WO2022261440A1 - Administration of resiniferatoxin for treatment of pancreatic cancer - Google Patents

Administration of resiniferatoxin for treatment of pancreatic cancer Download PDF

Info

Publication number
WO2022261440A1
WO2022261440A1 PCT/US2022/033011 US2022033011W WO2022261440A1 WO 2022261440 A1 WO2022261440 A1 WO 2022261440A1 US 2022033011 W US2022033011 W US 2022033011W WO 2022261440 A1 WO2022261440 A1 WO 2022261440A1
Authority
WO
WIPO (PCT)
Prior art keywords
mcg
meg
rtx
composition
administered
Prior art date
Application number
PCT/US2022/033011
Other languages
French (fr)
Inventor
Andreas Herrmann
Alexis Nahama
Henry Hongjun Ji
Original Assignee
Sorrento Therapeutics, Inc.
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 Sorrento Therapeutics, Inc. filed Critical Sorrento Therapeutics, Inc.
Publication of WO2022261440A1 publication Critical patent/WO2022261440A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions

Definitions

  • the present disclosure provides methods of treating pancreatic cancer comprising administering resiniferatoxin (RTX), and resiniferatoxin for use in such methods.
  • RTX resiniferatoxin
  • TRPV-1 receptor is ubiquitously expressed throughout the human body (Velasco et al. Handb Exp Pharmacol. 2015;231:449-72). Comparing patient biopsy normal versus malignant tissue, shows that TRPV-1 expression is elevated in tumor tissue. Tumor- associated TRPV-1 overexpression can be seen in biopsy immunohistochemistry (Fig. 1A) and Western blot analyses (Fig. IB).
  • Resiniferatoxin herein referred to as RTX
  • RTX is a TRPV-1 receptor agonist currently used for pain management.
  • the cytotoxic activity of RTX has been documented in vitro for pancreatic, lung and prostate cancer cells (Ziglioli et al. Acta Biomed. 80 (2009) 13- 20; Hartel et al. Gut 55 (2006) 519-528; Hail et al. Apoptosis 8 (2003) 251-262; Athanasiou et al. Biochem. Biophys. Res. Commun. 354 (2007) 50-55).
  • Resiniferatoxin acts as an ultrapotent analog of capsaicin, the pungent principal ingredient of the red pepper.
  • RTX is a tricyclic diterpene isolated from certain species of Eurphorbia. A homovanillyl group is an important structural feature of capsaicin and is the most prominent feature distinguishing resiniferatoxin from typical phorbol-related compounds.
  • Native RTX has the following structure:
  • RTX and analog compounds such as tinyatoxin and other compounds (20- homovanillyl esters of diterpenes such as 12-deoxyphorbol 13-phenylacetate 20- homovanillate and mezerein 20-homovanillate) are described in U.S. Patent Nos. 4,939,194; 5,021,450; and 5,232,684.
  • Other resiniferatoxin-type phorboid vanilloids have also been identified (Szallasi et al. (1999) Brit. J. Pharmacol. 128:428-434).
  • RTX is known as a TRPV-1 agonist.
  • TRPV-1 the transient receptor potential cation channel subfamily V member 1 (also known as Vanilloid receptor-1 (VR1)) is a multimeric cation channel prominently expressed in nociceptive primary afferent neurons (Caterina et al. (1997) Nature 389:816-824; Tominaga et al. (1998) Neuron 21:531-543).
  • Activation of TRPV-1 typically occurs at the nerve endings via application of painful heat and is up regulated during certain types of inflammatory stimuli.
  • Activation of TRPV-1 in peripheral tissues by a chemical agonist results in the opening of calcium channels and the transduction of a pain sensation (Szalllasi et al.
  • TRPV-1 agonists to the cell body of a neuron (ganglion) expressing TRPV-1 opens calcium channels and triggers a cascade of events leading to programmed cell death (“apoptosis”) (Karai et al. (2004) J. of Clin. Invest. 113:1344-1352).
  • pancreatic cancer accounts for about 3% of all cancers in the US and about 7% of all cancer deaths and is slightly more common in men than in women. Based on pancreatic cancer diagnoses between 2010 and 2016, the 5-year relative survival rate after diagnosis (all stage diagnosis combined) is -10%. (See www.cancer.org/cancer/pancreatic-cancer/about/key- statistics.html.). To date, chemotherapy has had limited efficacy, and improvement in pancreatic cancer patient survival has been unsatisfactory.
  • Embodiment 1 is a method of treating pancreatic cancer, comprising administering resiniferatoxin (RTX) to a subject in need of treatment of pancreatic cancer.
  • RTX resiniferatoxin
  • Embodiment 2 is a composition comprising resiniferatoxin (RTX) for use in a method of treating pancreatic cancer, the method comprising administering RTX to a subject in need of treatment of pancreatic cancer.
  • RTX resiniferatoxin
  • Embodiment 3 is the method or composition for use according to any one of the preceding embodiments, wherein the RTX is administered locally.
  • Embodiment 4 is the method or composition for use according to any one of the preceding embodiments, wherein the RTX is administered peritumorally.
  • Embodiment 5 is the method or composition for use according to any one of the preceding embodiments, wherein the pancreatic cancer is primary pancreatic cancer.
  • Embodiment 6 is the method or composition for use according to any one of the preceding embodiments, wherein the method comprises administering RTX at a concentration of 0.005 mcg/ml - 0.01 mcg/ml, 0.01 mcg/ml - 0.05 mcg/ml, 0.05 mcg/ml - 0.1 mcg/ml, 0.1 mcg/ml - 0.15 mcg/ml, 0.15 mcg/ml - 0.2 mcg/ml, 0.2 mcg/ml - 0.25 mcg/ml, 0.25 mcg/ml - 0.3 mcg/ml, 0.30 mcg/ml - 0.35 mcg/ml, 0.35 mcg/ml - 0.4 mcg/ml, 0.4 mcg/ml, 0.4 mcg/ml - 0.45 mc
  • Embodiment 7 is the method or composition for use according to any one of the preceding embodiments, wherein a dose of 0.05 meg to 0.10 meg, or 0.10 meg to 0.15 meg, or 0.15 meg to 0.25 meg, or 0.25 meg to 0.50 meg, or 0.50 meg to 0.75 meg, or 0.75 meg to 1.0 meg, or 1.0 meg to 1.1 meg, or 1.1 meg to 1.5 meg of RTX is administered.
  • Embodiment 8 is the method or composition for use according to embodiment 7, wherein the RTX is administered at a dose of at least about 0.1 meg.
  • Embodiment 9 is the method or composition for use according to embodiment 7, wherein the RTX is administered at a dose of at least about 0.5 meg.
  • Embodiment 10 is the method or composition for use according to embodiment 7, wherein the RTX is administered at a dose of at least about 1.0 meg.
  • Embodiment 11 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered in one dose.
  • Embodiment 12 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered in repeated doses.
  • Embodiment 13 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered daily.
  • Embodiment 14 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered every other day.
  • Embodiment 15 is the method or composition for use of any one of the preceding embodiments, wherein the subject is a mammal.
  • Embodiment 16 is the method or composition for use of embodiment 15, wherein the mammal is a human.
  • Embodiment 17 is the method or composition for use according to any one of the preceding embodiments, wherein the pancreatic cancer is pancreatic adenocarcinoma.
  • Embodiment 18 is the method or composition for use according to any one of the preceding embodiments, wherein the method comprises administering a pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier.
  • Embodiment 19 is the method or composition for use of embodiment 18, wherein the pharmaceutically acceptable carrier comprises water.
  • Embodiment 20 is the method or composition for use of embodiment 18 or 19, wherein the pharmaceutically acceptable carrier comprises polysorbate 80.
  • Embodiment 21 is the method or composition for use of any one of embodiments 18- 20, wherein the pharmaceutically acceptable carrier comprises a buffer, optionally wherein the buffer is phosphate buffer and/or the pH of the formulation is about 7.0-7.5 or about 7.2.
  • the pharmaceutically acceptable carrier comprises a buffer, optionally wherein the buffer is phosphate buffer and/or the pH of the formulation is about 7.0-7.5 or about 7.2.
  • Figure 1A shows biopsies of human pancreas adenocarcinoma immunostained to show TRPV-1 expression in tumor. Imaging was performed using a confocal laser scanning microscope LSM510Meta. Scale, 50 pm.
