WO2020251556A1 - Utilisation d'un agoniste de rxr dans le traitement de cancers her2 + - Google Patents

Utilisation d'un agoniste de rxr dans le traitement de cancers her2 + Download PDF

Info

Publication number
WO2020251556A1
WO2020251556A1 PCT/US2019/036594 US2019036594W WO2020251556A1 WO 2020251556 A1 WO2020251556 A1 WO 2020251556A1 US 2019036594 W US2019036594 W US 2019036594W WO 2020251556 A1 WO2020251556 A1 WO 2020251556A1
Authority
WO
WIPO (PCT)
Prior art keywords
her2
cancer
rxr agonist
therapeutic agent
rxr
Prior art date
Application number
PCT/US2019/036594
Other languages
English (en)
Inventor
M.D. PH.D. Powel H. BROWN
Martin E. Sanders
Original Assignee
Io Therapeutics, Inc.
The University Of Texas Md Anderson Cancer Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Io Therapeutics, Inc., The University Of Texas Md Anderson Cancer Center filed Critical Io Therapeutics, Inc.
Priority to BR112021024970A priority Critical patent/BR112021024970A2/pt
Priority to KR1020217043352A priority patent/KR20220019724A/ko
Priority to JP2021573710A priority patent/JP7438496B2/ja
Priority to PCT/US2019/036594 priority patent/WO2020251556A1/fr
Priority to CA3142076A priority patent/CA3142076A1/fr
Priority to AU2019450366A priority patent/AU2019450366A1/en
Publication of WO2020251556A1 publication Critical patent/WO2020251556A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • RXR retinoid X receptors
  • TARGRETIN ® (bexarotene), which is a retinoid X receptor (RXR) agonist with retinoic acid receptor (RAR) agonist activity as well, was approved by the U.S. Food and Drug Administration for the treatment, both oral and topical, of cutaneous manifestations of cutaneous T cell lymphoma in patients who are refractory to at least one prior systemic therapy. Encouraging results were obtained with TARGRETIN ® in several Phase II studies in NSCLC. However, the pivotal Phase III clinical study did not show increased survival.
  • RXR retinoid X receptor
  • RAR retinoic acid receptor
  • Treatments for cancer are ever evolving, gaining in specificity and sophistication. Early non-surgical cancer treatments generally targeted rapidly dividing cells which were more sensitive to radiological and chemical assault. Over time, more specific and less generally toxic treatments have been developed. Some treatments appear to have broad applicability, for example immune checkpoint inhibitors or rexinoids. Others are targeted to cancers that express a particular antigen or other biomarker involved in the regulation of proliferation or differentiation; including many monoclonal antibodies and kinase inhibitors. Yet as the variety of cancer treatments has grown, it has become ever harder to determine which treatments might be productively combined and for what indications.
  • inventions comprising treating a patient having a Her2 + tumor with a combination of a Her2-targeting therapeutic agent and a RXR agonist capable of inhibiting cancer growth.
  • the treatment combination further comprises thyroid hormone.
  • the RXR agonist is capable of activating RXR/Nurr1 heterodimeric receptors.
  • the RXR agonist is a compound of Formula I as disclosed herein below, or a pharmaceutically-acceptable salt thereof.
  • the RXR agonist is a compound of Formula II as disclosed herein below, or a pharmaceutically-acceptable salt thereof.
  • compounds of Formula I and Formula II, and their pharmaceutically-acceptable salts are referred to as means for activating RXR/Nurr1 heterodimeric receptors or rexinoid means for inhibiting tumor growth.
  • Her2-targeting therapeutic agent is an inhibitor of Her2 kinase activity or Her2-mediated signaling.
  • Her2-targeting therapeutic agent is therapeutic anti-Her2 antibody.
  • Therapeutic antibodies may mediate antibody-dependent cellular cytotoxicity (ADCC) instead of, or in addition to, inhibiting signaling (kinase activity).
  • ADCC antibody-dependent cellular cytotoxicity
  • Trastuzumab and pertuzumab are examples of therapeutic anti-Her2 antibodies, as disclosed herein below.
  • such antibodies are referred to as immunoglobulin means for inhibiting Her2 + tumor cell proliferation, means for mediating ADCC of Her2 + tumor cells, or immunoglobulin means for inhibiting Her2 signaling.
  • a Her2-targeting therapeutic agent is an antibody-drug conjugate comprising an anti-Her2 antibody.
  • the anti-Her2 antibody has therapeutic activity alone, while in other embodiments it does not, merely serving to deliver a cytotoxic agent to Her2 + cells.
  • Ado-trastuzumab emtansine is an example of a Her2-targeting antibody-drug conjugate, as disclosed herein below.
  • such antibody- drug conjugates are referred to as means for delivering a cytotoxic agent to Her2 + cells.
  • the inhibitor of Her2 kinase activity or Her2-mediated signaling is a small organic molecule (small drug) inhibitor of Her2 kinase activity.
  • small drug small organic molecule
  • Lapatinib and neratinib are examples of Her2 kinase inhibitors as disclosed herein below.
  • small drug inhibitor of Her2 kinase activity are referred to as small molecule means for inhibiting Her2 kinase activity.
  • the Her2 + cancer is a Her2 + breast cancer.
  • the Her2 + cancer is a Her2 + ovarian cancer, stomach cancer, adenocarcinoma of the lung, uterine cancer (such as serous endometrial carcinoma), gastric cancer, or salivary duct carcinoma.
  • the herein disclosed treatments are carried out concurrently with other pharmaceutical therapies or radiotherapies.
  • the herein disclosed treatments are the exclusive therapy in the time interval in which they are conducted.
  • the herein disclosed treatments serve as a debulking treatment in preparation for subsequent surgical removal of tumor.
  • the herein disclosed treatments are applied as adjuvant therapy subsequent to surgical removal of tumor to address any residual disease or potential recurrent disease.
  • combination drug compositions or formulations comprising at least one Her2-targeting therapeutic agent and at least one RXR agonist capable of inhibiting cancer growth.
  • the combination (used for treatment) may include more than one agent in one or the other classification (Her2 targeting agent and/or RXR agonist).
  • the combination may further include thyroid hormone.
  • the thyroid hormone is thyroxine.
  • FIGURE 1 is a three-dimensional plot depicting the growth inhibitory effects of IRX4204 and trastuzumab, alone and in combination, on breast cancer cell lines: MCF7 cells ( Figure 1 A) and SkBr3 cells ( Figure 1 B).
  • FIGURE 2 is a three-dimensional plot depicting the growth inhibitory effects of IRX4204 and lapatinib, alone and in combination, on breast cancer cell lines: MCF7 cells (Figure 2A), SkBr3 cells ( Figure 2B), BT474 cells (Figure 2C), and MDA-MB-361 cells ( Figure 2D).
  • FIGURE 3 is a three-dimensional plot depicting the growth inhibitory effect of IRX4204 and neratinib, alone and in combination, on breast cancer cell lines: MCF7 cells (Figure 3A), SkBr3 cells ( Figure 3B), BT474 cells (Figure 3C), and MDA-MB-361 cells ( Figure 3D).
  • the herein disclosed embodiments include methods of treating Her2 + cancers, such as Her2 + carcinoma of the breast, with a combination a retinoid X (rexinoid) receptor (RXR) agonist and a Her2-targeted anticancer agent. Some embodiment further comprise administration of thyroid hormone in conjunction with the RXR agonist.
  • Embodiments include a RXR agonist for use in combination with a Her2-targeted anticancer agent, or a thyroid hormone and a Her2-targeted anticancer agent, in the treatment of Her2 + cancers.
  • the Her2 + cancer is Her2 + breast cancer.
  • the methods of treatment involve the administration of a combination of two, three, or more therapeutic agents.
  • the administration of one of these agents may be described as being done in coordination or conjunction with another of these agents.
  • administration of in coordination or in conjunction with it is meant that the manner of administration of each of these agents is such that the physiologic effects of the agents overlap in time. This does not require that the agents be contained in the same composition or formulation, orthat they be administered as separate compositions at the same time, by the same route of administration, or on the same schedule, though in some embodiments any of the foregoing may be the case.
  • RXR agonists, thyroid hormone, and Her2 kinase inhibitors are more typically administered at intervals measured in weeks.
  • Her2-targeted treatments include monoclonal antibodies (mAbs), such as trastuzumab and pertuzumab, and inhibitors of Her2 kinase activity, such as lapatinib, neratinib, and afatinib (which all also inhibit epidermal growth factor receptor (EGFR)).
