Intracellular protein-protein interactions (PPIs) control many essential cellular pathways implicated in human diseases [1, 2], representing an important class of therapeutic targets that are considered to be The Holy Grail in drug discovery and development [3, 4]. Among various PPI inhibitors with therapeutic potential, small peptides, compared with low molecular weight compounds, often excel due to their high potency and selectivity and low toxicity [5, 6]. However, major pharmacological disadvantages of peptide inhibitors exist. For example, peptides are susceptible to enzymatic degradation because they generally do not possess a stable tertiary structure to confer resistance to proteolysis; peptides also lack the ability to actively traverse the cell membrane, thus failing to reach intracellular drug targets. Poor proteolytic stability and membrane permeability severely limit peptide bioavailability and therapeutic efficacy [5, 7]. Various elaborate medicinal chemistry approaches and peptide delivery techniques have been developed to overcome these pharmacological barriers [8-16]. While considerable success has been achieved in using peptides to target intracellular PPIs [17-21], much still remains to be done to fulfill their full therapeutic potential.
Nanotechnology has been widely used in the development of new strategies for drug delivery and cancer therapy [22, 23]. Nanoparticle-based traditional delivery tools include, but are not limited to, micelle, liposome, dendrimer, gold nanoshell, and polymer [24, 25]. As unique biopolymers in the nanoscale, proteins are superior in many aspects as a drug carrier to synthetic polymers [26, 27]. Protein-based drug carriers are attractive also because they are amenable to both biological and chemical modifications so that their properties such as molecular size, site of conjugation, and loading capacity can be controlled [28]. In addition, novel functionalities can be engineered into proteins to facilitate cellular uptake and improve targeting specificity. Albumin, a natural transport protein with multiple ligand binding sites, cellular receptor engagement, and a long circulatory half-life, represents a clinically proven platform for the delivery of various drug molecules [29, 30]. Despite the obvious advantages of protein-based drug delivery of low molecular weight compounds, it remains challenging to efficiently deliver peptide therapeutics to target intracellular PPIs.
Accordingly, new approaches are needed for intracellular delivery of peptide therapeutics, for example, to therapeutically disrupt intracellular protein-protein interactions involved in cancer and other diseases. The present description relates to the use of molecular grafting approaches to design a stable protein scaffold with multiple functionalities for the intracellular delivery of peptide therapeutics.
The present invention relates to a stable protein scaffold with multiple functionalities for delivering peptide therapeutics to the interiors of cells.
In one aspect, the invention relates to a protein comprising a protein scaffold and at least one therapeutic peptide grafted therein, wherein the protein scaffold is a disulfide-devoid tetramerization domain of chimeric oncoprotein Bcr/Abl protein of chronic myeloid leukemia, as defined in SEQ ID NO: 6.
In another aspect, the invention relates to a PMIBcr/Abl protein comprising a sequence as shown in SEQ ID NO: 5.
In still another aspect, the invention relates to a PMIBcr/Abl-R6 protein comprising a sequence as shown in SEQ ID NO: 3.
In yet another aspect, the invention relates to a method of inhibiting tumor cell growth in a mammal, said method comprising administering a protein to said mammal, wherein said protein comprises a protein scaffold and at least one therapeutic peptide grafted therein, wherein the protein scaffold is a disulfide-devoid tetramerization domain of chimeric oncoprotein Bcr/Abl protein of chronic myeloid leukemia, as defined in SEQ ID NO: 6.
In yet another aspect, the invention relates to a method of inhibiting tumor cell growth in a mammal, said method comprising administering a protein to said mammal, wherein said protein comprises a PMIBcr/Abl protein comprising a sequence as shown in SEQ ID NO: 5.
In still another aspect, the invention relates to a method of inhibiting tumor cell growth in a mammal, said method comprising administering a protein to said mammal, wherein said protein is a PMIBcr/Abl-R6 protein comprising a sequence as shown in SEQ ID NO: 3.
In yet another aspect, the invention relates to a method of inducing apoptosis of cancer cells in a mammal, said method comprising administering a protein to said mammal, wherein said protein comprises a protein scaffold and at least one therapeutic peptide grafted therein, wherein the protein scaffold is a disulfide-devoid tetramerization domain of chimeric oncoprotein Bcr/Abl protein of chronic myeloid leukemia, as defined in SEQ ID NO: 6.
In yet another aspect, the invention relates to a method of inducing apoptosis of cancer cells in a mammal, said method comprising administering a protein to said mammal, wherein said protein comprises a PMIBcr/Abl protein comprising a sequence as shown in SEQ ID NO: 5.
In still another aspect, the invention relates to a method of inducing apoptosis of cancer cells in a mammal, said method comprising administering a protein to said mammal, wherein said protein is a PMIBcr/Abl-R6 protein comprising a sequence as shown in SEQ ID NO: 3.
In yet another aspect, the invention relates to a method of treating Philadelphia chromosome-positive acute lymphocytic leukemia (ALL) and/or chronic myelogenous leukemia (CML) in a mammal, said method comprising administering a protein to said mammal, wherein said protein comprises a protein scaffold and at least one therapeutic peptide grafted therein, wherein the protein scaffold is a disulfide-devoid tetramerization domain of chimeric oncoprotein Bcr/Abl protein of chronic myeloid leukemia, as defined in SEQ ID NO: 6.
In yet another aspect, the invention relates to a method of treating Philadelphia chromosome-positive acute lymphocytic leukemia (ALL) and/or chronic myelogenous leukemia (CML) in a mammal, said method comprising administering a protein to said mammal, wherein said protein comprises a PMIBcr/Abl protein comprising a sequence as shown in SEQ ID NO: 5.
In still another aspect, the invention relates to a method of treating Philadelphia chromosome-positive acute lymphocytic leukemia (ALL) and/or chronic myelogenous leukemia (CML) in a mammal, said method comprising administering a protein to said mammal, wherein said protein is a PMIBcr/Abl-R6 protein comprising a sequence as shown in SEQ ID NO: 3.
In yet another aspect, the invention relates to a method of delivering a p53-activating compound for cancer treatment, said method comprising administering a protein to a mammal, wherein said protein comprises a protein scaffold and at least one therapeutic peptide grafted therein, wherein the protein scaffold is a disulfide-devoid tetramerization domain of chimeric oncoprotein Bcr/Abl protein of chronic myeloid leukemia, as defined in SEQ ID NO: 6.
In yet another aspect, the invention relates to a method of delivering a p53-activating compound for cancer treatment, said method comprising administering a protein to a mammal, wherein said protein comprises a PMIBcr/Abl protein comprising a sequence as shown in SEQ ID NO: 5.
In still another aspect, the invention relates to a method of delivering a p53-activating compound for cancer treatment, said method comprising administering a protein to a mammal, wherein said protein is a PMIBcr/Abl-R6 protein comprising a sequence as shown in SEQ ID NO: 3.
The present invention relates to a stable protein scaffold with multiple functionalities for delivering peptide therapeutics to the interiors of cells. More specifically, the present invention relates to protein-based peptide drug carrier derived from the tetramerization domain of the chimeric oncogenic protein Bcr/Abl of chronic myeloid leukemia (MVDPVGFAEAWKAQFPDSEPPRMELRSVGDIEQELERAKASIRRLEQEVNQERFRMIYLQTLLAKEK KSYDR; SEQ ID NO: 6).
The p53-MDM2/MDMX interaction has garnered much attention as an important intracellular drug target for the development of MDM2/MDMX antagonists or p53-activating agents for anticancer therapy [37, 55-57]. Small molecule antagonists are generally mono-specific for MDM2, and several are in clinical trials with promising early results [54, 58]. By contrast, peptide antagonists are often dual-specific for both MDM2 and MDMX, potentially affording more robust and sustained p53 activation. One notable example is ALRN-6924, a hydrocarbon-stapled peptide antagonist of MDM2 and MDMX kills tumor cells harboring wild-type p53 in phase 2 clinical trials for advanced solid tumors and lymphomas [59]. More recently, ALRN-6924 has been reported to be effective against acute myeloid leukemia in vitro and in vivo [60]. The hydrocarbon-stapling technique pioneered by Verdine and colleagues enables side-chain cross-linked and conformationally stabilized helical peptides to traverse the cell membrane with improved proteolytic stability and enhanced biological activity [61, 62]. Of note, a hydrocarbon- or dithiocarbamate-stapled PMI (p53-MDM2/MDMX inhibitor) has been shown to be a potent p53 activator in vitro and in vivo [63-65]. Despite these successes, it is worth noting that small peptides do not have a sufficiently long circulation half-life in vivo due to renal excretion (<20 KDa), thus adversely affecting their therapeutic efficacy. By contrast, the protein construct PMIBcr/Abl-R6 described herein, a stable tetramer of 35 KDa that can be readily prepared in large quantity via recombinant expression, is expected to have excellent bioavailability compared with small peptide therapeutics.
Most protein scaffolds used for peptide grafting are stabilized by disulfide bonds [31-33], and thus are unsuitable for targeting PPIs in the cytoplasmic space where the reducing environment can structurally destabilize disulfide-bridged proteins prompting their proteolytic degradation. To circumvent this severe limitation, the present inventors have identified the disulfide-devoid tetramerization domain of the chimeric oncoprotein Bcr/Abl of chronic myeloid leukemia (CML) [34], which forms a highly stable tetramer in solution, as a protein scaffold for molecular grafting of therapeutic peptides of an α-helical nature.
The present inventors introduced into the N-terminus of Bcr/Abl, a potent dodecameric peptide antagonist, termed PMI, of both MDM2 and MDMX—the two oncogenic proteins that functionally inhibit the tumor suppressor protein p53 in many tumor types [35, 36]. To antagonize intracellular MDM2/MDMX for p53 activation, PMIBcr/Abl was extended by a C-terminal Arg-repeating hexapeptide (R6) to facilitate its cellular uptake. The resultant tetrameric protein PMIBcr/Abl-R6 adopted an alpha-helical conformation in solution and bound to MDM2 at an affinity of 32 nM. PMIBcr/Abl-R6 effectively induced apoptosis of HCT116 p53+/+ cells in vitro in a p53-dependent manner and potently inhibited tumor growth in a nude mouse xenograft model by antagonizing MDM2/MDMX to reactivate the p53 pathway. This protein scaffold, Bcr/Abl-R6, can be used as a delivery tool for α-helical peptides to target a great variety of intracellular PPIs for disease intervention. In addition to being generally useful as a protein-based universal carrier for delivering peptide therapeutics for treatment of various diseases, the protein scaffold and methods described herein can be used specifically, e.g., to deliver PMIBcr/Abl as a p53-activating compound for cancer therapy, as well as to deliver PMIBcr/Abl as a p53-activating and Bcr/Abl-inhibiting compound for the treatment of Philadelphia chromosome-positive acute lymphocytic leukemia (ALL) and/or chronic myelogenous leukemia (CML) resistant to imatinib.
Accordingly, in one aspect, the instant application relates to a protein comprising a protein scaffold and at least one therapeutic peptide grafted therein, wherein the protein scaffold is a disulfide-devoid tetramerization domain of chimeric oncoprotein Ber/Abl protein of chronic myeloid leukemia, as defined in SEQ ID NO: 6. The therapeutic peptide can have an α-helical structure. The therapeutic peptide can be grafted into the N-terminus of the Bcr/Abl protein. The therapeutic peptide can be any p53-activating peptide of an alpha-helical nature and is useful for the treatment of any cancer harboring wild type-p53 and elevated MDM2/MDMX. Further, the therapeutic peptide can be any antitumor peptide of an alpha-helical nature and is useful for treating cancer in general. The therapeutic peptide can be linear or stapled. For example, in one embodiment, the therapeutic peptide is PMI, which antagonizes intracellular MDM2/MDMX, thereby activating p53. The PMI is grafted in place of residues 5-16 of the Bcr/Abl protein. The protein, regardless of the peptide grafted therein, can further comprise a C-terminal extension to allow the protein to traverse a cell membrane. For example, in one embodiment, the C-terminal extension is an Arg-repeating hexapeptide (R6).
In another aspect, the present application relates to a PMIBcr/Abl protein comprising a sequence as shown in SEQ ID NO: 5. The PMIBcr/Abl protein can further comprise a C-terminal extension to allow the protein to traverse a cell membrane. For example, in one embodiment, the C-terminal extension is an Arg-repeating hexapeptide (R6).
In still another aspect, the present application relates to a PMIBcr/Abl-R6 protein comprising a sequence as shown in SEQ ID NO: 3.
In addition to PMI, other therapeutic peptides that can be grafted into the protein scaffold of SEQ ID NO: 6 include, but are not limited to: MTide-01, sMTide-01, MTide-02, sMTide-02, sMTide-02A, sMTide-02B, as disclosed C. J. Brown et al., ACS Chem. Biol., 2013, 8, 506-512 [63], which is incorporated by reference herein in its entirety; PMI(1,5)-a, PMI(1,5)-b, PMI(2,6)-a, PMI(2,6)-b, PMI(4,8)-a, PMI(4,8)-b, PMI(5,9)-a, PMI(5,9)-b, PMI(8,12)-a, and PMI(8,12)-b, as disclosed in Xiang Li et al., Chem. Sci., 2079, 10, 1522, which is incorporated by reference herein in its entirety; and N8A-PMI and other truncated analogs of PMI, as disclosed in Chong Li et al., J. Mol. Biol., 2010, 398(2), 200-213 (doi: 10.1016/j.jmb.2010.03.005).
It should be appreciated that the proteins described herein can be present in a formulation that is suited for administration to the subject. Accordingly, in another aspect, the present application relates to a formulation, said formulation comprising the protein and at least one pharmaceutically acceptable excipient. The formulation can further comprise at least one additional active pharmaceutical ingredient (API) such as an anticancer agent.
The term “pharmaceutically acceptable excipient” refers to a carrier, diluent, or adjuvant which is administered with the proteins described herein. Such pharmaceutically acceptable excipients may be liquid-based, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. Water or aqueous salt solutions and aqueous solutions of dextrose and glycerol, particularly for injectable solutions, are preferably used as excipients. Additional pharmaceutically acceptable excipients include, but are not limited to, any and all solvents, buffering agents (such as a phosphate buffer, citrate buffer, and buffers made from other organic acids), dispersion media, surfactants, antioxidants (e.g., ascorbic acid), preservatives (e.g., antibacterial agents, antifungal agents), a polypeptide (such as serum albumin, gelatin, and an immunoglobulin), a hydrophilic polymer (such as polyvinylpyrrolidone), an amino acid (such as glycine, glutamine, asparagine, arginine, and/or lysine), a monosaccharide, a disaccharide, and/or other carbohydrates (including glucose, mannose, and dextrins), a chelating agent (e.g., ethylenediaminetetratacetic acid (EDTA)), a sugar alcohol (such as mannitol and sorbitol), a salt-forming counterion (e.g., sodium), an anionic surfactant (such as TWEEN, PLURONICS, and PEG), isotonic agents, absorption delaying agents, salts, drug stabilizers, gels, lubricants, sweetening agents, flavoring agents, dyes, and combinations thereof, as would be known to one of ordinary skill in the art (see, “Remington Pharmaceutical Sciences” by E W Martin, 21′ Edition, 2005). A pharmaceutically acceptable excipient suitable for use in the formulation and methods described herein is non-toxic to cells, tissues, or subjects at the dosages employed.
The proteins or formulations described herein can be used for ameliorating and/or treating a cancer. In one embodiment, the cancer is related to inactivation and/or mutation of p53. A non-limiting exemplary list of cancers includes, but is not limited to, breast cancer, prostate cancer, lymphoma, skin cancer, pancreatic cancer, colon cancer, melanoma, malignant melanoma, ovarian cancer, brain cancer, primary brain carcinoma, head-neck cancer, glioma, glioblastoma, liver cancer, bladder cancer, non-small cell lung cancer, head or neck carcinoma, breast carcinoma, ovarian carcinoma, lung carcinoma, small-cell lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, bladder carcinoma, pancreatic carcinoma, stomach carcinoma, colon carcinoma, prostatic carcinoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, myeloma, multiple myeloma, adrenal carcinoma, renal cell carcinoma, endometrial carcinoma, adrenal cortex carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, malignant hypercalcemia, cervical hyperplasia, leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia (CLL) including B-CLL, acute myelogenous leukemia, chronic myelogenous leukemia, chronic granulocytic leukemia, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, sarcoma such as liposarcoma, malignant fibrous histiocytoma, osteosarcoma, Ewing's sarcoma, leiomyosarcoma, and rhabdomyosarcoma, Kaposi's sarcoma, polycythemia vera, essential thrombocytosis, Hodgkin's disease, non-Hodgkin's lymphoma, soft-tissue sarcomas such as lipoma, and malignant Schwannoma, osteogenic sarcoma, primary macroglobulinemia, and retinoblastoma, and the like, T and B cell mediated autoimmune diseases; inflammatory diseases; infections; hyperproliferative diseases; AIDS; degenerative conditions, vascular diseases, and the like. In a preferred embodiment, the cancer ameliorated and/or treated is selected from at least one of melanoma, lung cancer, a sarcoma, colon cancer, prostate cancer, choriocarcinoma, breast cancer, retinoblastoma, stomach carcinoma, acute myeloid leukemia, a lymphoma, multiple myeloma, and a leukemia in a subject. In some embodiments, the cancer cells being treated are metastatic. In other embodiments, the cancer cells being treated are resistant to other anticancer agents.
Treating cancer includes, but is not limited to, reducing the number of cancer cells or the size of a tumor in the subject, reducing progression of a cancer to a more aggressive form, reducing proliferation of cancer cells or reducing the speed of tumor growth, killing of cancer cells, inducing apoptosis of cancer cells, reducing metastasis of cancer cells or reducing the likelihood of recurrence of a cancer in a subject. Treating a subject, as used herein, refers to any type of treatment that imparts a benefit to a subject afflicted with a disease or at risk of developing the disease, including improvement in the condition of the subject (e.g., in one or more symptoms), delay in the progression of the disease, delay the onset of symptoms or slowing the progression of symptoms, etc.
The proteins or formulations and methods related to treating cancer, as described herein, can be used to treat subjects such as mammals (e.g., humans) having cancer. Examples of mammals that can be treated as described herein include, without limitation, humans, monkeys, dogs, cats, cows, horses, pigs, rats, and mice.
The methods of treatment, as described herein, relate to the administration of a therapeutically effective amount of the proteins or formulations, as described herein, to a subject in need of said treatment. As used herein, an “effective amount” or a “therapeutically effective amount” means the amount of a protein that, when administered to a subject for treating cancer is sufficient to effect a treatment (as defined above). The therapeutically effective amount will vary depending on the protein, or formulation comprising same, the disease and its severity and the age, weight, physical condition and responsiveness of the subject to be treated.
It will be appreciated that the specific dosage administered in any given case will be adjusted in accordance with the protein or formulation being administered, the disease to be treated or inhibited, the condition of the subject, and other relevant medical factors that may modify the activity of the protein or formulation or the response of the subject, as is well known by those skilled in the art. For example, the specific dose for a particular subject depends on age, body weight, general state of health, diet, the timing and mode of administration, the rate of excretion, medicaments used in combination and the severity of the particular disorder to which the therapy is applied. Dosages for a given patient can be determined using conventional considerations, e.g., by customary comparison of the differential activities of the protein or formulation such as by means of an appropriate conventional pharmacological or prophylactic protocol.
The maximal dosage for a subject is the highest dosage that does not cause undesirable or intolerable side effects. The number of variables in regard to an individual treatment regimen is large, and a considerable range of doses is expected. The route of administration will also impact the dosage requirements. It is anticipated that dosages of the protein scaffold or formulation will reduce growth of the cancer by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% compared to a cancer left untreated.
Suitable effective dosage amounts for administering the protein or formulation may be determined by those of skill in the art, but typically range from about 1 microgram to about 10,000 micrograms per kilogram of body weight weekly, although they are typically about 1,000 micrograms or less per kilogram of body weight weekly. In some embodiments, the effective dosage amount ranges from about 10 to about 10,000 micrograms per kilogram of body weight weekly. In another embodiment, the effective dosage amount ranges from about 50 to about 5,000 micrograms per kilogram of body weight weekly. In another embodiment, the effective dosage amount ranges from about 75 to about 1,000 micrograms per kilogram of body weight weekly. The effective dosage amounts described herein refer to total amounts administered, that is, if more than one protein or formulation is administered, the effective dosage amounts correspond to the total amount administered. The protein or formulation can be administered as a single dose or as divided doses. For example, the protein or formulation may be administered two or more times separated by 4 hours, 6 hours, 8 hours, 12 hours, a day, two days, three days, four days, one week, two weeks, or by three or more weeks.
In various embodiments, the protein or formulation can be administered by intravenous, intraarterial, intrathecal, intradermal, intracavitary, oral, rectal, intramuscular, subcutaneous, intracisternal, intravaginal, intraperitonial, topical, buccal, and/or nasal routes of administration.
Accordingly, in another aspect, the present application relates to a method of inhibiting tumor cell growth in a mammal, said method comprising administering any of the proteins or formulations described herein to said mammal. The protein antagonizes intracellular MDM2/MDMX for p53 activation.
In still another aspect, the present application relates to a method of inducing apoptosis of cancer cells in a mammal, said method comprising administering any of the proteins or formulations described herein to said mammal. The protein antagonizes intracellular MDM2/MDMX for p53 activation.
In still another aspect, the present application relates to a method of treating Philadelphia chromosome-positive acute lymphocytic leukemia (ALL) and/or chronic myelogenous leukemia (CML) in a mammal, said method comprising administering any of the proteins or formulations described herein to said mammal. In one embodiment, the ALL and/or CML are resistant to imatinib.
In another aspect, the present application relates to a method of delivering a p53-activating compound for cancer treatment, said method comprising administering any of the proteins or formulations described herein to said mammal.
In yet another aspect, the invention also relates to the use of a protein or formulation as described herein for the manufacture of a medicament for the treatment of cancer.
In still another aspect, the invention relates to the use of the protein or the formulation as described herein as a medicament.
In yet another aspect, the present invention relates to a method of using recombinant techniques to make any of the proteins as described herein, as readily understood by the person skilled in the art.
It has been demonstrated that the tetrameric Bcr/Abl scaffold is an ideal protein-based nanocarrier of p53-activating peptides to target the p53-MDM2/MDMX interaction for cancer therapy. MDM2 and MDMX cooperate to persistently inhibit p53 function and target the tumor suppressor protein for proteasomal degradation, contributing to tumor development and progression. PMIBcr/Abl-R6 as a dual-specificity antagonist of MDM2 and MDMX and a powerful p53 activator in vitro and in vivo is superior in many aspects to mono-specific small molecule inhibitors of MDM2 as well as stapled peptide antagonists currently in clinical trials, promising a novel class of antitumor agents with significant therapeutic potential. Importantly, this protein-based nanocarrier is also suitable for the design of different classes of peptide therapeutics of an α-helical nature to target intracellular PPIs involved in many other human diseases.
It should be appreciated by the person skilled in the art that although reference herein is to the targeting of intracellular proteins, the protein-based nanocarrier is also suitable for the design of different classes of peptide therapeutics of an α-helical nature to target extracellular PPIs involved in many other human diseases as well.
The features and advantages of the invention are more fully shown by the illustrative examples discussed below.
In many tumor cells harboring wild type p53, the E3 ubiquitin ligase MDM2 and/or its homolog MDMX (also known as MDM4) block the transcriptional activity of p53 and target the tumor suppressor protein for proteasomal degradation, conferring tumor development and progression [35-37]. MDM2/MDMX antagonism has been validated as an effective therapeutic strategy for cancer treatment. Since MDMX potentiates MDM2 function in p53 inhibition, dual-specificity antagonists of both MDM2 and MDMX are particularly attractive as therapeutic agents for robust and sustained p53 activation [37]. The present inventors previously identified PMI, a series of high-affinity and dual-specificity dodecameric peptide antagonists of MDM2 and MDMX, through combinatorial library screening and structure-based rational design approaches [38, 39]. Although PMI peptides tightly bind, in an α-helical conformation, to the p53-binding pocket of MDM2 and MDMX at affinities ranging from high pM to low nM, they are not inhibitory per se against tumor growth due mainly to their inability to traverse the cell membrane [38, 39].
To carry therapeutic peptides of an α-helical nature for cancer therapy, it has been hypothesized that the protein must meet the following five criteria: (1) structurally amenable to peptide grafting with a pre-existing short α-helix, (2) sufficiently large in size (via oligomerization, for example) to alleviate renal excretion, (3) resistant to proteolytic degradation by adopting a stable structure with few flexible loops and disordered regions, (4) devoid of disulfide bonds, and (5) efficient in membrane permeabilization. Bcr/Abl tetramerization domain comprises 72 amino acid residues and forms a coiled-coil tetramer, with each monomer consisting of a short N-terminal α-helix, a connecting loop, and a long C-terminal α-helix [34]. This protein is thus ideally suited as a nano-carrier of PMI for cancer therapy because it readily meets the first four criteria defined. To enable its membrane permeability, however, additional modifications such as introduction of a cationic penetrating peptide sequence to Bcr/Abl tetramerization domain is warranted. The design strategy is schematically illustrated in
In
Structural studies indicate that the N-terminal α-helix of Bcr/Abl (residues 5-15) does not contribute to protein tetramerization, which is mediated predominantly by the elongated C-terminal α-helix (residues 28-67) [34] (see,
Bcr/Abl-R6 and PMIBcr/Abl-R6 were also characterized using size exclusion chromatography (
Isothermal titration calorimetry (ITC) data analysis was performed to measure the binding affinity of PMI and PMIBcr/Abl-R6 for MDM2. Assays were performed on a MicroCal ITC 200 at 25° C. Concentrations of PMI and MDM2 were 100 μM and 10 μM, respectively. For the binding of PMIBcr/Abl-R6 to MDM2, the concentrations 80 μM and 8 μM, respectively. The results are provided in Table 1, where it can be seen that despite a small net gain in entropy for PMIBcr/Abl-R6 as opposed to a large loss for PMI, an expected outcome instigated by molecular grafting, PMIBcr/Abl-R6 lost a substantial amount of enthalpy for binding, suggesting that structurally rigidified PMI in the context of Bcr/Abl was energetically suboptimal for MDM2 binding. Nevertheless, ITC-based binding assays clearly validated the molecular design at the functional level of a protein antagonist of MDM2.
PMIBcr/Abl-R6 for MDM2 by isothermal titration calorimetry (ITC).
PMIBcr/Abl-R6 for MDM2
PMIBcr/Abl-R6 with enhanced proteolytic stability efficiently permeabilizes HCT116 p53+/+ tumor cells via an endocytosis-independent pathway.
As was demonstrated previously [39], PMI was inactive in killing HCT116 p53+/+ cells due to its poor proteolytic stability and inability to traverse the cell membrane. Accordingly, the proteolytic stability of free PMI and PMIBcr/Abl-R6 was compared in the presence of human serum (mainly serine proteases) or the intracellular cysteine protease cathepsin B. Intact peptide or protein was identified by mass spectrometry and quantified by RP-HPLC. As shown in
Peptide/protein internalization and cytosolic release were also examined using both confocal microscopy and flow cytometry. As shown in
Cationic cell penetrating peptides as a carrier are known to promote cellular uptake of cargos primarily through the non-endocytic uptake pathway, or direct membrane translocation, which is inhibited by heparin but not by amiloride [43, 44]. To better understand the mechanism of cellular uptake of PMIBcr/Abl-R6, confocal microscopic analysis was performed on cells treated with heparin or amiloride. As shown in
PMIBcr/Abl-R6 Kills HCT116 p53+/+ Tumor Cells In Vitro by Reactivating the p53 Pathway.
To evaluate the tumor-killing activity of PMIBcr/Abl-R6 in vitro, isogenic HCT116 p53+/+ and HCT116 p53−/− cell lines expressing abundant MDM2 [45, 46] were treated with the PMIBcr/Abl-R6 protein at concentrations from 1.56 μM to 50 μM. PMI and Bcr/Abl-R6 were used as a negative control and Nutlin-3, an extensively studied small molecule antagonist of MDM2 [47], as a positive control. As expected, while neither PMI nor Bcr/Abl-R6 had any effect on the viability of HCT116 cells 48 h after treatment (
To investigate into the mechanisms of action of PMIBcr/Abl-R6, the expression of p53, p21, PUMA and NOXA in HCT116 p53+/+ cells 48 h after treatment was analyzed by Western blotting. As shown in
Nanoparticles can actively accumulate in solid tumors through leaky blood vessels in diseased tissues—a phenomenon known as the enhanced permeability and retention (EPR) effect [50, 51]. To examine the biodistribution of PMIBcr/Abl-R6, the protein was fluorescently labeled with BDP TR and subcutaneously injected into BALB/c nude mice with palpable tumors grown from subcutaneously inoculated HCT116 p53+/+ cells. The biodistribution of PMIBcr/Abl-R6 in the heart, lung, spleen, kidney, liver and tumor at three different time points (12, 24, and 48 h) was semi-quantitatively evaluated on an in vivo optical imaging system. As shown in
PMIBcr/Abl-R6 Potently Inhibits Tumor Growth in Xenograft Mice by Inducing p53-Dependent Apoptotic Responses In Vivo.
To evaluate the therapeutic efficacy of PMIBcr/Abl-R6 in vivo, a nude mouse xenograft model was established where animals were subcutaneously inoculated with HCT116 p53+/+ cells (3×106). Thirty-six tumor-bearing mice were randomly divided into 6 groups (n=6) and received a 3-week subcutaneous treatment with medium, Nutlin-3, free PMI, Bcr/Abl-R6, PMIBcr/Abl and PMIBcr/Abl-R6 at the same dose of 5 mg/Kg every other day. As shown in
Consistent with the above findings from the in vivo efficacy study, histopathological analysis using hematoxylin and eosin (H&E) (
Immunogenicity of peptide/protein therapeutics often impedes their clinical use. The immunogenicity of PMI and PMIBcr/Abl-R6 in immune-competent C57BL/6 mice was evaluated by measuring the level of the cytokines IL-2, TNF-α and erythropoietin (EPO) in the blood in response to subcutaneous treatments with PMI and PMIBcr/Abl-R6 for three weeks, every other day, at a dose of 5 mg/Kg. IL-2 and TNF-α were used as markers because T cell responses are known to play a critical role in the development of immunogenic responses to therapeutic peptides and proteins [52, 53]. Since biotherapeutics can potentially generate cross-reactive neutralizing antibodies that inhibit endogenous proteins such as EPO, leading to anemia known as antibody-mediated pure red-cell aplasia, EPO was also used as a marker for immunogenicity in the study. As shown in
Some small molecule antagonists of MDM2 have showed cytotoxicity against B lymphocytes and hematopoietic stem cells in clinical trials, resulting in side effects such as thrombocytopenia, leukopenia and neutropenia [54]. The cytotoxicity profile of PMI, Bcr/Abl-R6, PMIBcr/Abl, PMIBcr/Abl-R6 and Nutlin-3 was established at the end of the three-week treatment by counting white blood cells, lymphocytes, monocytes, granulocytes, red blood cells, and platelets in a complete blood cell analysis. As shown in
Since PMIBcr/Abl-R6 accumulates in the liver and kidney in addition to the solid tumor (
This application claims priority to U.S. Provisional Patent Application No. 62/814,574, filed 6 Mar. 2019 and entitled “Tetrameric Protein Scaffolds as Nano-Carriers of Therapeutic Peptides for Treating Cancer and Other Diseases” in the name of Wuyuan L U et al., incorporated herein by reference in its entirety.
This invention was made with government support under CA219150 and CA167296 awarded by The National Institutes of Health. The government has certain rights in the invention.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US20/21407 | 3/6/2020 | WO | 00 |
Number | Date | Country | |
---|---|---|---|
62814574 | Mar 2019 | US |