  • Figure IB shows western blot analyses illustrating TRPV-1 overexpression in carcinoma using commercially available tissue homogenates.
  • Figure 1C shows TRPV-1 expression by pancreatic carcinoma cell lines MiaPaCa-2 (left) and Panc-1 (right) (ATCC) assessed by flow cytometric analyses.
  • Figure ID shows the dose-dependent reduced proliferative activity of human pancreas carcinoma cell lines Panc-1 and MiaPaCa-2 upon RTX treatment assessed by commercially available Proliferation Assay (Promega). The bars through each data point represent the standard deviation in measurement.
  • FIG. 2 shows pancreas carcinoma tumor volume change over time in vivo in human pancreas adenocarcinoma cell line Pane- 1.
  • the bars through each data point represent the standard deviation in measurement.
  • Pantix cancer refers to any condition in which malignant cells are present in the pancreas.
  • Primary pancreatic cancer refers to pancreatic cancer in which the cancer cells arose from pancreatic tissue.
  • “Well-tolerated” as used herein refers to a response to administration of RTX in which the subject experiences little to no clinical adverse effects (e.g., only grade 2 or 1 adverse effects, or only grade 1 adverse effects).
  • Grade 1 means mild (e.g., minor event requiring no specific medical intervention; asymptomatic laboratory findings only; marginal clinical relevance);
  • grade 2 means moderate (e.g., event requiring only minimal intervention; local intervention; non-invasive intervention; transfusion; elective interventional radiological procedure; or therapeutic endoscopy or operation).
  • Adverse effects include unfavorable and unintended signs, symptoms, or diseases associated with the use of RTX. Examples of adverse effects include but are not limited to toxicity and distress indicated by, for example, high or low body temperature, hypotension, hypertension, hypocarbia, and ventricular arrhythmias, lethargy, responsiveness, loss of appetite, and/or weight loss.
  • compositions comprising RTX for use in a method of treating pancreatic cancer, the method comprising administering RTX to a subject in need of treatment of pancreatic cancer.
  • methods for treating pancreatic cancer comprising administering resiniferatoxin (RTX) to a subject in need of treatment of pancreatic cancer.
  • the methods are well-tolerated, e.g., having minimal side effects, or adverse events.
  • minimal side effects means no side effect or adverse events, or, if a side effect or adverse event is experienced, no medical intervention is required and/or the condition resolves on its own.
  • at least one side effect e.g., a side effect such as or related to toxicity
  • side effects include causing loss of activity or apoptosis in cells that do not overexpress TRPV-1 or causing loss of activity or apoptosis in non-cancerous cells.
  • toxic side effects include the subject experiencing one or more of the following: weight loss, distress (as evidenced, for example, by an irregular heart rate, irregular blood pressure, lack of responsiveness, loss of appetite, or other measure of emotional distress), or pain.
  • the standard of care for treating pancreatic cancer comprises administering 5-fluorouracil or gemcitabine.
  • the RTX is administered locally. In some embodiments the RTX is administered peritumorally. In some embodiments, the method comprises administering resiniferatoxin every other day. In some embodiments, the method comprises administering resiniferatoxin two times a week.
  • compositions and methods described herein are for use with any subject in whom RTX is effective, e.g., able to bind and activate TRPV-1 or a homolog thereof, and who is in need of treatment for pancreatic cancer.
  • the subject is a mammal.
  • the mammal is a human.
  • the mammal is a cat.
  • the mammal is a dog. 1. Dosage
  • the RTX is administered (e.g., peritumorally) at a dose of 0.05 meg to 0.10 meg, or 0.10 meg to 0.15 meg, or 0.15 meg to 0.25 meg, or 0.25 meg to 0.50 meg, or 0.50 meg to 0.75 meg, or 0.75 meg to 1.0 meg, or 1.0 meg to 1.1 meg, or 1.1 meg to 1.5 meg.
  • the RTX is administered at a dose of about 0.1 meg, or about 0.5 meg, or about 1.0 meg.
  • the RTX is delivered in a composition having a volume of 0.2 ml - 0.5 ml, 0.5 ml - 1.0 ml, 1 ml - 10 ml, 20 ml - 30 ml, 30 ml - 40 ml, 40 ml - 50 ml, 50 ml - 60 ml, 60 ml - 70 ml, 70 ml - 80 ml, 80 ml - 90 ml, or 90 ml - 100 ml.
  • the RTX is administered (e.g., systemically) at a dose of at least about 0.1 meg/kg, such as 0.1 meg/kg - 0.2 meg/kg, 0.2 meg/kg - 0.3 meg/kg, 0.3 meg/kg - 0.4 meg/kg, 0.4 meg/kg - 0.5 meg/kg, 0.5 meg/kg - 0.6 meg/kg, 0.6 meg/kg - 0.7 meg/kg, 0.7 meg/kg - 0.8 meg/kg, 0.8 meg/kg - 0.9 meg/kg, 0.9 meg/kg - 1 meg/kg, 1 meg/kg - 1.2 meg/kg, 1.2 meg/kg - 1.4 meg/kg, 1.4 meg/kg - 1.6 meg/kg, 1.6 meg/kg - 1.8 meg/kg, 1.8 meg/kg - 2.0 meg/kg, 2.0 meg/kg - -
  • the RTX is administered in one dose. In some embodiments, the RTX is administered in repeated doses. In some embodiments, the RTX is administered in 1, 2, 3, 4, or 5 doses.
  • the RTX is administered daily. In some embodiments, the RTX is administered every other day. In some embodiments, the RTX is administered weekly.
  • RTX is prepared for administration by dilution in saline.
  • the RTX which may be at the dosages discussed above, is administered with a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier comprises water.
  • the pharmaceutically acceptable carrier comprises saline.
  • the pharmaceutically acceptable carrier comprises polysorbate 80.
  • the pharmaceutically acceptable carrier comprises polyethylene glycol. In some embodiments, the pharmaceutically acceptable carrier comprises sugar or sugar alcohol. In some embodiments, the pharmaceutically acceptable carrier comprises mannitol. In some embodiments, the pharmaceutically acceptable carrier comprises dextrose. In some embodiments, the pharmaceutically acceptable carrier comprises a pharmaceutically acceptable buffer. In some embodiments, the pharmaceutically acceptable carrier comprises a phosphate buffer. In some embodiments, the pharmaceutically acceptable carrier comprises a pharmaceutically acceptable salt. In some embodiments, the pharmaceutically acceptable carrier comprises NaCl. In some embodiments, the pharmaceutically acceptable carrier comprises an organic solvent such as ethanol or DMSO, e.g., as a minority or residual component used as an aid in dissolving RTX before dilution in a primarily aqueous composition.
  • an organic solvent such as ethanol or DMSO, e.g., as a minority or residual component used as an aid in dissolving RTX before dilution in a primarily aqueous composition.
  • the concentration of RTX in the formulation may be any suitable value for delivery of the intended dose. In some embodiments, the concentration of RTX in the formulation may be any suitable value for storage and may be diluted to obtain a concentration that is suitable for delivery of the intended dose.
  • the concentration of RTX in the pharmaceutical formulation is in the range of 0.1 to 300 mcg/ml.
  • the concentration of RTX in the pharmaceutical formulation is in the range of 0.1-1 mcg/ml, 1-5 mcg/ml, 5-10 mcg/ml, 10-20 mcg/ml, 10-30 mcg/ml, 20-30 mcg/ml, 20-50 mcg/ml, 50-100 mcg/ml, 100-150 mcg/ml, 150-200 mcg/ml, 200-250 mcg/ml, or 250-300 mcg/ml.
  • the concentration of RTX in the pharmaceutical formulation is 0.005 mcg/ml - 0.01 mcg/ml, 0.01 mcg/ml - 0.05 mcg/ml, 0.05 mcg/ml - 0.1 mcg/ml, 0.1 mcg/ml - 0.15 mcg/ml, 0.15 mcg/ml - 0.2 mcg/ml, 0.2 mcg/ml - 0.25 mcg/ml, 0.25 mcg/ml - 0.3 mcg/ml, 0.30 mcg/ml - 0.35 mcg/ml, 0.35 mcg/ml - 0.4 mcg/ml, 0.4 mcg/ml, 0.4 mcg/ml - 0.45 mcg/ml, 0.45 mcg/ml - 0.5 mcg/ml,
  • the formulation may have any pH suitable for administration.
  • the pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier has a pH in the range of 6 to 7.6.
  • the pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier has a pH in the range of 6 to 6.4, 6.3 to 6.7, 6.4 to 6.8, 6.8 to 7.2, 7 to 7.4, or 7.2 to 7.6.
  • the pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier has a pH of 6.5 or 7.2.
  • the formulation comprises polysorbate 80.
  • the concentration of polysorbate 80 is 0.03-7% w/v.
  • the concentration of polysorbate 80 is 2-4% w/v.
  • the concentration of polysorbate 80 is 3% w/v.
  • the formulation may further comprise a buffer, such as phosphate buffer (e.g., sodium phosphate buffer).
  • phosphate buffer e.g., sodium phosphate buffer
  • the concentration of phosphate buffer is 10-50 mM.
  • the concentration of phosphate buffer is 10-30 mM.
  • the concentration of phosphate buffer is lOmM.
  • the concentration of phosphate buffer is 30 mM.
  • the formulation may have a pH in the range of 7-7.5, such as about 7.2.
  • the concentration of RTX may be 10-30 mcg/ml, such as 10 mcg/ml or 25 mcg/ml.
  • the formulation further comprises phosphate buffer, e.g., at a concentration and pH shown for phosphate buffer in Table 1.
  • the formulation further comprises NaCl, e.g., at a concentration shown for NaCl in Table 1.
  • the phosphate buffer and NaCl may be (but are not necessarily) present at a combination of concentrations and phosphate buffer pH shown for an individual formulation.
  • formulations in Table 1 include dextrose.
  • the concentration of dextrose is 0.05-5% w/v.
  • the concentration of dextrose is 0.8-5% w/v.
  • the concentration of dextrose is 0.05% w/v.
  • the concentration of dextrose is 0.8% w/v.
  • the concentration of dextrose is 3.0% w/v.
  • the concentration of dextrose is 5.0% w/v.
  • formulations in Table 1 include mannitol.
  • the concentration of mannitol is 0.8-3.0% w/v. In some embodiments, the concentration of mannitol is 0.8% w/v. In some embodiments, the concentration of mannitol is 3.0% w/v.
  • the dextrose or mannitol is omitted from a formulation shown in Table 1.
  • the concentration of RTX in a formulation shown in Table 1 is adjusted to any of the RTX concentrations or concentration ranges disclosed herein.
  • the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.3-200 mcg/ml.
  • the concentration of RTX in a formulation shown in Table 1 is 200 mcg/ml.
  • the concentration of RTX in a formulation shown in Table 1 is 0.3-100 mcg/ml.
  • the concentration of RTX in a formulation shown in Table 1 is 100 mcg/ml.
  • the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.3- 50 mcg/ml. In some embodiments, the concentration of RTX in a formulation shown in Table 1 is 25 mcg/ml. As another example, in some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.3-15 mcg/ml. As another example, in some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.5- 10 mcg/ml. As another example, in some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.6- 1.5 mcg/ml. The dextrose or mannitol is omitted from any such formulation having an adjusted RTX concentration.
  • formulations in Table 1 may be prepared according to the following exemplary methods, which are provided for formulations 3 and 5 but may be adapted to the other formulations by one skilled in the art.
  • Formulation 3 may be made by adding 46 mg sodium phosphate monobasic monohydrate, 94.7 mg sodium phosphate dibasic anhydrous, and 860 mg NaCl to a 100 ml volumetric flask. 50 ml of water for injection (WFI) is added to dissolve the components in the flask, followed by addition of 1.0 g of polysorbate 80, to form the aqueous component. 20 mg of RTX is added to the aqueous component in the volumetric flask, and pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2.
  • WFI water for injection
  • RTX will sometimes precipitate at the interface of aqueous solution and PEG initially, but will go back into solution upon sonication.
  • the full mixture in the flask is diluted to volume (100.00 ml) with water (WFI) and this is mixed by an inversion process.
  • WFI water
  • the full formulation is filtered through a 0.2 pm polytetrafluoroethylene (PTFE) filter.
  • Formulation 5 may be made by adding 138 mg sodium phosphate monobasic monohydrate, 284.1 mg sodium phosphate dibasic anhydrous, and 540 mg NaCl to a 100 ml volumetric flask. 50 ml of water for injection (WFI) is added to dissolve the components in the flask, followed by addition of 3.0 g of polysorbate 80, and 800 mg of dextrose to form the aqueous component. 20 mg of RTX is added to the aqueous component in the volumetric flask, and pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2. The solution is then sonicated to dissolve all the solids.
  • WFI water for injection
  • the RTX may be initially dissolved in a small volume of ethanol or DMSO, and this solution may then be added to the aqueous component.
  • the full mixture in the flask is diluted to volume (100.00 ml) with water (WFI) and this is mixed by an inversion process.
  • the full formulation is filtered through a 0.2 pm PTFE filter.
  • a formulation according to Formulation 11 is prepared using 200 meg RTX, 300 meg Polysorbate 80 (using commercially-available polysorbate 80); 5.4 mg of sodium chloride, 500 meg of dextrose, 1.38 mg sodium phosphate monobasic monohydrate, 2.84 mg sodium phosphate dibasic anhydrous, and water (WFI) to 1 mL, then pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2. As noted above, the dextrose may be omitted.
  • a formulation according to Formulation 13 is prepared using 25 meg RTX, 30 mg Polysorbate 80 (using commercially-available polysorbate 80); 5.4 mg of sodium chloride, 50 mg of dextrose, 1.38 mg sodium phosphate monobasic monohydrate, 2.84 mg sodium phosphate dibasic anhydrous, water (WFI) to 1 mL, then pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2. As noted above, the dextrose may be omitted.
  • RTX Resiniferatoxin
  • HBSS Hank’s Balanced Salt solution
  • SD Standard derivation
  • test compounds placed in vivo to assess the antitumoral activity of RTX administered locally at three different concentrations. Placebo formulation was administered to exclude undesired/unanticipated anti-tumor activity of placebo formulation.
  • RTX Anti-tumor activity of RTX was tested in vivo by local/peritumoral administration.
  • Human pancreas carcinoma cells Panc-1 (ATCC) were engrafted subcutaneously (s.c.) in the ventral flank of immune compromised NOD/SCID male mice.
  • the single-cell-suspension was mixed/resuspended in phenol-red free RPMI1640 media (GibCO) and growth factor containing Matrigel (Corning) at a 1:1 ratio.
  • mice underwent treatment with either placebo formulation (volume comparable to 1.0 pg/dose RTX administration dose preparation), or 0.1, 0.5, or 1.0 pg/dose of RTX diluted in HBSS every other day.
  • the initial test system showed that RTX treatment in vitro reduced proliferative activity of TRPV-1+ pancreas carcinoma.
  • RTX reduced and cytostatically suppressed human pancreas carcinoma tumor growth.
  • the antitumor activity was immediately effective upon initial administration.
  • Reduced pancreas carcinoma tumor growth was shown to be dose dependent with 0.1, 0.5 and 1.0 pg/dose showing antitumor efficacy.
  • the Placebo formulation similarly administered did not show any efficacy on tumor growth or on disease progression. No pain or distress, including weight loss, was observed in the tumor bearing mice.
  • Biopsies of human pancreas adenocarcinoma (Tissue Array obtained from BioMax, cat# HPan-Ade060CS-01) were immunostained (aTRPV-1, Thermo Fisher Scientific, cat# PA1-748). Elevated TRPV-1 expression was seen in tumor cells (Figure 1A) by confocal laser scanning microscope LSM510Meta.
  • Figure ID shows the dose-dependent reduction of proliferative activity of human pancreas carcinoma cell lines Panc-1 and MiaPaCa-2 upon RTX treatment assessed by commercially available Proliferation Assay (Promega). In this assay 10,000 cells/well were plated and treated for 2 consecutive days prior to assessing proliferative activity.
  • RTX administration dramatically reduced pancreas carcinoma growth in vivo, as measured by tumor volume shown in Figure 2.
  • RTX administration did not result in indications of toxicity nor were there signs of weight loss, distress or pain detectable in treated subjects.