  • mAbs monoclonal antibodies
  • inhibitors of Her2 kinase activity such as lapatinib, neratinib, and afatinib (which all also inhibit epidermal growth factor receptor (EGFR)
  • EGFR epidermal growth factor receptor
  • the rexinoid IRX4204 has shown activity against a variety of cancers in model systems (see for example, U.S. Patent Pub.
  • IRX4204 and a Her2-targeted therapeutic agent are particularly effective against Her2 + cancers, such as Her2 + breast cancer.
  • Some embodiments further include thyroid hormone or the administration of thyroid hormone.
  • Other embodiments exclude thyroid hormone or the administration of thyroid hormone.
  • a Her2-targeted therapeutic agent as used herein is a therapeutic agent that inhibits growth of cancer cells by inhibiting Her2 function.
  • Such agents include mAbs that bind to Her2, antibody-drug conjugates comprising such mAbs, and inhibitors of Her2 tyrosine kinase activity. Collectively, such agents can be referred to as means for therapeutically targeting Her2.
  • Some embodiments specifically include or are limited to one or more of these classes of agent, or one or more species within one or more of these classes of agent.
  • Some embodiments specifically exclude one or more of these classes of agent, or one or more snecies within one nr more nf these classes nf anent
  • means for therapeutically targeting Her2 are mAbs that bind to Her2, antibody-drug conjugates comprising mAbs that bind to Her2, and inhibitors or Her2 tyrosine kinase activity.
  • the mAb in an antibody-drug conjugate has therapeutic activity by itself, in other embodiments it does not.
  • Many embodiments comprise administration of a single anti-Her2 mAb, but some embodiments comprise administration of multiple anti-Her2 mAbs, for example, trastuzumab and pertuzumab. While the disclosed embodiments are generally described as using anti-Her2 mAbs, further embodiments can substitute anti-Her2 polyclonal antiserum for anti-Her2 mAb. Some embodiments can comprise administration of additional mAbs targeting other antigens, some embodiments specifically exclude administration of other antibodies.
  • Her2 is a growth factor receptor found on, and implicated in, a variety of cancers, especially breast cancer but also, for example, gastroesophageal cancer, ovarian cancer, stomach cancer, adenocarcinoma of the lung, uterine cancer (such as serous endometrial carcinoma), or salivary duct carcinoma.
  • Anti-Her2 antibodies are believed to work as anticancer agents through a combination of mechanisms: inhibition of signaling through Her2, antibody-dependent cellular cytotoxicity (ADCC), and mediating presentation of tumor antigen by antigen presenting cells (APC), such as macrophages; additional mechanisms may also exist.
  • ADCC antibody-dependent cellular cytotoxicity
  • APC antigen presenting cells
  • the best understood and most clinically advanced anti-Her2 antibodies are trastuzumab, pertuzumab, and margetuximab.
  • these antibodies are administered by intravenous infusion.
  • infusions may be done over 30-90 minutes and may occur at intervals of 1-3 weeks for as long as a year.
  • the antibody is administered at an initial higher dose and a subsequent lower dose.
  • the initial higher dosage is twice the subsequent lower dosage.
  • the initial dose is single administration.
  • the initial dosage is administered multiple times before switching to the lower subsequent dosage. Specific examples of dosages and dose regimens can be found in the prescribing information for HERCEPTIN® and PERJETA®, which are incorporated herein by reference in their entireties.
  • trastuzumab (sold as HERCEPTIN®) binds to domain IV of the extracellular segment of Her2. Trastuzumab inhibits the proliferation of cells that overexpress Her2 and mediates ADCC. Biosimilar antibodies to trastuzumab have been developed and are marketed in some jurisdictions.
  • Pertuzumab (sold as PERJETA®) recognizes the extracellular dimerization domain (domain II) of Her2, a different epitope than trastuzumab. By preventing ligand dependent dimerization, it inhibits signaling through Her2, leading to cell growth arrest and apoptosis. Pertuzumab also mediates ADCC. Pertuzumab augmented the activity of trastuzumab in tumor xenograft models that over express Her2.
  • Margetuximab (currently still in clinical development) recognizes the same epitope as trastuzumab, but possesses an engineered Fc region designed to increase Fc-dependent mechanisms of immune attack, such as ADCC.
  • the engineered Fc region confers increased binding to activating Fc-g receptors (CD16A) and reduced binding to inhibitory Fc-g receptors (CD16B) on immune effector cells, including monocytes, macrophages, dendritic cells and natural killer (NK) cells.
  • Further anti-Her2 mAbs include TrasGEX®, HM2, hertuzumab, and HT-19. TrasGEX® and HM2 are being developed as“biobetters” of trastuzumab, while the other two are independently derived. In HM2 a metai-binding motif has been incorporated into trastuzumab to aid conjugation. TrasGEX® is a glycosylation-optimized version of trastuzumab. Hertuzumab had a higher ELISA-based affinity for Her2 than trastuzumab. HT- 19 is an !gG1 antibody that is non-competitive for HER2 binding with trastuzumab and pertuzumab; that is, it binds a different epitope than either of those two mAbs.
  • Many embodiments comprise administration of a single anti-Her2 mAb, but some embodiments comprise administration of multiple anti-Her2 mAbs, for example, trastuzumab and pertuzumab. While the disclosed embodiments are generally described as using anti-Her2 mAbs, further embodiments can substitute anti-Her2 polyclonal antiserum for anti-Her2 mAb.
  • anti-Her2 mAbs In addition to use of anti-Her2 mAbs themselves as therapeutic agents, anti-Her2 mAbs have also been incorporated into antibody-drug conjugates.
  • One example is ado- trastuzumab emtansine (sold as KADCYLA®).
  • ALT-P7 (trastuzumab biobetter HM2 conjugated in a site-specific manner to monomethyl auristatin E)
  • ARX788 (a monoclonal HER2 targeting antibody site-specifically conjugated, via a non-natural amino acid linker para-acetyl-phenylalanine (pAcF), to monomethyl auristatin F)
  • DHES0815A (a monoclonal HER-2 targeting antibody linked to pyrrolo[2,1-c][1 ,4]benzodiazepine monoamide)
  • DS-8201 a (trastuzumab deruxtecan; trastuzumab, an enzymatically cleavable maleimide glycynglycyn-phenylalanyn-glycyn (GGFG) peptide linker and a topoisomerase I inhibitor)
  • RC48 humanized anti-HER2 antibody hertuzumab conjugated with monomethyl auri
  • the Her2-targeting component comprises, or is, an antibody- drug conjugate comprising an anti-Her2 antibody.
  • Her2 and EGFR are closely related protein tyrosine kinases and many drugs developed as an inhibitor of one also inhibit the other. At least four drugs that are irreversible inhibitors of these kinases are now marketed as a cancer treatment, though the indications vary: lapatinib, neratinib, afatinib, and dacomitinib. It should be noted that not all EGFR inhibitors are irreversible inhibitors or are known to cross-inhibit Her2. EGFR inhibitors that do not - or are not known to - inhibit Her2 should not be considered Her2 inhibitors as the term is used herein. In some embodiments, the Her2 inhibitor is an irreversible inhibitor. In some embodiments, the Her2 inhibitor is not a reversible inhibitor.
  • Lapatinib (sold as TYKERB®) can be administered, according to its prescribing instructions which are incorporated herein by reference in their entirety, on a 21 day treatment cycle.
  • Lapatinib plus capecitabine is taken on days 1 to 14.
  • Lapatinib aione is taken on days 15 to 21.
  • the treatment cycle should be repeated until disease progression or unacceptable toxicity occurs.
  • Capecitabine is administered orally in two doses approximately 12 hours apart at a dosage of 2000 mg/m 2 /day
  • Neratinib (sold as NERLYNX®) can be administered, according to its prescribing instructions which are incorporated herein by reference in their entirety, with food at an initial dose of 240 mg/day and taken daily for a year. If toxicity exceeds grade 1 , the dose can be reduced by 40 mg/day in stepwise fashion until toxicity is grade 1 or less. If the dose has been reduced to 120 mg/day and toxicity remains greater than grade 1 , treatment with Neratinib should be discontinued.
