WO2015051349A1 - Treating metastatic cancer with micellular paclitaxel - Google Patents

Treating metastatic cancer with micellular paclitaxel Download PDF

Info

Publication number
WO2015051349A1
WO2015051349A1 PCT/US2014/059207 US2014059207W WO2015051349A1 WO 2015051349 A1 WO2015051349 A1 WO 2015051349A1 US 2014059207 W US2014059207 W US 2014059207W WO 2015051349 A1 WO2015051349 A1 WO 2015051349A1
Authority
WO
WIPO (PCT)
Prior art keywords
stable
paclitaxel
dose
less stable
mice
Prior art date
Application number
PCT/US2014/059207
Other languages
French (fr)
Inventor
Korous MOTMED
Vuong Trieu
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 WO2015051349A1 publication Critical patent/WO2015051349A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • 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
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • A61K47/44Oils, fats or waxes according to two or more groups of A61K47/02-A61K47/42; Natural or modified natural oils, fats or waxes, e.g. castor oil, polyethoxylated castor oil, montan wax, lignite, shellac, rosin, beeswax or lanolin
    • 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

Definitions

  • the present disclosure provides a method for treating peritoneal metastatic cancers comprising administering a nanoparticulate formulation of paclitaxel intraperitoneally. More specifically, the present disclosure provides a method for treating metastatic cancers comprising intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising paclitaxel encapsulated in a micelle with a diblock copolymer.
  • Taxol is paclitaxel pharmaceutical formulations. Taxol is solvent based and should be carefully administered since it is an irritant. Side effects are common although the symptoms are either one or two in most cases. The most common side effects include hair loss, peripheral neuropathy, vomiting, diarrhea, myalagia, arthralagia, low blood counts and hypersensitivity. Taxol is a first generation paclitaxel formulation in which Cremophor EL (polyoxyethySated castor oil) is mixed with paclitaxel and given as an infusion for the treatment of ovarian cancer, lung cancer, head and neck cancer, bladder cancer. Taxol has adverse side effects such as anaphylactic shock. Abraxane is paciltaxel nanoparticle encapsulated by albumin.
  • Abraxane (nab-paclitaxel) is used in first and second line of treatment in metastatic breast cancer. Side effects of Abraxane include bone marrow suppression (primarily neutropenia) which is dose-dependent and a dose-limiting toxicity of Abraxane. In clinical studies, Grade 3-4 neutropenia occurred in 34% of patients with metastatic breast cancer (MBC) and 47% of patients with non-small cell lung cancer (NSCLC).
  • MBC metastatic breast cancer
  • NSCLC non-small cell lung cancer
  • IG-001 is a polymer bound nanoparticle paclitaxel.
  • Ovarian cancer is the fifth deadliest disease among American women, with more than 50% presenting with serous papillary histology.
  • High-grade serous epithelial ovarian cancer (SEOC) is associated with intraperitoneal spreading (carcinomatosis) and distant metastases.
  • SEOC serous epithelial ovarian cancer
  • Standard treatment is aggressive surgical resection followed by platinum-taxane
  • the present disclosure provides a method for treating peritoneal metastatic cancers comprising administering a nanoparticulate formulation of paclitaxel intraperitoneally. More specifically, the present disclosure provides a method for treating metastatic cancers comprising intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising paclitaxel encapsulated in a micelle with a diblock copolymer. The present disclosure provides a method for treating metastatic cancers comprising
  • the present disclosure provides a method for administering a nanoparticulate formulation of paclitaxel comprising infusing a nanoparticulate formulation intraperitoneally at a dose of from about 15 mg/kg/day to about 60 mg/kg/day. More preferably, the intraperitoneal dose of paclitaxel is from 25 mg/kg/day to 50 mg/kg/day. Most preferably, the intraperitoneal dose of paclitaxel is from 30 mg/kg/day to 50 mg/kg/day.
  • the nanoparticulate formulation of paclitaxel has a particle size average of 15 nm to 40 nm.
  • compositions comprising cancer drugs in a micelle where the composition is stable in protein-free medium and less stable in a protein containing medium.
  • the compositions of the present invention are conditionally stable in that they are more stable in one media than another, in particular the compositions are more stable in protein- free medium and less stable in a protein containing medium.
  • the present disclosure provides paclitaxel compositions where the paclitaxel is contained within a micelle and where the paclitaxel composition is stable in protein-free medium and less stable in a protein containing medium.
  • the present disclosure further provides micellular compositions where the composition comprises about 4 ⁇ g/ml to about 2000 ⁇ g/ml of paclitaxel.
  • the present disclosure provides micellular compositions where the composition is at least 20% more stable or 50% more stable or 100% more stable in a protein- free solution than in a solution containing protein.
  • the present disclosure provides micellular compositions where the composition is at least 20% more stable or 50% more stable or 100% more stable in a protein-free solution than nab-paclitaxel.
  • the present disclosure provides micellular compositions where the composition is at least 20% less stable or 50% less stable or 100% less stable in a protein containing solution such as serum than nab-paclitaxel.
  • the disclosure provides methods of treating patients with metastatic breast cancer by administering paclitaxel-containing micelles to patients.
  • the disclosure provides methods of treating patients with metastatic cancer by administering paclitaxel-containing micelles to patients where the micelles are comprised of a diblock copolymer including IG-001 which is a paclitaxel-containing micelle.
  • the present disclosure provides method of treating metastatic breast cancer wherein the amount of paclitaxel administered is at least 190mg/m 2 .
  • the present disclosure provides methods of treating metastatic breast cancer where the paclitaxel containing micelles are administered in at least 3 cycles.
  • the present disclosure provides method of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 3 cycles to 20 cycles.
  • the present disclosure provides methods of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 3 cycles to 15 cycles.
  • the present disclosure provides method of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 3 cycles to 10 cycles.
  • the present disclosure provides method of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 5 cycles to 10 cycles.
  • the present disclosure provides method of treating metastatic breast cancer where the overall response rate is greater than 20% or greater than 30% or greater than 40% or greater than 50% or from between about 20% to about 90% or from about 25% to about 75% or from about 30% to about 60% or from 30% to about 50% or from about 30% to about 40%.
  • the present disclosure provides method of treating metastatic breast cancer by administering paclitaxel containing micelles to the patient at least twice such that there is a first dose and a second dose.
  • the present disclosure provides method of treating metastatic breast cancer by administering paclitaxel containing micelles to the patient at least twice such that there is a first dose and a second dose where the first dose is about 300 mg/m 2 and the second dose is about 240 mg/m 2 .
  • the present disclosure provides method of treating patients with metastatic breast cancer comprising administering paclitaxel containing micelles to the patient comprising administering a dose of paclitaxel containing micelles containing about 300 mg/m 2 pacitaxel for at least one cycle; administering a dose of paclitaxel containing micelles containing about 260 mg/m 2 pacitaxel for at least one cycle; and administering a dose of paclitaxel containing micelles containing about 190 mg/m 2 pacitaxel for at least one cycle.
  • Figure 2 Plot of particle size versus paclitaxel concentration for IG-001 in phosphate buffered saline (PBS) and O.lx serum and lx serum.
  • PBS phosphate buffered saline
  • O.lx serum and lx serum O.lx serum
  • Figure 4 Plot of dose limiting toxicity curve for Taxol, nab-paclitaxel and IG-001.
  • Figure 7 Plot of overall response rate for IG-001, Taxol and nab-paclitaxel in Phase III study of metastatic breast cancer.
  • Figure 8 shows the effects of the treatments in Example 5 in terms of weight gains of the mice.
  • Figure 9 shows that treatment with either ip IG-001 or iv ABX (nab-paclitaxel) significantly increased the median survival of mice compared to untreated control
  • Figure 10 shows overall survival of control and experimental groups of mice in Example 5.
  • Figure 11 shows Kaplan Meier curves of the survival of IG-001 (Pax) treated group (left) and nab-paclitaxel (ABX) treated group (right).
  • Figure 12 shows (Left) Box plots of tumor burden arbitrary scores of mice treated with ip IG-001. And (Right) dot plots of the incidence and amount of ascitic fluid accumulating in tumor-bearing mice receiving the indicated doses of IG-001.
  • * Statistical analysis between the indicated groups was performed by Mann-Whitney non-parametric t- test. **Disease progression as a function of the indicated doses of IG-001 was analyzed by Kruskall Wallis test (non-parametric One Way ANOVA ).. The incidence of ascites development was analyzed by Chi square (X 2 ) test, # P ⁇ 0.0001. Ascites volume in treated mice compared to controls was analyzed by One way ANOVA and unpaired Student' s t-test.
  • Figure 13 shows (Left) Box plots of tumor burden arbitrary scores of mice treated with iv ABX. and (Right) Dot plots of the incidence and amount of ascitic fluid accumulating in tumor-bearing mice receiving the indicated doses of ABX. * Statistical analysis between the indicated groups was performed by Mann-Whitney non-parametric t-test. **Disease progression as a function of the indicated doses of ABX was analyzed by Kruskall Wallis test (non-parametric One Way ANOVA ). The incidence of ascites development was analyzed by Chi square (X 2 ) test, # P ⁇ 0.0001. Ascites volume in treated mice compared to controls was analyzed by One way ANOVA and unpaired Student's t-test.
  • the present disclosure provides a method for treating metastatic cancers comprising intraperitoneal or intravesicle administeration a nanoparticulate formulation of paclitaxel comprising paclitazel encapsulated in a micelle with a diblock copolymer. More specifically, the present disclosure provides a method for intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising infusing a nanoparticulate formulation intraperitoneally at a dose of from about 15 mg/kg/day to about 60 mg/kg/day. More preferably, the intraperitoneal dose of paclitaxel is from 25 mg/kg/day to 50 mg/kg/day.
  • the intraperitoneal dose of paclitaxel is from 30 mg/kg/day to 50 mg/kg/day.
  • the nanoparticulate formulation of paclitaxel has a particle size average of 15 nm to 40 nm.
  • the present disclosure provides paclitaxel-containing pharmaceutical compositions in a micelle, that is stable in protein-free medium and less stable in a protein containing medium.
  • the paclitaxel formulation includes amphiphilic block copolymer which may comprise a hydrophilic block (A) and a hydrophobic block (B) linked with each other in the form of A-B, A-B-A or B-A-B structure. Additionally, the amphiphilic block copolymer forma a core-shell type polymeric micelles in its aqueous solution state, wherein the hydrophobic block forms the core and the hydrophilic block forms the shell.
  • the hydrophilic block (A) of the amphiphilic block copolymer is polyethylene glycol (PEG) or monomethoxypolyethylene glycol (mPEG).
  • the hydrophilic block (A) may have a weight average molecular weight of 500-20,000 daltons, specifically 1,000-5,000 daltons, and more specifically 1,000-2,500 daltons.
  • the hydrophobic block (B) of the amphiphilic block copolymer is a water- insoluble, biodegradable polymer.
  • the hydrophobic block (B) is polylactic acid (PLA) or poly(lactic-co-glycolic acid) (PLGA).
  • the hydrophobic block (B) has a weight average molecular weight of 500-20,000 daltons, specifically 1,000-5,000 daltons, and more specifically 1,000-2,500 daltons. Hydroxyl end groups of the hydrophobic block (B) are protected with fatty acid groups, such as acetate, propionate, butyrate, stearate, palmitate groups, and the like.
  • the amphiphilic block copolymer comprising the hydrophilic block (A) and the hydrophobic block (B) are present in the pharmacrutical composition in an amount of 20-98 wt %, specifically 65-98 wt %, and more specifically 80-98 wt % based on the total dry weight of the composition.
  • the hydrophilic block (A) and the hydrophobic block (B) are present in the amphiphilic block copolymer in such a ratio that the copolymer comprises 40-70 wt %, specifically 50-60 wt % of the hydrophilic block (A) based on the weight of the copolymer.
  • the hydrophilic block (A) is present in a proportion less than 40%, the polymer has undesirably low solubility to water, resulting in difficulty in forming micelles.
  • the hydrophilic block (A) is present in a proportion greater than 70%, the polymer becomes too hydrophilic to form stable polymeric micelles, and thus the composition may not be used as a composition for solubilizing paclitaxel.
  • a preferred paclitaxel formulation is IG-001 (also referred to as Genexol-PM) which is a polymeric micelle formulation of paclitaxel.
  • IG-001 has a di-block copolymer composed of methoxy poly (ethylene glycol)-poly (lactide) to form nanoparticles with paclitaxel containing hydrophobic core and a hydrophilic shell.
  • the nanoparticles formed are around 25 nm in diameter, as contrasted with nab-paclitaxel (Abraxane) which has much larger particles of around 120 nm in diameter.
  • the micellular formulations are quite stable in protein-free media.
  • Sustained release micelles have been prepared in which polymers with very low CMC ( ⁇ 0.1 ⁇ g/ml) can be used for prolonging the circulation time before the micelle degrades.
  • the micelles Upon intravenous injection, the micelles undergo dilution in the body. If the CMC of the micelles is high, the concentration of the polymer or surfactant falls below the CMC upon dilution and hence, the micelles dissociate. Therefore, a higher concentration of the polymer or surfactant has to be used to prepare the micelles so that they withstand the dilution up to 5 1 in the blood.
  • the use of high concentrations might not be feasible due to toxicity related dose limitations.
  • concentrations as low as 5 mg/ml may be used to prepare a micelle formulation in order to counter the dilution effects in the blood.
  • a variety of polymers including diblock copolymers, triblock copolymers and graft copolymers have been synthesized to be stable even after intravenous administration.
  • the smaller nanoparticles are more stable in protein- free solutions than in solutions containing proteins such as serum.
  • the nanoparticles may be at least 20% more stable or 25% more stable or 30% more stable or 35% more stable or 40% more stable or 45% more stable or 50% more stable or 55% more stable or 60% more stable or 65% more stable or 70% more stable or 75% more stable or 80% more stable or 85% more stable or 90% more stable or 95% more stable or 100% more stable or 125% more stable or 150% more stable or 175% more stable or 200% more stable or 500% more stable or 1000% more stable or 5000% more stable or 10000% more stable in a protein free solution than in a solution containing protein.
  • the nanoparticles of the present invention may be between about 10% more stable to about 25000% more stable or about 10% more stable to about 15000% more stable or about 10% more stable to about 12500% more stable or about 10% more stable to about 10000% more stable or from about 10% more stable to about 9000% more stable or from about 10% more stable to about 8000% more stable or from about 10% more stable to about 7000% more stable or from about 10% more stable to about 6000% more stable or from about 1000% more stable to about 500% more stable or from about 10% more stable to about 400% more stable or from about 10% more stable to about 300% more stable or about 10% more stable to about 200% more stable or about 20% more stable to about 125% more stable or about 20% more stable to about 100% more stable or from about 20% more stable to about 90% more stable or from about 20% more stable to about 80% more stable or from about 20% more stable to about 70% more stable or from about 20% more stable to about 60% more stable or from about 20% more stable to about 50% more stable or from about 20% more stable to about 40% more stable or from about 50% more stable to about 2500% or from
  • Nanoparticles are less stable in solutions or media containing proteins such as serum than nab-paclitaxel.
  • the nanoparticles of the present invention may be at least 20% less stable or 25% less stable or 30% less stable or 35% less stable or 40% less stable or 45% less stable or 50% less stable or 55% less stable or 60% less stable or 65% less stable or 70% less stable or 75% less stable or 80% less stable or 85% less stable or 90% less stable or 95% less stable or 100% less stable or 125% less stable or 150% less stable or 175% less stable or 200% less stable or 500% less stable or 1000% less stable or 5000% less stable or 10000% less stable in a protein free solution than in a solution containing protein.
  • the nanoparticles of the present invention may be between about 10% less stable to about 25000% less stable or about 10% less stable to about 15000% less stable or about 10% less stable to about 12500% less stable or about 10% less stable to about 10000% less stable or from about 10% less stable to about 9000% less stable or from about 10% less stable to about 8000% less stable or from about 10% less stable to about 7000% less stable or from about 10% less stable to about 6000% less stable or from about 1000% less stable to about 500% less stable or from about 10% less stable to about 400% less stable or from about 10% less stable to about 300% more stable or about 10% more stable to about 200% more stable or about 20% more stable to about 125% more stable or about 20% more stable to about 100% more stable or from about 20% more stable to about 90% more stable or from about 20% more stable to about 80% more stable or from about 20% more stable to about 70% more stable or from about 20% more stable to about 60% more stable or from about 20% more stable to about 50% more stable or from about 20% more stable to about 40% less stable or from about 50% less stable to about 2500% or from
  • compositions show similar or the same pharmacokinetics in mice and humans as nab-paclitaxel.
  • Table 2 shows a comparison of mean non-compartmental pharmacokinetic parameters of nab-paclitaxel and IG-001 of a 3 hour infusion, 135 mg/m 2 dose.
  • IG-001 and nab-paclitaxel also show similar or the same clinical efficacy as measured by overall response rate in Phase III clinical trials for metastatic breast cancer (See Figure 7).
  • IG-001 exhibited significant instability in serum even at high paclitaxel
  • nab-paclitaxel ceased to exist as a nanoparticle starting at 200 ⁇ g/ml paclitaxel concentrations.
  • IG-001 had expanded PK proportionality and higher MTD as compared to nab-paclitaxel (See Figures 3 and 4).
  • IG-001 exhibits a remarkable stability in protein- free matrices even at low paclitaxel concentrations of 4 ⁇ g/ml.
  • nab-paclitaxel ceased to exist as a nanoparticle starting at 40 ⁇ g/ml paclitaxel concentrations.
  • IG-001 is suitable for intraperitoneal and/or intravesicle modes of drug delivery due to higher nanoparticle residence time and the reduced likelihood of paclitaxel precipitation.
  • IG-001 has 10-fold enhanced stability compared to Abraxane in protein-free matrices.
  • IG-001 exhibited remarkable stability (high CMC) in protein-free matrices even at low paclitaxel concentrations of 4 ug/ml.
  • Abraxane ceased to exist as a nanoparticle starting at 40 ⁇ g/ml paclitaxel concentrations.
  • Significance of these findings is the better suitability of IG-001 for intraperitoneal and/or intravesicle modes of drug delivery due to higher nanoparticle residence time and the reduced likelihood of paclitaxel precipitation.
  • IG- 001 exhibits exceptional stability in protein- free fluid- suitable for intraperitoneal administration for ovarian cancer and intravesicle administration for bladder cancer.
  • IG-001 is highly unstable in serum and therefore shows expanded PK-proportionality making a higher MTD possible.
  • a Phase 1 study of IG-001 a novel Cremophor-free, polymeric micelle formulation of paclitaxel with carboplatin as a primary treatment in patients with advanced ovarian carcinoma was conducted to determine the Maximum Tolerated Dose (MTD) and dosing for Phase 2 trial of IG-001 in combination with carboplatin.
  • MTD Maximum Tolerated Dose
  • Other objectives included overall survival, progress free survival, time to progression, duration of overall response and safety and toxicity.
  • Six patients/dose level were treated with 220, 260, and 300 mg/ in a manner shown in Table 1.
  • MTD was not determined in this phase 1 trial (over 300mg/m 2 )
  • Dose Limiting Toxicity was shown as grade 4 myalgia in one patient at 300 mg/m 2 .
  • the result indicated a response rate of 94.12%, adverse events: 20.64% and LT toxicity: 7.83%. No death related to the disease or treatment.
  • the recommended dose for Phase 2 trial of cremophor-free paclitaxel in combination with Carboplatin was 260mg/m 2 .
  • Ovarian trial design randomized, two-arm trial, primary advanced epithelial ovarian cancer consisting of 100 patients (50/each arm).
  • Control Arm Solvent based paclitaxel 175mg/m2 IV + Carboplatin 5 AUC IV, 3 weeks, 6 cycles and
  • Experimental Arm IG-001 260mg/m 2 IV + Carboplatin 5 AUC IV 3 weeks, 6 cycles.
  • IG-001 300 mg/m 2 with possible dose reductions in 2 stages based on hematological and non-hematological toxicities (except for alopecia) to 240 mg/m 2 and 190 mg/m 2 .
  • the mean age of the subjects was 50.31 years with a range of 29 to 75 years.
  • the disease duration ranged from 0.33 to 312 months with a mean duration of 64.44 months.
  • IG-001 nanoparticle-encapsulated paclitaxel
  • Abraxane nab- paclitaxel
  • IG- 001 was delivered intraperitoneally and Abraxane was delivered intravenously.
  • IGROV-1 Developmental therapeutic program, NCI, Frederick, MD
  • IGROV-1 Developmental therapeutic program, NCI, Frederick, MD
  • Five to six-weeks old athymic female nu + /nu + mice (Harlan) were used. All studies were done in compliance with the University of Virginia ACUC regulation under the approved protocol# 3879 (PI. Said).
  • mice were housed in specific pathogen- free environment with 12 hours light/dark cycles at the University of Virginia vivarium. Mice were injected intra-peritoneally (ip) with 2xl0 6 cells/1 ⁇ Dulbecco's phosphate buffered saline (DPBS). Mice were assigned into experimental groups initiated 7 days post tumor inoculation with the day of injection assigned ad Day 0:
  • mice were monitored twice/week for 12 weeks as follows:
  • % weight loss/gain weekly weight measurement, % weight change from day 0 (weight at the time of injection of tumor cells vs. post-treatment).
  • mice were assigned into the aforementioned cohorts. Examining the changes of the mice weights revealed that all mice receiving IG-001 (Pax) and Abraxane (ABX) experienced weight loss after initiation of treatment (-15-17% from the control untreated mice, ( Figure 8) with no significant difference between the mice receiving comparable doses of both drugs. Mice steadily gained weight after cessation of treatment. Experimental cohorts treated with higher doses of IG-001 90 m/kg/d dose exhibited profound weight loss, lethargy and triggering humane euthanasia at 3 days (8/13) and 5 days (5/13) after initiation of treatment (Table 8). Four mice (4/11) treated with 60 m/kg/d IG-001 also exhibited the same signs after 5 doses and did not survive the study. The latter were excluded from the study analyses vide infra.
  • mice weights did not represent the actual intraperitoneal tumor burden, we developed an arbitrary score for tumor burden to account for the intra-peritoneal spread to various organs as liver, diaphragm, omentum, mysentry as well as the ascitic fluid volume measured at the time of euthanasia. This score accounts for tumor size and multiplicity in a given organ. The total score of each animal is calculated and the mean values/cohort were analyzed.
  • Intraperitoneal injection of Pax significantly decreased IGROVl intraperitoneal tumor burden (Figure 11) in a dose-dependent manner.
  • the incidence of ascites development in mice treatment with ip IG-001 was significantly decreased after treatment with 15 mg/kg/d and 30 mg/kg/d ( Figure 11).
  • Ascites volume significantly decreased in mice treated with 15 mg/kg/d and 30 mg/kg/d compared to control untreated group. Mice treated with 60 mg/kg/d did not develop ascites. No significant difference was observed between cohorts treated with 15 and 30 mg/kg/d.
  • Intravenous Abraxane treatment significantly decreased intra-peritoneal tumor burden in a dose-dependent manner (Figure 12).
  • the incidence of ascites development in mice treatment with iv ABX significantly decreased only after treatment with 30mg/kg/d ( Figure 12).
  • Ascites volume significantly decreased in mice treated with 15 mg/kg/d and 30 mg/kg/d compared to controls. A significant difference was observed between ascites volumes from cohorts treated with 15 mg/kg/d and 30 mg/kg/d ( Figure 12).
  • tumor burden in mice treated with IG-001 was significantly less than those treated with the same doses of iv ABX ( Figure 13). Consistently, ascitic fluid volume in mice treated with 15 mg/kg/d ip IG-001 was significantly less than that in mice treated with the same dose of iv ABX ( Figure 13). However, no significant difference was observed in ascitic fluid volume in mice treated with 30 mg/kg/d of either drug.
  • Example 5 shows:
  • Intraperitoneal IG-001 was well tolerated at doses of 15-60 mg/kg/d but not at 90 mg/kg/d.
  • mice treated with intraperitoneal IG-001 exhibited significantly longer median survival than counterparts treated with the same doses of intravenous Abraxane. 3.
  • Intraperitoneal tumor burden and the development of ascites (incidence and volume) were significantly decreased in mice treated with intraperitoneal IG-001 compared to mice treated with similar doses of intravenous Abraxane.
  • a therapeutic dose of 60 mg/kg/d of intraperitoneal IG-001 was associated with better clinical outcome than 15 and 30 mg/kg/d; however, it was associated with morbidity and mortality in (4/11 mice, 36.4%) after initiation of treatment.

Abstract

There is disclosed a method for treating peritoneal metastatic cancers comprising administering a nanoparticulate formulation of paclitaxel intraperitoneally. More specifically, there is disclosed a method for treating metastatic cancers comprising intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising paclitaxel encapsulated in a micelle with a diblock copolymer.

Description

TREATING METASTATIC CANCER WITH MICELLULAR PACLITAXEL
Technical Field
The present disclosure provides a method for treating peritoneal metastatic cancers comprising administering a nanoparticulate formulation of paclitaxel intraperitoneally. More specifically, the present disclosure provides a method for treating metastatic cancers comprising intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising paclitaxel encapsulated in a micelle with a diblock copolymer.
Background
Abraxane (nab-paclitaxel) and Taxol (chremephore-solubilized paclitaxel) are paclitaxel pharmaceutical formulations. Taxol is solvent based and should be carefully administered since it is an irritant. Side effects are common although the symptoms are either one or two in most cases. The most common side effects include hair loss, peripheral neuropathy, vomiting, diarrhea, myalagia, arthralagia, low blood counts and hypersensitivity. Taxol is a first generation paclitaxel formulation in which Cremophor EL (polyoxyethySated castor oil) is mixed with paclitaxel and given as an infusion for the treatment of ovarian cancer, lung cancer, head and neck cancer, bladder cancer. Taxol has adverse side effects such as anaphylactic shock. Abraxane is paciltaxel nanoparticle encapsulated by albumin.
Abraxane (nab-paclitaxel) is used in first and second line of treatment in metastatic breast cancer. Side effects of Abraxane include bone marrow suppression (primarily neutropenia) which is dose-dependent and a dose-limiting toxicity of Abraxane. In clinical studies, Grade 3-4 neutropenia occurred in 34% of patients with metastatic breast cancer (MBC) and 47% of patients with non-small cell lung cancer (NSCLC).
IG-001 is a polymer bound nanoparticle paclitaxel.
Ovarian cancer is the fifth deadliest disease among American women, with more than 50% presenting with serous papillary histology. High-grade serous epithelial ovarian cancer (SEOC) is associated with intraperitoneal spreading (carcinomatosis) and distant metastases. Standard treatment is aggressive surgical resection followed by platinum-taxane
chemotherapy. Platinum-resistant cancer recurs in approximately 25% of patients within 6 months, and the overall 5-year survival probability is 31%. Therefore, there is a dire need to develop new therapeutics and enhance the delivery of current effective therapeutics. While there are cytotoxic drug compositions that are useful in the treatment of various cancers there exists a need for methods of treatment and drug administration that address metastatic cancers, particularly when there is widespread metastatic cancer in the peritoneum. Summary
The present disclosure provides a method for treating peritoneal metastatic cancers comprising administering a nanoparticulate formulation of paclitaxel intraperitoneally. More specifically, the present disclosure provides a method for treating metastatic cancers comprising intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising paclitaxel encapsulated in a micelle with a diblock copolymer. The present disclosure provides a method for treating metastatic cancers comprising
intraperitoneal or intravesicle administeration a nanoparticulate formulation of paclitaxel comprising paclitazel encapsulated in a micelle with a diblock copolymer. More specifically, the present disclosure provides a method for administering a nanoparticulate formulation of paclitaxel comprising infusing a nanoparticulate formulation intraperitoneally at a dose of from about 15 mg/kg/day to about 60 mg/kg/day. More preferably, the intraperitoneal dose of paclitaxel is from 25 mg/kg/day to 50 mg/kg/day. Most preferably, the intraperitoneal dose of paclitaxel is from 30 mg/kg/day to 50 mg/kg/day. Preferably, the nanoparticulate formulation of paclitaxel has a particle size average of 15 nm to 40 nm.
The present disclosure provides compositions comprising cancer drugs in a micelle where the composition is stable in protein-free medium and less stable in a protein containing medium. The compositions of the present invention are conditionally stable in that they are more stable in one media than another, in particular the compositions are more stable in protein- free medium and less stable in a protein containing medium. The present disclosure provides paclitaxel compositions where the paclitaxel is contained within a micelle and where the paclitaxel composition is stable in protein-free medium and less stable in a protein containing medium. The present disclosure further provides micellular compositions where the composition comprises about 4 μg/ml to about 2000 μg/ml of paclitaxel. The present disclosure provides micellular compositions where the composition is at least 20% more stable or 50% more stable or 100% more stable in a protein- free solution than in a solution containing protein. The present disclosure provides micellular compositions where the composition is at least 20% more stable or 50% more stable or 100% more stable in a protein-free solution than nab-paclitaxel. The present disclosure provides micellular compositions where the composition is at least 20% less stable or 50% less stable or 100% less stable in a protein containing solution such as serum than nab-paclitaxel.
The disclosure provides methods of treating patients with metastatic breast cancer by administering paclitaxel-containing micelles to patients. The disclosure provides methods of treating patients with metastatic cancer by administering paclitaxel-containing micelles to patients where the micelles are comprised of a diblock copolymer including IG-001 which is a paclitaxel-containing micelle. The present disclosure provides method of treating metastatic breast cancer wherein the amount of paclitaxel administered is at least 190mg/m2. The present disclosure provides methods of treating metastatic breast cancer where the paclitaxel containing micelles are administered in at least 3 cycles. The present disclosure provides method of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 3 cycles to 20 cycles. The present disclosure provides methods of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 3 cycles to 15 cycles. The present disclosure provides method of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 3 cycles to 10 cycles. The present disclosure provides method of treating metastatic breast cancer where the paclitaxel containing micelles are administered from 5 cycles to 10 cycles. The present disclosure provides method of treating metastatic breast cancer where the overall response rate is greater than 20% or greater than 30% or greater than 40% or greater than 50% or from between about 20% to about 90% or from about 25% to about 75% or from about 30% to about 60% or from 30% to about 50% or from about 30% to about 40%. The present disclosure provides method of treating metastatic breast cancer by administering paclitaxel containing micelles to the patient at least twice such that there is a first dose and a second dose. The present disclosure provides method of treating metastatic breast cancer by administering paclitaxel containing micelles to the patient at least twice such that there is a first dose and a second dose where the first dose is about 300 mg/m2 and the second dose is about 240 mg/m2. The present disclosure provides method of treating patients with metastatic breast cancer comprising administering paclitaxel containing micelles to the patient comprising administering a dose of paclitaxel containing micelles containing about 300 mg/m2 pacitaxel for at least one cycle; administering a dose of paclitaxel containing micelles containing about 260 mg/m2 pacitaxel for at least one cycle; and administering a dose of paclitaxel containing micelles containing about 190 mg/m2 pacitaxel for at least one cycle. Brief Description of the Figures Figure 1 - Plot of particle size versus paclitaxel concentration for nab-paclitaxel in phosphate buffered saline (PBS) and O.lx serum and lx serum.
Figure 2 - Plot of particle size versus paclitaxel concentration for IG-001 in phosphate buffered saline (PBS) and O.lx serum and lx serum.
Figure 3 - Plot of dose proportionality curve for Taxol, nab-paclitaxel and IG-001
Figure 4 - Plot of dose limiting toxicity curve for Taxol, nab-paclitaxel and IG-001.
Figure 5 - Plot of paclitaxel concentration of delivery of 30 mg/kg bolus by Abraxane and IG-001
Figure 6A and B - Plots of paclitaxel versus time for nab-paclitaxel and IG-001 in mice.
Figure 7 - Plot of overall response rate for IG-001, Taxol and nab-paclitaxel in Phase III study of metastatic breast cancer.
Figure 8 shows the effects of the treatments in Example 5 in terms of weight gains of the mice.
Figure 9 shows that treatment with either ip IG-001 or iv ABX (nab-paclitaxel) significantly increased the median survival of mice compared to untreated control
Figure 10 shows overall survival of control and experimental groups of mice in Example 5.
Figure 11 shows Kaplan Meier curves of the survival of IG-001 (Pax) treated group (left) and nab-paclitaxel (ABX) treated group (right).
Figure 12 shows (Left) Box plots of tumor burden arbitrary scores of mice treated with ip IG-001. And (Right) dot plots of the incidence and amount of ascitic fluid accumulating in tumor-bearing mice receiving the indicated doses of IG-001. * Statistical analysis between the indicated groups was performed by Mann-Whitney non-parametric t- test. **Disease progression as a function of the indicated doses of IG-001 was analyzed by Kruskall Wallis test (non-parametric One Way ANOVA ).. The incidence of ascites development was analyzed by Chi square (X2 ) test, #P<0.0001. Ascites volume in treated mice compared to controls was analyzed by One way ANOVA and unpaired Student' s t-test.
Figure 13 shows (Left) Box plots of tumor burden arbitrary scores of mice treated with iv ABX. and (Right) Dot plots of the incidence and amount of ascitic fluid accumulating in tumor-bearing mice receiving the indicated doses of ABX. * Statistical analysis between the indicated groups was performed by Mann-Whitney non-parametric t-test. **Disease progression as a function of the indicated doses of ABX was analyzed by Kruskall Wallis test (non-parametric One Way ANOVA ). The incidence of ascites development was analyzed by Chi square (X2 ) test, #P<0.0001. Ascites volume in treated mice compared to controls was analyzed by One way ANOVA and unpaired Student's t-test.
Detailed Description
The present disclosure provides a method for treating metastatic cancers comprising intraperitoneal or intravesicle administeration a nanoparticulate formulation of paclitaxel comprising paclitazel encapsulated in a micelle with a diblock copolymer. More specifically, the present disclosure provides a method for intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising infusing a nanoparticulate formulation intraperitoneally at a dose of from about 15 mg/kg/day to about 60 mg/kg/day. More preferably, the intraperitoneal dose of paclitaxel is from 25 mg/kg/day to 50 mg/kg/day. Most preferably, the intraperitoneal dose of paclitaxel is from 30 mg/kg/day to 50 mg/kg/day. Preferably, the nanoparticulate formulation of paclitaxel has a particle size average of 15 nm to 40 nm.
The present disclosure provides paclitaxel-containing pharmaceutical compositions in a micelle, that is stable in protein-free medium and less stable in a protein containing medium. The paclitaxel formulation includes amphiphilic block copolymer which may comprise a hydrophilic block (A) and a hydrophobic block (B) linked with each other in the form of A-B, A-B-A or B-A-B structure. Additionally, the amphiphilic block copolymer forma a core-shell type polymeric micelles in its aqueous solution state, wherein the hydrophobic block forms the core and the hydrophilic block forms the shell. Preferably, the hydrophilic block (A) of the amphiphilic block copolymer is polyethylene glycol (PEG) or monomethoxypolyethylene glycol (mPEG). The hydrophilic block (A) may have a weight average molecular weight of 500-20,000 daltons, specifically 1,000-5,000 daltons, and more specifically 1,000-2,500 daltons. The hydrophobic block (B) of the amphiphilic block copolymer is a water- insoluble, biodegradable polymer. Preferably, the hydrophobic block (B) is polylactic acid (PLA) or poly(lactic-co-glycolic acid) (PLGA). The hydrophobic block (B) has a weight average molecular weight of 500-20,000 daltons, specifically 1,000-5,000 daltons, and more specifically 1,000-2,500 daltons. Hydroxyl end groups of the hydrophobic block (B) are protected with fatty acid groups, such as acetate, propionate, butyrate, stearate, palmitate groups, and the like. The amphiphilic block copolymer comprising the hydrophilic block (A) and the hydrophobic block (B) are present in the pharmacrutical composition in an amount of 20-98 wt %, specifically 65-98 wt %, and more specifically 80-98 wt % based on the total dry weight of the composition. Alternatively, the hydrophilic block (A) and the hydrophobic block (B) are present in the amphiphilic block copolymer in such a ratio that the copolymer comprises 40-70 wt %, specifically 50-60 wt % of the hydrophilic block (A) based on the weight of the copolymer. When the hydrophilic block (A) is present in a proportion less than 40%, the polymer has undesirably low solubility to water, resulting in difficulty in forming micelles. On the other hand, when the hydrophilic block (A) is present in a proportion greater than 70%, the polymer becomes too hydrophilic to form stable polymeric micelles, and thus the composition may not be used as a composition for solubilizing paclitaxel.
A preferred paclitaxel formulation is IG-001 (also referred to as Genexol-PM) which is a polymeric micelle formulation of paclitaxel. IG-001 has a di-block copolymer composed of methoxy poly (ethylene glycol)-poly (lactide) to form nanoparticles with paclitaxel containing hydrophobic core and a hydrophilic shell. The nanoparticles formed are around 25 nm in diameter, as contrasted with nab-paclitaxel (Abraxane) which has much larger particles of around 120 nm in diameter.
The micellular formulations are quite stable in protein-free media. Sustained release micelles have been prepared in which polymers with very low CMC (< 0.1 μg/ml) can be used for prolonging the circulation time before the micelle degrades. Upon intravenous injection, the micelles undergo dilution in the body. If the CMC of the micelles is high, the concentration of the polymer or surfactant falls below the CMC upon dilution and hence, the micelles dissociate. Therefore, a higher concentration of the polymer or surfactant has to be used to prepare the micelles so that they withstand the dilution up to 5 1 in the blood.
However, the use of high concentrations might not be feasible due to toxicity related dose limitations. If the polymer or surfactant has a CMC lower than 0.1 μg/ml, concentrations as low as 5 mg/ml may be used to prepare a micelle formulation in order to counter the dilution effects in the blood. A variety of polymers including diblock copolymers, triblock copolymers and graft copolymers have been synthesized to be stable even after intravenous administration.
The smaller nanoparticles, such as IG-001, are more stable in protein- free solutions than in solutions containing proteins such as serum. The nanoparticles may be at least 20% more stable or 25% more stable or 30% more stable or 35% more stable or 40% more stable or 45% more stable or 50% more stable or 55% more stable or 60% more stable or 65% more stable or 70% more stable or 75% more stable or 80% more stable or 85% more stable or 90% more stable or 95% more stable or 100% more stable or 125% more stable or 150% more stable or 175% more stable or 200% more stable or 500% more stable or 1000% more stable or 5000% more stable or 10000% more stable in a protein free solution than in a solution containing protein. The nanoparticles of the present invention may be between about 10% more stable to about 25000% more stable or about 10% more stable to about 15000% more stable or about 10% more stable to about 12500% more stable or about 10% more stable to about 10000% more stable or from about 10% more stable to about 9000% more stable or from about 10% more stable to about 8000% more stable or from about 10% more stable to about 7000% more stable or from about 10% more stable to about 6000% more stable or from about 1000% more stable to about 500% more stable or from about 10% more stable to about 400% more stable or from about 10% more stable to about 300% more stable or about 10% more stable to about 200% more stable or about 20% more stable to about 125% more stable or about 20% more stable to about 100% more stable or from about 20% more stable to about 90% more stable or from about 20% more stable to about 80% more stable or from about 20% more stable to about 70% more stable or from about 20% more stable to about 60% more stable or from about 20% more stable to about 50% more stable or from about 20% more stable to about 40% more stable or from about 50% more stable to about 2500% or from about 50% more stable to about 1250% more stable or from about 50% more stable to about 1000% more stable in a protein free solution than in a solution containing protein.
Nanoparticles are less stable in solutions or media containing proteins such as serum than nab-paclitaxel. The nanoparticles of the present invention may be at least 20% less stable or 25% less stable or 30% less stable or 35% less stable or 40% less stable or 45% less stable or 50% less stable or 55% less stable or 60% less stable or 65% less stable or 70% less stable or 75% less stable or 80% less stable or 85% less stable or 90% less stable or 95% less stable or 100% less stable or 125% less stable or 150% less stable or 175% less stable or 200% less stable or 500% less stable or 1000% less stable or 5000% less stable or 10000% less stable in a protein free solution than in a solution containing protein. The nanoparticles of the present invention may be between about 10% less stable to about 25000% less stable or about 10% less stable to about 15000% less stable or about 10% less stable to about 12500% less stable or about 10% less stable to about 10000% less stable or from about 10% less stable to about 9000% less stable or from about 10% less stable to about 8000% less stable or from about 10% less stable to about 7000% less stable or from about 10% less stable to about 6000% less stable or from about 1000% less stable to about 500% less stable or from about 10% less stable to about 400% less stable or from about 10% less stable to about 300% more stable or about 10% more stable to about 200% more stable or about 20% more stable to about 125% more stable or about 20% more stable to about 100% more stable or from about 20% more stable to about 90% more stable or from about 20% more stable to about 80% more stable or from about 20% more stable to about 70% more stable or from about 20% more stable to about 60% more stable or from about 20% more stable to about 50% more stable or from about 20% more stable to about 40% less stable or from about 50% less stable to about 2500% or from about 50% less stable to about 1250% less stable or from about 50% less stable to about 1000% less stable in a protein free solution than in a solution containing protein.
The compositions show similar or the same pharmacokinetics in mice and humans as nab-paclitaxel.
Table 1 (in mice)
Figure imgf000009_0001
Table 2 shows a comparison of mean non-compartmental pharmacokinetic parameters of nab-paclitaxel and IG-001 of a 3 hour infusion, 135 mg/m2 dose.
Table 2 (in humans)
Figure imgf000009_0002
IG-001 and nab-paclitaxel also show similar or the same clinical efficacy as measured by overall response rate in Phase III clinical trials for metastatic breast cancer (See Figure 7).
IG-001 exhibited significant instability in serum even at high paclitaxel
concentrations of 2000 μg/ml. Conversely, nab-paclitaxel ceased to exist as a nanoparticle starting at 200 μg/ml paclitaxel concentrations. IG-001 had expanded PK proportionality and higher MTD as compared to nab-paclitaxel (See Figures 3 and 4). Moreover, IG-001 exhibits a remarkable stability in protein- free matrices even at low paclitaxel concentrations of 4 μg/ml. Conversely, nab-paclitaxel ceased to exist as a nanoparticle starting at 40 μg/ml paclitaxel concentrations. IG-001 is suitable for intraperitoneal and/or intravesicle modes of drug delivery due to higher nanoparticle residence time and the reduced likelihood of paclitaxel precipitation.
Example 1
A comparison of dissolution/instability profiles of nab-paclitaxel and IG-001 was conducted in serum-containing (IX FBS, 0.1 X FBS) and protein-free (IX PBS) matrices at 37°C using Dynamic Light Scattering (DLS) methodology (Malvern's Zetasizer Nano S and Wyatt's Nanostar). The results of the study are displayed in Figures 1 and 2. IG-001 had a 10-fold diminished stability versus Abraxane in serum. IG-001 exhibited significant instability in serum even at high paclitaxel concentrations of 2000 ug/ml. Conversely, Abraxane ceased to exist as a nanoparticle starting at about 200 ug/ml paclitaxel
concentrations. This data may explain the observed expanded PK proportionality and the higher maximum tolerated does (MTD) of IG-001 vs. Abraxane.
IG-001 has 10-fold enhanced stability compared to Abraxane in protein-free matrices. IG-001 exhibited remarkable stability (high CMC) in protein-free matrices even at low paclitaxel concentrations of 4 ug/ml. Conversely, Abraxane ceased to exist as a nanoparticle starting at 40 μg/ml paclitaxel concentrations. Significance of these findings is the better suitability of IG-001 for intraperitoneal and/or intravesicle modes of drug delivery due to higher nanoparticle residence time and the reduced likelihood of paclitaxel precipitation. IG- 001 exhibits exceptional stability in protein- free fluid- suitable for intraperitoneal administration for ovarian cancer and intravesicle administration for bladder cancer. IG-001 is highly unstable in serum and therefore shows expanded PK-proportionality making a higher MTD possible.
Example 2
Phase 1 study of IG-OOlwith Carboplatin as a Primary Treatment in Patients with Advanced Ovarian Carcinoma
A Phase 1 study of IG-001, a novel Cremophor-free, polymeric micelle formulation of paclitaxel with carboplatin as a primary treatment in patients with advanced ovarian carcinoma was conducted to determine the Maximum Tolerated Dose (MTD) and dosing for Phase 2 trial of IG-001 in combination with carboplatin. Other objectives included overall survival, progress free survival, time to progression, duration of overall response and safety and toxicity. Six patients/dose level were treated with 220, 260, and 300 mg/ in a manner shown in Table 1. MTD was not determined in this phase 1 trial (over 300mg/m2)
Table 3
Figure imgf000011_0001
Dose Limiting Toxicity was shown as grade 4 myalgia in one patient at 300 mg/m2. The result indicated a response rate of 94.12%, adverse events: 20.64% and LT toxicity: 7.83%. No death related to the disease or treatment. The recommended dose for Phase 2 trial of cremophor-free paclitaxel in combination with Carboplatin was 260mg/m2.
Example 3
Combination Therapy of IG-001 Plus Carboplatin Compared to Paclitaxel Plus Carboplatin as a 1st line Treatment in Patients with Epithelial Ovarian Cancer Ovarian trial design: randomized, two-arm trial, primary advanced epithelial ovarian cancer consisting of 100 patients (50/each arm). Control Arm: Solvent based paclitaxel 175mg/m2 IV + Carboplatin 5 AUC IV, 3 weeks, 6 cycles and Experimental Arm: IG-001 260mg/m2 IV + Carboplatin 5 AUC IV 3 weeks, 6 cycles.
Table 4
Figure imgf000011_0002
Table 5
Figure imgf000012_0001
The results indicate that the response rate was 88.00% vs. 77.8% (IG-001 vs.
paclitaxel) and the primary endpoint of noninferiority to paclitaxel was met. The one-sided 95% upper confidence limit was 4.95, which is lower than the non-inferiority threshold (16.3%), indicating that the study group is not inferior to the control group. Adverse events were similar to paclitaxel despite the higher dose.
Example 4
A total of 186 subjects were enrolled and had surveillance data sheets collected and efficacy and safety assessments were conducted. The initial dose of IG-001 was 300 mg/m2 with possible dose reductions in 2 stages based on hematological and non-hematological toxicities (except for alopecia) to 240 mg/m2 and 190 mg/m2. The mean age of the subjects was 50.31 years with a range of 29 to 75 years. The disease duration ranged from 0.33 to 312 months with a mean duration of 64.44 months. There were 180 subjects (96.77%) with stage IV disease and 6 subjects (3.23%) with stage III disease. There were 81 subjects (97.31%) with prior treatment for breast cancer including 174 subjects (96.13%) with prior chemotherapy.
The results of the efficacy study indicated that 36 of the 148 evaluable subjects (24.32%) responded to the drug treatment and were judged to be efficacious.
Table 6
Figure imgf000012_0002
Unevaluable 1 0.68
One factor which correlated with efficacy was the total number of drug treatment cycles (p <
0.0001)
Table 7
Figure imgf000013_0001
145 subjects (77.96%) reported at least one TEAE on study and there were a total of 710 adverse events. The most frequent TEAEs were neutropenia (23.12%), peripheral neuropathy (18.82%), nausea (16.13%), myalgia (13.98%), leukopenia (13.44%), peripheral sensory neuropathy (11.29%), tingling (10.22%), decreased hemoglobin (9.68%), alopecia (9.14%), loss of appetite (9.14%), neuropathy (8.60%), pruritus (5.91%), vomiting (5.91 %), general weakness (5.38%), dyspnea (5.38%) headache (5.38%), rash (4.84%) and cough (4.84%). Urticaria was reported in 1 subjects (0.54%). Hypotension occurred in 3 subjects (1.61 %). Thrombocytopenia occurred in 6 subjects (3.23%). Of the 710 TEAEs, 307 (43.24%) were Grade 1 in severity, 252 (35.49%) were Grade 2, 94 (13.24%) were Grade 3, 55 (7.75%) were Grade 4 and 2 (0.28%) were Grade 5. 620 TEAEs (87.32%) were related or unknown and 90 (12.68%) were not related to study drug.
Example 5
This example provides the results of a study designed to compare the in vivo efficacy of two formulations of nanoparticle-encapsulated paclitaxel: IG-001 and Abraxane (nab- paclitaxel) in a preclinical mouse model of human ovarian cancer peritoneal metastasis. IG- 001 was delivered intraperitoneally and Abraxane was delivered intravenously. One established ovarian cancer cell line (IGROV-1 ; Developmental therapeutic program, NCI, Frederick, MD) were used for tumor implantation studies. Five to six-weeks old athymic female nu+/nu+ mice (Harlan) were used. All studies were done in compliance with the University of Virginia ACUC regulation under the approved protocol# 3879 (PI. Said). Mice were housed in specific pathogen- free environment with 12 hours light/dark cycles at the University of Virginia vivarium. Mice were injected intra-peritoneally (ip) with 2xl06 cells/1 ΟΟμΙ Dulbecco's phosphate buffered saline (DPBS). Mice were assigned into experimental groups initiated 7 days post tumor inoculation with the day of injection assigned ad Day 0:
Group A: Control, injected with 50μ1 saline; n=12
Group B: Abraxane (ABX) 15 mg/kg/d, qdx5; n= 10— Administered intravenously (i.v.) Group C: Abraxane (ABX) 30 mg/kg/d, qdx5; n= 10— Administered i.v.
Group D: IG-001 (Pax) 15 mg/kg/d, qdx5; n= 10— Administered i.p.
Group E: IG-001 (Pax) 30 mg/kg/d, qdx5; n= 10— Administered i.p.
Group F: IG-001 (Pax) 60 mg/kg/d, qdx5; n=l l— Administered i.p.
Group G: IG-001 (Pax) 90 mg/kg/d, qdx5; n=l 3— Administered i.p.
Mice were monitored twice/week for 12 weeks as follows:
1. % weight loss/gain: weekly weight measurement, % weight change from day 0 (weight at the time of injection of tumor cells vs. post-treatment).
2. Survival Time (Kaplan-Meier, K-M plot).
3. Presence/extent of intra-peritoneal tumor growth and the development of ascites.
One week after tumor cell injection, mice were assigned into the aforementioned cohorts. Examining the changes of the mice weights revealed that all mice receiving IG-001 (Pax) and Abraxane (ABX) experienced weight loss after initiation of treatment (-15-17% from the control untreated mice, (Figure 8) with no significant difference between the mice receiving comparable doses of both drugs. Mice steadily gained weight after cessation of treatment. Experimental cohorts treated with higher doses of IG-001 90 m/kg/d dose exhibited profound weight loss, lethargy and triggering humane euthanasia at 3 days (8/13) and 5 days (5/13) after initiation of treatment (Table 8). Four mice (4/11) treated with 60 m/kg/d IG-001 also exhibited the same signs after 5 doses and did not survive the study. The latter were excluded from the study analyses vide infra.
Table 8
Figure imgf000014_0001
Figure imgf000015_0001
Effect of Treatment on the survival of experimental groups: Treatment with either ip IG-001 or iv ABX (nab-paclitaxel) significantly increased the median survival of mice compared to untreated control (Figure 9). Intraperitoneal treatment with IG-001 conferred significant increase in median survival compared to similar doses of iv ABX (Figure 9).
Analysis of Kaplan Meier survival curves (Mantel-Cox analysis) of IG-001 treated cohorts indicated significant dose-dependent survival advantage (Figure 10). ABX-treated cohorts did not exhibit dose-dependent survival advantage (Figure 10). Intraperitoneal IG- 001 treatment significantly conferred survival advantage over iv ABX at 15 and 30 mg/kg/d.
Evaluation of Intra-abdominal Tumor Burden: Because mice weights did not represent the actual intraperitoneal tumor burden, we developed an arbitrary score for tumor burden to account for the intra-peritoneal spread to various organs as liver, diaphragm, omentum, mysentry as well as the ascitic fluid volume measured at the time of euthanasia. This score accounts for tumor size and multiplicity in a given organ. The total score of each animal is calculated and the mean values/cohort were analyzed.
In vivo response of IGROVl tumor xenografts to IG-001 intraperitoneal treatment: Intraperitoneal injection of Pax significantly decreased IGROVl intraperitoneal tumor burden (Figure 11) in a dose-dependent manner. The incidence of ascites development in mice treatment with ip IG-001 was significantly decreased after treatment with 15 mg/kg/d and 30 mg/kg/d (Figure 11). Ascites volume significantly decreased in mice treated with 15 mg/kg/d and 30 mg/kg/d compared to control untreated group. Mice treated with 60 mg/kg/d did not develop ascites. No significant difference was observed between cohorts treated with 15 and 30 mg/kg/d.
In vivo response of IGROVl tumor xenografts to Abraxane intravenous treatment: Intravenous Abraxane treatment significantly decreased intra-peritoneal tumor burden in a dose-dependent manner (Figure 12). The incidence of ascites development in mice treatment with iv ABX significantly decreased only after treatment with 30mg/kg/d (Figure 12). Ascites volume significantly decreased in mice treated with 15 mg/kg/d and 30 mg/kg/d compared to controls. A significant difference was observed between ascites volumes from cohorts treated with 15 mg/kg/d and 30 mg/kg/d (Figure 12).
At the time of euthanasia, tumor burden in mice treated with IG-001 (ip) was significantly less than those treated with the same doses of iv ABX (Figure 13). Consistently, ascitic fluid volume in mice treated with 15 mg/kg/d ip IG-001 was significantly less than that in mice treated with the same dose of iv ABX (Figure 13). However, no significant difference was observed in ascitic fluid volume in mice treated with 30 mg/kg/d of either drug.
Accordingly, Example 5 shows:
1. Intraperitoneal IG-001 was well tolerated at doses of 15-60 mg/kg/d but not at 90 mg/kg/d.
2. Tumor-bearing mice treated with intraperitoneal IG-001 exhibited significantly longer median survival than counterparts treated with the same doses of intravenous Abraxane. 3. Intraperitoneal tumor burden and the development of ascites (incidence and volume) were significantly decreased in mice treated with intraperitoneal IG-001 compared to mice treated with similar doses of intravenous Abraxane.
4. A therapeutic dose of 60 mg/kg/d of intraperitoneal IG-001 was associated with better clinical outcome than 15 and 30 mg/kg/d; however, it was associated with morbidity and mortality in (4/11 mice, 36.4%) after initiation of treatment.

Claims

We claim:
1. A method for treating peritoneal metastatic cancers comprising administering a nanoparticulate formulation of paclitaxel intraperitoneally.
2. A method for treating metastatic cancers comprising intraperitoneal or intravesicle administration a nanoparticulate formulation of paclitaxel comprising paclitaxel encapsulated in a micelle with a diblock copolymer.
3. The method for treating metastatic cancers of claim 2, wherein the paclitaxel dose of from about 15 mg/kg/day to about 60 mg/kg/day.
4. The method for treating metastatic cancers of claim 3, wherein the paclitaxel dose is from 25 mg/kg/day to 50 mg/kg/day.
5. The method for treating metastatic cancers of claim 3, wherein the paclitaxel dose is from 30 mg/kg/day to 50 mg/kg/day.
6. The method for treating metastatic cancers of claim 2, wherein the nanoparticulate formulation of paclitaxel has a particle size average of 15 nm to 40 nm.
7. The method for treating metastatic cancers of claim 2, wherein the amount of paclitaxel administered is at least 190 mg/m2.
PCT/US2014/059207 2013-10-04 2014-10-04 Treating metastatic cancer with micellular paclitaxel WO2015051349A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361961093P 2013-10-04 2013-10-04
US61/961,093 2013-10-04

Publications (1)

Publication Number Publication Date
WO2015051349A1 true WO2015051349A1 (en) 2015-04-09

Family

ID=52779220

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2014/059207 WO2015051349A1 (en) 2013-10-04 2014-10-04 Treating metastatic cancer with micellular paclitaxel
PCT/US2014/000194 WO2016043690A1 (en) 2013-10-04 2014-10-06 Conditionally stable micelle compositions for metastatic breast cancer treatment

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2014/000194 WO2016043690A1 (en) 2013-10-04 2014-10-06 Conditionally stable micelle compositions for metastatic breast cancer treatment

Country Status (1)

Country Link
WO (2) WO2015051349A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017192573A1 (en) * 2016-05-02 2017-11-09 Massachusetts Institute Of Technology Nanoparticle conjugates of highly potent toxins and intraperitoneal administration of nanoparticles for treating or imaging cancer

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001021174A1 (en) * 1999-09-23 2001-03-29 Dabur Research Foundation Formulations of paclitaxel entrapped into nanoparticles of polymeric micelles
US20030157161A1 (en) * 2001-05-01 2003-08-21 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating inflammatory conditions utilizing protein or polysaccharide containing anti-microtubule agents
WO2005040247A1 (en) * 2003-10-24 2005-05-06 Samyang Corporation Polymeric composition for drug delivery
US20080160095A1 (en) * 1993-02-22 2008-07-03 Abraxis Bioscience, Llc Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20100069426A1 (en) * 2008-06-16 2010-03-18 Zale Stephen E Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
US20110158906A1 (en) * 2007-08-13 2011-06-30 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Targeted block copolymer micelles
US8043631B2 (en) * 2004-04-02 2011-10-25 Au Jessie L S Tumor targeting drug-loaded particles
WO2012070029A1 (en) * 2010-11-26 2012-05-31 University Of The Witwatersrand, Johannesburg A pharmaceutical composition
US20120231053A1 (en) * 2009-10-21 2012-09-13 Nippon Kayaku Kabushiki Kaisha Block Copolymer For Intraperitoneal Administration Containing Anti-Cancer Agent, Micelle Preparation Thereof, And Cancer Therapeutic Agent Comprising The Micelle Preparation As Active Ingredient
WO2012158960A2 (en) * 2011-05-17 2012-11-22 H. Lee Moffitt Cancer Center & Research Institute, Inc. Melanocortin 1 receptor ligands and methods of use
US8383136B2 (en) * 2009-09-25 2013-02-26 Wisconsin Alumni Research Foundation Micelle encapsulation of therapeutic agents

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014165829A2 (en) * 2013-04-05 2014-10-09 Igdrasol Nanoparticle formulations

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080160095A1 (en) * 1993-02-22 2008-07-03 Abraxis Bioscience, Llc Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
WO2001021174A1 (en) * 1999-09-23 2001-03-29 Dabur Research Foundation Formulations of paclitaxel entrapped into nanoparticles of polymeric micelles
US20030157161A1 (en) * 2001-05-01 2003-08-21 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating inflammatory conditions utilizing protein or polysaccharide containing anti-microtubule agents
WO2005040247A1 (en) * 2003-10-24 2005-05-06 Samyang Corporation Polymeric composition for drug delivery
US8043631B2 (en) * 2004-04-02 2011-10-25 Au Jessie L S Tumor targeting drug-loaded particles
US20110158906A1 (en) * 2007-08-13 2011-06-30 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Targeted block copolymer micelles
US20100069426A1 (en) * 2008-06-16 2010-03-18 Zale Stephen E Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
US8383136B2 (en) * 2009-09-25 2013-02-26 Wisconsin Alumni Research Foundation Micelle encapsulation of therapeutic agents
US20120231053A1 (en) * 2009-10-21 2012-09-13 Nippon Kayaku Kabushiki Kaisha Block Copolymer For Intraperitoneal Administration Containing Anti-Cancer Agent, Micelle Preparation Thereof, And Cancer Therapeutic Agent Comprising The Micelle Preparation As Active Ingredient
WO2012070029A1 (en) * 2010-11-26 2012-05-31 University Of The Witwatersrand, Johannesburg A pharmaceutical composition
WO2012158960A2 (en) * 2011-05-17 2012-11-22 H. Lee Moffitt Cancer Center & Research Institute, Inc. Melanocortin 1 receptor ligands and methods of use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
XIAO, K ET AL.: "A self-assembling nanoparticle for paclitaxel delivery in ovarian cancer", BIOMATERIALS, vol. 30, 2009, pages 6006 - 6016 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017192573A1 (en) * 2016-05-02 2017-11-09 Massachusetts Institute Of Technology Nanoparticle conjugates of highly potent toxins and intraperitoneal administration of nanoparticles for treating or imaging cancer
US10751423B2 (en) 2016-05-02 2020-08-25 Massachusetts Institute Of Technology Nanoparticle conjugates of highly potent toxins and intraperitoneal administration of nanoparticles for treating or imaging cancer
US11730826B2 (en) 2016-05-02 2023-08-22 Massachusetts Institute Of Technology Amphiphilic nanoparticles for delivery of CRISPR based therapy

Also Published As

Publication number Publication date
WO2016043690A1 (en) 2016-03-24

Similar Documents

Publication Publication Date Title
AU2017239541B2 (en) Use of polymeric excipients for lyophilization or freezing of particles
Bromberg Polymeric micelles in oral chemotherapy
Lee et al. Development of docetaxel-loaded intravenous formulation, Nanoxel-PM™ using polymer-based delivery system
ES2719093T3 (en) Compositions of low water soluble drugs with greater stability and methods for their preparation
US11090268B2 (en) Targeted hydrophobic anti-tumor drug nanoformulation and preparation method thereof
He et al. Poly (ethylene glycol)-block-poly (ε-caprolactone)–and phospholipid-based stealth nanoparticles with enhanced therapeutic efficacy on murine breast cancer by improved intracellular drug delivery
Jiang et al. Deoxycholic acid-modified chitooligosaccharide/mPEG-PDLLA mixed micelles loaded with paclitaxel for enhanced antitumor efficacy
CN109771663B (en) Preparation and application of acid-responsive anticancer nano-drug
Wang et al. Improving systemic circulation of paclitaxel nanocrystals by surface hybridization of DSPE-PEG2000
Liang et al. Nanocrystal-loaded liposome for targeted delivery of poorly water-soluble antitumor drugs with high drug loading and stability towards efficient cancer therapy
Andrade et al. Micelle-based systems for pulmonary drug delivery and targeting
CN110538328A (en) polypeptide compound, drug-loaded nanoparticle, preparation method of polypeptide compound and drug-loaded nanoparticle, drug composition and application of drug composition
Cheng et al. Carfilzomib and paclitaxel co-loaded protein nanoparticles an effective therapy against pancreatic adenocarcinomas
US20160128971A1 (en) Nanoparticle Compositions
WO2009121997A2 (en) Pegylated nanoparticles containing a biologically active molecule and use thereof
Lv et al. “Carrier–drug” layer-by-layer hybrid assembly of biocompatible polydopamine nanoparticles to amplify photo-chemotherapy
Mustafa et al. Nanoscale drug delivery systems for cancer therapy using paclitaxel—a review of challenges and latest progressions
WO2015051349A1 (en) Treating metastatic cancer with micellular paclitaxel
US20160346219A1 (en) Phospholipid-coated therapeutic agent nanoparticles and related methods
US20150366806A1 (en) Method of Engineering Nanoparticle
Lim et al. Dynamic drug release state and PEG length in PEGylated liposomal formulations define the distribution and pharmacological performance of drug
WO2015042234A1 (en) Conditionally stable micelle compositions for cancer treatment including ovarian cancer
TW201705941A (en) Phospholipid-coated therapeutic agent nanoparticlesn and related methods
Oh et al. Core/shell nanoparticles for pH-sensitive delivery of doxorubicin
Merkulova et al. Etoposide-loaded colloidal delivery systems based on biodegradable polymeric carriers

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: 14851023

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205N DATED 11/07/2016)

122 Ep: pct application non-entry in european phase

Ref document number: 14851023

Country of ref document: EP

Kind code of ref document: A1