Abstract

Disclosed herein are methods of administering resiniferatoxin (RTX) for treatment of pancreatic cancer.

Description

ADMINISTRATION OF RESINIFERATOXIN FOR TREATMENT OF
PANCREATIC CANCER
[0011 This application claims priority to United States Provisional Application No. 63/209,786, filed June 11, 2021, the disclosure of which is hereby incorporated by reference in its entirety.
[002J The present disclosure provides methods of treating pancreatic cancer comprising administering resiniferatoxin (RTX), and resiniferatoxin for use in such methods.
INTRODUCTION AND SUMMARY
[003] The TRPV-1 receptor is ubiquitously expressed throughout the human body (Velasco et al. Handb Exp Pharmacol. 2015;231:449-72). Comparing patient biopsy normal versus malignant tissue, shows that TRPV-1 expression is elevated in tumor tissue. Tumor- associated TRPV-1 overexpression can be seen in biopsy immunohistochemistry (Fig. 1A) and Western blot analyses (Fig. IB).
[004] Resiniferatoxin, herein referred to as RTX, is a TRPV-1 receptor agonist currently used for pain management. In addition, the cytotoxic activity of RTX has been documented in vitro for pancreatic, lung and prostate cancer cells (Ziglioli et al. Acta Biomed. 80 (2009) 13- 20; Hartel et al. Gut 55 (2006) 519-528; Hail et al. Apoptosis 8 (2003) 251-262; Athanasiou et al. Biochem. Biophys. Res. Commun. 354 (2007) 50-55). Recent studies using RTX illustrated anti-tumor activity of TRPV-1 agonists including RTX and RTX-derivates on human bladder cancer in a rodent xenograft tumor model (Rossi et al. Int J Mol Sci. 2019 Apr 18;20(8)).
[005] Resiniferatoxin (RTX) acts as an ultrapotent analog of capsaicin, the pungent principal ingredient of the red pepper. RTX is a tricyclic diterpene isolated from certain species of Eurphorbia. A homovanillyl group is an important structural feature of capsaicin and is the most prominent feature distinguishing resiniferatoxin from typical phorbol-related compounds. Native RTX has the following structure:
Figure imgf000003_0001
[006] RTX and analog compounds such as tinyatoxin and other compounds (20- homovanillyl esters of diterpenes such as 12-deoxyphorbol 13-phenylacetate 20- homovanillate and mezerein 20-homovanillate) are described in U.S. Patent Nos. 4,939,194; 5,021,450; and 5,232,684. Other resiniferatoxin-type phorboid vanilloids have also been identified (Szallasi et al. (1999) Brit. J. Pharmacol. 128:428-434).
[007] RTX is known as a TRPV-1 agonist. TRPV-1, the transient receptor potential cation channel subfamily V member 1 (also known as Vanilloid receptor-1 (VR1)) is a multimeric cation channel prominently expressed in nociceptive primary afferent neurons (Caterina et al. (1997) Nature 389:816-824; Tominaga et al. (1998) Neuron 21:531-543). Activation of TRPV-1 typically occurs at the nerve endings via application of painful heat and is up regulated during certain types of inflammatory stimuli. Activation of TRPV-1 in peripheral tissues by a chemical agonist results in the opening of calcium channels and the transduction of a pain sensation (Szalllasi et al. (1999) Mol. Pharmacol. 56:581-587). However, direct application of certain TRPV-1 agonists to the cell body of a neuron (ganglion) expressing TRPV-1 opens calcium channels and triggers a cascade of events leading to programmed cell death (“apoptosis”) (Karai et al. (2004) J. of Clin. Invest. 113:1344-1352).
[008] The American Cancer Society estimates that about 60,000 people will be diagnosed with and about 48,000 people will die of pancreatic cancer in the United States in 2021. Pancreatic cancer accounts for about 3% of all cancers in the US and about 7% of all cancer deaths and is slightly more common in men than in women. Based on pancreatic cancer diagnoses between 2010 and 2016, the 5-year relative survival rate after diagnosis (all stage diagnosis combined) is -10%. (See www.cancer.org/cancer/pancreatic-cancer/about/key- statistics.html.). To date, chemotherapy has had limited efficacy, and improvement in pancreatic cancer patient survival has been unsatisfactory. Though agents such as 5- fluorouracil (5-FU) and gemcitabine showed promising cytotoxic effects in vitro, that promise did not translate into effective treatment. The toxicity and inherent resistance to these agents in patients with pancreatic cancer does not allow for satisfactory survival rates. [009J Accordingly, there is a need for improved compositions that are safe and well tolerated, and methods and uses that are safe and well tolerated, for effective treatment of pancreatic cancer. The present disclosure aims to meet this need and/or provide other benefits and shows that RTX can be effective against pancreatic cancer in vivo.
[0010] Accordingly, the following exemplary embodiments are provided.
[0011] Embodiment 1 is a method of treating pancreatic cancer, comprising administering resiniferatoxin (RTX) to a subject in need of treatment of pancreatic cancer.
[0012] Embodiment 2 is a composition comprising resiniferatoxin (RTX) for use in a method of treating pancreatic cancer, the method comprising administering RTX to a subject in need of treatment of pancreatic cancer.
[0013] Embodiment 3 is the method or composition for use according to any one of the preceding embodiments, wherein the RTX is administered locally.
[0014] Embodiment 4 is the method or composition for use according to any one of the preceding embodiments, wherein the RTX is administered peritumorally.
[0015] Embodiment 5 is the method or composition for use according to any one of the preceding embodiments, wherein the pancreatic cancer is primary pancreatic cancer.
[0016] Embodiment 6 is the method or composition for use according to any one of the preceding embodiments, wherein the method comprises administering RTX at a concentration of 0.005 mcg/ml - 0.01 mcg/ml, 0.01 mcg/ml - 0.05 mcg/ml, 0.05 mcg/ml - 0.1 mcg/ml, 0.1 mcg/ml - 0.15 mcg/ml, 0.15 mcg/ml - 0.2 mcg/ml, 0.2 mcg/ml - 0.25 mcg/ml, 0.25 mcg/ml - 0.3 mcg/ml, 0.30 mcg/ml - 0.35 mcg/ml, 0.35 mcg/ml - 0.4 mcg/ml, 0.4 mcg/ml - 0.45 mcg/ml, 0.45 mcg/ml - 0.5 mcg/ml, 0.5 mcg/ml - 0.55 mcg/ml, 0.55 mcg/ml - 0.6 mcg/ml, 0.6 mcg/ml - 0.65 mcg/ml, 0.65 mcg/ml - 0.7 mcg/ml, 0.7 mcg/ml - 0.75 mcg/ml, 0.75 mcg/ml - 0.8 mcg/ml, 0.8 mcg/ml - 0.85 mcg/ml, 0.85 mcg/ml - 0.9 mcg/ml,
0.9 mcg/ml - 0.95 mcg/ml, 0.95 mcg/ml - 1.0 mcg/ml, 1.0 mcg/ml - 1.1 mcg/ml, or 1.1 mcg/ml - 1.2 mcg/ml.
[0017] Embodiment 7 is the method or composition for use according to any one of the preceding embodiments, wherein a dose of 0.05 meg to 0.10 meg, or 0.10 meg to 0.15 meg, or 0.15 meg to 0.25 meg, or 0.25 meg to 0.50 meg, or 0.50 meg to 0.75 meg, or 0.75 meg to 1.0 meg, or 1.0 meg to 1.1 meg, or 1.1 meg to 1.5 meg of RTX is administered.
[0018] Embodiment 8 is the method or composition for use according to embodiment 7, wherein the RTX is administered at a dose of at least about 0.1 meg.
[0019] Embodiment 9 is the method or composition for use according to embodiment 7, wherein the RTX is administered at a dose of at least about 0.5 meg. [0020] Embodiment 10 is the method or composition for use according to embodiment 7, wherein the RTX is administered at a dose of at least about 1.0 meg.
[0021] Embodiment 11 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered in one dose.
[0022] Embodiment 12 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered in repeated doses.
[0023] Embodiment 13 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered daily.
[0024] Embodiment 14 is the method or composition for use of any one of the preceding embodiments, wherein the RTX is administered every other day.
[0025] Embodiment 15 is the method or composition for use of any one of the preceding embodiments, wherein the subject is a mammal.
[0026] Embodiment 16 is the method or composition for use of embodiment 15, wherein the mammal is a human.
[0027] Embodiment 17 is the method or composition for use according to any one of the preceding embodiments, wherein the pancreatic cancer is pancreatic adenocarcinoma.
[0028] Embodiment 18 is the method or composition for use according to any one of the preceding embodiments, wherein the method comprises administering a pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier.
[0029] Embodiment 19 is the method or composition for use of embodiment 18, wherein the pharmaceutically acceptable carrier comprises water.
[0030] Embodiment 20 is the method or composition for use of embodiment 18 or 19, wherein the pharmaceutically acceptable carrier comprises polysorbate 80.
[0031] Embodiment 21 is the method or composition for use of any one of embodiments 18- 20, wherein the pharmaceutically acceptable carrier comprises a buffer, optionally wherein the buffer is phosphate buffer and/or the pH of the formulation is about 7.0-7.5 or about 7.2. BRIEF DESCRIPTION OF THE DRAWINGS
[0032] Figure 1A shows biopsies of human pancreas adenocarcinoma immunostained to show TRPV-1 expression in tumor. Imaging was performed using a confocal laser scanning microscope LSM510Meta. Scale, 50 pm.
[0033] Figure IB shows western blot analyses illustrating TRPV-1 overexpression in carcinoma using commercially available tissue homogenates.
[0034] Figure 1C shows TRPV-1 expression by pancreatic carcinoma cell lines MiaPaCa-2 (left) and Panc-1 (right) (ATCC) assessed by flow cytometric analyses. [0035] Figure ID shows the dose-dependent reduced proliferative activity of human pancreas carcinoma cell lines Panc-1 and MiaPaCa-2 upon RTX treatment assessed by commercially available Proliferation Assay (Promega). The bars through each data point represent the standard deviation in measurement.
[0036] Figure 2 shows pancreas carcinoma tumor volume change over time in vivo in human pancreas adenocarcinoma cell line Pane- 1. The bars through each data point represent the standard deviation in measurement. T-test: *) P<0.05, **) P O.Ol, ***) P O.001.
DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
[0037] Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying drawings. While the invention will be described in conjunction with the illustrated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents, which may be included within the invention as defined by the appended claims.
[0038] Before describing the present teachings in detail, it is to be understood that the disclosure is not limited to specific compositions or process steps, as such may vary. It should be noted that, as used in this specification and the appended claims, the singular form “a”, “an” and “the” include plural references unless the context clearly dictates otherwise. Thus, for example, reference to “a conjugate” includes a plurality of conjugates and reference to “a cell” includes a plurality of cells and the like.
[0039] Numeric ranges are inclusive of the numbers defining the range. Measured and measurable values are understood to be approximate, taking into account significant digits and the error associated with the measurement. Also, the use of “comprise”, “comprises”, “comprising”, “contain”, “contains”, “containing”, “include”, “includes”, and “including” are not intended to be limiting. It is to be understood that both the foregoing general description and detailed description are exemplary and explanatory only and are not restrictive of the teachings.
[0040] Unless specifically noted in the above specification, embodiments in the specification that recite “comprising” various components are also contemplated as “consisting of’ or “consisting essentially of’ the recited components; embodiments in the specification that recite “consisting of’ various components are also contemplated as “comprising” or “consisting essentially of’ the recited components; and embodiments in the specification that recite “consisting essentially of’ various components are also contemplated as “consisting of’ or “comprising” the recited components (this interchangeability does not apply to the use of these terms in the claims).
[0041] The section headings used herein are for organizational purposes only and are not to be construed as limiting the desired subject matter in any way. In the event that any literature incorporated by reference contradicts any term defined in this specification, this specification controls. While the present teachings are described in conjunction with various embodiments, it is not intended that the present teachings be limited to such embodiments. On the contrary, the present teachings encompass various alternatives, modifications, and equivalents, as will be appreciated by those of skill in the art.
A. Definitions
[0042] “Pancreatic cancer” refers to any condition in which malignant cells are present in the pancreas. “Primary pancreatic cancer” refers to pancreatic cancer in which the cancer cells arose from pancreatic tissue.
[0043] The terms “or a combination thereof’ and “or combinations thereof’ as used herein refers to any and all permutations and combinations of the listed terms preceding the term. For example, “A, B, C, or combinations thereof’ is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, ACB, CBA, BCA, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AAB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.
[0044] “Or” is used in the inclusive sense, i.e., equivalent to “and/or,” unless the context requires otherwise.
[0045] “Well-tolerated” as used herein refers to a response to administration of RTX in which the subject experiences little to no clinical adverse effects (e.g., only grade 2 or 1 adverse effects, or only grade 1 adverse effects). Grade 1 means mild (e.g., minor event requiring no specific medical intervention; asymptomatic laboratory findings only; marginal clinical relevance); grade 2 means moderate (e.g., event requiring only minimal intervention; local intervention; non-invasive intervention; transfusion; elective interventional radiological procedure; or therapeutic endoscopy or operation). Adverse effects include unfavorable and unintended signs, symptoms, or diseases associated with the use of RTX. Examples of adverse effects include but are not limited to toxicity and distress indicated by, for example, high or low body temperature, hypotension, hypertension, hypocarbia, and ventricular arrhythmias, lethargy, responsiveness, loss of appetite, and/or weight loss.
B. Exemplary methods and compositions for use
[0046] Provided herein are methods for treating pancreatic cancer, comprising administering resiniferatoxin (RTX) to a subject in need of treatment of pancreatic cancer. Also provided are compositions comprising RTX for use in a method of treating pancreatic cancer, the method comprising administering RTX to a subject in need of treatment of pancreatic cancer. Provided herein are methods for treating pancreatic cancer, comprising administering resiniferatoxin (RTX) to a subject in need of treatment of pancreatic cancer. In some embodiments, the methods are well-tolerated, e.g., having minimal side effects, or adverse events. In some embodiments, minimal side effects means no side effect or adverse events, or, if a side effect or adverse event is experienced, no medical intervention is required and/or the condition resolves on its own. In some embodiments, at least one side effect (e.g., a side effect such as or related to toxicity) is reduced compared to a side effect seen using the standard of care (e.g., 5-fluorouracil or gemcitabine) for treating pancreatic cancer. In some embodiments, side effects include causing loss of activity or apoptosis in cells that do not overexpress TRPV-1 or causing loss of activity or apoptosis in non-cancerous cells. In some embodiments, toxic side effects include the subject experiencing one or more of the following: weight loss, distress (as evidenced, for example, by an irregular heart rate, irregular blood pressure, lack of responsiveness, loss of appetite, or other measure of emotional distress), or pain. In some embodiments, the standard of care for treating pancreatic cancer comprises administering 5-fluorouracil or gemcitabine.
[0047] In some embodiments, the RTX is administered locally. In some embodiments the RTX is administered peritumorally. In some embodiments, the method comprises administering resiniferatoxin every other day. In some embodiments, the method comprises administering resiniferatoxin two times a week.
[0048] The compositions and methods described herein are for use with any subject in whom RTX is effective, e.g., able to bind and activate TRPV-1 or a homolog thereof, and who is in need of treatment for pancreatic cancer. In some embodiments, the subject is a mammal. In some embodiments, the mammal is a human. In some embodiments, the mammal is a cat. In some embodiments, the mammal is a dog. 1. Dosage
[0049J In some embodiments, the RTX is administered (e.g., peritumorally) at a dose of 0.05 meg to 0.10 meg, or 0.10 meg to 0.15 meg, or 0.15 meg to 0.25 meg, or 0.25 meg to 0.50 meg, or 0.50 meg to 0.75 meg, or 0.75 meg to 1.0 meg, or 1.0 meg to 1.1 meg, or 1.1 meg to 1.5 meg.
[0050] In some embodiments, the RTX is administered at a dose of about 0.1 meg, or about 0.5 meg, or about 1.0 meg.
[0051] In some embodiments, the RTX is delivered in a composition having a volume of 0.2 ml - 0.5 ml, 0.5 ml - 1.0 ml, 1 ml - 10 ml, 20 ml - 30 ml, 30 ml - 40 ml, 40 ml - 50 ml, 50 ml - 60 ml, 60 ml - 70 ml, 70 ml - 80 ml, 80 ml - 90 ml, or 90 ml - 100 ml.
[0052] In some embodiments, the RTX is administered (e.g., systemically) at a dose of at least about 0.1 meg/kg, such as 0.1 meg/kg - 0.2 meg/kg, 0.2 meg/kg - 0.3 meg/kg, 0.3 meg/kg - 0.4 meg/kg, 0.4 meg/kg - 0.5 meg/kg, 0.5 meg/kg - 0.6 meg/kg, 0.6 meg/kg - 0.7 meg/kg, 0.7 meg/kg - 0.8 meg/kg, 0.8 meg/kg - 0.9 meg/kg, 0.9 meg/kg - 1 meg/kg, 1 meg/kg - 1.2 meg/kg, 1.2 meg/kg - 1.4 meg/kg, 1.4 meg/kg - 1.6 meg/kg, 1.6 meg/kg - 1.8 meg/kg, 1.8 meg/kg - 2.0 meg/kg, 2.0 meg/kg - 2.2 meg/kg, 2.2 meg/kg - 2.4 meg/kg, 2.4 meg/kg - 2.6 meg/kg, 2.6 meg/kg - 2.8 meg/kg, 2.8 meg/kg - 3.0 meg/kg, 3.0 meg/kg - 3.2 meg/kg, 3.2 meg/kg - 3.4 meg/kg, 3.4 meg/kg - 3.6 meg/kg, 3.6 meg/kg - 3.8 meg/kg, 4.0 meg/kg - 4.2 meg/kg, 4.2 meg/kg - 4.4 meg/kg, 4.4 meg/kg - 4.6 meg/kg, 4.6 meg/kg - 4.8 meg/kg, 4.8 meg/kg - 5.0 meg/kg, 5.0 meg/kg - 5.2 meg/kg, 5.2 meg/kg - 5.4 meg/kg, 5.4 meg/kg - 5.6 meg/kg, 5.6 meg/kg - 5.8 meg/kg, or 5.8 meg/kg - 6.0 meg/kg.
[0053] In some embodiments, the RTX is administered in one dose. In some embodiments, the RTX is administered in repeated doses. In some embodiments, the RTX is administered in 1, 2, 3, 4, or 5 doses.
[0054] In some embodiments, the RTX is administered daily. In some embodiments, the RTX is administered every other day. In some embodiments, the RTX is administered weekly.
2. Formulations
[0055] Multiple examples of formulations of RTX are available in the literature. See, e.g., Ueda et al. (2008) J. of Cardiovasc. Pharmacol. 51:513-520, and US 2015/0190509 Al. Any suitable formulation of RTX for administration may be used. In some embodiments, RTX is prepared for administration by dilution in saline. [0056] In some embodiments, the RTX, which may be at the dosages discussed above, is administered with a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutically acceptable carrier comprises water. In some embodiments, the pharmaceutically acceptable carrier comprises saline. In some embodiments, the pharmaceutically acceptable carrier comprises polysorbate 80. In some embodiments, the pharmaceutically acceptable carrier comprises polyethylene glycol. In some embodiments, the pharmaceutically acceptable carrier comprises sugar or sugar alcohol. In some embodiments, the pharmaceutically acceptable carrier comprises mannitol. In some embodiments, the pharmaceutically acceptable carrier comprises dextrose. In some embodiments, the pharmaceutically acceptable carrier comprises a pharmaceutically acceptable buffer. In some embodiments, the pharmaceutically acceptable carrier comprises a phosphate buffer. In some embodiments, the pharmaceutically acceptable carrier comprises a pharmaceutically acceptable salt. In some embodiments, the pharmaceutically acceptable carrier comprises NaCl. In some embodiments, the pharmaceutically acceptable carrier comprises an organic solvent such as ethanol or DMSO, e.g., as a minority or residual component used as an aid in dissolving RTX before dilution in a primarily aqueous composition.
[0057] In some embodiments, the concentration of RTX in the formulation may be any suitable value for delivery of the intended dose. In some embodiments, the concentration of RTX in the formulation may be any suitable value for storage and may be diluted to obtain a concentration that is suitable for delivery of the intended dose.
[0058] In some embodiments, the concentration of RTX in the pharmaceutical formulation is in the range of 0.1 to 300 mcg/ml.
[0059] In some embodiments, the concentration of RTX in the pharmaceutical formulation is in the range of 0.1-1 mcg/ml, 1-5 mcg/ml, 5-10 mcg/ml, 10-20 mcg/ml, 10-30 mcg/ml, 20-30 mcg/ml, 20-50 mcg/ml, 50-100 mcg/ml, 100-150 mcg/ml, 150-200 mcg/ml, 200-250 mcg/ml, or 250-300 mcg/ml. In some embodiments, the concentration of RTX in the pharmaceutical formulation is 0.005 mcg/ml - 0.01 mcg/ml, 0.01 mcg/ml - 0.05 mcg/ml, 0.05 mcg/ml - 0.1 mcg/ml, 0.1 mcg/ml - 0.15 mcg/ml, 0.15 mcg/ml - 0.2 mcg/ml, 0.2 mcg/ml - 0.25 mcg/ml, 0.25 mcg/ml - 0.3 mcg/ml, 0.30 mcg/ml - 0.35 mcg/ml, 0.35 mcg/ml - 0.4 mcg/ml, 0.4 mcg/ml - 0.45 mcg/ml, 0.45 mcg/ml - 0.5 mcg/ml, 0.5 mcg/ml - 0.55 mcg/ml, 0.55 mcg/ml - 0.6 mcg/ml, 0.6 mcg/ml - 0.65 mcg/ml, 0.65 mcg/ml - 0.7 mcg/ml, 0.7 mcg/ml - 0.75 mcg/ml, 0.75 mcg/ml - 0.8 mcg/ml, 0.8 mcg/ml - 0.85 mcg/ml, 0.85 mcg/ml - 0.9 mcg/ml, 0.9 mcg/ml - 0.95 mcg/ml, 0.95 mcg/ml - 1.0 mcg/ml, 1.0 mcg/ml - 1.1 mcg/ml, or 1.1 mcg/ml - 1.2 mcg/ml.
[0060] The formulation may have any pH suitable for administration. In some embodiments, the pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier has a pH in the range of 6 to 7.6. In some embodiments, the pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier has a pH in the range of 6 to 6.4, 6.3 to 6.7, 6.4 to 6.8, 6.8 to 7.2, 7 to 7.4, or 7.2 to 7.6. In some embodiments, the pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier has a pH of 6.5 or 7.2.
[0061] In some embodiments, the formulation comprises polysorbate 80. In some embodiments, the concentration of polysorbate 80 is 0.03-7% w/v. In some embodiments, the concentration of polysorbate 80 is 2-4% w/v. In some embodiments, the concentration of polysorbate 80 is 3% w/v. The formulation may further comprise a buffer, such as phosphate buffer (e.g., sodium phosphate buffer). In some embodiments, the concentration of phosphate buffer is 10-50 mM. In some embodiments, the concentration of phosphate buffer is 10-30 mM. In some embodiments, the concentration of phosphate buffer is lOmM. In some embodiments, the concentration of phosphate buffer is 30 mM. The formulation may have a pH in the range of 7-7.5, such as about 7.2. In some embodiments, in any of the foregoing formulations, the concentration of RTX may be 10-30 mcg/ml, such as 10 mcg/ml or 25 mcg/ml. In some embodiments, the formulation further comprises phosphate buffer, e.g., at a concentration and pH shown for phosphate buffer in Table 1. In some embodiments, the formulation further comprises NaCl, e.g., at a concentration shown for NaCl in Table 1.
When both are present, the phosphate buffer and NaCl may be (but are not necessarily) present at a combination of concentrations and phosphate buffer pH shown for an individual formulation.
[0062] Exemplary formulations of RTX are shown in the following table.
[0063] Table 1. Exemplary RTX Solution Formulations
Figure imgf000011_0001
Figure imgf000012_0001
[0064] In some embodiments, formulations in Table 1 include dextrose. In embodiments, the concentration of dextrose is 0.05-5% w/v. In some embodiments, the concentration of dextrose is 0.8-5% w/v. In some embodiments, the concentration of dextrose is 0.05% w/v. In some embodiments, the concentration of dextrose is 0.8% w/v. In some embodiments, the concentration of dextrose is 3.0% w/v. In some embodiments, the concentration of dextrose is 5.0% w/v.
[0065] In some embodiments, formulations in Table 1 include mannitol. In some embodiments, the concentration of mannitol is 0.8-3.0% w/v. In some embodiments, the concentration of mannitol is 0.8% w/v. In some embodiments, the concentration of mannitol is 3.0% w/v.
[0066] In some embodiments, the dextrose or mannitol is omitted from a formulation shown in Table 1.
[0067] In some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to any of the RTX concentrations or concentration ranges disclosed herein. For example, in some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.3-200 mcg/ml. In some embodiments, the concentration of RTX in a formulation shown in Table 1 is 200 mcg/ml. In some embodiments, the concentration of RTX in a formulation shown in Table 1 is 0.3-100 mcg/ml. In some embodiments, the concentration of RTX in a formulation shown in Table 1 is 100 mcg/ml. In some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.3- 50 mcg/ml. In some embodiments, the concentration of RTX in a formulation shown in Table 1 is 25 mcg/ml. As another example, in some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.3-15 mcg/ml. As another example, in some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.5- 10 mcg/ml. As another example, in some embodiments, the concentration of RTX in a formulation shown in Table 1 is adjusted to 0.6- 1.5 mcg/ml. The dextrose or mannitol is omitted from any such formulation having an adjusted RTX concentration.
[0068] The formulations in Table 1 may be prepared according to the following exemplary methods, which are provided for formulations 3 and 5 but may be adapted to the other formulations by one skilled in the art. Formulation 3 may be made by adding 46 mg sodium phosphate monobasic monohydrate, 94.7 mg sodium phosphate dibasic anhydrous, and 860 mg NaCl to a 100 ml volumetric flask. 50 ml of water for injection (WFI) is added to dissolve the components in the flask, followed by addition of 1.0 g of polysorbate 80, to form the aqueous component. 20 mg of RTX is added to the aqueous component in the volumetric flask, and pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2. Then 30 mL of PEG 300 is added and the solution is sonicated to dissolve the solids. It should be noted that RTX will sometimes precipitate at the interface of aqueous solution and PEG initially, but will go back into solution upon sonication. The full mixture in the flask is diluted to volume (100.00 ml) with water (WFI) and this is mixed by an inversion process. The full formulation is filtered through a 0.2 pm polytetrafluoroethylene (PTFE) filter.
[0069] Formulation 5 may be made by adding 138 mg sodium phosphate monobasic monohydrate, 284.1 mg sodium phosphate dibasic anhydrous, and 540 mg NaCl to a 100 ml volumetric flask. 50 ml of water for injection (WFI) is added to dissolve the components in the flask, followed by addition of 3.0 g of polysorbate 80, and 800 mg of dextrose to form the aqueous component. 20 mg of RTX is added to the aqueous component in the volumetric flask, and pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2. The solution is then sonicated to dissolve all the solids. (Alternatively, the RTX may be initially dissolved in a small volume of ethanol or DMSO, and this solution may then be added to the aqueous component.) The full mixture in the flask is diluted to volume (100.00 ml) with water (WFI) and this is mixed by an inversion process. The full formulation is filtered through a 0.2 pm PTFE filter.
[0070] A formulation according to Formulation 11 is prepared using 200 meg RTX, 300 meg Polysorbate 80 (using commercially-available polysorbate 80); 5.4 mg of sodium chloride, 500 meg of dextrose, 1.38 mg sodium phosphate monobasic monohydrate, 2.84 mg sodium phosphate dibasic anhydrous, and water (WFI) to 1 mL, then pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2. As noted above, the dextrose may be omitted. [0071] A formulation according to Formulation 13 is prepared using 25 meg RTX, 30 mg Polysorbate 80 (using commercially-available polysorbate 80); 5.4 mg of sodium chloride, 50 mg of dextrose, 1.38 mg sodium phosphate monobasic monohydrate, 2.84 mg sodium phosphate dibasic anhydrous, water (WFI) to 1 mL, then pH is adjusted with hydrochloric acid/sodium hydroxide to 7.2. As noted above, the dextrose may be omitted.
[0072] Further details on techniques for formulation and administration may be found in Gennaro, A., Ed., Remington's Pharmaceutical Sciences, 18th Ed. (1990) (Mack Publishing Co., Easton, Pa.).
EXAMPLES
A. Anti-Pancreatic Carcinoma Efficacy of Resiniferatoxin
Materials and Methods [0073] Resiniferatoxin (RTX), from Eurofins AmatsiGroup, with > 95% purity, was diluted/dissolved in placebo formulation, shown in Table 2. RTX solutions with a concentraion of 25 mg/mL in Hank’s Balanced Salt solution (HBSS) were prepared for injection. The placebo formulation was prepared using HBSS for injection. All solutions were stored at 4°C.
[0074] Table 2: Placebo Formulation
Figure imgf000015_0001
x/y/z dimensions were protocolled in [mm]. Tumor volumes were then calculated following Vol.=xyz [mm3]. All data were fed to Prism Graph to generate a tumor growth kinetics graph. Standard derivation (SD) was inserted and significance was determined based on 2-tailed student’s T-test.
Study Designs
[0076] An initial test system was assessed in vitro using commercially available human pancreas carcinoma cell lines Panc-1 (and MiaPaCa-2 for in vitro assessment only) (ATCC). Cell proliferation assay (Promega) was used to assess cell division activity upon increasing treatment doses with RTX. Local administration of test compounds (placebo, RTX) was performed in vivo to assess the antitumoral activity of RTX administered locally at three different concentrations. Placebo formulation was administered to exclude undesired/unanticipated anti-tumor activity of placebo formulation.
[0077] Anti-tumor activity of RTX was tested in vivo by local/peritumoral administration. Human pancreas carcinoma cells Panc-1 (ATCC) were engrafted subcutaneously (s.c.) in the ventral flank of immune compromised NOD/SCID male mice. To secure proper engraftment, the single-cell-suspension was mixed/resuspended in phenol-red free RPMI1640 media (GibCO) and growth factor containing Matrigel (Corning) at a 1:1 ratio. Each mouse received 5 x 106 cells in a total volume of 100 pL. Tumor volumes (volume=length*width*height [mm3]) were assessed using calipers. Once tumors reached volumes > 200 mm3 mice underwent treatment with either placebo formulation (volume comparable to 1.0 pg/dose RTX administration dose preparation), or 0.1, 0.5, or 1.0 pg/dose of RTX diluted in HBSS every other day.
Results
[0078] The initial test system showed that RTX treatment in vitro reduced proliferative activity of TRPV-1+ pancreas carcinoma. In vivo , when administered locally, RTX reduced and cytostatically suppressed human pancreas carcinoma tumor growth. The antitumor activity was immediately effective upon initial administration. Reduced pancreas carcinoma tumor growth was shown to be dose dependent with 0.1, 0.5 and 1.0 pg/dose showing antitumor efficacy. The Placebo formulation similarly administered did not show any efficacy on tumor growth or on disease progression. No pain or distress, including weight loss, was observed in the tumor bearing mice.
[0079] Biopsies of human pancreas adenocarcinoma (Tissue Array obtained from BioMax, cat# HPan-Ade060CS-01) were immunostained (aTRPV-1, Thermo Fisher Scientific, cat# PA1-748). Elevated TRPV-1 expression was seen in tumor cells (Figure 1A) by confocal laser scanning microscope LSM510Meta.
[0080] Western blot analyses using commercially available tissue homogenates (Biotechne/Novus Biologicals NB820-59244, NB820-59443) also showed TRPV-1 overexpression in carcinoma. (Figure IB).
[0081] Flow cytometric analyses of TRPV-1 expression by pancreatic carcinoma cell lines Panc-1 and MiaPaCa-2 (ATCC) are shown in Figure 1C.
[0082] Figure ID shows the dose-dependent reduction of proliferative activity of human pancreas carcinoma cell lines Panc-1 and MiaPaCa-2 upon RTX treatment assessed by commercially available Proliferation Assay (Promega). In this assay 10,000 cells/well were plated and treated for 2 consecutive days prior to assessing proliferative activity.
[0083] RTX administration dramatically reduced pancreas carcinoma growth in vivo, as measured by tumor volume shown in Figure 2.
Summary
[0084] Human tumor tissue overexpresses RTX cognate receptor TRPV-1 representing a viable target for RTX treatment.
[0085] Locally constrained administration via peritumoral route s.c. given every other day is well tolerated by tumor bearing immune-compromised mice.
[0086] RTX administration did not result in indications of toxicity nor were there signs of weight loss, distress or pain detectable in treated subjects.

Claims

What is claimed is:
1. A method of treating pancreatic cancer, comprising administering resiniferatoxin (RTX) to a subject in need of treatment of pancreatic cancer.
2. A composition comprising resiniferatoxin (RTX) for use in a method of treating pancreatic cancer, the method comprising administering RTX to a subject in need of treatment of pancreatic cancer.
3. The method or composition for use according to any one of the preceding claims, wherein the RTX is administered locally.
4. The method or composition for use according to any one of the preceding claims, wherein the RTX is administered peritumorally.
5. The method or composition for use according to any one of the preceding claims, wherein the pancreatic cancer is primary pancreatic cancer.
6. The method or composition for use according to any one of the preceding claims, wherein the method comprises administering RTX at a concentration of 0.005 mcg/ml - 0.01 mcg/ml, 0.01 mcg/ml - 0.05 mcg/ml, 0.05 mcg/ml - 0.1 mcg/ml, 0.1 mcg/ml - 0.15 mcg/ml, 0.15 mcg/ml - 0.2 mcg/ml, 0.2 mcg/ml - 0.25 mcg/ml, 0.25 mcg/ml - 0.3 mcg/ml, 0.30 mcg/ml - 0.35 mcg/ml, 0.35 mcg/ml - 0.4 mcg/ml, 0.4 mcg/ml - 0.45 mcg/ml, 0.45 mcg/ml - 0.5 mcg/ml, 0.5 mcg/ml - 0.55 mcg/ml, 0.55 mcg/ml - 0.6 mcg/ml, 0.6 mcg/ml - 0.65 mcg/ml, 0.65 mcg/ml - 0.7 mcg/ml, 0.7 mcg/ml - 0.75 mcg/ml, 0.75 mcg/ml - 0.8 mcg/ml, 0.8 mcg/ml - 0.85 mcg/ml, 0.85 mcg/ml - 0.9 mcg/ml, 0.9 mcg/ml - 0.95 mcg/ml, 0.95 mcg/ml - 1.0 mcg/ml, 1.0 mcg/ml - 1.1 mcg/ml, or 1.1 mcg/ml - 1.2 mcg/ml.
7. The method or composition for use according to any one of the preceding claims, wherein a dose of 0.05 meg to 0.10 meg, or 0.10 meg to 0.15 meg, or 0.15 meg to 0.25 meg, or 0.25 meg to 0.50 meg, or 0.50 meg to 0.75 meg, or 0.75 meg to 1.0 meg, or 1.0 meg to 1.1 meg, or 1.1 meg to 1.5 meg of RTX is administered.
8. The method or composition for use according to claim 7, wherein the RTX is administered at a dose of about 0.1 meg.
9. The method or composition for use according to claim 7, wherein the RTX is administered at a dose of about 0.5 meg.
10. The method or composition for use according to claim 7, wherein the RTX is administered at a dose of about 1.0 meg.
11. The method or composition for use of any one of the preceding claims, wherein the RTX is administered in one dose.
12. The method or composition for use of any one of the preceding claims, wherein the RTX is administered in repeated doses.
13. The method or composition for use of any one of the preceding claims, wherein the RTX is administered daily.
14. The method or composition for use of any one of the preceding claims, wherein the RTX is administered every other day.
15. The method or composition for use of any one of the preceding claims, wherein the subject is a mammal.
16. The method or composition for use of claim 15, wherein the mammal is a human.
17. The method or composition for use according to any one of the preceding claims, wherein the pancreatic cancer is pancreatic adenocarcinoma.
18. The method or composition for use according to any one of the preceding claims, wherein the method comprises administering a pharmaceutical formulation comprising the RTX and a pharmaceutically acceptable carrier.
19. The method or composition for use of claim 18, wherein the pharmaceutically acceptable carrier comprises water.
20. The method or composition for use of claim 18 or 19, wherein the pharmaceutically acceptable carrier comprises polysorbate 80.
21. The method or composition for use of any one of claims 18-20, wherein the pharmaceutically acceptable carrier comprises a buffer, optionally wherein the buffer is phosphate buffer and/or the pH of the formulation is about 7.0-7.5 or about 7.2.
PCT/US2022/033011 2021-06-11 2022-06-10 Administration of resiniferatoxin for treatment of pancreatic cancer WO2022261440A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163209786P 2021-06-11 2021-06-11
US63/209,786 2021-06-11

Publications (1)

Publication Number Publication Date
WO2022261440A1 true WO2022261440A1 (en) 2022-12-15

Family

ID=84426359

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/033011 WO2022261440A1 (en) 2021-06-11 2022-06-10 Administration of resiniferatoxin for treatment of pancreatic cancer

Country Status (1)

Country Link
WO (1) WO2022261440A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080214574A1 (en) * 2004-07-19 2008-09-04 Novo Nordisk A/S Inhibition of the Activity of the Capsaicin Receptor in the Treatment of Obesity or Obesity-Related Diseases and Disorders
WO2009045504A1 (en) * 2007-10-04 2009-04-09 President And Fellows Of Harvard College Methods and compositions for treating cancer and modulating signal transduction and metabolism pathways
US10350239B2 (en) * 2009-10-22 2019-07-16 Propanc Pty Ltd Pharmaceutical composition for treating cancer comprising trypsinogen and/or chymotrypsinogen and an active agent selected from a selenium compound, a vanilloid compound and a cytoplasmic glycolysis reduction agent
US20200262877A1 (en) * 2017-10-09 2020-08-20 Blaze Bioscience, Inc. Ion channel-binding peptides and methods of use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080214574A1 (en) * 2004-07-19 2008-09-04 Novo Nordisk A/S Inhibition of the Activity of the Capsaicin Receptor in the Treatment of Obesity or Obesity-Related Diseases and Disorders
WO2009045504A1 (en) * 2007-10-04 2009-04-09 President And Fellows Of Harvard College Methods and compositions for treating cancer and modulating signal transduction and metabolism pathways
US10350239B2 (en) * 2009-10-22 2019-07-16 Propanc Pty Ltd Pharmaceutical composition for treating cancer comprising trypsinogen and/or chymotrypsinogen and an active agent selected from a selenium compound, a vanilloid compound and a cytoplasmic glycolysis reduction agent
US20200262877A1 (en) * 2017-10-09 2020-08-20 Blaze Bioscience, Inc. Ion channel-binding peptides and methods of use thereof

Similar Documents

Publication Publication Date Title
US20140037715A1 (en) Disulfiram formulation and uses thereof
US9549922B2 (en) Use of eribulin and lenvatinib as combination therapy for treatment of cancer
CN110339159A (en) The reduction of adipose tissue
US20130022609A1 (en) Method of treating androgen independent prostate cancer
EP3370706B1 (en) Disulfiram formulation
US20100216755A1 (en) Anti-tumor agent
EP2424516A2 (en) Pentamidine combinations for treating cancer
JP2017507151A (en) Use of eribulin and mTOR inhibitors as combination therapy for the treatment of cancer
WO2007092414A2 (en) Use of phosphatases to treat tumors overexpressing n-cor
US20180169055A1 (en) Composition containing carboplatin and use
CN109475554A (en) ARDS treatment based on Liponucleotide
UA126029C2 (en) Pharmaceutical combinations for treating cancer
JP2016528217A (en) Treatment of pancreatic cancer using a combination of hypoxia activated prodrug and taxane
Ahmed et al. Repurposing of metformin for cancer therapy: Updated patent and literature review
US9937261B2 (en) Combination therapy comprising a liposomal prodrug of mitomycin C and radiotherapy
EP4098251A1 (en) Use of mitoxantrone hydrochloride liposome for treating breast cancer
JP6462147B2 (en) HSP90 inhibitory peptide conjugate and its application in tumor therapy
WO2022261440A1 (en) Administration of resiniferatoxin for treatment of pancreatic cancer
US20230046869A1 (en) Therapeutic combinations of orally administered docetaxel and a p-gp inhibitor for the treatment of cancer
US7863255B2 (en) Methods of administering antitumor agent comprising deoxycytidine derivative
WO2022173916A1 (en) Administration of resiniferatoxin for treatment of prostate cancer
JP2014028841A (en) Intravesical apaziquone administration following transurethral resection for treating cancer
BR112019020519A2 (en) use of melatonin for the treatment of tumors
US20230255993A1 (en) Treatment of triple negative breast cancer
WO2023133508A1 (en) Methods for treating pancreatic cancer

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: 22821110

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE