principle evidence that uptake of FDHT in metastatic breast cancer is related to tumor AR expression. The data also supports the potential role of FDHT-PET/CT as a systemic non-invasive imaging biomarker that can be used
For use in larger, well-designed clinical trials to optimize strategies for modulating AR signaling to treat metastatic breast cancer.
Example 32
Efficacy and safety of formula IX on metastatic or locally advanced ER+/AR+ Breast Cancer (BC) in postmenopausal women
Study design: this is an open-label, multicentric, multinational, randomized, parallel design phase 2 study and was used to evaluate the efficacy and safety of formula IX in postmenopausal subjects with er+/ar+bc. Subjects were randomized to receive daily oral administration of 9mg or 18mg of formula IX for up to 24 months. Each dose group was treated independently and the efficacy of each dose group was assessed using a Simon two-stage (Optimal) design (Simon r., "Optimal two-stage designs for Phase 2clinical trials.," Controlled Clinical Trials, 1989; volume 10: pages 1-10). Subjects were randomized to one of the two dose groups in a 1:1 fashion.
Randomization was stratified by subjects exhibiting only bone metastases and all other subjects, and further stratified by setting immediately prior to treatment (adjuvant setting or metastasis setting) in order to balance the proportion of subjects with these manifestation characteristics in each dose group. A statistical comparison of the two dose groups was not intended, but rather a determination was made as to whether one or both doses would produce acceptable Clinical Benefit Remission (CBR), which was defined as the proportion of subjects that could be evaluated (i.e., subjects with centrally confirmed ar+ and who received at least one study drug) to CR, PR or SD according to RECIST 1.1 at week 24, while maintaining acceptable safety profiles. Considering such results, future exploration of formula IX in ER+/AR+BC will be warranted at this dose level.
Thirty-six to eighty-eight (36-88) subjects with centrally confirmed ar+ who received at least one dose of study drug (evaluable subjects) would be required for primary efficacy analysis purposes and would be a subset of the Full Analysis Set (FAS). These criteria were met by 50 patients in the 9mg cohort and 52 patients in the 18mg cohort. One hundred seventy-two (172) subjects (including surrogate subjects) were randomized in a 1:1 fashion to receive daily oral doses of 9mg or 18mg of formula IX. 30 of the above subjects can be considered as surrogate subjects to address the lack of concentrated confirmation of ar+ status, or randomized but not study drug-receiving rare subjects (assuming 25% of enrolled subjects are not rated for primary efficacy analysis). Other statistical parameters as part of the sample size calculation were α=0.025 (one-sided) and test efficacy=90%. The first phase in each study group will be evaluated in the first 18 evaluable subjects. Subjects exceeding 3/18 reached CB (defined as CR, PR or SD) at week 24, and both groups proceeded to the second phase of recruitment until a total of 44 evaluable subjects per group. Otherwise, the group will be interrupted due to lack of efficacy. If at least 9/44 of the subjects in this group reached CB at week 24, then a statistical significance would be declared, i.e., it is more likely to reject the zero hypothesis of unacceptably low CBR+.10%, while supporting an alternative hypothesis indicating a higher ratio+.30%.
Subjects not centrally confirming ar+ continue to participate in the trial but do not participate in the primary efficacy analysis-these subjects contribute to the secondary and tertiary analysis.
Subjects in the 18mg treatment group who experienced Adverse Events (AEs) of intensity no less than grade 3 (national cancer institute-adverse events common terminology standard [ NCI-CTCAE ], version 4.0) and/or intolerance, based on the medical judgment of the researcher and confirmed by the research medical inspector, can either reduce the dose from 18mg daily to 9mg or undergo drug discontinuation. Drug discontinuation can last for up to 5 days after which the subject must re-prime or discontinue the study with study drug (18 mg or 9 mg). In the case of dose reduction, once AE has resolved or intensity reduced to grade 1, the subject may be re-challenged with 18mg or maintained at 9mg at the discretion of the investigator.
Subjects experiencing an AE of intensity > 3 grade (NCI-CTCAE 4.0) and/or intolerance in the 9mg treatment group may be subjected to drug disruption based on the medical judgment of the researcher and confirmed by the research medical inspector. Drug discontinuation may last for up to 5 days after which the subject must re-prime or discontinue the study with study drug (9 mg).
For safety analysis, subjects were analyzed in the treatment group where dosing was started. For efficacy analysis, subjects were analyzed according to their randomized treatment group groups.
Subjects exhibiting CB received treatment for up to 24 months from the randomized date (as long as they continued to exhibit CB during treatment during these 24 months). Subjects who continue to exhibit CB in study treatment at 24 months would be proposed to continue safety extension studies according to a separate regimen. For safety purposes, all subjects will receive a follow-up for one month after receiving the last dose of formula IX.
For safety purposes, all subjects received a follow-up for one month after receiving the last dose of formula IX.
Target population: adult postmenopausal women with metastatic or recurrent locally advanced er+/ar+bc.
Duration of study: the duration of the study was estimated to be 3 years.
Description of the agent or intervention: three (3) capsules of formula IX 3.0mg were orally administered with or without food at about the same time per day at a daily dose of 9mg, or six (6) capsules of formula IX 3.0mg at a daily dose of 18mg.
Potential benefits: based on the trial of example 9, 9mg of formula IX was studied once daily in 22 postmenopausal women with metastatic er+bc and previously responsive to hormone therapy. Primary endpoints of 17 AR positive subjects were assessed. 6 of these 17 subjects exhibited CB (SD) at 6 months. In a subject exhibiting SD (RECIST 1.1), 27% tumor regression was confirmed in a single target lesion. A total of 7 subjects (1 subject with unknown AR status) reached CB at 6 months. Of the 7 subjects who reached CB at 6 months, the Time To Progress (TTP) was estimated to be 10.2 months. The results also show that after a median duration of 81 days of study, 41% (9/22) of all subjects reached CB as optimal relief, and also had increased PSA, which appears to be an indicator of AR activity. By the time the protocol was finalized, the study was still in progress, with the disease in one subject remaining stable after 336 days.
Preclinical data of formula IX indicate that it is also anabolic in bone and reduces bone turnover markers. Treatment with formula IX may reduce bone turnover compared to other hormone therapies used to treat hormone receptor positive BC. A stronger bone microenvironment may reduce metastasis to bone or delay the time of a bone related event.
Therapeutic efficacy targets
The main therapeutic objective of this test was to estimate CBR (defined as complete relief [ CR ], partial relief [ PR ] or SD) (according to RECIST 1.1) at week 24 of daily oral administration of 9mg of formula IX and 18mg of formula IX in subjects with estrogen receptor positive and androgen receptor positive (er+/ar+) BC and with a centrally confirmed ar+ status.
The secondary efficacy objective was to estimate CBR (according to RECIST 1.1) at week 24 given 9mg and 18mg of formula IX in all randomized subjects (full analysis set [ FAS ]) receiving at least one dose of study drug, regardless of AR status as determined by the central laboratory.
Additional secondary efficacy targets apply to both centrally confirmed ar+ subjects (an evaluable subset of FAS) and to all subjects in FAS: (a) Objective remission rates (ORR; defined as CR or PR) at week 24 (according to RECIST 1.1) were estimated at 9mg and 18mg of formula IX; (b) Estimating the optimal overall remission rate (BOR) of 9mg and 18mg of formula IX; (c) Estimating Progression Free Survival (PFS) of subjects receiving 9mg and 18mg of formula IX; (d) estimating TTP in subjects receiving 9mg and 18mg of formula IX; (e) The duration of remission (time from recording tumor remission to disease progression or death) was estimated for subjects receiving 9mg and 18mg of formula IX.
Tertiary targets apply to both centrally confirmed ar+ subjects (an evaluable subset of FAS) and all subjects in FAS: (a) assessing the effect of 9mg and 18mg of formula IX on serum PSA; (b) Assessing the effect of 9mg and 18mg of formula IX on quality of life (QoL) as measured by EQ-5D-5L; (c) Assessing the effect of 9mg and 18mg of formula IX on Circulating Tumor Cells (CTCs); (d) evaluating the effect of the duration of the previous CB on the result; (e) Evaluating the effect of time to transition from diagnosis to randomization on the result; (f) describes the effect of 9mg and 18mg of formula IX on tumor volume; (g) The effect of plasma concentrations of glucuronide of formula IX and formula IX on CBR was assessed at 24 weeks.
The safety objective is to describe the safety profile of 9mg and 18mg per day of formula IX orally in subjects with er+/ar+bc and with centrally confirmed ar+ and in all randomized and treated subjects.
Pharmacokinetic targets: the plasma concentrations of the glucuronides of formula IX and formula IX at each evaluation time point are described.
Preparing, packaging and labeling: the 3.0mg soft capsule of formula IX will be provided in the form of an opaque, white to off-white, oval-shaped No. 5 soft capsule containing 3.0mg of formula IX. The liquid soft capsule fill consists of formula IX dissolved in polyethylene glycol 400. 3.0mg of formula IX soft capsules are packaged in blister packs. Each blister pack contains enough study medication to be administered for one (1) week. At randomization (visit 2) and visit 3, visit 4 and visit 5, subjects were provided with a carton containing 7 blister packs of study medication, corresponding to a 7 week dose. At visit 6, 8, 9, 10, 11, 12 and 13, subjects received two cartons of study medication (7 blister packs per carton) to cover 14 weeks of study treatment in order to accommodate a visit schedule every 12 weeks (+ -7 days). At each visit, the subject was asked to carry a carton containing all blister packs.
Each blister pack consists of an appropriate number of blister strips (1 blister for the 9mg treatment group and 2 blisters for the 18mg treatment group) enclosed in a child resistant heat seal card. The blister strip consists of a PVC/ACLAR substrate and an aluminum foil/PVC/PVAC copolymer and a polymethacrylate (product contact) cover. The perforations on the back of the heat seal card overlie the foil cover. To remove the study drug, the subject releases the appropriate perforation by pressing a release button on the inside of the card. Once released, the perforations may be removed and the study drug pushed through the foil.
Pharmacokinetic assessment
Blood samples for pharmacokinetic assessment were collected at baseline (pre-dose), at visit 3 (week 6), at visit 5 (week 18) and at visit 6 (week 24). On each of these days, one blood sample was collected in 6mL of K2-ethylenediamine tetraacetic acid (EDTA) blood collection tube. The exact time (hh: mm) and date of each blood sample collection was recorded on an electronic case report form. At baseline visit, blood samples were collected before subjects received their first dose of formula IX. At visit 3 (week 6), visit 5 (week 18) and visit 6 (week 24), the date and approximate time of administration of the last dose of formula IX prior to blood sample collection should be recorded; that is, it should be noted whether the subject took the previous dose in the morning or in the evening of the previous day. Immediately after collection, the tube was gently inverted several times to mix the anticoagulant with the blood sample.
The blood sample is kept on wet ice (ice bags in a water bath are also acceptable) for up to 20 minutes until processing. The plasma fraction was isolated by placing the collection tube in a centrifuge and centrifuging at 1,500xg for 10 minutes. The plasma fraction was removed by pipette and split into two 2mL polypropylene transfer bottles (each tube receiving approximately equal aliquots).
All sample collection tubes and freezer tubes were clearly labeled in a manner that identified the subject, study number, visit number, and freezer tube sample letters. The label is secured to the freezer pipe in a manner that prevents the label from falling off after freezing. The samples were stored in a refrigerator at-20 ℃ or lower. The samples were transported in insulated containers with enough dry ice to ensure that they remained frozen.
Any plasma sample remaining after completion of the pharmacokinetic analysis outlined in the protocol can be used to identify and quantify the metabolites of formula IX.
Results
Table 13 depicts a subset of the total number of patients. These are patients who were evaluable (ar+) at the beginning of the study and had measurable disease and received palbociclib as the previous therapy.
TABLE 13
9mg patient number Results Patient number of 18mg Results
7004-8120 6003-8133
7019-8066 CR 7001-8001 PR
7026-8083 7001-8118
7004-8100
7019-8087 CR
7022-8078
According to table 13, in group 9mg of formula IX, 3 patients were previously treated with palbociclib and failed. Of these 3 patients, one patient (33%) had Complete Remission (CR). In the 18mg group, 6 patients were previously treated with palbociclib and failed. Among these 6 patients, one CR and one PR achieved a remission rate of 33%. These data support that formula IX is active in patients who are resistant/unresponsive to CDK 4/6 inhibitors.
Example 33
Re-sensitization to CDK 4/6 inhibitors by formula IX (SARM) in a CDK 4/6 resistance model
Many of the embodiments described above cumulatively provide clinical and preclinical studies using clinically relevant in vitro and in vivo model systems, providing powerful support for the use of formula IX in a variety of situations of advanced ER-positive breast cancer, including many estrogen endocrine resistance models. Furthermore, example 32 demonstrates that formula IX maintains activity in patients resistant/unresponsive to CDK 4/6 inhibitors (CDKi) palbociclib. The combination of inhibitors of cyclin-dependent kinase 4/6 with estrogenic endocrine therapy has become the standard treatment for women with advanced breast cancer, and failure of these treatments represents an increasing unmet need.
Advances in palbociclib (i.e., resistance thereto) from ER positive, PR negative and HER2 negative (ER + 、PR - 、HER2 - ) Tissue samples of patients were implanted into animal (NSG mice) models and grown as patient-derived xenografts (PDX). This GAR15-13 PDX model of CDKi resistance showed a response to treatment with formula IX (SARM) (FIG. 45A). Furthermore, as expected based on the patient's treatment history, they are relatively refractory to palbociclib (Palbo) as a single agent. Interestingly, when treated with both formula IX and pamoxnib (Combo), the treatment was synergistic and the tumor was essentially absent. Thus, formula IX treatment appears to re-sensitize the tumor to drugs that it previously had resistance to. This human tumor model also amplified cyclin D1 (CCND 1) Amp ) Genes that are thought to be driving factors for resistance to the treatment of CDKi. Furthermore, formula IX (SARM), alone or in combination, nonetheless has activity and further, formula IX re-sensitizes tumors to palbociclib. This was also observed in other in vitro tested models including palbociclib resistant MCF-7 (MCF 7 palbo R ) Cell line (FIG. 45B). MCF7 palbo R Cell (Palb) R ) There was a compensatory increase in CDK2 expression, indicating resistance to CDK 4/6 inhibitors, but maintaining expression of AR and ER comparable to the parental cell line (data not shown). Lundberg, a. Et al, (2019), breast Cancer res., volume 21, page 34.
FIG. 45B demonstrates that either formula IX (SARM) alone or palbociclib (Palbo) alone is used in Palb R Has moderate activity in cells, but combination therapy (SARM+Palbo) is performed in MCF7 Palbo R In cellsHas synergistic activity; another model is provided wherein CDKi resistance can be overcome by a combination therapy of formula IX and pamoxnib. These findings, i.e., that formula IX (SARM) can restore sensitivity to CDKi in these estrogen endocrine and CDKi resistance models, are important and unexpected findings that expand the scope of ER positive breast cancers suitable for treatment with formula IX. For example, this data supports that formula IX, alone or in combination with a CDKi, can be used even after the patient has acquired resistance to the CDKi and estrogen-targeted endocrine combination therapy. For example, CDKi is approved for use with SERMs, aromatase inhibitors, and SERDs such as fulvestrant, but the ultimate failure of treatment results in a population resistant to CDKi and estrogen endocrine therapy, for which few treatment options are available. This data supports the combined use of formula IX with CDKi in this population, extending the time that ER positive advanced breast cancer can be treated with hormone and kinase therapies rather than chemotherapy. Furthermore, new evidence is provided that formula IX may be more effective than existing estrogenic endocrine therapies (e.g., tamoxifen) or new CDKi (e.g., pamoxnib) standard therapies, and in the latter case, may be combined to unexpectedly enhance growth inhibition.
While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those skilled in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.
Sequence listing
<110> UNIVERSITY OF tennessee research FOUNDATION (UNIVERSITY OF TENNESSEE RESEARCH FOUNDATION)
Michel Shi Taina (STEINER Mitchell)
La Mei Shen Nala (NARAYANAN, ramesh)
An Chunzhou (AHN, sunjoo)
James, dorton (DALTON, james)
<120> pharmaceutical compositions for treating breast cancer and methods of use thereof
<130> P-74233-PC2
<150> 17/151,108
<151> 2021-01-15
<150> 17/066,487
<151> 2020-10-09
<150> 16/242,950
<151> 2019-01-08
<150> 16/051,042
<151> 2018-07-31
<150> 15/371,104
<151> 2016-12-06
<150> 15/075,373
<151> 2016-03-21
<150> 14/798,208
<151> 2015-07-13
<150> 14/293,632
<151> 2014-06-02
<150> 13/953,492
<151> 2013-07-29
<150> 13/789,005
<151> 2013-03-07
<150> 61/726,274
<151> 2012-11-14
<150> 61/671,366
<151> 2012-07-13
<160> 5
<170> patent In 3.5 version
<210> 1
<211> 49
<212> DNA
<213> artificial sequence
<220>
<223> response element motif
<400> 1
agtgaagcta gtccatggct aatgccgatt acgtactcga cttcatcga 49
<210> 2
<211> 43
<212> DNA
<213> artificial sequence
<220>
<223> response element motif
<400> 2
cagtagtgaa gtcacagttg catacgcgat tacgtatcag cta 43
<210> 3
<211> 21
<212> DNA
<213> artificial sequence
<220>
<223> response element motif
<400> 3
atagctatgg atcacacact a 21
<210> 4
<211> 48
<212> DNA
<213> artificial sequence
<220>
<223> response element motif
<400> 4
acgtagtgta gatgaccgta gtggtacgat gcattagaac tgcatcat 48
<210> 5
<211> 21
<212> DNA
<213> artificial sequence
<220>
<223> response element motif
<400> 5
atagctatgg atcacacact a 21

Claims (35)

1. A pharmaceutical composition comprising a Selective Androgen Receptor Modulator (SARM) compound and a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor, wherein the SARM compound is represented by the structure of formula I:
Wherein the method comprises the steps of
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 (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, CN, NO 2 、COR、COOH、CONHR、CF 3 、Sn(R) 3 Or R is 3 Together with the benzene ring to which it is attached, form a fused ring system represented by the structure:
z is NO2, CN, COR, COOH or CONHR;
y is CF 3 F, br, cl, I, CN 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 A, B or C:
n is an integer from 1 to 4; and is also provided with
m is an integer of 1 to 3, or
Optical isomers, racemic mixtures, pharmaceutically acceptable salts, pharmaceutical products, hydrates, N-oxides or crystals thereof.
2. The pharmaceutical composition of claim 1, wherein the SARM compound is represented by the structure of formula II:
wherein the method comprises the steps of
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 A, B or C:
r is C 1 -C 4 Alkyl, aryl, phenyl, alkenyl, hydroxy,C 1 -C 4 Haloalkyl, halogen or haloalkenyl; and is also provided with
R 1 Is CH 3 、CF 3 、CH 2 CH 3 Or CF (CF) 2 CF 3
3. The pharmaceutical composition of claim 1, wherein the SARM compound is represented by the structure of formula VIII, IX, X, XI, XII, XIII or XIV:
4. a pharmaceutical composition comprising formula IX or an optical isomer, racemic mixture, pharmaceutically acceptable salt, pharmaceutical product, hydrate, N-oxide or crystal thereof and a CDK 4/6 inhibitor,
5. pharmaceutical composition according to any one of claims 1 to 4, wherein the CDK 4/6 inhibitor is palbociclib, rebabociclib, ly Luo Xili, trazoxili or abbe-cilide.
6. Pharmaceutical composition according to any one of claims 1 to 4, wherein the CDK 4/6 inhibitor is palbociclib.
7. Pharmaceutical composition according to any one of claims 1 to 4, wherein the CDK 4/6 inhibitor is arbeli.
8. 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 palbociclib,
9. 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 arbeli,
10. the pharmaceutical composition according to any one of claims 1 to 9, wherein the composition is in the form of a pill, tablet, capsule, solution, suspension, emulsion, elixir, gel, cream, suppository or parenteral formulation.
11. A method for treating a subject having breast cancer, the method comprising administering to the subject the pharmaceutical composition of any one of claims 1-10.
12. The method of claim 11, wherein the breast cancer is 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 an inhibitor employing a 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), rebaciclesonidine (Kisqali), abbe-zenio, trazopride, ly Luo Xili), apilimus (piqry) (inhibitors of phosphatidylinositol-3-kinase subunit α (PI 3), mTOR inhibitor (evergreen), polysaccharase (PARP) inhibitor (ADP) or (guanylate), anti-panzotinib (Herceptin), anti-panzotinib (HER 2), anti-bead (panzotinib) or anti-panoramide (HER 2), anti-bead (HER 2, panoramide (HER 2) antibody (HER 2) Enmetrastuzumab (Kadcyla) or pertuzumab/trastuzumab/hyaluronidase-zzxf (Phesgo)), actlizumab (Tecentriq) (PD-L1 blocking antibody), pembrolizumab (Keystuda) (PD-L1 blocking antibody), gor Sha Tuozhu mab (Trodelvy) (antibody drug conjugate for TNBC), and/or bevacizumab (Avastin) treat failed breast cancer.
13. The method of claim 11, wherein the breast cancer is AR positive metastatic breast cancer.
14. The method of claim 11, wherein the breast cancer is AR positive refractory breast cancer.
15. The method of claim 11, wherein the breast cancer fails treatment with a Selective Estrogen Receptor Modulator (SERM).
16. The method of claim 15, wherein the SERM is tamoxifen, toremifene, or raloxifene.
17. The method of claim 11, wherein the breast cancer fails treatment with a cyclin-dependent kinase 4/6 (CDK 4/6) inhibitor.
18. The method of claim 17, wherein the subject is resistant to or non-responsive to a CDK 4/6 inhibitor.
19. The method of claim 17 or claim 18, wherein the CDK 4/6 inhibitor is palbociclib (Ibrance), rebabociclib (Kisqali), traracilli, ly Luo Xili, or abbe cili (Vorzenio).
20. A method according to claim 17 or claim 18 wherein the CDK 4/6 inhibitor is palbociclib (Ibrance).
21. A method according to claim 17 or claim 18 wherein the CDK 4/6 inhibitor is arbitide (Vorzenio).
22. A method according to any one of claims 11 to 21 wherein the composition re-sensitises the breast cancer to treatment with a CDK 4/6 inhibitor.
23. The method of any one of claims 11-21, wherein the composition overcomes estrogen endocrine resistance.
24. A method according to any one of claims 11 to 21 wherein the composition overcomes resistance to a combination therapy of estrogenic endocrine and CDK 4/6 inhibitors.
25. The method of claim 23 or claim 24, wherein the estrogenic endocrine therapy comprises at least one of tamoxifen, toremifene, raloxifene, exemestane, letrozole, anastrozole, and fulvestrant.
26. A method according to claim 23 or claim 24 wherein the CDK 4/6 inhibitor is at least one of palbociclib (Ibrance), rebabociclib (Kisqali), trarasilli, ly Luo Xili and abbe cili (Vorzenio).
27. The method of claim 11, wherein the breast cancer fails to be treated with an mTOR inhibitor.
28. The method of claim 27, wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, or ground phosphorus limus (ridafarolimus).
29. The method of claim 12, wherein the ER-positive breast cancer is AR-positive and ER-positive breast cancer, or AR-negative and ER-positive breast cancer.
30. The method of claim 12, wherein 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 positive and HER2 negative; ER positive, PR negative and HER2 positive; or ER positive, PR positive and HER2 positive.
31. The method of claim 11, wherein the method further increases the survival of the subject with breast cancer or increases the progression free survival of the subject with breast cancer.
32. The method of claim 11, wherein the composition is administered intravenously, intra-arterially, intramuscularly, subcutaneously, orally, or topically.
33. The method of any one of claims 11-32, wherein the dosage of the selective androgen receptor modulator is 1mg to 50mg per day.
34. The method of any one of claims 11-32, wherein the dose of the selective androgen receptor modulator is 9mg per day.
35. The method of any one of claims 11-32, wherein the dose of the selective androgen receptor modulator is 18mg per day.
CN202280021307.2A 2021-01-15 2022-01-14 Pharmaceutical composition for treating breast cancer and method of use thereof Pending CN116981451A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US17/151,108 2021-01-15
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
CN116981451A true CN116981451A (en) 2023-10-31

Family

ID=82448697

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280021307.2A Pending CN116981451A (en) 2021-01-15 2022-01-14 Pharmaceutical composition for treating breast cancer and method of use thereof

Country Status (9)

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

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115369089A (en) * 2022-08-11 2022-11-22 中山大学孙逸仙纪念医院 Two ER positive HER-2 negative breast cancer CDK4/6 inhibitor drug-resistant strains and construction method and application thereof
CN117982655A (en) * 2023-11-21 2024-05-07 北京大学人民医院 Method for constructing animal model of pain related to aromatase inhibitor treatment and application

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009506119A (en) * 2005-08-31 2009-02-12 ユニバーシティ オブ テネシー リサーチ ファウンデーション Treatment of kidney disease, burns, wounds and spinal cord injury using 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)
WO2017223115A1 (en) * 2016-06-22 2017-12-28 Radius Health, Inc. Ar+ breast cancer treatment methods

Also Published As

Publication number Publication date
KR20230163362A (en) 2023-11-30
JP2024503718A (en) 2024-01-26
EP4277613A1 (en) 2023-11-22
BR112023014247A2 (en) 2023-10-03
CA3205212A1 (en) 2022-07-21
IL304467A (en) 2023-09-01
AU2022208712A1 (en) 2023-08-10
WO2022155543A1 (en) 2022-07-21

Similar Documents

Publication Publication Date Title
JP6386120B2 (en) Method of treating androgen receptor (AR) positive breast cancer with a selective androgen receptor modulator (SARM)
US10987334B2 (en) Method of treating ER mutant expressing breast cancers with selective androgen receptor modulators (SARMs)
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)
EP4039253A1 (en) Methods of treating cancer
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)
CN116981451A (en) Pharmaceutical composition for treating breast cancer and method of use 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)
US20230172897A1 (en) Methods of treating breast cancers with selective androgen receptor modulators and additional therapeutic agents
US20210228529A1 (en) Pharmaceutical compositions for treating breast cancers and methods of uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination