OA18218A - Carrier-antibody compositions and methods of making and using the same - Google Patents

Carrier-antibody compositions and methods of making and using the same Download PDF

Info

Publication number
OA18218A
OA18218A OA1201700009 OA18218A OA 18218 A OA18218 A OA 18218A OA 1201700009 OA1201700009 OA 1201700009 OA 18218 A OA18218 A OA 18218A
Authority
OA
OAPI
Prior art keywords
cancer
nanoparticle
abx
tumor
antibodies
Prior art date
Application number
OA1201700009
Inventor
Svetomir N. Markovic
Wendy K. NEVALA
Original Assignee
Mayo Foundation For Medical Education And Research
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 Mayo Foundation For Medical Education And Research filed Critical Mayo Foundation For Medical Education And Research
Publication of OA18218A publication Critical patent/OA18218A/en

Links

Abstract

Described herein are compositions of antibodies and carrier proteins and methods of making and using the same, in particular, as a cancer therapeutic. Also described are lyophilized compositions of antibodies and carrier proteins and methods of making and using the same, in particular, as a cancer therapeutic.

Description

CARRIER-ANTIBODY COMPOSITIONS AND METHODS OF MAKING AND USING THE SAME
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of the filing date of U.S. Provisional Patent Application No. 62/060,484, filed October 6, 2014; and U.S. Provisional Patent Application Nos 62/206,770; 62/206,771; and 62/206,772 filed August 18, 2015. The foregoing are incorporated by reference in their entireties.
FIELD OF THE INVENTION
This disclosure relates to novel compositions of antibodies and carrier proteins and methods of making and using the same, in particular, as a cancer therapeutic.
STATE OF THE ART
Chemotherapy remains a mainstay for systemic therapy for many types of cancer, including melanoma. Most chemotherapeutics are only slightly sélective to tumor cells, and toxicity to healthy proliferating cells can be high (Allen TM. (2002) Cancer 2:750-763), often requiring dose réduction and even discontinuation of treatment. In theory, one way to overcome chemotherapy toxicity issues as well as improve drug efficacy is to target the chemotherapy drug to the tumor using antibodies that are spécifie for proteins selectively expressed (or overexpressed) by tumors cells to attract targeted drugs to the tumor, thereby altering the biodistribution of the chemotherapy and resulting in more drug going to the tumor and less affecting healthy tissue. Despite 30 years of research, however, spécifie targeting rarely succeeds in the therapeutic context.
Conventional antibody dépendent chemotherapy (ADC) is designed with a toxic agent linked to a targeting antibody via a synthetic protease-cleavable linker. The efficacy of such ADC therapy is dépendent on the ability of the target cell to bind to the antibody, the linker to be cleaved, and the uptake of the toxic agent into the target cell. Schrama, D. et al. (2006) Nature reviews. Drug discovery 5:147-159.
Antibody-targeted chemotherapy promised advantages over conventional therapy because it provides combinations of targeting ability, multiple cytotoxic agents, and improved therapeutic capacity with potentially less toxicity. Despite extensive research, clinically effective antibodytargeted chemotherapy remains elusive: major hurdles include the instability of the linkers between the antibody and chemotherapy drug, reduced tumor toxicity of the chemotherapeutic agent when bound to the antibody, and the inability of the conjugate to bind and enter tumor cells. In addition, these thérapies did not allow for control over the size of the antibody-drug conjugates.
There remains a need in the art for antibody-based cancer therapeutics that retain cytoxic effect for targeted drug delivery to provide reliable and improved anti-tumor efficacy over prior therapeutics. In addition, as to any therapeutic application, there also remains a need for the composition to be stable in its physical, chemical and biological properties.
Lyophilization, or freeze drying, removes water from a composition. In the process, the material to be dried is first frozen and then the ice or frozen solvent is removed by sublimation in a vacuum environment. An excipient may be included in pre-lyophilized formulations to enhance stability during the freeze-drying process and/or to improve stability of the lyophilized product upon storage. Pikal, M. Biopharm. 3(9)26-30 (1990) and Arakawa et al., Pharm. Res. 8(3):285-291 (1991). While proteins may be lyophilized, the process of lyophilization and reconstitution may affect the properties of the protein. Because proteins are larger and more complex than traditional organic and inorganic drugs (i.e. possessing multiple functional groups in addition to complex three-dimensional structures), the formulation of such proteins poses spécial problems. For a protein to remain biologically active, a formulation must preserve intact the conformational integrity of at least a core sequence of the protein’s amino acids while at the same time protecting the protein’s multiple functional groups from dégradation. Dégradation pathways for proteins can involve chemical instability (i.e. any process which involves modification of the protein by bond formation or cleavage resulting in a new chemical entity) or physical instability (i.e. changes in the higher order structure of the protein). Chemical instability can resuit from deamidation, racemization, hydrolysis, oxidation, beta élimination or disulfide exchange. Physical instability can resuit from dénaturation, aggregation, précipitation or adsorption, for example. The three most common protein dégradation pathways are protein aggregation, deamidation and oxidation. Cleland, et al., Critical Reviews in Therapeutic Drug Carrier Systems 10(4): 307-377 (1993).
In the présent invention, the composition comprises nanoparticles which contain (a) carrier protein (b) antibody and (c) optionally a therapeutic agent. The antibody is believed to be bound to the carrier protein through hydrophobie interactions which, by their nature, are weak. The lyophilization and reconstitution of such a composition must, therefore, not only preserve the activity of the individual components, but also their relative relationship in nanoparticle.
Further challenges are imposed because the nanoparticles are used in therapy.
For example, rearrangement of the hydrophobie components in the nanoparticle may be mitigated through covalent bonds between the components. However, such covalent bonds pose challenges for the therapeutic use of nanoparticles in cancer treatment. The antibody, carrier protein, and additional therapeutic agent typically act at different locations in a tumor and through different 2 mechanisms. Non-covalent bonds permit the components of the nanoparticle to dissociate at the tumor. Thus, while a covalent bond may be advantageous for lyophilization, it may be disadvantageous for therapeutic use.
The size of the nanoparticles, and the distribution ofthe size, is also important. The nanoparticles of the invention may behave differently according to their size. At large sizes, the nanoparticles or the agglomération of these particles may block blood vessels either of which can affect the performance and safety of the composition.
Finally, cryoprotectants and agents that assist in the lyophilization process must be safe and tolerated for therapeutic use.
SUMMARY
In one aspect, provided herein are nanoparticle compositions comprising nanoparticles wherein each of the nanoparticles comprises a carrier protein, between about 100 to about 1000 antibodies, and optionally at least one therapeutic agent, wherein the antibodies are arranged outward from the surface of the nanoparticles and wherein the nanoparticles are capable of binding to a predetermined epitope in vivo.
When administered intravenously, large particles (e.g. greater than lpm) are typically disfavored because they can become lodged in the microvasculature of the lungs. At the same time, larger particles can accumulate in the tumor or spécifie organs. See e.g. 20-60 micron glass particle that is used to inject into the hepatic artery feeding a tumor of the liver, called “therasphere” (in clinical use for liver cancer).
Therefore, for intravenous administration, particles under lpm are used. Particles over lpm are, more typically, administered directly into a tumor (“direct injection”) or into an artery feeding into the site of the tumor.
In another aspect, provided herein are nanoparticle compositions comprising nanoparticles wherein each of the nanoparticles comprises a carrier protein that is not albumin, between about 100 to about 100 antibodies, preferably about 400 to about 800 antibodies, and optionally at least one therapeutic agent, wherein the antibodies are arranged on an outside surface of the nanoparticles and wherein the nanoparticles are capable of binding to a predetermined epitope in vivo. When nanoparticles multimerize, the number of antibodies is increased proportionally. For example, if a 160 nm nanoparticle contains 400 antibodies, a 320 nm dimer contains about 800 antibodies.
In another aspect, provided herein are nanoparticle compositions comprising nanoparticles, wherein each of the nanoparticles comprises carrier protein, between about 400 to about 800 antibodies, and optionally at least one therapeutic agent that is not paclitaxel, wherein the antibodies are arranged on 3 a surface of the nanoparticles such that the binding portion of the antibody is directed outward from that surface and wherein the nanoparticles are capable of binding to a predetermined epitope in vivo. In other embodiments, the nanoparticles multimerize, e.g. dimerize. Multimerization may be observed as multiples of the weight or size of the unit molécule, e.g. 160 nm particles multimerize to about 320 nm, 480 nm, 640 nm, etc. In some embodiments, less than 20% of the population are multimers. In some embodiments, more than 80% of the population are multimers.
In one embodiment, the weight ratio of camer-bound drug to antibody (e.g. albumin-bound paclitaxel to bevacizumab) is between about 5:1 to about 1:1. In one embodiment, the weight ratio of carrier-bound drug to antibody is about 10:4. In one embodiment, the antibody is a substantially single layer of antibodies on ail or part of the surface of the nanoparticle. In one embodiment, less than 0.01% of nanoparticles in the composition hâve a size selected from the group consisting of greater than 200 nm, greater than 300 nm, greater than 400 nm, greater than 500 nm, greater than 600 nm, greater than 700 nm and greater than 800 nm. Larger sizes are believed to be the resuit of multimerization of several nanoparticles, each comprising a core and antibody coating on ail or part of the surface of each nanoparticle. .
The invention further includes lyophilized compositions, and lyophilized compositions that do not materially differ from, or are the same as, the properties of freshly-prepared nanoparticles. In particular, the lypholized composition, upon resuspending in aqueous solution, is similar or identical to the fresh composition in terms of particle size, particle size distribution, toxicity for cancer cells, antibody affinity, and antibody specificity. The invention is directed to the surprising finding that lyophilized nanoparticles retain the properties of freshly-made nanoparticles notwithstanding the presence of two different protein components in these particles.
In one aspect, this invention relates to a lyophilized nanoparticle composition comprising nanoparticles, wherein each of the nanoparticles comprises a carrrier-bound drug core and an amount of antibody arranged on a surface of the core such that the binding portion of the antibody is directed outward from that surface, wherein the antibodies retain their association with the outside surface of the nanoparticle upon reconstitution with an aqueous solution. In one embodiment, the lyophilized composition is stable at room température for at least 3 months. In one embodiment, the reconstituted nanoparticles retain the activity of the therapeutic agent and are capable of binding to the target in vivo.
In one embodiment, the average reconstituted nanoparticle size is from about 130 nm to about 1 pm. In a preferred embodiment, the average reconstituted nanoparticle size is from about 130 nm to about 200 nm, and more preferably about 160 nm. In one embodiment, in the average reconstituted 4 nanoparticle size is from greater than 800 nm to about 3.5 gm, comprising multimers of smaller nanoparticles, e.g. multimers of 100-200 nm nanoparticles. In one embodiment, the weight ratio of core to antibody is from greater than 1:1 to about 1:3.
In one aspect, this disclosure relates to a lyophilized nanoparticle composition comprising nanoparticles, wherein each of the nanoparticles comprises: (a) an albumin-bound paclitaxel core and (b) between about 400 to about 800 molécules of bevacizumab arranged on a surface of the albumin-bound paclitaxel core such that the binding portion of the antibody is directed outward from that surface, wherein the antibodies retain their association with the surface of the nanoparticle upon reconstitution with an aqueous solution, provided that said lyophilized composition is stable at about 20 °C to about 25 °C for at least 3 months and the reconstituted nanoparticles are capable of binding to VEGF in vivo.
In other aspects, this disclosure relates to a lyophilized nanoparticle composition comprising nanoparticles, wherein each of the nanopaiticles comprises: (a) an albumin-bound paclitaxel core and (b) an amount of bevacizumab arranged on a surface of the albumin-bound paclitaxel core such that the binding portion of the antibody is directed outward from that surface, wherein the antibodies retain their association with the surface of the nanoparticle upon reconstitution with an aqueous solution, provided that said lyophilized composition is stable at about 20 °C to about 25 °C for at least 3 months and the reconstituted nanoparticles are capable of binding to VEGF in vivo, and further wherein the average reconstituted nanopaiticle size is not substantially different from the particle size of the freshly prepared nanoparticles. In some embodiments, the particle sizes are between 200 and 800 nm, including 200, 300, 400, 500, 600, 700 or 800nm. In other embodiments, the particles are larger, e.g. from greater than 800 nm to about 3.5 gm. In some embodiments, the particles are multimers of nanoparticles.
In some embodiments, the weight ratio of albumin-bound paclitaxel to bevacizumab is between about 5:1 to about 1:1. In other embodiments, the weight ratio of albumin-bound paclitaxel to bevacizumab is about 10:4. In further embodiments, the weight ratio of albumin-bound paclitaxel to bevacizumab is from greater than 1:1 to about 1:3.
In some embodiments, the core is albumin-bound paclitaxel, and the antibodies are selected from antibodies that selectively recognize VEGF (e.g. bevacizumab/Avastin), antibodies that selectively recognize CD20 (e.g. rituximab/Rituxin) and antibodies that selectively recognize Her2 (Trastuzumab/Herceptin).
In some embodiments, the at least one therapeutic agent is located inside the nanoparticle. In other embodiments, the at least one therapeutic agent is located on the outside surface of the nanoparticle.
In yet other embodiments, the at least one therapeutic agent is located inside the nanoparticle and on the outside surface of the nanoparticle.
In some embodiments, the nanoparticle contains more than one type of therapeutic agent. For example, a taxane and a platinum drug, e.g. paclitaxel and cisplatin.
In some embodiments, the antibodies are selected from the group consisting of ado-trastuzumab emtansine, alemtuzumab, bevacizumab, cetuximab, denosumab, dinutuximab, ipilimumab, nivolumab, obinutuzumab, ofatumumab, panitumumab, pembrolizumab, pertuzumab, rituximab, and trastuzumab. In some embodiments, the antibodies are a substantially single layer of antibodies on ail or part of the surface of the nanoparticle.
In further embodiments, the antibodies are less glycosylated than normally found in naturel human antibodies. Such glycosylation can be influenced by e.g. the expression system, or the presence of glycosylation inhibitors during expression. In some embodiments, the glycosylation status of an antibody is altered through enzymatic or chemical action.
In some embodiments, the at least one therapeutic agent is selected from the group consisting of abiraterone, bendamustine, bortezomib, carboplatin, cabazitaxel, cisplatin, chlorambucil, dasatinib, docetaxel, doxorubicin, epirubicin, erlotinib, etoposide, everolimus, gefïtinib, idarubicin, imatinib, hydroxyurea, imatinib, lapatinib, leuprorelin, melphalan, methotrexate, mitoxantrone, nedaplatin, nilotinib, oxaliplatin, paclitaxel, pazopanib, pemetrexed, picoplatin, romidepsin, satraplatin, sorafenib, vemurafenib, sunitinib, teniposide, triplatin, Vinblastine, vinorelbine, vincristine, and cyclophosphamide.
In some emobodiments, the nanoparticle fùrther comprises at least one additional therapeutic agent that is not paclitaxel or bevacizumab.
In some embodiments, the antibodies, carrier protein and, when présent, therapeutic agent, are bound through non-covalent bonds.
In some embodiments, the carrier protein is selected from the group consisting of gelatin, elastin, gliadin, legumin, zein, a soy protein, a milk protein, and a whey protein. In other embodiments, the carrier protein is albumin, for example, human sérum albumin.
In some embodiments, the composition is formulated for intravenous delivery. In other embodiments, the composition is formulated for direct injection or perfusion into a tumor.
In some embodiments, the average nanoparticle size in the composition is from greater than 800 nm to about 3.5 pm.
In some embodiments, the nanoparticles hâve a dissociation constant between about 1 x 10'11 M and about 1 x ΙΟ'9 M.
In another aspect, provided herein are methods of making nanoparticle compositions, wherein said methods comprise contacting the carrier protein and the optionally at least one therapeutic agent with the antibodies in a solution having a pH of between 5.0 and 7.5 and a température between about 5°C and about 60°C, between about 23°C and about 60°C, or between about 55°C and about 60°C under conditions and ratios of components that will allow for formation of the desired nanoparticles. In one embodiment, the nanoparticle is made at 55-60°C and pH 7.0. In another aspect, provided herein are methods of making the nanoparticle compositions, wherein said method comprises (a) contacting the carrier protein and optionally the at least one therapeutic agent to form a core and (b) contacting the core with the antibodies in a solution having a pH of about 5.0 to about
7.5 at a température between about 5°C and about 60°C, between about 23°C and about 60°C, or between about 55°C and about 60°C under conditions and ratios of components that will allow for formation of the desired nanoparticles.
The amount of components (e.g., carrier protein, antibodies, therapeutic agents, combinations thereof) is controlled in order to provide for formation of the desired nanoporaticles. A composistion wherein the amount of components is too dilute will not form the nanoparticles as desirbed herein. In a prefered embodiment, weight ratio of carrier protein to antibody is 10:4. In some embodiments, the amount of carrier protein is between about 1 mg/mL and about 100 mg/mL. In some embodiments, the amount of antibody is between about 1 mg/mL and about 30 mg/mL. For example, in some embodiments, the ratio of carrier protein:antibody:solution is approximately 9 mg of carrier protein (e.g., albumin) to 4 mg of antibody (e.g., BEV) in 1 mL of solution (e.g., saline). An amount of therapeutic agent (e.g., taxol) can also be added to the carrier protein.
In further embodiments, the nanoparticles are made as above, and then lyophilized.
In another aspect, provided herein are methods for treating a cancer cell, the method comprising contacting the cell with an effective amount of a nanoparticle composition disclosed herein to treat the cancer cell.
In another aspect, provided herein are methods for treating a tumor in a patient in need thereof, the method comprising contacting the cell with an effective amount of a nanoparticle composition disclosed herein to treat the tumor. In some embodiments, the size of the tumor is reduced. In other embodiments, the nanoparticle composition is administered intravenously. In yet other embodiments, the nanoparticle composition is administered by direct injection or perfusion into the tumor.
In some embodiments, the methods provided herein include the steps of: a) administering the nanoparticle composition once a week for three weeks; b) ceasing administration ofthe nanoparticle composition for one week; and c) repeating steps a) and b) as necessary to treat the tumor.
In related embodiments, the treatment comprises administration of the targeting antibody prior to administration of the nanoparticles. In one embodiment, the targeting antibody is administered between about 6 and 48, or 12 and 48 hours prior to administration ofthe nanoparticles. In another embodiment, the targeting antibody is administered between 6 and 12 hours prior to administration of the nanoparticles. In yet another embodiment, the targeting antibody is administered between 2 and 8 hours prior to administration of the nanoparticles. In still other embodiments, the targeting antibody is administered a week prior to administration of the nanoparticles. For example, administration of a dose of BEV 24 hours prior to administration of AB 160. In another example, prior administration of rituximab prior to administering AR nanoparticles. The antibody administered prior to the nanoparticle may be administered as a dose that is subtherapeutic, such as 1/2, 1/10Λ or 1/20 the amount normally considered therapeutic. Thus, in man, pretreatment with BEV may comprise administration of lmg/kg BEV which is 1/10th the ususual dose, followed by administration of AB 160.
In some embodiments, the therapeutically effective amount comprises about 75 mg/m2 to about 175 mg/m2 of the carrier protein (i.e., milligrams carrier protein per m2 of the patient). In other embodiments, the therapeutically effective amount comprises about 75 mg/m2 to about 175 mg/m2 of therapeutic agent (e.g., paclitaxel). In other embodiments, the therapeutically effective amount comprises about 30 mg/m2 to about 70 mg/m2 of the antibody. In yet other embodiments, the therapeutically effective amount comprises about 30 mg/m2 to about 70 mg/m2 bevacizumab.
In one spécifie embodiment, the lypholized composition comprises from about 75 mg/m2 to about 175 mg/m2 of the carrier protein which is preferably albumin; from about 30 mg/m2 to about 70 mg/m2 of the antibody which is preferably bevacizumab; and from about about 75 mg/m2 to about 175 mg/m of paclitaxel.
BRIEF DESCRIPTION OF THE DRAWINGS
The following figures are représentative only of the invention and are not intended as a limitation. For the sake of consistency, the nanoparticles of this invention using Abraxane® and bevacizumab employ the acronym “AB” and the number after AB such as AB 160 is meant to confer the average particle size of these nanoparticles (in nanometers). Likewise, when the antibody is rituximab, the acronym is “AR” while the number thereafter remains the same.
FIG. IA shows flow cytometry scatterplots including: Abraxane® (ABX - commercially available from Celgene Corporation, Summit, NJ 07901) stained with secondary antibody only (top left panel), ABX stained with goat anti-mouse IgGl Fab plus secondary antibody (top right panel), AB 160 (which is a nanoparticle of albumin-bound paclitaxel to bevacizumab in a ratio of about 10:4 and hâve an average particle size of 160 nm) stained with secondary antibody only (bottom left panel), or AB 160 stained with goat anti-mouse IgGl Fab plus secondary antibody (bottom right panel).
FIG. IB shows a représentative électron micrograph after incubation of AB 160 with gold partielelabeled anti-human IgG Fc.
FIG. IC shows a pie chart (top) indicating the percentages of total paclitaxel in AB 160 fractions (particulate, proteins greater than 100 kD and proteins less than 100 kD); and a Western blot with antibodies against mouse IgG Fab (BEV) and paclitaxel to verify co-localization (bottom).
FIG. ID represents the activity of paclitaxel in an in vitro toxicity assay with A375 human melanoma cells, compared to ABX alone. The results are represented by the average (+/- SEM) prolifération index, which is the percentage of total prolifération of untreated cells. This data represents 3 experiments and différences were not significant.
FIG. 1E represents results from a VEGF ELISA of supematant after co-incubation of VEGF with ABX and AB 160 to détermine binding of the ligand, VEGF, by the antibody. The results are shown as the average percentage +/- SEM of VEGF that was unbound by the 2 complexes. The data represents 3 experiments ** P< 0.005.
FIG. 2A shows the size of the complexes (determined by light scattering technology) formed by adding BEV (bevacizumab) to ABX under conditions where nanoparticles and higher are formed. Increasing concentrations of BEV (0-25 mg) were added to 10 mg of ABX and the size of the complexes formed was determined. The average size of the complexes (146 nm to 2,166 nm ) increased as the concentration of BEV was increased. The data is displayed as volume of sample/size and graphs show the size distribution of the particles. This data is représentative of 5 separate drug préparations. As a comparison, ABX, by itself, has an average particle size of about 130 nm.
FIG. 2B shows affinity of the binding of ABX and BEV (as determined by light absorption (BLItz) technology). The data is displayed as dissociation constant (Kd). The binding affinity of particles made at four pH levels (3, 5, 7, 9) and 3 températures (RT, 37°C and 58°C) was assessed, and the data are représentative of 5 experiments.
FIG. 2C shows the stability of the nanoparticle complexes from Figure 2B in sérum as determined by a nanoparticle tracking analysis (NTA) on Nanosight 300 (NS300). The data are displayed as the number of particles/mg of ABX and compares AB 160 prepared at RT and pH 7 (AB 16007; particle size, pH), 58 °C and pH 7 (AB1600758; particle size, pH, température) and 58 °C and pH 5 (AB 1600558; particle size, pH, température), relative to ABX alone under each condition. Once particles were prepared, they were added to human AB sérum for 15, 30, and 60 minutes to détermine stability in sérum over time.
FIG. 3A shows in vivo testing of AB nanoparticles in athymie nude mice injected with 1 x 106 A375 human melanoma cells in the right flank and treated with PBS, 12 mg/kg BEV, 30 mg/kg ABX, 12 mg/kg BEV + 30 mg/kg ABX, or AB 160 (having about 12 mg/kg BEV and about 30 mg/kg ABX) at tumor size between approximately 600 mm3 to 900 mm3. Data is represented at day 7-post treatment as the percent change in tumor size from baseline (the size of the tumor on the day of treatment). Student’s t-test was used to détermine significance. The p-values for the AB particles were ail significantly different than PBS, the individual drugs alone and the 2 drugs given sequentially.
FIG. 3B shows Kaplan-Meier curves generated for médian survival of the mice analyzed in FIG. 3 A. Significance was determined using the Mantle-Cox test comparing survival curves.
FIG. 3C shows the percent change from baseline for mice treated when tumors were less than or o greater than 700 mm , to ascertain whether the size of the tumor affected tumor response for the ABX only and AB 160 groups. The Student’s t-test was used to détermine significance; the ABX only groups showed no significant différence (p = 0.752) based on tumor size, while the AB 160 groups were significantly different (p = 0.0057).
FIG. 3D shows in vivo testing of AB nanoparticles in athymie nude mice injected with 1 x ΙΟ6 A375 human melanoma cells in the right flank and treated with PBS, 30 mg/kg ABX, or 45 mg/kg BEV and AB 160, AB580 (nanoparticle of albumin-bound paclitaxel to bevacizumab having an average particle size of 580 nm) or AB 1130 (nanoparticle of albumin-bound paclitaxel to bevacizumab o having an average particle size of 1130 nm) at tumor size between approximately 600 mm to 900 mm3. Data is represented at day 7-post treatment as the percent change in tumor size from baseline (the size of the tumor on the day of treatment). Student’s t-test was used to détermine significance. The p-values for the AB particles were ail significantly different than PBS, the individual drugs alone and the 2 drugs given sequentially. The différence among the AB particles of different sizes was not significant.
FIG. 3E shows Kaplan-Meier curves generated for médian survival of the mice analyzed in FIG. 3D. Significance was determined using the Mantle-Cox test comparing survival curves.
FIG. 4A demonstrates blood paclitaxel concentration displayed in line graph with y-axis in log scale, based on blood and tumor samples taken from non-tumor and tumor bearing mice at 0-24 hours after IV injection with 30 mg/kg of paclitaxel in the context of ABX or AB 160 and measured by LC-MS. Mice were IV injected at time 0, with blood samples taken and the mice sacrificed at time points of 0, 4, 8, 12, and 24 hours. There were at least 3 mice per time point. Student’s t-test was utilized to détermine if any différences in concentrations between ABX and AB 160 were significant.
FIG. 4B demonstrates the blood paclitaxel concentration from FIG. 4A, displayed in line graph with y-axis in numeric scale.
FIG. 4C shows the Cmax, half-life and AUC values calculated from the blood concentration data provide in FIGs 4A and 4B.
FIG. 4D demonstrates blood paclitaxel concentration displayed in line graph with y-axis in log scale from a second phannacokinetic experiment using earlier time points (2 to 8 hours).
FIG. 4E demonstrates the blood paclitaxel concentration from FIG. 4D, displayed in line graph with y-axis in numeric scale.
FIG. 4F shows blood paclitaxel concentration in mice in which the tumors were allowed to grow to a larger size before ABX and AB 160 injections.
FIG. 4G shows the Cjnax and the AUC calculated from the data in FIG. 4F.
FIG. 4H shows paclitaxel concentrations in the tumors from the second mouse experiment as determined by LC-MS. Data are displayed as pg of paclitaxel/mg of tumor tissue. Student’s t-test was utilized to détermine if différences were significant.
FIG. 41 shows 1-125 radioactivity levels in mice treated with AB 160 relative to ABX alone.
FIG. 4 J shows a graphical represenatation of the 1-125 radioactivity levels shown in FIG. 4L
FIG. 5A shows particle size measurements and affînity of nanoparticles made with rituximab. 10 mg/ml of ABX was incubated with rituximab (RIT) at 0-10 mg/ml and light scatter technology (Mastersizer 2000) was used to détermine resulting particle sizes. Data are displayed as the percent volume of particles at each size and the curves represent particle size distributions (top). The table (bottom) shows the sizes of the resulting particles at each concentration of antibody.
FIG. 5B shows particle size measurements and affînity of nanoparticles made with trastuzumab. 10 mg/ml of ABX was incubated with trastuzumab (HER) at 0-22 mg/ml and light scatter technology (Mastersizer 2000) was used to détermine resulting particle sizes. Data are displayed as the percent 11 volume of particles at each size and the curves represent particle size distributions (top). The table (bottom) shows the sizes of the resulting particles at each concentration of antibody.
FIG. 5C shows the binding affinity of rituximab and trastuzumab as compared to ABX at pH 3, 5, 7 and 9, determined by biolayer interferometry (BLItz) technology. The dissociation constants are displayed for each interaction.
FIG. 6A shows in vitro toxicity of AR160 as tested with the CD20-positive Daudi human lymphoma cell line. The data are displayed in a graph of the prolifération index, which is the percent of FITC positive cells in treated wells relative to FITC positive cells in the untreated well (the highest level of prolifération).
FIG. 6B shows in vivo tumor efficacy in athymie nude mice injected with 5 x 106 Daudi human lymphoma cells in the right flank. The tumors were allowed to grow to 600 mm3 to 900 mm3 and the mice were treated with PBS, 30 mg/kg ABX, 12 mg/kg rituximab, 12 mg/kg rituximab + 30 mg/kg ABX, or AR160. Tumor response was determined at day 7 post-treatment by the percent change in tumor size from the first day of treatment. Significance was determined by Student’s ttest; the percent change from baseline was significantly different between the AR160 treated mice and ail other groups (p <0.0001).
FIG. 6C shows Kaplan-Meier survival curves generated from the experiment shown in FIG. 6B. Médian survival for each treatment group is shown. A Mantle-Cox test was used to détermine whether survival curve différences were significant.
FIG. 7A demonstrates addition of another chemotherapy drug (cisplatin) cisplatin to AB 160. ABX (5 mg/ml) was incubated with cisplatin (0.5 mg/ml) at room température for 30 minutes and free cisplatin was measured by HPLC in the supematant after ABX particulate was removed. The quantity of free cisplatin was subtracted from the starting concentration to détermine the quantity of cisplatin that bound to the ABX. The data are displayed in a column graph, along with the starting concentration (cisplatin).
FIG. 7B shows the toxicity of cisplatin-bound ABX (AC) in a prolifération assay of A375 human melanoma cells. After 24 hours of drug exposure and EdU incoiporation, the cells were fixed, permeabilized and labeled with a FITC conjugated anti-EdU antibody. The data is displayed in a graph of the prolifération index, which is the percent of FITC positive cells in treated wells compared to FITC positive cells in the untreated well (the highest level of prolifération).
FIG. 7C shows in vivo tumor efficacy of AC (ABC complex; cisplatin-bound ABX) in athymie nude mice injected with 1 x 106 A375 human melanoma cells in the right flank. The tumors were allowed to grow to 600 mm3 to 900 mm3 and the mice were treated with PBS, 30 mg/kg ABX, 2 12 mg/kg cisplatin, AB160, 2 mg/kg cisplatin + AB160 or ABC160. Tumor response was determined at day 7 post-treatment by the percent change in tumor size from the day of treatment. Significance was determined by Student’s t-test; the percent change from baseline was significantly different between the ABC160 treated mice and PBS-, cisplatin-, or ABX-treated mice (p <0.0001). There was no significant différence between the AB 160, AB 160 + cisplatin, and ABC 160 treated groups for day 7 post-treatment percent change from baseline.
FIG. 7D shows Kaplan-Meier survival curves generated based on the experiment shown in FIG. 7C and médian survival for each treatment group is shown. A Mantle-Cox test was used to détermine whether survival curve différences were significant.
FIG. 8A shows the size distribution of AB 160 nanoparticles that were lyophilized, stored at room température for one month, and reconstituted, as compared to fresh AB 160 or ABX alone.
FIG. 8B shows the ligand (VEGF) binding ability of AB 160 nanoparticles that were lyophilized, stored at room température for one month, and reconstituted, as compared to fresh AB 160 or ABX alone.
FIG. 8C shows in vitro cancer cell toxicity of AB 160 nanoparticles that were lyophilized, stored at room température for one month, and reconstituted, as compared to fresh AB 160 or ABX alone.
FIG. 8D shows the size distribution of AB 160 nanoparticles that were lyophilized, stored at room température for ten months, and reconstituted, as compared to fresh AB 160 or ABX alone.
FIG. 8E shows the ligand (VEGF) binding ability of AB 160 nanoparticles that were lyophilized, stored at room température for ten months, and reconstituted, as compared to fresh AB 160 or ABX alone.
FIG. 8F shows in vitro cancer cell toxicity of AB 160 nanoparticles that were lyophilized, stored at room température for ten months, and reconstituted, as compared to fresh AB 160 or ABX alone. FIGs. 9A-C show the size distributions of the ABX-BEV complexes at I.V. infusion conditions (ABX final concentration of 5 mg/mL) incubated in saline at room température for up to 24 hours (FIGs. A and B). By 4 hours at room température, there is some evidence of complex breakdown by ELISA (20%, FIG. C).
FIG. 10 shows in vitro incubation for 30 seconds of ABX (top panel) or AB 160 (bottom panel) in saline or heparinized human plasma at relative volume ratios of 9:1 or 1:1.
FIGs. 11A-E show in vivo testing of athymie nude mice injected with 1 x ΙΟ6 A375 human melanoma cells in the right flank and treated with (Fig 11A) PBS, (Fig 11B) 12 mg/kg BEV, (Fig 11C) 30 mg/kg ABX, (Fig 11D) AB160, or (Fig 11E) pretreated with 01.2 mg/kg BEV and, 24hr later, AB 160. Data is represented at day 7-post and 10-day treatment as tumor volume in mm .
FIG. 11F summarizes the day 7-post treatment data from FIGs. 11A-E.
FIG. 11G summarizes the day 10-post treatment data from FIGs. 11A-E.
DETAILED DESCRIPTION
After reading this description it will become apparent to one skilled in the art how to implement the invention in various alternative embodiments and alternative applications. However, ail the various embodiments of the présent invention will not be described herein. It will be understood that the embodiments presented here are presented by way of an example only, and not limitation. As such, this detailed description of various alternative embodiments should not be construed to limit the scope or breadth of the présent invention as set forth below.
Before the présent invention is disclosed and described, it is to be understood that the aspects described below are not limited to spécifie compositions, methods of preparing such compositions, or uses thereof as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting. The detailed description of the invention is divided into various sections only for the reader’s convenience and disclosure found in any section may be combined with that in another section. Titles or subtitles may be used in the spécification for the convenience of a reader, which are not intended to influence the scope of the présent invention.
Définitions
Unless defined otheiwise, ail technical and scientific terms used herein hâve the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In this spécification and in the claims that follow, reference will be made to a number of terms that shall be defined to hâve the following meanings:
The terminology used herein is for the puipose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the singular forms a, an and the are intended to include the plural forms as well, unless the context clearly indicates otherwise. “Optional” or “optionally” means that the subsequently described event or circumstance can or cannot occur, and that the description includes instances where the event or circumstance occurs and instances where it does not.
The term “about” when used before a numerical désignation, e.g., température, time, amount, concentration, and such other, including a range, indicates approximations which may vary by ( + ) or ( - ) 10%, 5%, 1%, or any subrange or subvalue there between. Preferably, the term “about” when used with regard to a dose amount means that the dose may vary by +/- 10%. For example, “about
400 to about 800 antibodies” indicates that an outside surface of a nanoparticles contain an amount of antibody between 360 and 880 particles.
“Comprising” or “comprises” is intended to mean that the compositions and methods include the recited éléments, but not excluding others. “Consisting essentially of ’ when used to defîne compositions and methods, shall mean excluding other éléments of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the éléments as defined herein would not exclude other materials or steps that do not materially affect the basic and novel characteristic(s) of the claimed invention. “Consisting of ’ shall mean excluding more than trace éléments of other ingrédients and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this invention.
The term “nanoparticle” as used herein refers to particles having at least one dimension which is less than 5 microns. In preferred embodiments, such as for intravenous administration, the nanoparticle is less than 1 micron. For direct administration, the nanoparticle is larger. Even larger particles are expressly contemplated by the invention.
In a population of particles, the size of individual particles are distributed about a mean. Particle sizes for the population can therefore be represented by an average, and also by percentiles. D50 is the particle size below which 50% of the particles fall. 10% of particles are smaller than the D10 value and 90% of particles are smaller than D90. Where unclear, the “average” size is équivalent to D50. So, for example, AB160 refers to nanoparticles having an average size of 160 nanometers. The term “nanoparticle” may also encompass discrète multimers of smaller unit nanoparticles. For example, a 320 nm particle comprises a dimer of a unit 160 nm nanoparticle. For 160 nm nanoparticles, multimers would therefore be approximately 320 nm, 480 nm, 640 nm, 800 nm, 960 nm, 1120 nm, and so on.
The term “carrier protein” as used herein refers to proteins that function to transpoit antibodies and/or therapeutic agents. The antibodies of the présent disclosure can reversibly bind to the carrier proteins. Exemplary carrier proteins are discussed in more detail below.
The term “core” as used herein refers to central or inner portion of the nanoparticle which may be comprised of a carrier protein, a carrier protein and a therapeutic agent, or other agents or combination of agents. In some embodiments, a hydrophobie portion of the antibody may be incorporated into the core.
The term “therapeutic agent” as used herein means an agent which is therapeutically useful, e.g., an agent for the treatment, remission or atténuation of a disease state, physiological condition, symptoms, or etiological factors, or for the évaluation or diagnosis thereof. A therapeutic agent may 15 be a chemotherapeutic agent, for example, mitotic inhibitors, topoisomerase inhibitors, steroids, anti-tumor antibiotics, antimetabolites, alkylating agents, enzymes, protéasome inhibitors, or any combination thereof.
The term “antibody” or “antibodies” as used herein refers to immunoglobulin molécules and immunologically active portions of immunoglobulin molécules (i.e., molécules that contain an antigen binding site that immuno-specifically bind an antigen). The term also refers to antibodies comprised of two immunoglobulin heavy chains and two immunoglobulin light chains as well as a variety of forms including full length antibodies and portions thereof; including, for example, an immunoglobulin molécule, a monoclonal antibody, a chimeric antibody, a CDR-grafted antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody (dAb), a diabody, a multispecifîc antibody, a dual spécifie antibody, an anti-idiotypic antibody, a bispecifîc antibody, a functionally active epitope-binding fragment thereof, bifunctional hybrid antibodies (e.g., Lanzavecchia et al., Eur. J. Immunol. 17, 105 (1987)) and single chains (e.g., Huston et al., Proc. Natl. Acad. Sci. U.S.A., 85, 5879-5883 (1988) and Bird et al., Science 242, 423426 (1988), which are incoiporated herein by reference). (See, generally, Hood et al., Immunology, Benjamin, N.Y., 2ND ed. (1984); Harlow and Lane, Antibodies. A Laboratory Manual, Cold Spring Harbor Laboratory (1988); Hunkapiller and Hood, Nature, 323, 15-16 (1986), which are incorporated herein by reference). The antibody may be of any type (e.g., IgG, IgA, IgM, IgE or IgD). Preferably, the antibody is IgG. An antibody may be non-human (e.g., from mouse, goat, or any other animal), fully human, humanized, or chimeric.
The term “dissociation constant,” also referred to as Ka, refers to a quantity expressing the extent to which a particular substance séparâtes into individual components (e.g., the protein carrier, antibody, and optional therapeutic agent).
The terms “lyophilized,” “lyophilization” and the like as used herein refer to a process by which the material (e.g., nanoparticles) to be dried is first frozen and then the ice or frozen solvent is removed by sublimation in a vacuum environment. An excipient is optionally included in pre-lyophilized formulations to enhance stability of the lyophilized product upon storage. In some embodiments, the nanoparticles can be formed from lyophilized components (carrier protein, antibodiy and optional therapeutic) prior to use as a therapeutic. In other embodiments, the carrier protein, antibody, and optional therapeutic agent are first combined into nanoparticles and then lyophilized. The lyophilized sample may further contain additional excipients.
The term “bulking agents” comprise agents that provide the structure of the ffeeze-dried product. Common examples used for bulking agents include mannitol, glycine, lactose and sucrose. In addition to providing a pharmaceutically élégant cake, bulking agents may also impart useful qualifies in regard to modifying the collapse température, providing freeze-thaw protection, and enhancing the protein stability over long-term storage. These agents can also serve as tonicity modifiers.
The term “buffer” encompasses those agents which maintain the solution pH in an acceptable range prior to lyophilization and may include succinate (sodium or potassium), histidine, phosphate (sodium or potassium), Tris(tris(hydroxymethyl)aminomethane), diethanolamine, citrate (sodium) and the like. The buffer of this invention has a pH in the range from about 5.5 to about 6.5; and preferably has a pH of about 6.0. Examples of buffers that will control the pH in this range include succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers. The term “cryoprotectants” generally includes agents which provide stability to the protein against freezing-induced stresses, presumably by being preferentially excluded from the protein surface. They may also offer protection during primary and secondary drying, and long-tenu product storage. Examples are polymers such as dextran and polyethylene glycol; sugars such as sucrose, glucose, trehalose, and lactose; surfactants such as polysorbates; and amino acids such as glycine, arginine, and serine.
The term “lyoprotectant” includes agents that provide stability to the protein during the drying or ‘déhydration’ process (primary and secondary drying cycles), presumably by providing an amorphous glassy matrix and by binding with the protein through hydrogen bonding, replacing the water molécules that are removed during the drying process. This helps to maintain the protein conformation, minimize protein dégradation during the lyophilization cycle and improve the longterm products. Examples include polyols or sugars such as sucrose and trehalose.
The term “pharmaceutical formulation” refers to préparations which are in such form as to permit the active ingrédients to be effective, and which contains no additional components which are toxic to the subjects to which the formulation would be administered.
“Pharmaceutically acceptable” excipients (vehicles, additives) are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingrédient employed. “Reconstitution time” is the time that is required to rehydrate a lyophilized formulation with a solution to a particle-free clarified solution.
A “stable” formulation is one in which the protein therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage.
The term “epitope” as used herein refers to the portion of an antigen which is recognized by an antibody. Epitopes include, but are not limited to, a short amino acid sequence or peptide (optionally 17 glycosylated or otherwise modified) enabling a spécifie interaction with a protein (e.g., an antibody) or ligand. For example, an epitope may be a part of a molécule to which the antigen-binding site of an antibody attaches.
The term treating or treatment covers the treatment of a disease or disorder (e.g., cancer), in a subject, such as a human, and includes: (i) inhibiting a disease or disorder, i.e., arresting its development; (ii) relieving a disease or disorder, i.e., causing régression of the disorder; (iii) slowing progression of the disorder; and/or (iv) inhibiting, relieving, or slowing progression of one or more symptoms of the disease or disorder. In some embodiments “treating” or “treatment” refers to the killing of cancer cells.
The term “kill” with respect to a cancer treatment is directed to include any type of manipulation that will lead to the death of that cancer cell or at least of portion of a population of cancer cells. Additionally, some terms used in this spécification are more specifically defined below.
Overview
The current invention is predicated, in part, on the surprising discovery that optionally lyophilized nanoparticles comprising a carrier protein, an antibody, and a therapeutic agent provide targeted therapy to a tumor while minimizing toxicity to the patient. The nanoparticles as described herein are thus a significant improvement versus conventional ADCs.
For conventional ADCs to be effective, it is critical that the linker be stable enough not to dissociate in the systemic circulation but allow for sufficient drug release at the tumor site. Alley, S.C., et al. (2008) Bioconjug Chem 19:759-765. This has proven to be a major hurdle in developing effective drug conjugate (Julien, D.C., et al. (2011) MAbs 3:467-478; Alley, S.C., et al. (2008) Bioconjug Chem 19:759-765); therefore, an attractive feature of the nano-immune conjugate is that a biochemical linker is not required.
Another shortcoming of current ADCs is that higher drug pénétration into the tumor has not been substantively proven in human tumors. Early testing of ADCs in mouse models suggested that tumor targeting with antibodies would resuit in a higher concentration of the active agent in the tumor (Deguchi, T. et al. (1986) Cancer Res 46: 3751-3755); however, this has not correlated in the treatment of human disease, likely because human tumors are much more heterogeneous in permeability than mouse tumors. Jain, R.K. et al. (2010) Nat Rev Clin Oncol 7:653-664. Also, the size of the nanoparticle is critical for extravasation from the vasculature into the tumor. In a mouse study using a human colon adenocarcinoma xenotransplant model, the vascular pores were permeable to liposomes up to 400 nm. Yuan, F., et al. (1995) Cancer Res 55: 3752-3756. Another study of tumor pore size and permeability demonstrated that both characteristics were dépendent on 18 tumor location and growth status, with regressing tumors and cranial tumors permeable to particles less than 200 nm. Hobbs, S.K., et al. (1998) Proc NatlAcadSci USA 95:4607-4612. The nanoimmune conjugate described herein overcomes this issue by the fact that the large complex, which is less than 200 nm intact, is partially dissociated in systemic circulation into smaller functional units that are easily able to permeate tumor tissue. Furthermore, once the conjugate arrives to the tumor site, the smaller toxic payload can be released and only the toxic portion needs to be taken up by tumor cells, not the entire conjugate.
The advent of antibody- (i.e. AVASTIN®) coated albumin nanoparticles containing a therapeutic agent (i.e., ABRAXANE®) has led to a new paradigm of directional delivery of two or more therapeutic agents to a predetermined site in vivo. See PCT Patent Publication Nos. WO 2012/154861 and WO 2014/055415, each of which is incoiporated herein by reference in its entirety.
When compositions of albumin and an antibody are admixed together in an aqueous solution at spécifie concentrations and ratios, the antibodies spontaneously self-assemble into and onto the albumin to form nanoparticles having multiple copies of the antibody (up to 500 or more). Without being limited to any theory, it is contemplated that the antigen receptor portion of the antibody is positioned outward from the nanoparticle while the hydrophobie tail in integrated into the albumin by hydrophobie — hydrophobie interactions.
While protein compositions comprising a single source protein are commonly stored in lyophilized form where they exhibit significant shelf-life, such lyophilized compositions do not contain a selfassembled nanoparticle of two different proteins integrated together by hydrophobie-hydrophobie interactions. Moreover, the nanoparticle configuration wherein a majority of the antibody binding portions are exposed on the surface of the nanoparticles lends itself to being susceptible to dislodgement or reconfiguration by conditions which otheiwise would be considered benign. For example, during lyophilization, ionic charges on the proteins are dehydrated thereby exposing the underlying charges. Exposed charges allow for charge-charge interactions between the two proteins which can alter the binding affinity of each protein to the other. In addition, the concentration of the nanoparticles increases significantly as the solvent (e.g., water) is removed. Such increased concentrations of nanoparticles could lead to irréversible oligomérization. Oligomérization is a known property of proteins that reduces the biological properties of the oligomer as compared to the monomeric form and increases the size of the particle sometimes beyond 1 micron.
On the other hand, a stable form of a nanoparticle composition is required for clinical and/or commercial use where a shelf-life of at least 3 months is required and shelf-lives of greater than 6 19 months or 9 months are preferred. Such a stable composition must be readily available for intravenous injection, must retain its self-assembled form upon intravenous injection so as to direct the nanoparticle to the predetermined site in vivo, must hâve a maximum size of less than 1 micron so as to avoid any ischémie event when delivered into the blood stream, and finally must be compatible with the aqueous composition used for injection.
Compounds
As will be apparent to the skilled artisan upon reading this disclosure, the présent disclosure relates to compositions of nanoparticles containing a carrier protein, antibodies, and optionally at least one therapeutic agent, wherein said compositions are optionally lyophilized.
In some embodiments, the carrier protein can be albumin, gelatin, elastin (including topoelastin) or elastin-derived polypeptides (e.g., α-elastin and elastin-like polypeptides (ELPs)), gliadin, legumin, zein, soy protein (e.g., soy protein isolate (SPI)), milk protein (e.g., β-lactoglobulin (BLG) and casein), or whey protein (e.g., whey protein concentrâtes (WPC) and whey protein isolâtes (WPI)). In preferred embodiments, the carrier protein is albumin. In preferred embodiments, the albumin is egg white (ovalbumin), bovine seium albumin (BSA), or the like. In even more preferred embodiments, the carrier protein is human sérum albumin (HSA). In some embodiments, the carrier protein is a generally regarded as safe (GRAS) excipient approved by the United States Food and Drug Administration (FDA).
In some embodiments, the antibodies are selected from the group consisting of ado-trastuzumab emtansine, alemtuzumab, bevacizumab, cetuximab, denosumab, dinutuximab, ipilimumab, nivolumab, obinutuzumab, ofatumumab, panitumumab, pembrolizumab, pertuzumab, rituximab, and trastuzumab. In som embodiments, the antibodies are a substantially single layer of antibodies on ail or part of the surface of the nanoparticle.
Table 1 depicts a list of non-limiting list of antibodies.
Table 1: Antibodies
Antibodies
Biologie Treatment(s)/T arget(s)
Monoclonal antibodies Rituximab (Rituxan ®) Non-Hodgkin lymphoma
Antibodies
Biologie Treatment(s)/T arget(s)
(MAbs) Alemtuzumab (Campath®) Chronic lymphocytic leukemia (CLL)
Ipilimumab (Yervoy®) Metastatic melanoma
Bevacizumab (Avastin®) Colon cancer, lung cancer, rénal cancer, ovarian cancer, glioblastoma multiforme
Cetuximab (Erbitux®) Colorectal cancer, non-small cell lung cancer, head and neck cancer, cervical cancer, glioblastoma, ovarian epithelia, fallopian tube or primary peritoneal cancer, rénal cell cancer
Panitumumab (Vectibix®) Colorectal cancer
Trastuzumab (Herceptin ®) Breast cancer, Adenocarcinoma
9UY-ibritumomab tiuxetan (Zevalin ®) Non-Hodgkin lymphoma
Ado-trastuzumab emtansine (Kadycla®, also called TDM-1) Breast cancer
Brentuximab vedotin (Adcetris®) Hodgkin lymphoma, Anaplastic large cell lymphoma
Blinatumomab (Blincyto) Acute lymphocytic leukemia (ALL)
Pembrolizumab (Keytruda®) PD-1 (melanoma, non-small cell lung cancer)
Nivolumab (Opdivo®) PD-1 (melanoma, non-small cell lung cancer)
Ofatumumab (Arzerra®) Chronic lymphocytic leukemia (CLL)
Pertuzumab (Perjeta®) Breast cancer
Antibodies
Biologie Treatment(s)/T arget(s)
Obinutuzumab (Gazyva®) Lymphoma
Dinutuximab (Unituxin®) Neuroblastoma
Denosumab (Prolia®) Bone métastasés, multiple myeloma, giant cell tumor of bone
In some embodiments, the at least one therapeutic agent is selected from the group consisting of abiraterone, bendamustine, bortezomib, carboplatin, cabazitaxel, cisplatin, chlorambucil, dasatinib, docetaxel, doxorubicin, epirubicin, erlotinib, etoposide, everolimus, gefitinib, idarubicin, imatinib, 5 hydroxyurea, imatinib, lapatinib, leuprorelin, melphalan, methotrexate, mitoxantrone, nedaplatin, nilotinib, oxaliplatin, paclitaxel, pazopanib, pemetrexed, picoplatin, romidepsin, satraplatin, sorafenib, vemurafenib, sunitinib, teniposide, triplatin, Vinblastine, vinorelbine, vincristine, and cyclophosphamide.
Table 2 depicts a list of non-limiting list of cancer therapeutic agents.
Table 2: Cancer therapeutic agents
Cancer Drugs
Drug Target(s)
Abitrexate (Methotrexate) Acute lymphoblastic leukemia; breast cancer; gestational trophoblastic disease, head and neck cancer; lung cancer; mycosis fungoides; non-Hodgkin lymphoma; osteosarcoma
Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation) Breast cancer; non-small cell lung cancer; pancreatic cancer
ABVD (Adriamycin, bleomycin, Vinblastine sulfate, dacarbazine) Hodgkin lymphoma
AB VE (Adriamycin, bleomycin, vincristine sulfate, etoposide) Hodgkin lymphoma (in children)
ABVE-PC(Adriamycin, bleomycin, vincristine sulfate, etoposide, prednisone, cyclophosphamide) Hodgkin lymphoma (in children)
AC (Adriamycin cyclophosphamide) Breast cancer
AC-T (Adriamycin, cylclophosphamide, Taxol) Breast cancer
Adcetris (Brentuximab Vedotin) Anaplastic large cell lymphoma; Hodgkin lymphoma
ADE (Cytarabine (Ara-C), Daunorubicin Hydrochloride, Etoposide) Acute myeloid leukemia (in children)
Ado-Trastuzumab Emtansine Breast cancer
Adriamycin (Doxorubicin Hydrochloride) Acute lymphoblastic leukemia; acute myeloid leukemia; breast cancer, gastric (stomach) cancer; Hodgkin lymphoma; neuroblastoma; non-Hodgkin lymphoma; ovarian cancer; small cell lung cancer; soft tissue and bone sarcomas; thyroid cancer; transitional cell bladder cancer; Wilms tumor
Adrucil (Fluorouracil) Basal cell carcinoma; breast cancer; colorectal cancer; gastric (stomach) adenocarcinoma; pancreatic cancer; squamous cell carcinoma of the head and neck
Afatinib Dimaleate Non-small cell lung cancer
Afïnitor (Everolimus) Breast cancer, pancreatic cancer; rénal cell carcinoma; subependymal giant cell astrocytoma
Alimta (Pemetrexed Disodium) Malignant pleural mesothelioma; non-small cell lung cancer
Ambochlorin (Chlorambucil) Chronic lymphocytic leukemia; Hodgkin lymphoma; non-Hodgkin lymphoma
Anastrozole Breast cancer
Aredia (Pamidronate Disodium) Breast cancer; multiple myeloma
Arimidex (Anastrozole) Breast cancer
Aromasin (Exemestane) Advanced breast cancer; early-stage breast cancer and estrogen receptor positive
Arranon (Nelarabine) T-cell acute lymphoblastic leukemia; T-cell lymphoblastic lymphoma
Azacitidine Myelodysplastic syndromes
BEACOPP Hodgkin lymphoma
Becenum (Carmustine) Brain tumors; Hodgkin lymphoma; multiple myeloma; non-Hodgkin lymphoma
Beleodaq (Belinostat) Peripheral T-cell lymphoma
BEP Ovarian germ cell tumors; testicular germ cell tumors
Bicalutamide Prostate cancer
BiCNU (Carmustine) Brain tumors; Hodgkin lymphoma; multiple myeloma; non-Hodgkin lymphoma
Bleomycin Hodgkin lymphoma; non-Hodgldn lymphoma; penile cancer; squamous cell carcinoma of the cervix; squamous cell carcinoma of the head and neck; squamous cell carcinoma of the vulva; testicular cancer
Bosulif (Bosutinib) Chronic myelogenous leukemia
Brentuximab Vedotin Anaplastic large cell lymphoma; Hodgkin lymphoma
Busulfan Chronic myelogenous leukemia
Busulfex (Busulfan) Chronic myelogenous leukemia
Cabozantinib-S-Malate Medullary thyroid cancer
CAF Breast cancer
Camptosar (Irinotecan Hydrochloride) Colorectal cancer
CAPOX Colorectal cancer
Carfilzomib Multiple myeloma
Casodex (Bicalutamide) Prostate cancer
CeeNU (Lomustine) Brain tumors; Hodgkin lymphoma
Ceritinib Non-small cell lung cancer
Cerubidine (Daunorubicin Hydrochloride) Acute lymphoblastic leukemia; acute myeloid leukemia
Chlorambucil Chronic lymphocytic leukemia; Hodgkin lymphoma; non-Hodgkin lymphoma
CHLORAMBUCIL-PREDNISONE Chronic lymphocytic leukemia
CHOP Non-Hodgkin lymphoma
Cisplatin Bladder cancer; cervical cancer; malignant mesothelioma; non-small cell lung cancer; ovarian cancer; squamous cell carcinoma of the head and neck; testicular cancer
Clafen (Cyclophosphamide) Acute lymphoblastic leukemia; acute myeloid leukemia; breast cancer; chronic lymphocytic leukemia; chronic myelogenous leukemia; Hodgkin lymphoma; multiple myeloma; mycosis fungoides; neuroblastoma; nonHodgkin lymphoma; ovarian cancer; retinoblastoma
Clofarex (Clofarabine) Acute lymphoblastic leukemia
CMF Breast cancer
Cometriq (Cabozantinib-S-Malate) Medullary thyroid cancer
COPP Hodgkin lymphoma; non-Hodgkin lymphoma
COPP-ABV Hodgkin lymphoma
Cosmegen (Dactinomycm) Ewing sarcoma; gestational trophoblastic disease; rhabdomyosarcoma; solid tumors; testicular cancer; Wilms tumor
CVP Non-Hodgkin lymphoma; chronic lymphocytic leukemia
Cyclophosphamide Acute lymphoblastic leukemia; acute myeloid leukemia; breast cancer; chronic lymphocytic leukemia; chronic myelogenous leukemia; Hodgkin lymphoma; multiple myeloma; mycosis fungoides; neuroblastoma; nonHodgkin lymphoma; ovarian cancer; retinoblastoma.
Cyfos (Ifosfamide) Testicular germ cell tumors
Cyramza (Ramucirumab) Adenocarcinoma; colorectal cancer; non-small cell lung cancer
Cytarabine Acute lymphoblastic leukemia; acute myeloid leukemia; chronic myelogenous leukemia; meningeal leukemia
Cytosar-U (Cytarabine) Acute lymphoblastic leukemia; acute myeloid leukemia; chronic myelogenous leukemia; meningeal leukemia
Cytoxan (Cyclophosphamide) Acute lymphoblastic leukemia; acute myeloid leukemia; breast cancer; chronic lymphocytic leukemia; chrome myelogenous leukemia; Hodgkin lymphoma; multiple myeloma; mycosis fungoides; neuroblastoma; nonHodgkin lymphoma; ovarian cancer; retinoblastoma
Dacarbazine Hodgkin lymphoma; melanoma
Dacogen (Decitabine) Myelodysplastic syndromes
Dactinomycin Ewing sarcoma; gestational trophoblastic disease; rhabdomyosarcoma; solid tumors; testicular cancer; Wilms tumor
Daunorubicin Hydrochloride Acute lymphoblastic leukemia; acute myeloid leukemia
Degarelix Prostate cancer
Denileukin Diftitox Cutaneous T-cell lymphoma
Denosumab Giant cell tumor of the bone; breast cancer, prostate cancer
DepoCyt (Liposomal Cytarabine) Lymphomatous meningitis
DepoFoam (Liposomal Cytarabine) Lymphomatous meningitis
Docetaxel Breast cancer; adenocarcinoma of the stomach or gastroesophageal junction; non-small cell lung cancer; prostate cancer; squamous cell
carcinoma of the head and neck
Doxil (Doxorubicin Hydrochloride Liposome) AIDS-related Kaposi sarcoma; multiple myeloma; ovarian cancer
Doxorubicin Hydrochloride Acute lymphoblastic leukemia; acute myeloid leukemia; breast cancer; gastric (stomach) cancer; Hodgkin lymphoma; neuroblastoma; non-Hodgkin lymphoma; ovarian cancer; small cell lung cancer; soft tissue and bone sarcomas; thyroid cancer; transitional cell bladder cancer; Wilms tumor.
Dox-SL (Doxorubicin Hydrochloride Liposome) AIDS-related Kaposi sarcoma; multiple myeloma; ovarian cancer
DTIC-Dome (Dacarbazine) Hodgkin lymphoma; melanoma
Efudex (Fluorouracil) Basal cell carcinoma; breast cancer; colorectal cancer; gastric (stomach) adenocarcinoma; pancreatic cancer; squamous cell carcinoma of the head and neck
Ellence (Epirubicin Hydrochloride) Breast cancer
Eloxatin (Oxaliplatin) Colorectal cancer; stage III colon cancer
Emend (Aprepitant) Nausea and vomiting caused by chemotherapy and nausea and vomiting after surgery
Enzalutamide Prostate cancer
Epirubicin Hydrochloride Breast cancer
EPOCH Non-Hodgkin lymphoma
Erbitux (Cetuximab) Colorectal cancer; squamous cell carcinoma of the head and neck
Eribulin Mesylate Breast cancer
Erivedge (Vismodegib) Basal cell carcinoma
Erlotinib Hydrochloride Non-small cell lung cancer; pancreatic cancer
Erwinaze (Asparaginase Erwinia chrysanthemi) Acute lymphoblastic leukemia
Etopophos (Etoposide Phosphate) Small cell lung cancer; testicular cancer
Evacet (Doxorubicin Hydrochloride Liposome) AJDS-related Kaposi sarcoma; multiple myeloma; ovarian cancer
Everolimus Breast cancer; pancreatic cancer; rénal cell carcinoma; subependymal giant cell astrocytoma
Evista (Raloxifene Hydrochloride) Breast cancei·
Exemestane Breast cancer
Fareston (Toremifene) Breast cancer
Farydak (Panobinostat) Multiple myeloma
Faslodex (Fulvestrant) Breast cancer
FEC Breast cancer
Femara (Letrozole) Breast cancer
Filgrastim Neutropenia
Fludara (Fludarabine Phosphate) Chronic lymphocytic leukemia
Fluoroplex (Fluorouracil) Basal cell carcinoma; breast cancer; colorectal cancer; gastric (stomach) adenocarcinoma; pancreatic cancer; squamous cell carcinoma of the head and neck
Folex (Methotrexate) Acute lymphoblastic leukemia; breast cancer; gestational trophoblastic disease; head and neck cancer; lung cancer; mycosis fungoides; non-Hodgkin lymphoma; osteosarcoma
FOLFIRI Colorectal cancer
FOLFIRI-BEVACIZUMAB Colorectal cancer
FOLFIRI-CETUXIMAB Colorectal cancer
FOLFIRINOX Pancreatic cancer
FOLFOX Colorectal cancer
Folotyn (Pralatrexate) Peripheral T-cell lymphoma
FU-LV Colorectal cancer; esophageal cancer; gastric cancer
Fulvestrant Breast cancer
Gefïtinib Non-small cell lung cancer
Gemcitabine Hydrochloride Breast cancer; non-small cell lung cancer; ovarian cancer; pancreatic cancer
GEMCITABINE-CISPLATIN Biliary tract cancer; bladder cancer; cervical cancer; malignant mesothelioma; non-small cell lung cancer; ovarian cancer; pancreatic cancer
GEMCITABINE-OXALIPLATIN Pancreatic cancer
Gemtuzumab Ozogamicin (antibody drug conjugate) Acute myeloid leukemia
Gemzar (Gemcitabine Hydrochloride) Breast cancer; non-small cell lung cancer; ovarian cancer; pancreatic cancer
Gilotrif (Afatinib Dimaleate) Non-small cell lung cancer
Gleevec (Imatinib Mesylate) Acute lymphoblastic leukemia; chronic éosinophilie leukemia or hyperéosinophilie syndrome; chronic myelogenous leukemia; dermatofibrosarcoma protuberans; gastrointestinal stromal tumor; myelodysplastic/myeloproliferative neoplasms; systemic mastocytosis.
Gliadel (Carmustine Implant) Glioblastoma multiforme; malignant glioma
Goserelin Acetate Breast cancer; prostate cancer
Halaven (Eribulin Mesylate) Breast cancer
Hycamtin (Topotecan Hydrochloride) Cervical cancer; ovarian cancer; small cell lung cancer
Hyper-CVAD Acute lymphoblastic leukemia; non-Hodgkin lymphoma
Ibrance (Palbociclib) Breast cancer
Ibrutinib Chronic lymphocytic leukemia; mantel cell lymphoma;
ICE Hodgkin lymphoma; non-Hodgkin lymphoma
Iclusig (Ponatinib Hydrochloride) Acute lymphoblastic leukemia; Chronic myelogenous leukemia
Idamycin (Idarubicin Hydrochloride) Acute myeloid leukemia
Imatinib Mesylate Acute lymphoblastic leukemia; chronic éosinophilie leukemia or hyperéosinophilie syndrome; chronic myelogenous leukemia; dermatofibrosarcoma protuberans; gastrointestinal stromal tumor; myelodysplastic/myeloproliferative neoplasms; systemic mastocytosis.
Imbruvica (Ibnitinib) Chronic lymphocytic leukemia; mantle cell lymphoma; Waldenstrôm macroglobulinemia
Inlyta (Axitinib) Rénal cell carcinoma
Iressa (Gefitinib) Non-small cell lung cancer
Irinotecan Hydrochloride Colorectal cancer
Istodax (Romidepsin) Cutaneous T-cell lymphoma
Ixempra (Ixabepilone) Breast cancer
Jevtana (Cabazitaxel) Prostate cancer
Keoxifene (Raloxifene Hydrochloride) Breast cancer
Kyprolis (Carfilzomib) Multiple myeloma
Lenvima (Lenvatinib Mesylate) Thyroid cancer
Letrozole Breast cancer
Leucovorin Calcium Colorectal cancer
Leukeran (Chlorambucil) Chronic lymphocytic leukemia; Hodgkin lymphoma; non-Hodgkin lymphoma
Leuprolide Acetate Prostate cancer
Linfolizin (Chlorambucil) Chronic lymphocytic leukemia; Hodgkin lymphoma; non-Hodgkin lymphoma
LipoDox (Doxorubicin Hydrochloride Liposome) AIDS-related Kaposi sarcoma; multiple myeloma; ovarian cancer
Lomustine Brain tumors; Hodgkin lymphoma
Lupron (Leuprolide Acetate) Prostate cancer
Lynparza (Olaparib) Ovarian cancer
Marqibo (Vincristine Sulfate Liposome) Acute lymphoblastic leukemia
Matulane (Procarbazine Hydrochloride) Hodgkin lymphoma
Mechlorethamine Hydrochloride Bronchogenic carcinoma; chronic lymphocytic leukemia; chronic myelogenous leukemia; Hodgkin lymphoma; malignant pleural effusion, malignant pericardial effusion, and malignant peritoneal effusion; mycosis fungoides; non-Hodgkin lymphoma
Megace (Megestrol Acetate) Breast cancer; endométrial cancer
Mekinist (Trametinib) Melanoma
Mercaptopurine Acute lymphoblastic leukemia
Mesnex (Mesna) Hémorrhagie cystitis
Methazolastone (Temozolomide) Anaplastic astrocytoma; glioblastoma multiforme
Mexate (Methotrexate) Acute lymphoblastic leukemia; breast cancer; gestational trophoblastic disease; head and neck cancer; lung cancer; mycosis fungoides; non-Hodgkin lymphoma; osteosarcoma
Mexate-AQ (Methotrexate) Acute lymphoblastic leukemia; breast cancer; gestational trophoblastic disease; head and neck cancer; lung cancer; mycosis fungoides; non-Hodgkin lymphoma; osteosarcoma
Mitoxantrone Hydrochloride Acute myeloid leukemia; prostate cancer
Mitozytrex (Mitomycin C) Gastric (stomach) and pancreatic adenocarcinoma
MOPP Hodgkin lymphoma
Mozobil (Plerixafor) Multiple myeloma; non-Hodgkin lymphoma
Mustargen (Mechlorethamine Hydrochloride) Bronchogenic carcinoma; chronic lymphocytic leukemia; chronic myelogenous leukemia; Hodgkin lymphoma; malignant pleural effusion, malignant pericardial effusion, and malignant peritoneal effusion; mycosis fungoides; non-Hodgkin lymphoma
Myleran (Busulfan) Chronic myelogenous leukemia
Mylotarg (Gemtuzumab Ozogamicin) Acute myeloid leukemia
Nanoparticle Paclitaxel (Paclitaxel Albuminstabilized Nanoparticle Formulation) Breast cancer; Non-small cell lung cancer; Pancreatic cancer
Navelbine (Vinorelbine Tartrate) Non-small cell lung cancer
Nelarabine T-cell acute lymphoblastic leukemia
Neosar (Cyclophosphamide) Acute lymphoblastic leukemia; Acute myeloid leukemia; Breast cancer; Chronic lymphocytic leukemia; Chronic myelogenous leukemia; Hodgkin lymphoma; Multiple myeloma; Mycosis fungoides; Neuroblastoma; NonHodgkin lymphoma; Ovarian cancer; Retinoblastoma
Nexavar (Sorafenib Tosylate) Hepatocellular carcinoma; Rénal cell carcinoma; Thyroid cancer
Nilotinib Chronic myelogenous leukemia
Nivolumab Melanoma; Squamous non-small cell lung cancer
Nolvadex (Tamoxifen Citrate) Breast cancer
Odomzo (Sonidegib) Basal cell carcinoma
OEPA Hodgkin lymphoma
OFF Pancreatic cancer
Olaparib Ovarian cancer
Oncaspar (Pegaspargase) Acute lymphoblastic leukemia
OPPA Hodgkin lymphoma
Oxaliplatin Colorectal cancer; Stage III colon cancer
Paclitaxel AIDS-related Kaposi sarcoma; Breast cancer; Non-small cell lung cancer; Ovarian cancer
Paclitaxel Albumin-stabilized Nanoparticle Formulation Breast cancer; Non-small lung cancer; Pancreatic cancer
PAD Multiple myeloma
Palbociclib Breast cancer
Pamidronate Disodium Breast cancer; Multiple myeloma
Panitumumab Colorectal cancer
Panobinostat Multiple myeloma
Paraplat (Carboplatin) Non-small cell lung cancer; Ovarian cancer
Paraplatin (Carboplatin) Non-small cell lung cancer; Ovarian cancer
Pazopanib Hydrochloride Rénal cell carcinoma; Soft tissue sarcoma
Pegaspargase Acute lymphoblastic leukemia
Pemetrexed Disodium Malignant pleural mesothelioma; Non-small cell lung cancer
Platinol (Cisplatin) Bladder cancer; Cervical cancer; Malignant mesothelioma; Non-small cell lung cancer; Ovarian cancer; Squamous cell carcinoma of the head and neck; Testicular cancer
Platinal-AQ (Cisplatin) Bladdei· cancer; Cervical cancer; Malignant mesothelioma; Non-small cell lung cancer; Ovarian cancer; Squamous cell carcinoma of the head and neck; Testicular cancer
Plerixafor Multiple myeloma; Non-Hodgkin lymphoma
Pomalidomide Multiple myeloma
Pomalyst (Pomalidomide) Multiple myeloma
Pontinib Hydrochloride Acute lymphoblastic leukemia; Chronic myelogenous leukemia
Pralatrexate Peripheral T-cell lymphoma
Prednisone Acute lymphoblastic leukemia; Chronic lymphocytic leukemia; Hodgkin lymphoma; Multiple myeloma; Non-Hodgkin lymphoma; Prostate cancer; Thymoma and thymie carcinoma
Procarbazine Hydrochloride Hodgkin lymphoma
Provenge (Sipuleucel-T) Prostate cancer
Purinethol (Mercaptopurine) Acute lymphoblastic leukemia
Radium 223 Dichloride Prostate cancer
Raloxifene Hydrochloride Breast cancer
R-CHOP Non-Hodgkin lymphoma
R-CVP Non-Hodgkin lymphoma
Regorafenib Colorectal cancer; Gastrointestinal stromal tumor
R-EPOCH B-cell non-Hodgkin lymphoma
Revlimid (Lenalidomide) Mantle cell lymphoma; Multiple myeloma; Anémia
Rheumatrex (Methotrexate) Acute lymphoblastic leukemia; Breast cancer; Gestational trophoblastic disease; Head and neck cancer; Lung cancer; Non-Hodgkin lymphoma; Osteosarcoma
Romidepsin Cutaneous T-cell lymphoma
Rubidomycin (Daunorubicin Hydrochloride) Acute lymphoblastic leukemia; Acute myeloid leukemia
Sipuleucel-T Prostate cancer
Somatuline Depot (Lanreotide Acetate) Gastroenteropancreatic neuroendocrine tumors
Sonidegib Basal cell carcinoma
Sorafenib Tosylate Hepatocellular carcinoma; Rénal cell carcinoma; Thyroid cancer
Sprycel (Dasatinib) Acute lymphoblastic leukemia; Chronic myelogenous leukemia
STANFORD V Hodgkin lymphoma
Stivarga (Regorafenib) Colorectal cancer; Gastrointestinal stromal tumor
Sunitnib Malate Gastronintestinal stromal tumor; Pancreatic cancer; Rénal cell carcinoma
Sutent (Sunitinib Malate) Gastronintestinal stromal tumor; Pancreatic cancer; Rénal cell carcinoma
Synovir (Thalidomide) Multiple myeloma
Synribo (Omacetaxine Mepesuccinate) Chronic myelogenous leukemia
TAC Breast cancer
Tafinlar (Dabrafenib) Melanoma
Tamoxifen Citrate Breast cancer
Tarabine PFS (Cytarabine) Acute lymphoblastic leukemia; Acute myeloid leukemia; Chronic myelogenous leukemia
Tarceva (Erlotinib Hydrochloride) Non-small cell lung cancer; Pancreatic cancer
Targretin (Bexarotene) Skin problème caused by cutaneous T-cell lymphoma
Tasigna (Niltinib) Chronic myelogenous leukemia
Taxol (Paclitaxel) AIDS-related Kaposi sarcoma; Breast cancer; Non-small cell lung cancer; Ovarian cancer
Taxotere (Docetaxel) Breast cancer; Adenocarcinoma; Non-small cell lung cancer; Prostate cancer; Squamous cell carcinoma of the head and neck
Temodar (Temozolomide) Anaplastic astrocytoma; Glioblastoma multiforme
Temozolomide Anaplastic astrocytoma; Glioblastoma multiforme
Thiotepa Bladder cancer; Breast cancer; Malignant pleural effusion, malignant pericardial effusion, and malignant peritoneal effusion; Ovarian cancer
Toposar (Etoposide) Small cell lung cancer; Testicular cancer
Topotecan Hydrochloride Cervical cancer; Ovarian cancer; Small cell lung cancer
Toremifene Breast cancer '
Torisel (Temsirolimus) Rénal cell carcinoma
TPF Squamous cell carcinoma of the head and neck; Gastric (stomach) cancer
Trastuzumab Adenocarcinoma; Breast cancer
Treanda (Bendamustine Hydrochloride) B-cell non-Hodgkin lymphoma; Chronic lymphocytic leukemia
Trisenox (Arsenic Trioxide) Acute promyelocytic leukemia
Tykerb (Lapatinib Ditosylate) Breast cancer
Vandetabib Medullary thyroid cancer
VAMP Hodgkin lymphoma
VelP Ovarian germ cell; Testicular cancer
Velban (Vinblastine Sulfate) Breast cancer; Choriocarcinoma; Hodgkin lymphoma; Kaposi sarcoma; Mycosid fungoides; Non-Hodgkin lymphoma; Testicular cancer
Velcade (Bortezomib) Mulitple myeloma; Mantle cell lymphoma
Velsar (Vinblastine Sulfate) Breast cancer; Choriocarcinoma; Hodgkin lymphoma; Kaposi sarcoma; Mycosis fungoides; Non-Hodgkin lymphoma; Testicular cancer
VePesid (Etoposide) Small cell lung cancer; Testicular cancer
Viadur (Leuprolide Acetate) Prostate cancer
Vidaza (Azacitidine) Myelodysplastic syndromes
Vincasar PFS (Vincristine Sulfate) Acute leukemia; Hodgkin lymphoma; Neuroblastoma; Non-Hodgkin lymphoma; Rhabdomyosarcoma; Wilms tumor
Vincristine Sulfate Liposome Acute lymphoblastic leukemia
Vinorelbine Tartrate Non-small cell lung cancer
VIP Testicular cancer
Visbodegib Basal cell carcinoma
Voraxaze (Glucarpidase) Toxic blood levels of the anticancer drug methotrexate
Votrient (Pazopanib Hydrochloride) Rénal cell carcinoma; Soft tissue sarcoma
Wellcovorin (Leucovorin Calcium) Colorectal cancer; Anémia
Xalkori (Crizotinib) Non-small cell lung cancer
Xeloda (Capecitabine) Breast cancer; Colorectal cancer
XELIRI Colorectal cancer; Esophageal cancer; Gastric (stomach) cancer
XELOX Colorectal cancer
Xofîgo (Radium 223 Dichloride) Prostate cancer
Xtandi (Enzalutamide) Prostate cancer
Zaltrap (Ziv-Aflibercept) Colorectal cancer
Zelboraf (Vemurafenib) Melanoma
Ziv-Aflibercept Colorectal cancer
Zoladex (Goserelin Acetate) Breast cancer; Prostate cancer
Zolinza (Vorinostat) Cutaneous T-cell lymphoma
Zometa (Zoledronic Acid) Multiple myeloma
Zydelig (Idelalisib) Chronic lymphocytic leukemia; Non-Hodgkin lymphoma (Follicula B-cell non Hodgkin lymphoma and Small lymphocytic lymphoma)
Zykadia (Certinib) Non-small cell lung cancer
Zytiga (Abiraterone Acetate) Prostate cancer
It is to be understood that the therapeutic agent may be located inside the nanoparticle, on the outside surface of the nanoparticle, or both. The nanoparticle may contain more than one therapeutic agent, for example, two therapeutic agents, three therapeutic agents, four therapeutic agents, five therapeutic agents, or more. Furthermore, a nanoparticle may contain the same or different therapeutic agents inside and outside the nanoparticle.
In some embodiments any carrier protein, antibody, therapeutic agent, or any combination thereof is expressly excluded. For example in some embodiments a composition may comprise any carrier protein and chemotherapeutic except Abraxane® and/or any targeting antibody except bevacizumab. In one aspect, the nanoparticle comprises at least 100 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises at least 200 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises at least 300 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises at least 400 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises at least 500 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises at least 600 antibodies non-covalently bound to the surface of the nanoparticle.
In one aspect, the nanoparticle comprises between about 100 and about 1000 antibodies noncovalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises between about 200 and about 1000 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises between about 300 and about 1000 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises between about 400 and about 1000 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises between about 500 and about 1000 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises between about 600 and about 1000 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises between about 200 and about 800 antibodies non-covalently bound to the surface of the nanoparticle. In one aspect, the nanoparticle comprises between about 300 and about 800 antibodies non-covalently bound to the surface of the nanoparticle. In preferred embodiments, the nanoparticle comprises between about 400 and about 800 antibodies non-covalently bound to the surface of the nanoparticle. Contemplated values include any value or subrange within any of the recited ranges, including endpoints.
In one aspect, the average particle size in the nanoparticle composition is less than about 1 pm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 1 pm. In one aspect, the average paiticle size in the nanoparticle composition is between about 130 nm and about 900 nm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 800 nm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 700 nm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 600 nm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 500 nm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 400 nm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 300 nm. In one aspect, the average particle size in the nanoparticle composition is between about 130 nm and about 200 nm. In a preferred embodiment, the average particle size in the nanoparticle composition is between about 150 nm and about 180 nm. In an especially preferred embodiment, the mean particle size in the nanoparticle composition is about 160 nm. Contemplated values include any value, subrange, or range within any ofthe recited ranges, including endpoints.
In one aspect, the nanoparticle composition is formulated for intravenous injection. In order to avoid an ischémie event, the nanoparticle composition formulated for intravenous injection should comprise nanoparticles with an average particle size of less than about 1 pm.
In one aspect, the average particle size in the nanoparticle composition is greater than about 1 pm. In one aspect, the average particle size in the nanoparticle composition is between about 1 pm and about 5 pm. In one aspect, the average particle size in the nanoparticle composition is between about 1 pm and about 4 pm. In one aspect, the average particle size in the nanoparticle composition is between about 1 pm and about 3 pm. In one aspect, the average particle size in the nanoparticle composition is between about 1 pm and about 2 pm. In one aspect, the average particle size in the nanoparticle composition is between about 1 pm and about 1.5 pm. Contemplated values include any value, subrange, or range within any of the recited ranges, including endpoints.
In one aspect, the nanoparticle composition is formulated for direct injection into a tumor. Direct injection includes injection into or proximal to a tumor site, perfusion into a tumor, and the like. When formulated for direct injection into a tumor, the nanoparticle may comprise any average particle size. Without being bound by theory, it is believed that larger particles (e.g., greater than 500 nm, greater than 1 pm, and the like) are more likely to be immobilized within the tumor, thereby providing a bénéficiai effect. Larger paiticles can accumulate in the tumor or spécifie organs. See, e.g., 20-60 micron glass particle that is used to inject into the hepatic artery feeding a tumor ofthe liver, called “TheraSphere®” (in clinical use for liver cancer). Therefore, for intravenous administration, particles under 1 pm are typically used. Particles over 1 pm are, more typically, administered directly into a tumor (“direct injection”) or into an artery feeding into the site of the tumor.
In one aspect, less than about 0.01% of the nanoparticles within the composition hâve a particle size greater than 200 nm, greater than 300 nm, greater than 400 nm, greater than 500 nm, greater than 600 nm, greater than 700 nm, or greater than 800 nm. In one aspect, less than about 0.001% of the nanoparticles within the composition hâve a particle size greater than 200 nm, greater than 300 nm, greater than 400 nm, greater than 500 nm, greater than 600 nm, greater than 700 nm, or greater than 800 nm. In a preferred embodiment, less than about 0.01% of the nanoparticles within the composition hâve a particle size greater than 800 nm. In a more preferred embodiment, less than about 0.001% of the nanoparticles within the composition hâve a particle size greater than 800 nm. In a preferred aspect, the sizes and size ranges recited herein relate to particle sizes of the reconstituted lyophilized nanoparticle composition. That is, after the lyophilized nanoparticles are resuspended in an aqueous solution (e.g., water, other pharmaceutically acceptable excipient, buffer, etc.), the particle size or average particle size is within the range recited herein.
In one aspect, at least about 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, or 99.9% of the nanoparticles are présent in the reconstituted composition as single nanoparticles. That 39 is, fewer than about 50%, 40%, 30%, etc. of the nanoparticles are dimerized or multimerized (oligomerized).
In some embodiments, the size of the nanoparticle can be controlled by the adjusting the amount (e.g., ratio) of carrier protein to antibody. The size of the nanoparticles, and the size distribution, is also important. The nanoparticles of the invention may behave differently according to their size. At large sizes, an agglomération may block blood vessels. Therefore, agglomération of nanoparticles can affect the performance and safety of the composition. On the other hand, larger particles may be more therapeutic under certain conditions (e.g., when not administered intravenously).
In one aspect, the nanoparticle composition comprises at least one additional therapeutic agent. In one embodiment, the at least one additional therapeutic agent is non-covalently bound to the outside surface of the nanoparticle. In one embodiment, the at least one additional therapeutic agent is arranged on the outside surface of the nanoparticle. In one embodiment, the at least one additional therapeutic agent is selected from the group consisting of abiraterone, bendamustine, bortezomib, carboplatin, cabazitaxel, cisplatin, chlorambucil, dasatinib, docetaxel, doxorubicin, epirubicin, erlotinib, etoposide, everolimus, gemcitabine, gefïtinib, idarubicin, imatinib, hydroxyurea, imatinib, lapatinib, leuprorelin, melphalan, methotrexate, mitoxantrone, nedaplatin, nilotinib, oxaliplatin, pazopanib, pemetrexed, picoplatin, romidepsin, satraplatin, sorafenib, vemurafenib, sunitinib, teniposide, triplatin, Vinblastine, vinorelbine, vincristine, and cyclophosphamide. In one embodiment, the at least one additional therapeutic agent is an anti-cancer antibody.
Methods of Making Nanoparticles
In some aspects, the current invention relates to methods of making nanoparticle compositions as described herein.
In one aspect, the nanoparticles of the nanoparticle composition are formed by contacting the carrier protein or carrier protein-therapeutic agent particle with the antibody at a ratio of about 10:1 to about 10:30 carrier protein particle or carrier protein-therapeutic agent particle to antibody. In one embodiment, the ratio is about 10:2 to about 10:25. In one embodiment, the ratio is about 10:2 to about 1:1. In a preferred embodiment, the ratio is about 10:2 to about 10:6. In an especially preferred embodiment, the ratio is about 10:4. Contemplated ratios include any value, subrange, or range within any of the recited ranges, including endpoints.
In one embodiment, the amount of solution or other liquid medium employed to form the nanoparticles is particularly important. No nanoparticles are formed in an overly dilute solution of the carrier protein (or carrier protein-therapeutic agent) and the antibodies. An overly concentrated 40 solution will resuit in unstructured aggregates. In some embodiments, the amount of solution (e.g., stérile water, saline, phosphate buffered saline) employed is between about 0.5 mL of solution to about 20 mL of solution. In some embodiments, the amount of carrier protein is between about 1 mg/mL and about 100 mg/mL. In some embodiments, the amount of antibody is between about 1 mg/mL and about 30 mg/mL. For example, in some embodiments, the ratio of carrier protein:antibody:solution is approximately 9 mg of carrier protein (e.g., albumin) to 4 mg of antibody (e.g., BEV) in 1 mL of solution (e.g., saline). An amount of therapeutic agent (e.g., taxol) can also be added to the carrier protein. For example, 1 mg of taxol can be added 9 mg of carrier protein (10 mg carrier protein-therapeutic) and 4 mg of antibody in 1 mL of solution. When using a typical i.v. bag , for example, with the solution of approximately 1 liter one would need to use lOOOx the amount of carrier protein/cairier protein-therapeutic agent and antibodies compared to that used in 1 mL. Thus, one cannot form the présent nanoparticles in a standard i.v. bag. Furthermore, when the components are added to a standard i.v. bag in the therapeutic amounts of the présent invention, the components do not self-assemble to form nanoparticles.
In one embodiment, the carrier protein or carrier protein-therapeutic agent particle is contacted with the antibody in a solution having a pH between about 4 and about 8. In one embodiment, the carrier protein or carrier protein-therapeutic agent particle is contacted with the antibody in a solution having a pH of about 4. In one embodiment, the carrier protein or carrier protein-therapeutic agent particle is contacted with the antibody in a solution having a pH of about 5. In one embodiment, the carrier protein or carrier protein-therapeutic agent particle is contacted with the antibody in a solution having a pH of about 6. In one embodiment, the carrier protein or carrier proteintherapeutic agent particle is contacted with the antibody in a solution having a pH of about 7. In one embodiment, the carrier protein or carrier protein-therapeutic agent particle is contacted with the antibody in a solution having a pH of about 8. In a preferred embodiment, the carrier protein or carrier protein-therapeutic agent particle is contacted with the antibody in a solution having a pH between about 5 and about 7.
In one embodiment, the carrier protein particle or carrier protein-therapeutic agent particle is incubated with the antibody at a température of about 5 °C to about 60 °C, or any range, subrange, or value within that range including endpoints. In a preferred embodiment, the carrier protein particle or carrier protein-therapeutic agent particle is incubated with the antibody at a température of about 23 °C to about 60 °C.
Without being bound by theory, it is believed that the stability of the nanoparticles within the nanoparticle composition is, at least in part, dépendent upon the température and/or pH at which the 41 nanoparticles are formed, as well as the concentration of the components (i.e., carrier protein, antibody, and optionally therapeutic agent) in the solution. In one embodiment, the Kj of the nanoparticles is between about 1 x 10’11 M and about 2 x ΙΟ'5 M. In one embodiment, the Kj of the nanoparticles is between about 1 x 10’11 M and about 2 x 10'8 M. In one embodiment, the Ka of the nanoparticles is between about 1 x 10’11 M and about 7 x ΙΟ'9 M. In a preferred embodiment, the Ka of the nanoparticles is between about 1 x 10’11 M and about 3 x 10’8 M. Contemplated values include any value, subrange, or range within any of the recited ranges, including endpoints.
Lyophilization
The lyophilized compositions of this invention are prepared by standard lyophilization techniques with or without the presence of stabilizers, buffers, etc. Surprisingly, these conditions do not alter the relatively fragile structure of the nanoparticles. Moreover, at best, these nanoparticles retain their size distribution upon lyophilization and, more impoitantly, can be reconstituted for in vivo administration (e.g., intravenous delivery) in substantially the same form and ratios as if freshly made.
Formulations
In one aspect, the nanoparticle composition is formulated for direct injection into a tumor. Direct injection includes injection into or proximal to a tumor site, perfusion into a tumor, and the like. Because the nanopaiticle composition is not administered systemically, a nanoparticle composition is formulated for direct injection into a tumor may comprise any average particle size. Without being bound by theory, it is believed that larger particles (e.g., greater than 500 nm, greater than 1 pm, and the like) are more likely to be immobilized within the tumor, thereby providing what is believed to be a better bénéficiai effect.
In another aspect, provided herein is a composition comprising a compound provided herein, and at least one pharmaceutically acceptable excipient.
In general, the compounds provided herein can be formulated for administration to a patient by any of the accepted modes of administration. Various formulations and drug delivery Systems are available in the art. See, e.g., Gennaro, A.R., ed. (1995) Remington ’s Pharmaceutical Sciences, 18th ed., Mack Publishing Co..
In general, compounds provided herein will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parentéral (e.g., intramuscular, intravenous or subcutaneous) administration.
The compositions are comprised of, in general, a compound of the présent invention in combination with at least one pharmaceutically acceptable excipient. Acceptable excipients are non-toxic, aid 42 administration, and do not adversely affect the therapeutic benefît of the claimed compounds. Such excipient may be any solid, liquid, semi-solid or, in the case of an aérosol composition, gaseous excipient that is generally available to one of skill in the art.
Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnésium stéarate, sodium stéarate, glycerol monostearate, sodium chloride, dried skim milk and the like. Liquid and semisolid excipients may be selected from glycerol, propylene glycol, water, éthanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, minerai oil, sesame oil, etc. Preferred liquid carriers, particularly for injectable solutions, include water, saline, aqueous dextrose, and glycols. Other suitable pharmaceutical excipients and their formulations are described in Remington's Pharmaceutical Sciences, edited by E. W. Martin (Mack Publishing Company, 18th ed., 1990).
The présent compositions may, if desired, be presented in a pack or dispenser device containing one or more unit dosage forms containing the active ingrédient. Such a pack or device may, for example, comprise métal or plastic foil, such as a blister pack, or glass, and rubber stoppers such as in vials. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
Treatment Methods
The nanoparticle compositions as described herein are useful in treating cancer cells and/or tumors in a mammal. In a preferred embodiment, the mammal is a human (i.e., a human patient). Preferably, the lyophilized nanoparticle composition is reconstituted (suspended in an aqueous excipient) prior to administration.
In one aspect is provided a method for treating a cancer cell, the method comprising contacting the cell with an effective amount of nanoparticle composition as described herein to treat the cancer cell. Treatment of a cancer cell includes, without limitation, réduction in prolifération, killing the cell, preventing metastasis of the cell, and the like.
In one aspect is provided a method for treating a tumor in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a nanoparticle composition as described herein to treat the tumor. In one embodiment, the size of the tumor is reduced. In one embodiment, the tumor size does not increase (i.e. progress) for at least a period of time during and/or after treatment.
In one embodiment, the nanoparticle composition is administered intravenously. In one embodiment, the nanoparticle composition is administered directly to the tumor. In one embodiment, the nanoparticle composition is administered by direct injection or perfusion into the tumor.
In one embodiment, the method comprises:
a) administering the nanoparticle composition once a week for three weeks;
b) ceasing administration of the nanoparticle composition for one week; and
c) optionally repeating steps a) and b) as necessary to treat the tumor.
In one embodiment, the therapeutically effective amount of the nanoparticles described herein comprises about 50 mg/m2 to about 200 mg/m2 carrier protein or carrier protein and therapeutic agent. In a preferred embodiment, the therapeutically effective amount comprises about 75 mg/m2 to about 175 mg/m2 carrier protein or carrier protein and therapeutic agent. Contemplated values include any value, subrange, or range within any of the recited ranges, including endpoints.
In one embodiment, the therapeutically effective amount comprises about 20 mg/m2 to about 90 mg/m2 antibody. In a preferred embodiment, the therapeutically effective amount comprises 30 mg/m2 to about 70 mg/m2 antibody. Contemplated values include any value, subrange, or range within any of the recited ranges, including endpoints.
Cancers or tumors that can be treated by the compositions and methods described herein include, but are not limited to: biliary tract cancer; brain cancer, including glioblastomas and medulloblastomas; breast cancer; cervical cancer; choriocarcinoma; colon cancer; endométrial cancer; esophageal cancer, gastric cancer; hematological neoplasms, including acute lymphocytic and myelogenous leukemia; multiple myeloma; AIDS associated leukemias and adult T-cell leukemia lymphoma; intraepithélial neoplasms, including Bowen's disease and Paget's disease; liver cancer (hepatocarcinoma); lung cancer; lymphomas, including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer, including squamous cell carcinoma; ovarian cancer, including those arising from épithélial cells, stromal cells, germ cells and mesenchymal cells; pancréas cancer; prostate cancer; rectal cancer; sarcomas, including leiomyosarcoma, rhabdomyosarcoma, liposarcoma, fîbrosarcoma and osteosarcoma; skin cancer, including melanoma, Kaposi's sarcoma, basocellular cancer and squamous cell cancer; testicular cancer, including germinal tumors (seminoma, non-seminoma[teratomas, choriocarcinomas]), stromal tumors and germ cell tumors; thyroid cancer, including thyroid adenocarcinoma and medullar carcinoma; and rénal cancer including adenocarcinoma and Wilms tumor. In important embodiments, cancers or tumors include breast cancer, lymphoma, multiple myeloma, and melanoma.
In general, the compounds of this invention will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities. The actual amount of the compound of this invention, i.e., the nanoparticles, will dépend upon numerous factors such as the severity of the disease to be treated, the âge and relative health of the subject, the potency of the compound used, the route and form of administration, and other factors well known to the skilled artisan.
An effective amount of such agents can readily be determined by routine expérimentation, as can the most effective and convenient route of administration, and the most appropriate formulation. Various foimulations and drug delivery Systems are available in the art. See, e.g., Gennaro, A.R., ed. (1995) Remington ’s Pharmaceutical Sciences, 18th ed., Mack Publishing Co..
An effective amount or a therapeutically effective amount or dose of an agent, e.g., a compound of the invention, refers to that amount of the agent or compound that results in amelioration of symptoms or a prolongation of survival in a subject. Toxicity and therapeutic efficacy of such molécules can be determined by standard pharmaceutical procedures in cell cultures or experimental animais, e.g., by determining the LD50 (the dose léthal to 50 % of the population) and the ED50 (the dose therapeutically effective in 50 % of the population). The dose ratio of toxic to therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ ED50. Agents that exhibit high therapeutic indices are preferred.
The effective amount or therapeutically effective amount is the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. Dosages may vary within this range depending upon the dosage form employed and/or the route of administration utilized. The exact formulation, route of administration, dosage, and dosage interval should be chosen according to methods known in the art, in view of the spécifies of a subject’s condition.
Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety that are sufficient to achieve the desired effects; i.e., the minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from, for example, in vitro data and animal experiments. Dosages necessary to achieve the MEC will dépend on individual characteristics and route of administration. In cases of local administration or sélective uptake, the effective local concentration of the drug may not be related to plasma concentration.
EXAMPLES
The présent disclosure is illustrated using nanoparticles composed of albumin-bound paclitaxel (i.e., Abraxane®) or cisplatin as core, and bevacizumab (i.e., Avastin®) or Rituximab (i.e., Rituxan®) as antibodies.
One skilled in the art would understand that making and using the nanoparticles of the Examples are for the sole purpose of illustration, and that the présent disclosure is not limited by this illustration.
Any abbreviation used herein, has normal scientifïc meaning. Ail températures are °C unless otheiwise stated. Herein, the following ternis hâve the following meanings unless otheiwise defined:
ABX = Abraxane®/(albumin-b ound paclitaxel)
AC = cisplatin-bound ABX
ACN = acetonitrile
ADC = antibody dépendent chemotherapy
BEV = bevacizumab
BSA = bovine sérum albumin
dH2O = distilled water
DMEM = Dulbecco’s Modified Eagle’s Medium
nM = nano molar
EdU = 5-ethynyl-2'-deoxyuridine
EM = électron microscopy
FCB = flow cytometry buffer
FITC = Fluorescein
kD = kilo-dalton
I<d = dissociation constant
kg = kilogram
KV = kilo-volts
L/hr = liter/hour
LC-MS = liquid chromatography-mass spectrometry
M = molar
mCi = millicuries
mg = milligram
ml or mL milliliter square meters mm3 cubic millimeter
microgram microliter pm micrometer/micron
PBS pK
Phosphate buffered saline pharmacokinetics
RT room temperate rpm rotations per minute volts xg times gravity
Example 1: Nanoparticle Préparation
Abraxane (ABX) (10 mg) was suspended in bevacizumab (BEV) (4 mg [160 μΐ] unless otherwise indicated), and 840 μΐ of 0.9% saline was added to give a final concentration of 10 mg/ml and 2 mg/ml of ABX and BEV, respectively. The mixture was incubated for 30 minutes at room température (or at the température indicated) to allow particle formation. For Mastersizer experiments to measure particle size of ABX:BEV complexes, 10 mg of ABX was suspended in BEV at concentrations of 0 to 25 mg/ml. Complexes of ABX with rituximab (0-10 mg/ml) or trastuzumab (0-22 mg/ml) were formed under similar conditions.
For use in humans, the ABX:BEV complexes may be prepared by obtaining the dose appropriate number of 4 mL vials of 25 mg/mL BEV and diluting each vial per the following directions to 4 mg/mL. The dose appropriate number of 100 mg vials of ABX can be prepared by reconstituting to a final concentration containing 10 mg/mL ABX nanoparticles. Using a stérile 3 mL syringe, 1.6 mL (40 mg) of bevacizumab (25 mg/mL) can be withdrawn and slowly injected, over a minimum of 1 minute, onto the inside wall of each of the vials containing 100 mg of ABX. The bevacizumab solution should not be injected directly onto the lyophilized cake as this will resuit in foaming. Then, using a stérile 12 mL stérile syringe, 8.4 mL 0.9% Sodium Chloride Injection, USP, can be withdrawn and slowly injected, over a minimum of 1 minute, 8.4 mL onto the inside wall of each vial containing ABX 100 mg and BEV 40 mg. Once the addition of BEV 1.6 mL and 0.9% Sodium Chloride Injection, USP 8.4 mL is completed, each vial can be gently swirled and/or inverted slowly for at least 2 minutes until complété dissolution of any cake/powder occurs. Génération of foam should be avoided. At this point, the concentration of each vial should be 100 mg/10 mL ABX and 40 mg/10 mL BEV. The vials containing the ABX and BEV should sit for 60 minutes. The vial(s) should be gently swirled and/or inverted every 10 minutes to continue to mix the complex. Aftei· 60 minutes has elapsed, the calculated dosing volume of ABX and BEV should be withdrawn from each vial and slowly added to an empty viaflex bag. An equal volume of 0.9% Sodium Chloride Injection, USP is then added to make the final concentration of ABX 5 mg/mL and BEV 2 mg/mL. The bag should then be gently swirled and/or inverted slowly for 1 minute to mix. The ABX:BEV nanoparticles can be stored for up to 4 hours at room température following final diluation.
Example 2: Binding of ABX and BEV in vitro
To détermine whether ABX and BEV interact, the nanoparticles formed in Example 1 were analyzed by flow cytometry and électron microscopy.
Methods
Flow Cytometry: AB 160 was produced as described in Example 1 above. To détermine binding of BEV to ABX, visualization of AB 160 was performed on an Accuri C6 flow cytometer (BD Franklin Lakes, NJ) and data analysis was done using Accuri C6 software. Biotinylated (5pg) goat antimouse IgG (Abcam, Cambridge, MA) was labeled with 5 pg of streptavidin PE (Abcam, Cambridge, MA). The goat anti-mouse IgG was chosen to label AB160 because the Fab portion of the BEV is mouse derived. ABX and AB 160 were incubated with the PE-labeled goat anti-mouse IgG for 30 minutes at room température, washed and visualized by flow cytometery.
Electron Microscopy: Five μΐ ABX, dissolved in PBS at 6 mg/ml, was added to a 300-mesh parlodian-carbon coated copper grid and allowed to sit for 1 minute. A pointed piece of filter paper was touched to the drop to remove excess liquid, leaving a thin film on the grid. The grids were allowed to dry. To dissolve the buffer crystals left on the dried grid, the sample was washed three times in dH2O. A small drop of 1% phosphotungstic acid (PTA), pH 7.2, was added to the grid. The grid was then again touched by a pointed piece of filter paper to remove excess liquid, leaving a thin film on the grid and allowed to dry. BEV (Genentech) at 25 mg/ml in 0.9% sodium chloride solution was diluted with PBS at 1:10 ratio. Five μΐ of BEV was loaded on nickel formvar-coated grid and allowed to air dry for 30 minutes to 1 hour. For the AB 160, 10 mg/ml ABX, dissolved in PBS, and 4mg/ml BEV, in 0.9% sodium chloride solution, were mixed at 2.5:1 ratio. The complex was further diluted with PBS at 1:5. Five μΐ of the complex was loaded on nickel formvar-coated grid and air dried for 30 minutes to 1 hour. Both samples were incubated for 1 hour in goat anti48 mouse IgG with 6 nm gold-conjugated paiticles (Electron Microscopy Sciences), diluted 1:30 with 10% FCB/PBS, washed 6 times with PBS (each 2 minutes), 6 times with dhRO, then stained with the mixture of 2% methylcellulose and 4% UA (9:1) for 5 minutes. Filter paper was used to drain the stain and the grid was air dried for 1 hour. Both samples were incubated overnight in donkey anti-mouse IgG with 6 nm gold-conjugated paiticles (Jackson ImmunoResearch) diluted 1:25 with 10% FCB/PBS, washed 6 times with PBS (each 2 minutes), 6 times with dH^O water, stained with 1% PTA for 5 minutes, air dried, covered with 2% methylcellulose, and air dried for 1 hour. The micrographs were taken on a JEOL1400 at operating at 80 KV.
Results
ABX (10 mg/ml) was co-incubated with 4 mg/ml BEV in vitro and found that they formed 160 nm nanoparticles (referred to herein as AB 160). Because the Fab portion of the IgGl (BEV) is of mouse origin, particles containing BEV were selectively labeled with purified goat anti-mouse IgG followed by anti-goat PE as a secondary antibody. As a négative control, samples were stained with the anti-goat PE only. Particles were visualized by flow cytometry and demonstrated a bright signal of anti-mouse IgGl binding to AB160 (41.2% positive) relative to ABX (6.7% positive) alone (FIG. IA). To validate binding of BEV to ABX, the BEV were labeled with gold-labeled mouse antihuman IgG and the particles were visualized with électron microscopy (FIG IB). Surprisingly, the EM pictures suggest a monolayer of BEV surrounding ABX nanoparticles.
To détermine what protein (albumin or BEV) the paclitaxel remains bound to when the complex breaks down, AB 160 were made and collected fractions: the particulate (nanoAB160), proteins greater than 100 kD and proteins less than 100 kD. Paclitaxel was measured in each fraction by liquid chromatography-mass spectrometry (LC-MS). Roughly 75% of the paclitaxel remained within the particulate, and the majority of the remaining paclitaxel was associated with the fraction containing proteins 100 kD or greater (FIG. IC, top), suggesting that when the particulate dissociâtes the paclitaxel is bound to BEV alone or a BEV and albumin heterodimer. This indicates that the dissociated complexes contain the chemotherapy drug with the antibody, which would still traffic to the high-VEGF tumor microenvironment. These findings were confirmed by Western blot analysis of the supernatants from AB 160, which showed that BEV and paclitaxel co-localize at approximately 200 kD, a size consistent with a paclitaxel-BEV-albumin protein complex (FIG. IC, bottom).
Example 3: Function of AB160 in vitro
Confirmation that the two key éléments in the complexes, the antibody and the paclitaxel, retained their function when présent in the complexes was demonstrated.
Methods
In vitro toxicity: The A375 human melanoma cell line (ATCC Manassa, VA) and Daudi B-cell lymphoma line (ATCC Manassa, VA) were cultured in DMEM with 1% PSG and 10% FBS. Cells were harvested and plated at 0.75 x 106 cells per well in 24 well plates. Cells were exposed to ABX or AB 160 at paclitaxel concentrations from 0 to 200 pg/ml ovemight at 37 °C and 5% CO2. To measure prolifération, the Click-iT EdU (Molecular Probes, Eugene, OR) kit was utilized. Briefly, 10 mM EdU was added to the wells and incubated ovemight with the cells and ABX or AB 160. The cells were penneabilized with 1% saponin and intercalated EdU was labeled with a FITCconjugated antibody. The prolifération index was determined by dividing the FITC positive cells from each treatment by the maximum prolifération of untreated EdU labeled cells.
λ/EGF ELISA: To détermine whether BEV can still bind its ligand, λ/EGF, when bound to ABX, a standard VEGF ELISA (R and D Systems, Minneapolis, MN) was employed. AB 160 was prepared as described and 2000 pg/ml VEGF was added to the AB 160 complex or ABX alone. The VEGF was incubated with the nanoparticles for 2 hours at room température. The suspension was spun at 6000 rpm for 15 minutes, supematants were collected and free VEGF was measured by ELISA. Briefly, ELISA plates were coated with capture antibody ovemight at 4°C. Plates were washed, blocked and standards and samples were added. After washing, détection antibody was added and plates were developed with substrate (R and D Systems, Minneapolis, MN). Absorbance was measured at 450 nm using a Versamax ELISA plate reader (Molecular Devices, Sunnyvale, CA). The concentration of unbound VEGF was detennined with a standard curve from 0 to 2000 pg/ml. Results
AB 160 has similar toxicity relative to ABX alone in an in vitro toxicity assay with the human melanoma cell line, A375, suggesting that the paclitaxel functions equally in either formulation (FIG. ID).
To test the binding of VEGF to BEV in the AB 160 complex, AB 160 or ABX was co-incubated with VEGF, the particulate removed, and the supernatant tested for λ/EGF content. The lack of VEGF in the supernatant measured from AB 160 (<10% λ/EGF unbound) indicated that the VEGF was bound by the BEV in the AB 160 complex, while it remained free when incubated with the ABX (>80% λ/EGF unbound) alone (FIG. IE).
Importantly, these assays demonstrated that the paclitaxel in AB 160 retains its toxicity to tumor cells and the bound BEV maintains the ability to bind its ligand, VEGF.
Example 4: Particle Size and Protein Affinity
To understand the characteristics of the nanoparticles formed when binding BEV to ABX, the size of the ABX:BEV complexes was determined relative to ABX.
Methods
Mastersizer and Nanosight: The particle size of ABX and antibody-ABX drug complexes were measured by dynamic light scattering on a Mastersizer 2000 (Malvem Instruments, Westborough, MA). To measure particle size, 2 ml (5 mg/ml) of Abraxane or complex was added to the sample chamber. Data were analyzed with Malvem software and particle size distributions were displayed by volume. The particle sizes and stability were later validated using the Nanosight System (Malvem Instruments, Westborough, MA). The ABX or complex particles were diluted to the appropriate range to accurately measure particle sizes. Data was displayed by particle size distribution; however, the nanoparticle tracking analysis uses Brownian motion to détermine particle size.
Binding Assay: Biotinylated BEV, rituximab or trastuzumab at 100 pg/ml was bound to the streptavidin probe (ForteBio Corp. MenloPark, CA). The binding of ABX was measured by light absorbance on the BLItz system (ForteBio Coip. MenloPark, CA) at 1000, 500 and 100 mg/ml. The association and dissociation constants were calculated using the BLItz software.
Bio-Layer Interferometry (BLItz) technology was utilized to assess the binding affmity of BEV to ABX. Biotinylated BEV was bound to the streptavidin probe and exposed to ABX (1000, 500, and o
100 pg/ml). The dissociation constant (Kd) of BEV and ABX is 2.2 x 10’ M at room température and pH 7, consistent with a strong non-covalent interaction. The binding affmity of BEV and ABX is within the range of dissociation constants observed between albumin and natural or engineered albumin-binding domains of some bacterial proteins. Nilvebrant, J. et al. (2013) Comput Struct Biotechnol J 6:e201303009.
Results
ABX:BEV nanoparticles were consistently larger (approximately 160 nm) than the 130 nm ABX alone (Fig 2a). The size of the nanoparticle created directly correlated to the concentration of BEV used, with médian sizes ranging from 0.157 to 2.166 pm. (FIG. 2A). With the goal of these studies being a Phase I clinical trial, the smallest ABX:BEV particle (AB 160) were focused on because it is the most similar to the 130 nm ABX. The size of the AB 160 particle was consistent with ABX plus a monolayer of BEV surrounding it and with the EM image of the particle (see FIG. IB).
To détermine whether intravenous administration conditions affect nanoparticle size distributions, the particle size distributions of AB 160 (or ABX) incubated in saline for up to 24 hours at room température were evaluated. AB 160 size distribution does not significantly change for up to 24 51 hours (FIGs. 9A and 9B). However, by 4 hours at room température, there is some evidence of AB 160 breakdown by ELISA (FIG. 9C).
To détermine the stability of AB 160 in plasma, ABX or AB 160 was incubated in saline or heparinized human plasma at relative volume ratios of 9:1 or 1:1. Notably, no particles (0.01 to 1 pm) were detected when either ABX (FIG. 10, top panel) or AB 160 (FIG. 10, bottom panel) is incubated in plasma at equal volumes (1:1).
Western blot (data not shown) indicated that, in saline or heparinized human plasma, the AB 160 dissociated into smaller protein conjugates that still contain the tumor-targeting antibody, albumin and the cytotoxic agent, paclitaxel. These protein conjugates retain their ability to target the tumor and, once at the tumor site, can quickly dissolve and release the cytotoxic payload to effectively initiate tumor régression without internalization of the entire nanoparticle by tumor cells.
Next, the ABX was suspended in BEV and the mixture diluted with saline at pH 3, 5, 7, or 9 prior to incubation at various températures (RT, 37 °C and 58 °C) to allow particle formation in order to test whether binding affinity was pH- and/or temperature-dependent. The binding affinity of ABX and BEV is both pH- and temperature-dependent, with the highest binding affinity observed when the particles are formed at pH 5 and 58 °C (FIG. 2B).
To détermine if the higher affinity binding of BEV and ABX at 58 °C and pH 5 translated into stability of the complex, various préparations were compared by nanoparticle tracking analysis (Nanosight). The stability of AB160 prepared at 58 °C and pH 5 (AB1600558), room température and pH 7 (AB 16007), or 58 °C and pH 7 (AB 1600758) was compared to ABX exposed to the same conditions (ABX0558, ABX07, and ABX0758, respectively) after incubation in human AB sérum for 0, 15, 30, or 60 minutes.
The particles made under higher affinity conditions (pH 7 and 58 °C) were also more stable, as indicated by the number of particles présent per mg ABX after exposure to human AB sérum. The AB 160 particles exhibited increased stability in human sérum that correlated with their binding affinities. In particular, AB16007 and AB1600558 were more stable in both saline and human sérum than ABX alone, as determined by size and number of particles measured per mg ABX (FIG. 2C and Table 3). This shows that the stability of AB 160 particles can be manipulated by changing the conditions under which the AB 160 particles are formed.
Table 3: Stability of AB160 and ABX in human AB sérum
Saline Human AB Sérum
0 min 15 min 30 min 60 min
ABX07 221.5 54.4 85.2 84 32.1
AB16007 2500 516 508 756 296
ABX0758 236 182.4 155.4 54 66
AB1600758 2460 436 236 260 176
ABX0558 348 510 86.8 90 64
AB1600558 7296 2200 1224 1080 960
I---------------------------------------------------1—------------------------------------1-----------------------------------------1----------------------------------------1----------------------------------------1-----------------------------------------1
Particles per mg ABX x 10
These data demonstrated that BEV binds to ABX with affinity in the picomolar range, indicating a strong non-covalent bond, and demonstrated a particle size distribution consistent with ABX surrounded by a monolayer of antibody molécules; the size of the particles created is dépendent on the antibody concentration.
Example 5: Efficacy of AB160 in Mice
A xenograft model of A3 75 human melanoma cells implanted into athymie nude mice was employed to test AB 160 efficacy in vivo.
Methods
In vivo experiments were performed at least 2 times. The number of mice required for those experiments was determined by power analysis. Mouse tumors were measured 2-3 times/week and mice were sacrificed when the tumor was 10% by weight. Mice that had complété tumor responses were monitored for 60-80 days post-treatment. The end point of the mouse studies was médian survival. Kaplan-Meier curves were generated and Mantle-Cox test was performed to détermine significance of médian survival between treatment groups. The in vitro results presented are représentative of at least 5 repeated experiments. Statistical analyses of in vitro and in vivo percent change from baseline experiments were done using the Student’s t-test.
Mouse Model: To test tumor efficacy, 1 x ΙΟ6 A375 human melanoma cells were implanted into the right flank of athymie nude mice (Harlan Sprague Dawley, Indianapolis, IN). When the tumors had reached a size of about 700 mm3, the mice were randomized and heated with PBS, ABX (30 mg/kg), BEV (12 mg/kg), BEV followed by ABX, or AB 160 at the above concentrations. For the mouse experiments testing bigger AB particles, the highest dose of BEV (45 mg/kg) necessary to create the larger particles was used in the BEV-only treatment group. Tumor size was monitored 3 times/week and tumor volume was calculated with the following équation: (length*width2)/2. Mice were sacrificed when the tumor size equaled 10% of the mouse body weight or about 2500 mm3. The day 7 percent change from baseline was calculated as follows: [(tumor size on treatment day53 tumor size on day 7)/tumor size on treatment day]*100. The in vivo testing of the AR160 was similar except 5x106 Daudi cells were injected into the right flank of athymie nude mice.
Results
AB 160 was tested relative to PBS, the single drugs alone, and the drugs administered sequentially. Mice treated with AB 160 had significantly reduced tumor size compared to ali other treatment groups (p=0.0001 to 0.0089) at day 7 post-treatment, relative to baseline (FIG. 3 A). Tumors in ail of the mice treated with AB 160 had regressed at day 7, and this tumor response translated into significantly increased médian survival of the AB 160 group relative to ail other groups (FIG. 3B), with a survival of 7, 14, 14, 18 and 33 days for the PBS (p<0.0001), BEV (p=0.003), ABX (p=0.0003), BEV + ABX (p=0.0006) and AB 160 groups, respectively.
It is likely that larger tumors hâve a higher local VEGF concentration. When data were analyzed based on the size of the tumor on day of treatment (<700mm and >700mm ), the larger tumors were shown to hâve a greater response to AB 160, suggesting that higher tumor VEGF concentration attracts more BEV-targeted ABX to the tumor. The différence in the percent change from baseline was significant for the AB 160 groups (p=0.0057). This observation was not seen in the ABX only (p=0.752) group, where the ABX has no targeting capability (FIG. 3C).
Particles of increasing size were prepared using increasing BEV ABX ratios as shown in FIG. 2A. Tumor régression and médian survival positively correlated with increasing particle size, indicating that biodistribution of larger particles may be altered relative to the smaller ones (FIGs. 3D and 3E). Full toxicity studies were performed on the mice and no toxicities were noted.
Example 6: Paclitaxel Pharmakokinetics in Mice
To compare the pharmacokinetics (pk) of AB 160 and ABX, plasma paclitaxel concentrations were measured in mice administered AB 160 or ABX at 0, 4, 8, 12 and 24 hours.
Methods
Paclitaxel Pharmacokinetics: The liquid chromatographie séparation of paclitaxel and d5 paclitaxel were accomplished using an Agilent Poroshell 120 EC-C18 precolumn (2.1 x 5 mm, 2.7 pm, Chrom Tech, Apple Valley, MN) attached to an Agilent Poroshell 120 EC-C18 analytical column (2.1 x 100 mm, 2.7 pm Chrom Tech, Apple Valley, MN) at 40 °C, eluted with a gradient mobile phase composed of water with 0.1% formic acid (A) and ACN with 0.1% formic acid (B) with a constant flow rate of 0.5 ml/minute. The elution was initiated at 60% A and 40% B for 0.5 minutes, then B was linearly increased from 40-85% for 4.5 minutes, held at 85% B for 0.2 minutes, and retumed to initial conditions for 1.3 minutes. Autosampler température was 10 °C and sample injection volume was 2 pl. Détection of paclitaxel and the internai standard d5-paclitaxel were accomplished using 54 the mass spectrometer in positive ESI mode with capillary voltage 1.75 kV, source temp 150 °C, desolvation temp 500 °C, cône gas flow 150 L/hr, desolvation gas flow 1000 L/hr, using multiple reaction monitoring (MRM) Scan mode with a dwell time of 0.075 seconds. The cône voltages and collision energies were determined by MassLynx-Intellistart, v4.1, software and varied between 616 V and 12-60 eV, respectively. The MRM precursor and product ions were monitored at m/z 854.3>105.2 for paclitaxel and 859.3>291.2 for d5 paclitaxel. The primary stock solutions of paclitaxel (1 mg/ml in EtOH) and d5 paclitaxel (1 mg/ml in EtOH) were prepared in 4 ml amber silanized glass vials and stored at -20 °C. Working standards were prepared by dilution of the stock solution with ACN in 2 ml amber silanized glass vials and stored at -20 °C. Plasma samples were extracted as follows, 100 μΐ plasma sample was added to a 1.7 ml micro centrifuge tube containing d5 paclitaxel (116.4 nM or 100 ng/ml) and 300 μΐ ACN, vortexed, incubated at room température for 10 minutes to precipitate proteins, and centrifuged (14,000 rpm) for 3 minutes. The supernatant was filtered on an Agilent Captiva NDlipids plate (Chrom Tech, Apple Valley, MN), collected in a deep 96-well plate, and dried using nitrogen gas. The samples were reconstituted using 100 μΐ ACN and shaken on a plate shaker (high speed) for 5 minutes. Plasma standard curves were prepared daily containing paclitaxel (0.59-5855 nM or 0.5-5000 ng/ml) and d5 paclitaxel (116.4 nM) for paclitaxel quantitation. Mouse tumors were thawed on ice, weighed, and diluted 2 parts (weight to volume) in lx PBS. Tumors were then homogenized using a PRO200 tissue homogenizer using the saw tooth probe (5 mm x 75 mm). Tumor homogenate was than processed the same as the human plasma samples.
Mouse Imagina: Avastin and IgG control solutions were prepared and 1-125 labeled per protocol (Imanis Life Sciences). Briefly, Tris Buffer (0.125 M Tris-HCI, pH 6.8, 0.15 M NaCl) and 5 mCi Na125I were added directly to iodination tubes (ThermoFischer Scientifïc, Waltham, MA). The iodide was allowed to activate and was swirled at room température. Activated iodide was mixed with the protein solution. 50 μΐ of Scavenging Buffer (10 mg tyrosine/mL in PBS, pH 7.4) was added and incubated for five minutes. After addition of Tris/BSA buffer and mixing, samples were applied in 10K MWCO dialysis cassettes against pre-cooled PBS for 30 minutes, 1 hour, 2 hours, and overnight at 4 °C. Radioactivity was determined by Gamma counter, then disintegrations per minute (DPM) and spécifie activity were calculated. Mice were injected in their tail vein with Avastin 1-125, Abraxane-Avastin 1-125, Abraxane-human IgG 1-125, or Abraxane only. Animais were imaged at 3, 10, 24 and 72 hours post-administration via SPECT-CT imaging using the USPECT-II/CT scanner (MILabs, Utrecht, The Netherlands). SPECT reconstruction was performed using a POSEM (pixelated ordered subsets by expectation maximization) algorithme CT data were 55 reconstructed during the Feldkamp algorithm. Co-registered images were further rendered and visualized using PMOD software (PMOD Technologies, Zurich, Switzerland). Animais were sacrificed and dissected at 72 hours post- injection. Selected tissues and organs of interest were measured using radioisotope dose calibrator (Capintec CRC-127R, Capintec Inc.).
Results
Results of the first pk experiment are provided in FIGs. 4A and 4B. The area under the curve (AUC) and maximum sérum concentration (Cmax) were calculated in A375 tumor bearing and non-tumor bearing mice. In the first pk experiment the Cmax and AUC were very similar in the non-tumor bearing mice for AB160 and ABX (63.3+/-39.4 vs. 65.5+/-14.4 and 129 vs. 133 pg/ml, respectively). However, in the tumor bearing mice, the Cniax and AUC for the treatment groups were different (55.7+/-21.2 vs 63.3+/-17.3 and 112 vs 128 pg/ml, respectively) (FIG. 4C). Although this différence was not statistically signifïcant, it is consistent with superior targeting by AB 160, relative to ABX.
A second pk experiment was performed with additional early time points and large versus small tumor sizes (FIGs. 4D-4F). The results of this experiment demonstrated smaller AUC in tumor bearing mice relative to non-tumor bearing mice, with the lowest blood values of paclitaxel in the large tumor mice relative to the small tumor mice (80.4+/- 2.7, 48.4+/-12.3, and 30.7+/-5.2 for ABX-treated non-tumor, small tumor and large tumor bearing mice, respectively; 66.1+/-19.8, 44.4+/-12.1 and 22.8+/-6.9 for AB160-treated). Similarly, the Cmax dropped in both treatment groups in mice with larger tumors (47.2, 28.9 and 19.7 pg/ml for ABX and 40.1, 26.9 and 15.3 pg/ml for AB 160) (FIG. 4G). The AUC and Cmax of paclitaxel in blood were lower in AB160-treated mice relative to ABX-treated mice. Although not statistically signifïcant, this data is further consistent with higher déposition of paclitaxel in the tumors treated with AB 160.
To directly test this hypothesis, tumor paclitaxel concentrations by LC-MS were measured. The tumor paclitaxel concentration was significantly higher in tumors treated with AB 160 relative to ABX at the 4 hour (3473 pg/mg of tissue +/-340 vs 2127 pg/mg of tissue +/- 3.5; p=0.02) and 8 hour (3005 pg/mg of tissue +/- 146 vs 1688 pg/mg of tissue +/- 146; p=0.01) time points, suggesting paclitaxel stays in the tumor longer when targeted by the antibody (FIG. 4H). This explains the blood pk and is consistent with redistribution of drug to tissues including the tumor.
Live in vivo imaging of 1-125 labeled AB 160 (Abx-AvtI125) and IgG isotype bound ABX (AbxIgGI125) confirmed the results of the LC-MS, with higher levels of 1-125 in the tumor of mice treated with AB160 relative to IgG-ABX at 3 hours (32.2 uCi/g +/- 9.1 vs 18.5 uCi/g +/- 1.65; p=0.06) and 10 hours (41.5 uCi/g +/- 6.4 vs 28.7 uCi/g +/- 2.66; p=0.03) post injection (FIGs. 41 and 56
4J). Taken together, these data demonstrate that binding BEV to ABX alters blood pk, and this alteration is due to a redistribution of the diug to the tumor tissue as shown by both LC-MS of paclitaxel and 1-125 labeling of BEV relative to an isotype matched IgGl.
Without being bound by theory, it is believed that by binding a tumor-targeted antibody to ABX, the pk is altered more dramatically than ABX alone, lowering the Cmax and AUC in the blood because of redistribution of AB 160 to the tumor tissue. These results from mouse blood paclitaxel pk, tumor tissue levels of paclitaxel, and 1-125 radioactivity levels in mice treated with AB 160 relative to ABX alone suggest that antibody targeting of the ABX alters biodistribution of paclitaxel such that increased levels reach the tumor and are retained there for a longer period of time, yielding enhanced tumor régression.
Example 7: Binding of Other Therapeutic Antibodies
The binding of the anti-human CD20 antibody (ntuxamab) and the anti-HER2/neu receptor antibody (trastuzumab) to ABX was tested to détermine if other IgG therapeutic antibodies also exhibit binding to ABX when combined ex vivo.
Methods
Nanoparticles containing rituximab or trastuzumab were prepared and tested as described in the above examples.
Results
The particle size of the complexes with both BEV and trastuzumab (HER) were very similar, with average sizes ranging from 0.157 to 2.166 pm (FIG. 2A) and 0.148 to 2.868 pm (FIG. 5B), respectively. In contrast, particles formed with rituximab became much larger at lower antibody:ABX ratios, with particle sizes ranging from 0.159 to 8.286 pm (FIG. 5A).
The binding affinities of rituximab and trastuzumab with ABX were determined by BLItz under variable pH. Both antibodies bind with relatively high affinity in the picomolar range (FIG. 5C). The rituximab affinity to ABX decreased with higher pH, but trastuzumab affinity to ABX was unaffected bypH (FIG. 5C).
The efficacy of the 160 nm particle made with rituximab (AR160) was tested in vitro and in vivo. In vitro, the B-cell lymphoma cell line Daudi was treated with AR160, ABX, or rituximab alone at increasing concentrations (0 to 200 pg/ml) of paclitaxel. AR160 (IC5o=lOpg/ml) significantly inhibited prolifération of Daudi cells treated for 24 hours (p=0.024) compared to either ABX (IC5o>2OOpg/ml) or rituximab (ICso>2OOpg/ml) alone (FIG. 6A).
In vivo, a xenotransplant model of Daudi cells was established in athymie nude mice. Once tumors were established, mice were treated with PB S, ABX, rituximab, ABX and rituximab given 57 sequentially, or AR160. On day 7 post treatment, tumors were measured and the percent change in tumor size from baseline was calculated. AR160-treated tumors regressed or remained stable, while tumors in ail other treatment groups progressed (FIG. 6B). The percent change from baseline tumor size in the AR160 group compared to ail other groups was significant (p<0.0001). The mice treated with AR160 had a significantly longer médian survival of greater than 60 days compared to 12, 16, and 12 days for mice treated with PBS (p<0.0001), ABX (p<0.0001), or rituximab (p=0.0002), respectively (FIG. 6C). However, the différence in médian survival was not significant between AR160 and the sequentially treated groups (p=0.36). This may be because the rituximab binds to the tumor cells and remains on the cell surface, allowing the subsequently-administered ABX to bind to the antibody when it enters the tumor site, unlike BEV which binds a soluble target and not a cell surface marker.
Example 8: Binding of Other Chemotherapy Drugs to AB160
The efficacy of other chemotherapy drugs to form functional nanoparticles was evaluated.
Methods
Nanoparticles containing cisplatin were prepared and tested as described in the above examples. Results
To test if another chemotherapy drug could bind to the AB 160 particles, cisplatin and ABX were coincubated and the amount of free cisplatin remaining in the supernatant was measured by HPLC. Approximately 60% (i.e., only 40% remains in the supernatant) of the cisplatin bound to the ABX (FIG. 7A).
Next, tumor toxicity of AC relative to ABX and cisplatin alone was tested using A375 cells. The complexes were centrifuged to remove highly toxic unbound cisplatin, and reconstituted in media to ensure that any additional toxicity of AC relative to ABX is due only to ABX-bound cisplatin. For parity, the ABX only was centrifuged in a similar manner. AC (ICso=9Opg/ml) inhibited prolifération of A375 cells to a greater extent than ABX alone (IC5o>1000pg/ml) (FIG. 7B). The diminished toxicity in this experiment relative to other toxicity experiments is due to some loss of drug in the centrifugation step, but the comparison of ABX to AC remains relevant.
To détermine the tumor toxicity of cisplatin-containing AB 160 complexes, AB 160 was co-incubated with cisplatin to form cisplatin containing particles (ABC complex). The ABC complex was tested in the A375 melanoma xenotrasplant model relative to each drug alone and AB 160. Tumors treated with AB 160, AB 160 + cisplatin given sequentially, and the ABC complex ail showed régression in tumor size at 7 days post treatment (FIG. 7C), but the ABC complex conferred the longest médian survival (35 days, relative to AB160 and AB160 + cisplatin at 24 and 26 days, respectively).
Although the différence was not statistically significant (p= 0.82 and 0.79) (FIG. 7D), the data is consistent with benefîts of the ABC complex to long-term survival rates.
These data demonstrated that the albumin portion of the ABX provides a platform for other therapeutic antibodies to bind, such as rituximab and trastuzumab, as well as other chemotherapy agents (e.g., cisplatin), which ail had similar efficacy in vitro and in vivo as AB 160.
Together these data demonstrate a simple way to construct a versatile nano-immune conjugate, which allows multiple proteins or cytotoxic agents to be bound to a single albumin scaffold. Improved efficacy of the targeted drag relative to the single agents alone was demonstrated in the mouse model, which is at least in part due to altered pk of the antibody-targeted drug. Furthermore, without being bound by theory, it is believed that the versatility of the presently disclosured nanoimmune conjugate that does not require a linker or target cell intemalization will overcome the obstacles faced by other nanomedicines in translating results from mice to humans.
Example 9: Lyophilization of AB160
AB 160 was synthesized by adding 8mg (320μ1) of bevacizumab to 20mg of Abraxane. 1.66ml of 0.9% saline was then added for a final volume of 2ml for a final concentration of 4mg/ml bevacizumab and lOmg/ml Abraxane, and the mixture was allowed to incubate at room température for 30 minutes in a 15ml polypropylene conical tube.
After the 30 minute room température incubation, the mixture was diluted 1:2 in 0.9% saline to 2mg/ml and 5mg/ml bevacizumab and Abraxane, respectively. These are the concentrations of the 2 drugs when prepared by the pharmacy for administration to patients.
AB 160 was divided into twenty 200μ1 aliquots in 1.5 ml polypropylene eppendorfs and frozen at -80 °C.
Once frozen, the aliquots were lyophilized ovemight with the Virtis 3L benchtop lyophilizer (SP Scientifîc, Warmister, PA) with the réfrigération on. A lyophilized préparation was generated.
The dried aliquots were stored at room température in the same 1.5ml polypropylene eppendorfs. These samples were readily reconstituted in saline at room température for 30 minutes, followed by centrifugation for 7 minutes at 2000x g. The resulting sample was then resuspended in the appropriate buffer, as needed.
By comparison, a sample that was dried with heat and a speed vacuum was impossible to reconstitute.
Example 10: Testing of lyophilized préparations
Samples were reconstituted at different time points after lyophilization and tested for their physical properties against ABX, and freshly made AB 160.
Paiticle size distribution was evaluated as described above.
VEGF binding was evaluated by incubation of the sample with VEGF for 2 hours at room température, centrifuged at 2000 x g for 7 minutes. The amount of VEGF bound to the pellet (corresponding to the nanoparticles) or remaining in the supematant was measured with ELISA. Paclitaxel activity was assessed by cytotoxicity against A375 cells in vitro.
Suiprisingly, lyophilization did not signifîcantly affect either the particle size, VEGF binding, or the activity of paclitaxel as shown by the ability to inhibit cancer cell prolifération. This resuit held for lyophilized samples stored for 1 month (FIGs. 8A-8C) or 10 months (FIGs. 8D-8F).
Further surprising is that these results were observed with nanoparticles lyophilized without the use of cryoprotectants or other agents that may adversely effect human therapeutic use.
Example 11: Efficacy of AB160 in Humans
AB 160 was tested in a phase 1, fîrst-in-man, clinical trial testing the safety of AB 160 administered to patients with metastatic malignant melanoma that hâve failed prior thérapies. The study utilizes a classical 3+3, phase 1 clinical trial design, testing 3 different doses of AB 160 in the following schéma:
Table 4
Dose Level AB-complex Both drugs MUST be reduced
ABX dose Accompanying BEV dose
3 175 mg/m2 70 mg/m2
2 150 mg/m2 60 mg/m2
1* 125 mg/m2 50 mg/m2
-1 100 mg/m2 40 mg/m2
-2 75 mg/m2 30 mg/'m2
*Dose level 1 refers to the starting dose.
The doses were selected relevant to doses of Abraxane currently used in clinical practice. AB 160 was made prior to each treatment dose. Treatments were administered as a 30 minute intravenous infusion on days 1, 8 and 15 of a 28-day treatment cycle. Treatments were continued until intolérable toxicity, tumor progression or patient refusai. Prior to every treatment cycle, patients were evaluated for toxicity; tumor évaluations were performed every other cycle (RECIST).
The study is accompanied by formai (in-patient) pharmacokinetic studies associated with dose 1 of cycles 1 and 2 of therapy.
Five patients hâve been administered AB 160, at 100 mg/m2 of ABX and 40 mg/m2 of BEV, of which four hâve been analyzed.
PFS to
Table 5: Disease course in Phase I study
Disease Course: Dose Level 100 mg/m2
Patient number of cycles response PFS time off, treatment reasons follow-up time
1 8 stable 238 off, progression 444+
2 6 stable 400+ off, toxicity 400+
3 1 182+ off, toxicity 182+
4 6 stable 181 off, progression 203+
refers médian progression free survival, i.e. the number of days of treatment before the cancer· recurred. Adverse events are listed below. There was no dose limiting toxicity (DLT), i.e. the adverse events were not linked to the dose of AB 160. More detail is provided in Table 6
Table 6: Adverse events in Phase I study
patient toxicity DLT
1 grade 2 lymphopenia NO
2 grade 3 neutropenia and leukopenia grade 2 hypertension and anémia NO
3 grade 2 colonie perforation, fatigue, and blood bilirubin increase NO
4 grade 2 neutropenia NO
TABLE 7 Treatment Course: Dose Level 100 mg/m2
patient number of cycles administered number of cycles where day 15 omitted cycles where day 15 omitted reasons day 15 omitted number of dose réductions taken cycles where dose réduction taken reason for dose réductions status
1 8 0 1 4 grd 2 sensory neuropathy off, progression
2 6 3 1,2,4 grd 3 neutropenia and leukopeniaall 3 cycles 2 3,5 cycle 3: grade 3 neutropenia and leukopenia cycle 5: grade 3 neutropenia, leukopenia, and fatigue and grd 2 sensory neuropathy off toxicity persistent grd 2 sensory neuropathy
3 1 off toxicity grd 2 colonie perforation
4 6 2 3,5 grd 2 sensory neuropathy-both cycles off, progression
The mean PFS was 7.6 months and the médian was 7.0 months.
Comparison with other clinical trials
The following table shows other published clinical studies for taxane therapy for metastatic melanoma.
Table 8: Taxane therapy for metastatic melanoma
Study or Author N Rx regimens PFS OS
Hauschild 135 C = AUC6(q21) P = 225mg/m2; DI (q21) 4.5 10.5
Flaherty 411 C = AUC6(q21) P = 225ms/m2:Dl ta2D 4.9 11.3
N057E 41 35 C = AUC2;D1, 8 15 (q28) A= 100mg/m2 ; Dl, 8,15 (q28) 4.5 4.1 11.1 10.9
N047A 53 C = AUC 6; DI (q28) P = 80mg/m2; Dl, 8,15 (q28) n - ru 6.0 12.0
BEAM 71 C = AUC5;D1 (q21) P = 175ms/m2; Dl ία211 4.2 8.6
143 C = AUC5;D1 (q21) P = 175mg/m2; Dl (q21) D- 1C^rv/l<rr. Dl /«OH 5.6 12.3
N0775 51 C = AUC6(5);D1 (q28) A = 100 (80) mg/m2 ; Dl, 8,15 (q28) r> — i m i c 6.2 13.9
Spitler 50 A = 150mg/m2; Dl, 8, 15 (q28) B = lOme/ke: Dl. 15 ία28) 7.6 15.6
C=carboplatin, P=paclitaxel, A=nab-paclitaxel, B=bevacizumab
References: Hauschild: Hauschild et al., (2009) J Clin Oncol. 27(17):2823-30 Flaherty: Flaherty et al., (2010) J Clin Oncol. 28:15s (suppl; abstr 8511) N057E: Kottschade et al., (2010) Cancer 117(8):1704-10 N057A: Perez étal., (2009) Cancer 115(1):119-27 BEAM: Kim et al., (2012) J Clin Oncol. 30(1):34-41 N0775: Kottschade et al., (2013) Cancer 119(3):586-92 Spitler: Boasberg et al., (2011) J Clin Oncol. 29 (suppl; abstr 8543)
In the current trial, administration of AB 160 particles is équivalent to a dose of 100 mg/m2 of abraxane, and 40 mg/m2 of bevacizumab. The only study that used BEV and ABX alone was Spitler. Spitler, however, used a higher dose of ABX. The présent study also used less than than
10% of the dose of BEV reported in previous studies, if the doses are adjusted to the average patient (assumed to hâve a surface area of 1.9 m2 and a mass of 90 kg).
Spilter also examined patients who had not been previously treated, while the current study examined patients who had failed previous treatments. Ineffective prior treatment not only takes time from the expected PFS, but selects for cancer cells that are more résistant to treatment, and typically leaves a patient in poorer physical condition. Thus, the PFS for a population of patients on a “rescue” therapy (as here, with AB 160) is expected to hâve a lower PFS than a naïve population. This canbe seen in a Phase 2 clinical trial (Hersh et al., Cancer, January 2010, 116:155) that examined both rescue and naïve patients with Abraxane alone. For previously treated patients with
Abraxane alone, the PFS was 3.5 months. Hersh et al. Ann. Oncol 2015, (epub Septembei' 26, 2015), reported a 4.8 month PFS for naïve patients treated with ABX alone.
Table 9: Performance of AB160 in a limited study against published data
Study Prior treatment ABX dose in average patient (relative dose) BEV dose in average patient (relative dose) PFS (months)
AB 160 Yes 190 mg/patient (100 mg/m2) 76 mg/patient (40 mg/m2) 7.0
Spitler No 285 mg/patient (150 mg/m2) 900 mg/patient (10 mg/kg) 8.3
Hersh 2010 Yes 190 mg/patient (100 mg/m2) 3.5
Hersh 2010 No 285 mg/patient (150 mg/m2) 4.5
Hersh 2015 No 285 mg/patient (150 mg/m2) 4.8
Thus, early results of the Phase I clinical trial with AB 160 indicate an increase in PFS in late-stage metastatic malignant melanoma in previously treated patients. This increase is particularly surprising given that the PFS was greater than those in Spitler, who were chemotherapy naïve and were given a higher dose of Abraxane, and an almost 12 fold higher dose of bevacizumab. The dose of BEV used in AB 160 is far lower than any other study, so the best comparison is not Spitler, but Hersh.
Thus, the ABX/BEV complex (AB 160) is superior to sequential administration of ABX and BEV, or ABX alone, and achieves this superior resuit with a very low effective dose of BEV. The data is therefore consistent with AB 160 having improved targeting of the chemotherapeutics to the tumor, and that this targeting is mediated by BEV. It is possible that the ABX nanoparticle aids in targeting the BEV to the tumor, as albumin is selectively taken up by tumors. It is also possible that the existence of the BEV/ABX complex shows greater stability in vivo than Abraxane.
Example 12: Follow up study to investigate whether pretreatment with BEV improves targeting
Following the general protocol above, athymie nude mice were injected with 1 x 106 A375 human melanoma cells in the right flank and then treated with PBS, 12 mg/kg BEV, 30 mg/kg ABX, AB 160, or pretreated with 1.2 mg/kg BEV and, 24hr later, AB 160. Data is represented at day 7-post and day 10-post treatment as tumor volume in mm3. F 11A-E track tumor size over 10 days. Only mice treated with AB 160 (with or without pretreatment with BEV) showed a réduction in average tumor volume. See also FIG. 11F and FIG. 1 IG.
The day 7-post treatment data, as summarized in Figure 11F, show that pretreatment with BEV was associated with a stastically significant réduction in tumor volume over control or BEV alone (p<0.0001), or ABX alone (p<0.0001).
The day 10-post treatment data, as summarized in Figure 11 G, again show that pretreatment with BEV was associated with a stastically significant réduction in tumor volume over control or BEV alone (p<0.0001), or ABX alone (p<0.0001). Pretreatment with BEV before AB160 was also associated with a réduction in tumor volume over AB 160 alone (p=0.02), with complété response in two mice.
In this experiment, a 12 mg/kg dose of BEV was not therapeutic. The amount of BEV added in the pretreatment group was only 1.2 mg/kg, which is 1/10 the usual dose in mice. Yet pretreatment with a subtherapeutic dose appears to show improved efficacy of the AB 160 nanoparticle. This data support the idea that pretreatment with a subtherapeutic amount of BEV can clear systemic levels of VEGF, leaving a greater relative concentration at the tumor such that tumor-associated VEGF targeting by the AB 160 nanoparticles is more effective.
Example 13: Alternative means of delivering nanoparticles
It is contemplated that nanoparticles of this invention can be directly delivered to the tumor. For example, nanoparticles can be delivered via intra-arterial cannula or by direct injection into the turmor. In such embodiments, it is contemplated that large nanoparticles (e.g., 580 nm or 1130 nm) can be delivered by direct injection into or proximate to a tumor.

Claims (14)

1. A lyophilized composition comprising nanoparticle complexes, wherein each of the nanoparticle complexes comprises:
a. albumin,
b. between about 100 to about 1000 antibodies having an antigen-binding portion, and
c. paclitaxel, said nanoparticle complexes being lyophilized, and wherein upon reconstitution with an aqueous solution, the nanoparticle complexes remain capable of binding to the antigen in vivo.
2. The lyophilized nanoparticle composition of claim 1, wherein the antibodies recognize an antigen on a cancer cell.
3. The lyophilized nanoparticle composition of claim 1 or 2, wherein each of the nanoparticle complexes comprises between about 400 and about 800 antibodies.
4. The lyophilized composition of any one of the above claims, wherein the antibodies are ado-trastuzumab emtansine, alemtuzumab, bevacizumab, cetuximab, denosumab, dinutuximab, ipilimumab, nivolumab, obinutuzumab, ofatumumab, panitumumab, pembrolizumab, pertuzumab, rituximab, or trastuzumab.
5. The lyophilized composition any one of the above claims, wherein the antibodies arrange on ail or part of the outer surface of the nanoparticle complexes.
6. The lyophilized composition of any one of the above claims, wherein the weight ratio of albumin-bound paclitaxel to antibody is between 10:1 and 10:30.
7. The lyophilized composition of any one of the above claims, wherein the albumin is human sérum albumin.
8. The lyophilized composition of any one of the above claims, wherein said nanopailicle complexes hâve an average size of less than 1 pm.
9. The lyophilized composition of any one of the above claims, wherein the antibodies and/or paclitaxel are associated with the albumin through non-covalent bonds.
10. A reconstituted nanoparticle composition comprising a lyophilized composition of any one of claims 1-9 and a pharmaceutically acceptable excipient for use in treating cancer in a subject.
11. The reconstituted nanoparticle composition of claim 10, wherein a therapeutically effective amount of said compostion comprises about 20 mg/m2 to about 90 mg/m2 of antibody and/or about
5 50 mg/m2 to about 200 mg/m2 of albumin and paclitaxel.
12. A method of making the lyophilized nanoparticle composition of any one of claims 1-9, the method comprising mixing aqueous albumin-paclitaxel nanoparticles with the antibodies in vitro under conditions to form the nanoparticle complexes, and lyophilizing the nanoparticle complexes to form the lyophilized nanoparticle composition; such that the nanoparticle complexes retain binding
10 specificity to the antigen on the cancer in vivo when reconstituted with an aqueous solution.
13. The method of claim 12, wherein the aqueous albumin-paclitaxel nanoparticles are mixed with the antibodies in a solution having a pH of between about 4 and about 8.
14. The method of claim 12 or 13, wherein the aqueous albumin-paclitaxel nanoparticles are incubated with the antibodies at a température of about 5 °C to about 60 °C.
OA1201700009 2014-10-06 2015-10-06 Carrier-antibody compositions and methods of making and using the same OA18218A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US62/060,484 2014-10-06
US62/206,772 2015-08-18
US62/206,770 2015-08-18
US62/206,771 2015-08-18

Publications (1)

Publication Number Publication Date
OA18218A true OA18218A (en) 2018-09-04

Family

ID=

Similar Documents

Publication Publication Date Title
AU2020201289B2 (en) Carrier-antibody compositions and methods of making and using the same
US10596111B2 (en) Methods of making lyophilized compositions comprising albumin-VEGF paclitaxel nanoparticle complexes
AU2016308337B2 (en) Carrier-binding agent compositions and methods of making and using the same
OA18218A (en) Carrier-antibody compositions and methods of making and using the same