  • Afatinib (sold as GILOTRIF® ⁇ can be administered, according to its prescribing instructions which are incorporated herein by reference in their entirety, orally without food once daily at 40 mg/day until disease progression or no longer tolerated by the patient.
  • Dacomitinib (sold as VIZIMPRO®) can be administered, according to its prescribing instructions which are incorporated herein by reference in their entirety, orally with or without food, once daily at 45 mg/day until disease progression or unacceptable toxicity occurs. Upon occurrence of unacceptable toxicity, the dosage can be reduced in stepwise fashion to 30 or 15 mg/day.
  • RXR Retinoid X Receptors
  • RARs Retinoic Acid Receptors
  • RXR Retinoic Acid Receptors
  • a RAR biomarker is a distinctive biological, biochemical or biologically derived indicator that signifies patient RAR activity.
  • RAR biomarkers include, but are not limited to, CYP26 levels, CRBPI levels, and the like, and combinations thereof.
  • the RAR activation threshold means one or more of a CYP26 level which is 25% increased over baseline and a CRBPI level 25% increased over baseline.
  • the RARs form heterodimers with RXRs and these RAR/RXR heterodimers bind to specific response elements in the promoter regions of target genes. The binding of RAR agonists to the RAR receptor of the heterodimer results in activation of transcription of target genes leading to retinoid effects. On the other hand, the disclosed RXR agonists do not activate RAR/RXR heterodimers.
  • RXR heterodimer complexes like RAR/RXR can be referred to as non-permissive RXR heterodimers as activation of transcription due to ligand-binding occurs only at the non-RXR protein (e.g., RAR); activation of transcription does not occur due to ligand binding at the RXR.
  • non-RXR protein e.g., RAR
  • RXRs also interact with nuclear receptors other than RARs and RXR agonists may elicit some of its biological effects by binding to such RXR/receptor complexes.
  • RXR/receptor complexes can be referred to as permissive RXR heterodimers as activation of transcription due to ligand-binding could occur at the RXR, the other receptor, or both receptors.
  • permissive RXR heterodimers include, without limitation, peroxisome proliferator activated receptor/RXR (PPAR/RXR), farnesyl X receptor/RXR (FXR/RXR), nuclear receptor related- 1 protein (Nurr1/RXR) and liver X receptor/RXR (LXR/RXR).
  • RXRs may form RXR/RXR homodimers which can be activated by RXR agonists leading to rexinoid effects.
  • RXRs interact with proteins other than nuclear receptors and ligand binding to an RXR within such protein complexes can also lead to rexinoid effects.
  • RXR agonists and RAR agonists elicit distinct biological outcomes and even in the instances where they mediate similar biological effects, they do so by different mechanisms.
  • the unwanted side effects of retinoids such as pro-inflammatory responses or mucocutaneous toxicity, are mediated by activation of one or more of the RAR receptor subtypes.
  • biological effects mediated via RXR pathways would not induce pro-inflammatory responses, and thus, would not result in unwanted side effects.
  • aspects of the present specification provide, in part, a RXR agonist.
  • RXR agonist is synonymous with“selective RXR agonist” and refers to a compound that selectively binds to one or more RXR receptors like a RXRa, a RXRp, or a RXRy in a manner that elicits gene transcription via an RXR response element.
  • the term“selectively binds,” when made in reference to a RXR agonist, refers to the discriminatory binding of a RXR agonist to the indicated target receptor like a RXRa, a RXRp, or a RXRy such that the RXR agonist does not substantially bind with non-target receptors like a RARa, a RARp or a RARy.
  • the term“RXR agonist” includes esters of RXR agonist.
  • RXR agonists can be compounds having the structure of Formula I
  • R is H, or lower alkyl of 1 to 6 carbons; or the agonist is a pharmaceutically acceptable salt of the compounds.
  • esters of RXR agonists may be derived from a carboxylic acid of C1 , or an ester may be derived from a carboxylic acid functional group on another part of the molecule, such as on a phenyl ring. While not intending to be limiting, an ester may be an alkyl ester, an aryl ester, or a heteroaryl ester.
  • alkyl has the meaning generally understood by those skilled in the art and refers to linear, branched, or cyclic alkyl moieties.
  • Ci- 6 alkyl esters are particularly useful, where alkyl part of the ester has from 1 to 6 carbon atoms and includes, but is not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, sec- butyl, /so- butyl, f-butyl, pentyl isomers, hexyl isomers, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and combinations thereof having from 1-6 carbon atoms, etc.
  • the RXR agonist is the ethyl ester of formula I.
  • the RXR agonist is 3,7-dimethyl-6(S),7(S)-methano,7- [1 ,1 ,4,4-tetramethyl-1 ,2,3,4-tetrahydron-aphth-7-yl]2(E), 4(E) heptadienoic acid, also known as IRX4204, and has the following chemical structure:
  • RXR agonists can also be used in the disclosed embodiments.
  • Acids commonly employed to form acid addition salts from RXR agonists with basic groups are inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenyl-sulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like
  • organic acids such as p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenyl-sulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
  • salts include the sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caproate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1 ,4- dioate, hexyne-1 ,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, sulfonate, xylenesulfonate, phenylacetate, phenylpropionate, phenylbuty
  • Bases commonly employed to form base addition salts from RXR agonists with acidic groups include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or trialkylamines; dicyclohexylamine; tributyl amine; pyridine; N-methyl,N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris- (2- hydroxy-lower alkyl amines), such as mono-, bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy- tert-butylamine, or tris-(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxy lower alkyl
  • IRX4204 like some other RXR ligands, does not activate non-permissive heterodimers such as RAR/RXR. However, IRX4204, is unique in that it specifically activates the Nurr1/RXR heterodimer and does not activate other permissive RXR heterodimers such as PPAR/RXR, FXR/RXR, and LXR/RXR. Other RXR ligands generally activate these permissive RXR heterodimers. Thus, all RXR ligands cannot be classified as belonging to one class. IRX4204 belongs to a unique class of RXR ligands which specifically activate RXR homodimers and only one of the permissive RXR heterodimers, namely the Nurr1/RXR heterodimer.
  • the selective RXR agonist does not activate to any appreciable degree the permissive heterodimers PPAR/RXR, FXR/RXR, and LXR/RXR.
  • the selective RXR agonist activates the permissive heterodimer Nurr1/RXR.
  • a selective RXR agonist is 3,7-dimethyl-6(S),7(S)-methano,7-[1 ,1 ,4,4- tetramethyl-1 ,2,3,4-tetrahydronaphth-7-yl]2(E),4(E) heptadienoic acid (IRX4204) disclosed herein, the structure of which is shown in Formula II.
  • the RXR agonists activates the permissive heterodimers PPAR/RXR, FXR/RXR, or LXR/RXR by 1 % or less, 2% or less, 3% or less, 4% or less, 5% or less, 6% or less, 7% or less, 8% or less, 9% or less, or 10% or less relative to the ability of activating agonists to the non-RXR receptor to activate the same permissive heterodimer.
  • RXR agonists which activates one or more of PPAR/RXR, FXR/RXR, or LXR/RXR include LGD1069 (bexarotene) and LGD268.
  • Binding specificity is the ability of a RXR agonist to discriminate between a RXR receptor and a receptor that does not contain its binding site, such as a RAR receptor.
  • Particular embodiments provide methods of treating cancer comprising administering to a patient in need of such treatment a RXR agonist at a level below an RAR activating threshold and at or above an RXR activating threshold.
  • the RAR ECio (the concentration effective to cause a 10% of maximal activation of the RAR) is 300 nM for the a isoform and 200 nM for the b and g isoforms. Thus, in some embodiments, concentrations not exceeding 200 nM are considered to be below an RAR activating concentration.
  • the RXR EC 90 (the concentration effective to cause a 90% of maximal activation of the RXR) is 0.1 nM for the a and g isoforms and 1 nM for the b isoform. Thus, in some embodiments concentrations of at least 0.1 nM are considered to be above an RXR activating threshold.
  • IRX4204 Based on studies in humans, oral dosages of IRX4204 of 20 mg/m 2 /day will produce systemic concentrations that remain below 200 nM. Similarly, it is estimated that an oral dosage in the range of 0.01 to 0.02 mg/m 2 /day will produce systemic concentrations of 0.1 nM or greater. Thus, in various individual embodiments a dosage of IRX4204 is at least 0.01 , 0.02, 0.03, 0.05, 0.1 , 0.3, 0.5, 1 , 3 or 5 mg/m 2 /day and does not exceed 150, 200, or 300 mg/m 2 /day, or any range bound by a pair of these values.
  • the dosage for a human adult of the RXR agonist is from 0.2 to 300 mg/day, such as in individual embodiments, from 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 mg/day, but not to exceed 10, 15, 20, 50, or 100 mg/day, or any range bound by a pair of these values.
  • the RXR agonist can be administered to a mammal using standard administration techniques, including parenteral, oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration.
  • parenteral includes intravenous, intramuscular, subcutaneous, rectal, vaginal, and intraperitoneal administration.
  • the RXR agonist preferably is suitable for oral administration, for example as a pill, tablet or capsule. Administration may be continuous or intermittent.
  • the total daily dosage of RXR agonist can be administered as a single dose or as two doses administered with a 24 hour period spaced 8 to 16, or 10 to 14, hours apart.
  • thyroid hormone Both biologically sourced and synthetic thyroid hormones have been used in medicine.
  • the major forms of thyroid hormone are referred to a T 3 (triiodothyronine) and T (thyroxine).
  • Thyroxine is less active but has a longer half-life and is sometimes considered a prohormone of triiodothyronine.
  • thyroid hormone refers to thyroxine and triiodothyronine.
  • Thyroxine thyroid hormone T 4 , levothyroxine sodium
  • T 4 thyroid hormone
  • the synthetic form of T levothyroxine
  • the administered thyroid hormone is specifically thyroxine.
  • the administered thyroid hormone is triiodothyronine.
  • RXR agonists may lead to the suppression of serum thyroid hormones and possibly to clinical hypothyroidism and related conditions.
  • thyroid hormone is not co-administered (or is not primarily co-administered) to remediate a suppression of serum thyroid hormone levels.
  • Coadministration of thyroid hormone with an RXR agonist improves the RXR agonist's anticancer efficacy, as compared to the effect of the RXR agonist alone, likely through multiple mechanisms of action.
  • the co-administered thyroid hormone can also mitigate the hypothyroid-inducing effects of the RXR agonist, thereby improving the clinical safety and tolerability of the treatment.
  • thyroid hormone is coadministered with an RXR agonist to improve the efficacy of the treatment, whether or not administration of the RXR agonist has caused, or is expected to cause, clinical hypothyroidism.
  • administration of thyroid hormone in coordination or in conjunction with the RXR agonist it is meant that the manner of administration of each of these two agents is such that the physiologic effects of the two agents overlap in time. This does not require that the RXR agonist and thyroid hormone be contained in the same composition or formulation, or that they be administered as separate compositions at the same time, by the same route of administration, or on the same schedule, though in some embodiments any of the foregoing may be the case.
  • Suitable thyroxine doses are generally from about 5 pg/day to about 250 pg/day orally initially with an increase in dose every 2-4 weeks as needed.
  • the suitable thyroxine dose is from about 5 pg/day to about 225 pg/day, from about 7.5 pg/day to about 200 pg/day, from about 10 pg/day to about 175 pg/day, from about 12.5 pg/day to about 150 pg/day, from about 15 pg/day to about 125 pg/day, from about 17.5 pg/day to about 100 pg/day, from about 20 pg/day to about 100 pg/day, from about 22.5 pg/day to about 100 pg/day, from about 25 pg/day to about 100 pg/day, from about 5 pg/day to about 200 pg/day, from about 5 pg/day to about 100 pg/day, from about 5 pg/
  • Increases in dose are generally made in increments of about 5 uo/dav about 7 5 uo/dav about 10 uo/dav about 12 5 uo/day, about 15 pg/day, about 20 pg/day, or about 25 pg/day.
  • the suitable thyroid hormone dose is a dose able to produce serum levels of T in the top 50%, the top 60%, the top 70%, the top 80%, or the top 90% of the normal range for the testing laboratory.
  • the target T levels are based on normal ranges determined for each particular testing laboratory.
  • compositions or formulations may be a liquid formulation, semi-solid formulation, or a solid formulation.
  • a formulation disclosed herein can be produced in a manner to form one phase, such as, e.g., an oil or a solid.
  • a formulation disclosed herein can be produced in a manner to form two phases, such as, e.g., an emulsion.
  • a pharmaceutical composition disclosed herein intended for such administration may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions.
  • Liquid formulations suitable for parenteral injection or for nasal sprays may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Formulations suitable for nasal administration may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions.
  • aqueous and nonaqueous carriers, diluents, solvents or vehicles examples include water, ethanol, polyols (propylene glycol, polyethyleneglycol (PEG), glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • PEG polyethyleneglycol
  • glycerol glycerol
  • suitable mixtures thereof such as vegetable oils (such as olive oil)
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • a pharmaceutical composition disclosed herein can optionally include a pharmaceutically acceptable carrier that facilitates processing of an active compound into pharmaceutically acceptable compositions.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem comDlications commensurate with a reasonable benefit/risk ratio. (This definition is also application to the phrase “pharmaceutically-acceptable salts”).
  • the term “pharmacologically acceptable carrier” is synonymous with “pharmacological carrier” and refers to any carrier that has substantially no long term or permanent detrimental effect when administered and encompasses terms such as “pharmacologically acceptable vehicle, stabilizer, diluent, additive, auxiliary, or excipient.”
  • a carrier generally is mixed with an active compound or permitted to dilute or enclose the active compound and can be a solid, semi-solid, or liquid agent. It is understood that the active compounds can be soluble or can be delivered as a suspension in the desired carrier or diluent.
  • aqueous media such as, e.g., water, saline, glycine, hyaluronic acid and the like
  • solid carriers such as, e.g., starch, magnesium stearate, mannitol, sodium saccharin, talcum, cellulose, glucose, sucrose, lactose, trehalose, magnesium carbonate, and the like
  • solvents dispersion media; coatings; antibacterial and antifungal agents; isotonic and absorption delaying agents; or any other inactive ingredient.
  • Selection of a pharmacologically acceptable carrier can depend on the mode of administration.
  • any pharmacologically acceptable carrier is incompatible with the active compound, its use in pharmaceutically acceptable compositions is contemplated.
  • Non-limiting examples of specific uses of such pharmaceutical carriers can be found in Pharmaceutical Dosage Forms and Drug Delivery Systems (Howard C. Ansel et al., eds., Lippincott Williams & Wilkins Publishers, 7 th ed. 1999); Remington: The Science and Practice of Pharmacy (Alfonso R. Gennaro ed., Lippincott, Williams & Wilkins, 20 th ed. 2000); Goodman & Gilman's The Pharmacological Basis of Therapeutics (Joel G.
  • a pharmaceutical composition disclosed herein can optionally include, without limitation, other pharmaceutically acceptable components (or pharmaceutical components), including, without limitation, buffers, preservatives, tonicity adjusters, salts, antioxidants, osmolality adjusting agents, physiological substances, pharmacological substances, bulking agents, emulsifying agents, wetting agents, sweetening or flavoring agents, and the like.
  • buffers include, without limitation, acetate buffers, borate buffers, citrate buffers, phosphate buffers, neutral buffered saline, and phosphate buffered saline.
  • antioxidants include, without limitation, sodium metabisulfite, sodium thiosulfate, acetylcysteine, butylated hydroxyanisole, and butylated hydroxytoluene.
  • Useful preservatives include, without limitation, benzalkonium chloride, chlorobutanol, thimerosal, phenylmercuric acetate, phenylmercuric nitrate, a stabilized oxy chloro composition, such as, e.g., sodium chlorite and chelants, such as, e.g., DTPA or DTPA- bisamide, calcium DTPA, and CaNaDTPA-bisamide.
  • Tonicity adjustors useful in a pharmaceutical composition include, without limitation, salts such as, e.g., sodium chloride, potassium chloride, mannitol or glycerin and other pharmaceutically acceptable tonicity adjustor.
  • the pharmaceutical composition may be provided as a salt and can be formed with many acids, including but not limited to, hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms. It is understood that these and other substances known in the art of pharmacology can be included in a pharmaceutical composition useful in the invention.
  • Fine particle dusts or mists which may be generated by means of various types of metered, dose pressurized aerosols, nebulizers, or insufflators.
  • Semi-solid formulations suitable for topical administration include, without limitation, ointments, creams, salves, and gels.
  • the active compound may be admixed with at least one inert customary excipient (or carrier) such as, a lipid and/or polyethylene glycol.
  • Solid formulations suitable for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound may be admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol and silicic acid, (b) binders, as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose and acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate, (e) solution retarders, as for example, paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g)
  • the dosage forms may also comprise buffering agents.
  • the small molecule components of the various embodiments that is, the RXR agonist, thyroid hormone, and Her2 kinase inhibitors are capable of being formulated in solid, oral dosage forms.
  • the antibody components are generally formulated as liquids typically for intravenous infusion.
  • the antibody components may be supplied in lyophilized form for reconstitution as a liquid locally at the site of treatment, where they are also typically infused intravenously into the patient. While intravenous infusion is typical, in alternative embodiments the antibody may be administered by another route of administration, such as subcutaneous injection or infusion.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect. This may be observed directly as a slowing of tumor growth, stabilization of disease, or a partial or complete response (that is, tumor regression or elimination of tumors), or extended overall or disease-free survival. Treatment may also be observed as an amelioration or reduction of symptoms related to the underlying cancer.
  • the disclosed embodiments aim and mechanism is directed to inhibiting, stabilizing, or reducing tumor growth (including metastases), or partially or completely eliminating tumors, or extending overall or disease-free survival; effects on other cancer symptoms are secondary.
  • treating cancer is not within the scope of treating cancer as used herein. That is, treating a symptom, for example, cachexia in a cancer patient is not treating cancer. However, an agent that treats cancer (e.g., has an impact on the growth and/or spread of cancer) may also ameliorate a symptom, such as cachexia, either indirectly, through its effect on the cancer, or directly, through a pleiotropic effect.
  • a "therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
  • the therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the Her2- targeted therapy, RXR agonist, and if used, thyroid hormone, to elicit a desired response in the individual.
  • the dose administered to a mammal, particularly a human, in the context of the present methods should be sufficient to effect a therapeutic response in the mammal over a reasonable timeframe.
  • the selection of the exact dose and composition and the most appropriate delivery regimen will also be influenced by inter alia the pharmacological properties of the formulation, the nature and severity of the condition being treated, and the physical condition and mental acuity of the recipient, as well as the potency of the specific compound, the age, condition, body weight, sex and response of the patient to be treated, and the stage/severity of the disease.
  • Treatment activity includes the administration of the medicaments, dosage forms, and pharmaceutical compositions described herein to a patient, especially according to the various methods of treatment disclosed herein, whether by a healthcare professional, the patient his/herself, or any other person.
  • Treatment activities include the orders, instructions, and advice of healthcare professionals such as physicians, physician’s assistants, nurse practitioners, and the like that are then acted upon by any other person including other healthcare professionals or the patient his/herself.
  • treatment activity can also include encouraging, inducing, or mandating that a particular medicament, or combination thereof, be chosen for treatment of a condition - and the medicament is actually used - by approving insurance coverage for the medicament, denying coverage for an alternative medicament, including the medicament on, or excluding an alternative medicament, from a drug formulary, or offering a financial incentive to use the medicament, as might be done by an insurance company or a pharmacy benefits management company, and the like.
  • treatment activity can also include encouraging, inducing, or mandating that a particular medicament be chosen for treatment of a condition - and the medicament is actually used - by a policy or practice standard as might be established by a hospital, clinic, health maintenance organization, medical practice or physicians group, and the like.
  • embodiments include methods of treatment comprising or consisting of administering RXR agonist of Formula I (or pharmaceutically acceptable salt thereof) and a Her2-targeted therapeutic to a patient having a Her2 + cancer. Some embodiments further comprise administration of thyroid hormone in coordination with administration of the RXR agonist. In some embodiments the Her2 + cancer is a Her2 + breast cancer.
  • the Her2 + cancer is a Her2 + gastroesophageal cancer, ovarian cancer, stomach cancer, adenocarcinoma of the lung, uterine cancer (such as serous endometrial carcinoma), or salivary duct carcinoma. Some embodiments specifically include one or more of these cancers. Other embodiments specifically exclude one or more of these cancers.
  • the herein disclosed treatments may be applied as a primary therapy, as a debulking therapy prior to surgical removal of tumor, or as an adjuvant therapy subsequent to any mode of primary therapy (especially surgery) to address residual disease and/or lower the risk of recurrent cancer.
  • the patient having a Her2 + cancer has not been previously treated with either RXR agonist of Formula I (or pharmaceutically acceptable salt thereof) or a Her2-targeted therapeutic.
  • the patient has been previously treated with RXR agonist of Formula I (or pharmaceutically acceptable salt thereof) and has achieved stable disease or a partial response (in some embodiments, as defined by RECIST or iRECIST criteria) - that is, the cancer is sensitive to RXR agonist of Formula I (or pharmaceutically acceptable salt thereof) - and a Her2-targeted therapeutic is added to the treatment regimen.
  • the patient has been previously treated with a Her2-targeted therapeutic and has achieved stable disease or a partial response (in some embodiments, as defined by RECIST or iRECIST criteria) - that is, the cancer is sensitive to a Her2-targeted therapeutic - and RXR agonist of Formula I (or pharmaceutically acceptable salt thereof) is added to the treatment regimen.
  • some embodiments entail administration of an RXR agonist to a patient with a Her2 + tumor who has received, is receiving, or is scheduled to receive, a Her2-targeted therapeutic agent. Some embodiments entail administration of an RXR agonist to a patient in whom a Her2-targeted therapeutic agent has had some therapeutic effect (less than a complete response), that is administration of the RXR agonist is added to the therapeutic regimen for the Her2-targeted therapeutic agent.
  • Some embodiments entail administration of a Her2-targeted therapeutic agent to a patient in whom an RXR agonist (or RXR agonist in conjunction with thyroid hormone) has had some therapeutic effect (less than a complete response), that is administration of the Her2-targeted therapeutic agent is added to the therapeutic regimen for the RXR agonist.
  • Therapeutic efficacy can be monitored by periodic assessment of treated patients. For repeated administrations over several days or longer, the treatment can be repeated until a desired suppression of disease or disease symptoms occurs. However, other dosage regimens may be useful and are within the scope of the present disclosure. Antibodies typically have a much longer half-life in the body than the other active agents used in these methods and therefore there will typically be substantially longer intervals (measured in weeks) between administrations.
  • the effectiveness of cancer therapy is typically measured in terms of "response.”
  • the techniques to monitor responses can be similar to the tests used to diagnose cancer such as, but not limited to:
  • a lump or tumor involving some lymph nodes can be felt and measured externally by physical examination. • Some internal cancer tumors will show up on an x-ray or CT scan and can be measured with a ruler.
  • a tumor marker test can be done for certain cancers.
  • test whether blood test, cell count, or tumor marker test, it is repeated at specific intervals so that the results can be compared to earlier tests of the same type.
  • Levels of a tumor marker may have fallen (or increased, based on the tumor marker, as an indication of decreased tumor burden) but evidence of disease remains.
  • a tumor marker (if applicable) has not changed significantly.
  • Other measures of the efficacy of cancer treatment include intervals of overall survival (that is time to death from any cause, measured from diagnosis or from initiation of the treatment being evaluated)), cancer-free survival (that is, the length of time after a complete response cancer remains undetectable), and progression-free survival (that is, the length of time after disease stabilization or partial response that resumed tumor growth is not detectable).
  • the tumor burden of a treated patient is reduced by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55% about 60%, about 65%, about 70%, about 75%, about 80%, about 90%, about 95%, about 100%, or any range bound by these values.
  • the 1-year survival rate of treated subjects is increased by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55% about 60%, about 65%, about 70%, about 75%, about 80%, about 90%, about 95%, about 100%, or any range bound by these values.
  • the 5-year survival rate of treated subjects is increased by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55% about 60%, about 65%, about 70%, about 75%, about 80%, about 90%, about 95%, about 100%, or any range bound by these values.
  • the 10-year survival rate of treated subjects is increased by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55% about 60%, about 65%, about 70%, about 75%, about 80%, about 90%, about 95%, about 100%, or any range bound by these values.
  • the subject has a sustained remission of at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 1 1 months, at least 12 months, at least 14 months, at least 16 months, at least 18 months, at least 20 months, at least 22 months, at least 24 months, at least 27 months, at least 30 months, at least 33 months, at least 36 months, at least 42 months, at least 48 months, at least 54 months, or at least 60 months or more.
  • the methods may additionally help to treat or alleviate conditions, symptoms, or disorders related to cancer.
  • these conditions or symptoms may include, but are not limited to, anemia, asthenia, cachexia, Cushing’s Syndrome, fatigue, gout, gum disease, hematuria, hypercalcemia, hypothyroidism, internal bleeding, hair loss, mesothelioma, nausea, night sweats, neutropenia, paraneoplastic syndromes, pleuritis, polymyalgia rheumatica, rhabdomyolysis, stress, swollen lymph nodes, thrombocytopenia, Vitamin D deficiency, or weight loss. While a cancer treatment may reduce or treat associated symptoms, treating symptoms associated with cancer, is not treating cancer if there is no expectation that tumor will be reduced or eliminated or their growth or spread will be inhibited.
  • Toxicities and adverse events are sometimes graded according to a 5 point scale.
  • a grade 1 or mild toxicity is asymptomatic or induces only mild symptoms; may be characterized by clinical or diagnostic observations only; and intervention is not indicated.
  • a grade 2 or moderate toxicity may impair activities of daily living (such as preparing meals, shopping, managing money, using the telephone, etc.) but only minimal, local, or non-invasive interventions are indicated.
  • Grade 3 toxicities are medically significant but not immediately life- threatening; hospitalization or prolongation of hospitalization is indicated; activities of daily living related to self-care (such as bathing, dressing and undressing, feeding oneself, using the toilet, taking medications, and not being bedridden) may be impaired.
  • Grade 4 toxicities are life-threatening and urgent intervention is indicated.
  • Grade 5 toxicity produces an adverse event-related death.
  • use of an RXR agonist, or RXR agonist and thyroid hormone reduces the grade of a toxicity that would otherwise be associated with use of the Her2-targered therapy, by allowing a lower dose to be used without substantial sacrifice of efficacy.
  • use of an RXR agonist, or RXR agonist and thyroid hormone, in combination with the Her2-targeted therapeutic agent limits a toxicity to grade 1 or less, or produces no observation of the toxicity, without substantial reduction of efficacy as would be expected from the Her2-targeted therapeutic agent alone.
  • the combined use of the Her2-targeted therapeutic agent and the RXR agonist, or RXR agonist and thyroid hormone allows continued use of the Her2-targeted therapeutic agent at a lower dosage with therapeutic effect in instances where treatment with the Her2-targeted therapeutic agent would have had to have been discontinued due to unacceptable toxicity.
  • the Her2-targeted therapeutic agent comprises a Her2 kinase inhibitor.
  • the combination of the disclosed RXR agonists and the Her2 targeted therapeutics are synergistic in effect. That is, they interact in a positive manner to produce a greater inhibition of tumor cell growth, than would be expected from the independent (non-interacting) effects of the two. Thus, some embodiments produce improved efficacy. Other embodiments allow for the reduction of dosage in order to reduce toxicity while still achieving at least similar efficacy as provided by an individual therapeutic agent. In some embodiments, both reduced tnxinitv and imnrnvad affinanv fas nnmnamd to tha mom toxic single agent) is achieved.
  • each method of treatment there are further parallel embodiments related to the foregoing methods directed to use of the RXR agonist in conjunction with a Her2-targeted therapeutic agent, or a Her2-targeted therapeutic agent and thyroid hormone, to treat Her2 + cancer; directed to use of the RXR agonist in the manufacture of a medicament for use in combination with a Her2-targeted therapeutic agent, or a Her2-targeted therapeutic agent and thyroid hormone, to treat Her2 + cancer.
  • Further embodiments include a combination comprising a RXR agonist as herein described and a Her2-targeted therapeutic agent. Some embodiments further comprise a thyroid hormone. In some embodiments, the Her2-targeted therapeutic agent is an anti-Her2 antibody. In some embodiments the Her2-targeted therapeutic agent is a Her2 kinase inhibitor.
  • kits comprising the above combinations.
  • the kits may additionally comprise solvents, diluents, injectors, and the like that may facilitate administration of one or more of the therapeutic agents.
  • the kits may further comprise instructions for the coordinated use of the therapeutic agents utilized in the disclosed methods, whether or not any particular agent is supplied in the kit.
  • Embodiment 1 A method of treating a patient with Her2 + cancer comprising administering a RXR agonist of Formula I,
  • Embodiment 2 A method of treating a patient with Her2+ cancer comprising administering a RXR agonist of Formula I,
  • Embodiment 3 A method of treating a patient with Her2+ cancer undergoing treatment with a RXR agonist of Formula I,
  • R is H, or lower alkyl of 1 to 6 carbons; or a pharmaceutically-acceptable salt thereof,
  • Embodiment 4 A method of treating a patient with Her2+ cancer undergoing treatment with a Her2-targeted therapeutic agent, wherein there is evidence of therapeutic effect that is less than a complete response, comprising continuing treatment with the Her2- targeted therapeutic agent and initiating treatment with a RXR agonist of Formula I,
  • Embodiment 5 A method of treating a patient with Her2 + cancer comprising administering a RXR agonist of Formula I, (Formula I) wherein R is H, or lower alkyl of 1 to 6 carbons; or a pharmaceutically-acceptable salt thereof, to the patient,
  • Embodiment 6 The method of Embodiment 5, wherein the means for therapeutically targeting Her2 are:
  • immunoglobulin means for inhibiting Her2 signaling
  • small molecule means for inhibiting Her2 kinase activity.
  • Embodiment 7 A method of treating a patient with Her2 + cancer comprising administering means for activating RXR/Nurr1 heterodimeric receptors or rexinoid means for inhibiting tumor growth, wherein the patient has received, is receiving, is scheduled to receive a Her2-targeted therapeutic agent.
  • Embodiment 8 The method of any one of Embodiments 1 -7, further comprising administering thyroid hormone in conjunction with the RXR agonist.
  • Embodiment 9 The method of Embodiment 8, wherein the thyroid hormone is thyroxine.
  • Embodiment 10 The method of any one of Embodiments 1-9, wherein the RXR agonist, the means for activating RXR/Nurr1 heterodimeric receptors, or the rexinoid means for inhibiting tumor growth, is a compound of Formula I.
  • Embodiment 1 1. The method of any one of Embodiments 1-9, wherein the RXR agonist, the means for activating RXR/Nurr1 heterodimeric receptors, or the rexinoid means for inhibiting tumor growth, is a pharmaceutically-acceptable salt of a compound of Formula I.
  • Embodiment 13 The method of any one of Embodiments 1-12, wherein:
  • the immunoglobulin means for inhibiting Her2 signaling, or
  • Embodiment 14 The method of Embodiment 13, wherein the therapeutic antibody is trastuzumab or pertuzumab.
  • Embodiment 15 The method of Embodiment 13, wherein the therapeutic antibody is margetuximab, TrasGEX, HM2, hertuzumab, or HT-19
  • Embodiment 16 The method of any one of Embodiments 1-13, wherein the Her2- targeted therapeutic agent, the means for inhibiting Her2 + tumor cell proliferation, or the immunoglobulin means for inhibiting Her2 signaling, comprises an antibody-drug conjugate wherein the antibody is an anti-Her2 antibody.
  • Embodiment 17 The method of Embodiment 16, wherein the antibody-drug conjugate or the means for delivering a cytotoxic agent to Her2 + cells, is ado-trastuzumab emtansine.
  • Embodiment 18 The method of Embodiment 16, wherein the antibody-drug conjugate or the means for delivering a cytotoxic agent to Her2 + cells, is A166, ALT-P7, ARX788, DHES0815A, DS-8201 a, RC48, SYD985, MEDI4276, or XMT-1522.
  • Embodiment 19 The method of any one of Embodiments 1-12, wherein the Her2- targeted therapeutic agent, or the small molecule means for inhibiting Her2 kinase activity, comprises a Her2 kinase inhibitor.
  • Embodiment 20 The method of Embodiment 19, wherein the Her2 kinase inhibitor is lapatinib or neratinib.
  • Embodiment 21 The method of Embodiment 19, wherein the Her2 kinase inhibitor is afatinib or dacomitinib.
  • Embodiment 22 The method of any one of Embodiments 1-21 , wherein the treatment is applied as a debulking therapy.
  • Embodiment 23 The method of any one of Embodiments 1-21 , wherein the treatment is applied as adjuvant therapy.
  • Embodiment 24 The method of any one of Embodiments 1 -23, wherein the Her2 + cancer is Her2 + breast cancer.
  • Embodiment 25 The method of any one of Embodiments 1-23, wherein the Her2 + cancer is Her2 + gastroesophageal cancer, ovarian cancer, stomach cancer, adenocarcinoma of the lung, uterine cancer (such as serous endometrial carcinoma), or salivary duct carcinoma.
  • Her2 + cancer is Her2 + gastroesophageal cancer, ovarian cancer, stomach cancer, adenocarcinoma of the lung, uterine cancer (such as serous endometrial carcinoma), or salivary duct carcinoma.
  • Embodiment 26 The method of any one of Embodiments 1-25, wherein a therapeutic response to the RXR agonist and the Her2-targeted therapeutic agent is greater than to the response to either of the agents alone.
  • Embodiment 27 The method of Embodiment 26, wherein the greater therapeutic response is a slowing of tumor growth, stabilization of disease, a partial response, a complete response, extended overall survival, or disease-free survival.
  • Embodiment 28 The method of Embodiment 26 or 27, wherein response is evaluated according to RECIST or iRECIST criteria.
  • Embodiment 29 The method of any one of Embodiments 26-28, comprising a reduction or amelioration of secondary symptoms.
  • Embodiment 30 A combination comprising a Her2-targeted therapeutic agent, or means for therapeutically targeting Her2, and a RXR agonist of Formula I, (Formula I) wherein R is H, or lower alkyl of 1 to 6 carbons; or a pharmaceutically-acceptable salt thereof.
  • Embodiment 31 The combination of Embodiment 30, wherein the Her2-targeted therapeutic agent, or means for therapeutically targeting Her2, comprises:
  • immunoglobulin means for inhibiting Her2 signaling
  • small molecule means for inhibiting Her2 kinase activity.
  • Embodiment 32 The combination of Embodiment 30 or 31 , further comprising thyroid hormone.
  • Embodiment 33 The combination of any one of Embodiments 30-32, wherein the RXR agonist is a compound of Formula II
  • Embodiment 34 The combination of any one of Embodiments 30-33, wherein: the Her2-targeted therapeutic agent,
  • the immunoglobulin means for inhibiting Her2 signaling, or
  • Embodiment 35 The combination of Embodiment 34, wherein the therapeutic antibody is trastuzumab or pertuzumab.
  • Embodiment 36 The combination of any one of Embodiments 30-34, wherein the Her2-targeted therapeutic agent, the means for inhibiting Her2 + tumor cell proliferation, or the immunoglobulin means for inhibiting Her2 signaling, comprises an antibody-drug conjugate wherein the antibody is an anti-Her2 antibody.
  • Embodiment 37 The combination of Embodiment 36, wherein the antibody-drug conjugate is ado-trastuzumab emtansine.
  • Embodiment 38 The combination of any one of Embodiments 30-33, wherein the Her2-targeted therapeutic agent, or the small molecule means for inhibiting Her2 kinase activity, comprises a Her2 kinase inhibitor.
  • Embodiment 39 The combination of Embodiment 38, wherein the Her2 kinase inhibitor is lapatinib or neratinib.
  • Embodiment 40 A kit comprising the combination of any one of Embodiments 30-39.
  • Embodiment 41 A RXR agonist of Formula I,
  • Embodiment 42 A RXR agonist of Formula I,
  • Embodiment 44 A pharmaceutical combination for treating Her2 + cancer comprising a RXR agonist of Formula I,
  • Embodiment 45 A RXR agonist of Formula I,
  • Embodiment 46 A RXR agonist of Formula I,
  • Embodiment 47 A Her2-targeted therapeutic agent for treating Her2+ cancer in a patient with Her2+ cancer, with a therapeutic response less than a complete response to treatment with a RXR agonist of Formula I,
  • Embodiment 48 A RXR agonist of Formula I,
  • each or Embodiments 41-48 can be modified in a manner similar to the modification of Embodiments 1 -5 and 7 by Embodiments 5 and 8-29.
  • Example 1 The following non-limiting examples are provided for illustrative purposes only in order to facilitate a more complete understanding of representative embodiments now contemplated. These examples should not be construed to limit any of the embodiments described in the present specification, Example 1
  • MCF7 Two breast cancer cell lines, MCF7 and SkBr3 were cultured in the presence of 0, 10, 100, or 1000 nM IRX4204 and 0, 0.1 , 1 , or 10 pg/ml trastuzumab.
  • MCF7 is an EFT PR + Her2 cell line.
  • SkBr3 is an ER PR Her2 + cell line.
  • Cells were plated in 96 well optical plates and IRX-4204 and trastuzumab were added 24 hours after cell plating. After a further 6 days, the cells were fixed with 4% paraformaldehyde in phosphate buffered saline (PBS). Nuclei were then stained with DAPI and imaged with the MetaXpress® microscope (Molecular Devices, San Jose, CA).
  • Example 1 The experiment of Example 1 was repeated using the Her2 kinase inhibitors lapatinib or neratinib instead of trastuzumab at 0, 0.1 , 1 , or 10 nM. Additionally the panel of breast cancer cell lines was expanded to also include the ER + PR + Her2 + cell line BT474 and the ER + PR Her2 + cell line MDA-MB-361. The general pattern seen above, that the Her2 MCF7 cell line showed generally marginal response to the treatments and that the Her + cell lines exhibited a greater degree of inhibition to the combination than either agent alone, was again observed (see Figures 2 and 3).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Medicinal Preparation (AREA)

Abstract

La présente invention concerne des combinaisons d'agents actifs pour le traitement amélioré de cancers Her2 + et des procédés de traitement associés. Les combinaisons comprennent un agoniste de RXR et un agent thérapeutique ciblant Her2 et peuvent éventuellement comprendre en outre une hormone thyroïdienne.
PCT/US2019/036594 2019-06-11 2019-06-11 Utilisation d'un agoniste de rxr dans le traitement de cancers her2 + WO2020251556A1 (fr)

Priority Applications (6)

Application Number Priority Date Filing Date Title
BR112021024970A BR112021024970A2 (pt) 2019-06-11 2019-06-11 Uso de um agonista rxr no tratamento de cânceres her2+
KR1020217043352A KR20220019724A (ko) 2019-06-11 2019-06-11 Her2+ 암 치료에서의 rxr 작용제의 용도
JP2021573710A JP7438496B2 (ja) 2019-06-11 2019-06-11 Her2+がんの治療におけるrxrアゴニストの使用
PCT/US2019/036594 WO2020251556A1 (fr) 2019-06-11 2019-06-11 Utilisation d'un agoniste de rxr dans le traitement de cancers her2 +
CA3142076A CA3142076A1 (fr) 2019-06-11 2019-06-11 Utilisation d'un agoniste de rxr dans le traitement de cancers her2 +
AU2019450366A AU2019450366A1 (en) 2019-06-11 2019-06-11 Use of an RXR agonist in treating Her2+ cancers

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2019/036594 WO2020251556A1 (fr) 2019-06-11 2019-06-11 Utilisation d'un agoniste de rxr dans le traitement de cancers her2 +

Publications (1)

Publication Number Publication Date
WO2020251556A1 true WO2020251556A1 (fr) 2020-12-17

Family

ID=73780879

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/036594 WO2020251556A1 (fr) 2019-06-11 2019-06-11 Utilisation d'un agoniste de rxr dans le traitement de cancers her2 +

Country Status (6)

Country Link
JP (1) JP7438496B2 (fr)
KR (1) KR20220019724A (fr)
AU (1) AU2019450366A1 (fr)
BR (1) BR112021024970A2 (fr)
CA (1) CA3142076A1 (fr)
WO (1) WO2020251556A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023108012A1 (fr) * 2021-12-07 2023-06-15 Io Therapeutics, Inc. Utilisation d'un agoniste de rxr et de taxanes dans le traitement de cancers her2+

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080102069A1 (en) * 2006-09-15 2008-05-01 Thomas Friess Tumor therapy with a combination of anti-her2 antibodies
WO2017075612A1 (fr) * 2015-10-31 2017-05-04 Io Therapeutics, Inc. Traitement du cancer avec des combinaisons d'agonistes de rxr et d'hormones thyroïdiennes
US20170136026A1 (en) * 2008-03-18 2017-05-18 Genentech, Inc. Combinations of anti-her2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
US20180369181A1 (en) * 2015-10-31 2018-12-27 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of rxr agonists and thyroid hormones

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018201016A1 (fr) 2017-04-28 2018-11-01 Cascadian Therapeutics, Inc. Traitement des cancers positifs à her2

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080102069A1 (en) * 2006-09-15 2008-05-01 Thomas Friess Tumor therapy with a combination of anti-her2 antibodies
US20170136026A1 (en) * 2008-03-18 2017-05-18 Genentech, Inc. Combinations of anti-her2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
WO2017075612A1 (fr) * 2015-10-31 2017-05-04 Io Therapeutics, Inc. Traitement du cancer avec des combinaisons d'agonistes de rxr et d'hormones thyroïdiennes
US20180369181A1 (en) * 2015-10-31 2018-12-27 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of rxr agonists and thyroid hormones

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CORRIGAN, PA ET AL.: "Ado-trastuzumab emtansine: a HER2-positive targeted antibody-drug conjugate", THE ANNALS OF PHARMACOTHERAPY, vol. 48, no. Issue 11, 31 July 2014 (2014-07-31), pages 1484 - 1493, XP055264705 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023108012A1 (fr) * 2021-12-07 2023-06-15 Io Therapeutics, Inc. Utilisation d'un agoniste de rxr et de taxanes dans le traitement de cancers her2+

Also Published As

Publication number Publication date
CA3142076A1 (fr) 2020-12-17
JP7438496B2 (ja) 2024-02-27
BR112021024970A2 (pt) 2022-01-25
AU2019450366A1 (en) 2022-01-06
JP2022543524A (ja) 2022-10-13
KR20220019724A (ko) 2022-02-17

Similar Documents

Publication Publication Date Title
US11224583B2 (en) Use of an RXR agonist in treating HER2+ cancers
US11648223B2 (en) Receptor subtype and function selective retinoid and rexinoid compounds in combination with immune modulators for cancer immunotherapy
Agero et al. Dermatologic side effects associated with the epidermal growth factor receptor inhibitors
US8828391B2 (en) Method for EGFR directed combination treatment of non-small cell lung cancer
JP2022017295A (ja) Hdac阻害剤とpd-l1阻害剤との組み合わせ療法
JP2014132009A5 (fr)
KR102088469B1 (ko) Mek 억제제 및 igf1r 억제제의 병용 요법
Bastholt et al. Phase I/II clinical and pharmacokinetic study evaluating a fully human monoclonal antibody against EGFr (HuMax-EGFr) in patients with advanced squamous cell carcinoma of the head and neck
JP7438496B2 (ja) Her2+がんの治療におけるrxrアゴニストの使用
US20230172890A1 (en) Use of an rxr agonist in treating drug resistant her2+ cancers
US11896558B2 (en) Use of an RXR agonist and taxanes in treating Her2+ cancers
Gamez et al. Corticorelin acetate, a synthetic corticotropin-releasing factor with preclinical antitumor activity, alone and with bevacizumab, against human solid tumor models
Yu et al. 1190TiP Pharmacokinetics, efficacy, and safety of patritumab deruxtecan (HER3-DXd) in EGFR inhibitor-resistant, EGFR-mutated (EGFRm) non-small cell lung cancer (NSCLC)
EP3493793A1 (fr) Combinaison d'inhibiteurs du protéasome et d'anticorps anti-cd30
Boni et al. CX-2009, a CD166-directed PROBODY drug conjugate (PDC): results from the first-in-human study in patients with advanced cancer including breast cancer
JP2024515816A (ja) Adamts結合免疫グロブリンの投与の方法

Legal Events

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

Ref document number: 19933094

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3142076

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021573710

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021024970

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217043352

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019450366

Country of ref document: AU

Date of ref document: 20190611

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112021024970

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20211209

122 Ep: pct application non-entry in european phase

Ref document number: 19933094

Country of ref document: EP

Kind code of ref document: A1