Macrophages are distributed across all major organs, play central roles in normal immune homeostasis and disease progression, such as cancer, and are key regulators in the tumor microenvironment. In many tumors, anti-inflammatory macrophages (alternatively activated or M2 subtype), also known as tumor-associated macrophages (TAMs), are responsible for generating an immunosuppressive tumor microenvironment, which prevents the recognition and elimination of tumor cells by the immune system.
Described herein are molecules which preferentially bind to and target tumor associated macrophages (TAMs), pharmaceutical compositions thereof, and methods of treating and diagnosing the cancer and the immunosuppressive tumor microenvironment. Further provided, are pharmaceutical compositions comprising a tumor associated macrophage (TAM) binding molecule conjugated to a moiety, and a delivery agent and methods relating to the use of such pharmaceutical compositions for the treatment of cancer wherein the pharmaceutical composition is cytolytic to TAMs, removes, reduces and/or neutralizes TAMs, or repolarizes TAMs from an M2 phenotype to an M1 phenotype, methods relating to the use of such a pharmaceutical for the detection of cancer, and methods relating to the use of such a pharmaceutical for the detection of TAMs or a tumor microenvironment.
In one aspect, pharmaceutical compositions are provided. In some embodiments, the pharmaceutical compositions comprise a TAM binding molecule. In some embodiments, the pharmaceutical compositions comprise a TAM binding molecule conjugated to a moiety. In some embodiments, the pharmaceutical compositions comprise a TAM binding molecule conjugated to a moiety, and a binding agent. In some embodiments, the TAM binding molecule binds to retinoid X receptor beta on the TAM. In some embodiments, the TAM binding molecule is a peptide, ligand, antibody, non-IG domain, or small molecule entity. In some embodiments, the TAM binding molecule is an antibody or antigen-binding fragment thereof. In some embodiments, the antibody is an IgG, IgA, or IgM antibody. In some embodiments, the antibody is a single domain antibody. In some instances, the antibody is a chimeric, humanized, or human antibody. In other instances, the antigen binding fragment is a Fab, Fab′, Fab′-SH, Fv, scFv, F(ab′)2, or a diabody. In other embodiments, the TAM binding molecule is a peptide. In some instances, the TAM binding peptide is cyclic. In yet other instances, the cyclic TAM binding peptide comprises a) CRVLRSGSC, or b) CRVLRSGSC with at least one conservative amino acid substitution. In some embodiments, the moiety which is conjugated to the TAM binding molecule is a therapeutic agent or a diagnostic agent. In some instances, the moiety is a therapeutic agent wherein the therapeutic agent is a cytotoxic agent, a chemotherapeutic agent, a protein, a peptide, an antibody, a growth inhibitory agent, a nucleic acid, or an anti-hormonal agent. In other instances, the therapeutic is a cytotoxic agent wherein the cytotoxic agent is a ribosome inactivating protein, a histone deacetylase (HDAC) inhibitor, a tubulin inhibitor, an alkylating agent, an antibiotic, an antineoplastic agent, an antiproliferative agent, an antimetabolite, a topoisomerase I or II inhibitor, a hormonal agonist or antagonist, an immunomodulator, a DNA minor groove binder, or a radioactive agent. In other embodiments, the moiety is a diagnostic agent wherein the diagnostic agent is a label. In some instances, the diagnostic agent is a label wherein the label is a fluorescent label, a chromogenic label, or a radiolabel. In some embodiments, the pharmaceutical composition is comprised of the TAM binding molecule directly conjugated to the moiety. In other embodiments, the pharmaceutical composition is comprised of the TAM binding molecule indirectly conjugated to the moiety via a linker. In some instances, the delivery agent is a liposome, microsphere, nanoparticle, microemulsion, microcapsule, polymer matrix, hydrogel, or viral vector.
In another aspect, a method of treating cancer is provided. The method generally includes administering to a subject a pharmaceutical composition as described above. In some embodiments, the TAM binding molecule is cytolytic to tumor cells. In some embodiments, the TAM binding molecule inhibits tumor growth. In some embodiments, the method of treating cancer is provided wherein the cancer is selected from the group consisting of brain cancer, renal cancer, ovarian cancer, prostate cancer, lymphoma, breast cancer colon cancer, lung cancer, squamous cell carcinoma of the head and neck, and melanoma. In some embodiments, the method is performed wherein the pharmaceutical composition is administered topically, subcutaneously, intravenously, intradermally, intraperitoneally, orally, intramuscularly, or intracranially. In some embodiments, the method is performed wherein the pharmaceutical composition is administered in combination with a second therapeutic agent. In further embodiments, the method is performed in combination with a second therapeutic agent wherein the second therapeutic agent is a cancer chemotherapeutic agent, radiation therapy, a cytotoxic agent, another antibody, a NSAID, a corticosteroid, a dietary supplement (e.g. an antioxidant), or a combination thereof.
In another aspect, a method of reducing the number of TAMS in a tumor microenvironment in a subject having cancer is provided. This method generally includes a pharmaceutical composition described above wherein the pharmaceutical composition is cytolytic to TAMs.
In another aspect, a method of removing immunosuppression in a tumor microenvironment in a subject having cancer is provided. This method generally includes a pharmaceutical composition described above wherein the pharmaceutical composition removes, reduces, and/or neutralizes TAMs.
In another aspect, a method of repolarizing TAMS from an M2 phenotype to an M1 phenotype in a subject having cancer is provided. This method generally includes a pharmaceutical composition described above wherein the pharmaceutical composition repolarizes TAMs from an M2 phenotype to an M1 phenotype.
In another aspect, a method of detecting cancer in a subject is provided. This method generally includes administering to a subject thereof a pharmaceutical composition as described.
In another aspect, a method of detecting TAMS in a tumor microenvironment in a subject having cancer is provided. This method generally includes administering to a subject thereof a pharmaceutical composition as described above.
In another aspect, a method of detecting a tumor microenvironment in a subject is provided. This method generally includes administering to a subject thereof a pharmaceutical composition as described above.
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
TABLE 1 is a summary of tumors tested for CRV in vivo homing.
In vivo, the tumor microenvironment is a complex milieu containing multiple cell types including tumor cells, vascular cells such as endothelial cells, and stromal cells, such as fibroblasts. In addition, in vivo, these cells are exposed to blood flow and various biological transport conditions. In vivo, microvascular cells in a tumor are affected by blood flow and communicate with tumor and non-tumor cells, both physically and through diffusible factors. In addition, the tumor vasculature is abnormal, characterized by chaotic branching, a low flow rate, and leaky vessels, and thus serves as a major transport barrier to anticancer therapies that target tumor cells. The interplay between tumor cells, endothelial cells, and stromal cells affects each cell type, leading to increased angiogenesis and tumor cell proliferation, and this crosstalk may be an important factor in determining the responsiveness of tumor cells to anticancer drugs.
In the tumor microenvironment cells experience oxygen and nutrient deprivation. Hypoxic stress alters the metabolism of tumor cells and macrophages within tumors with subsequent changes in the microenvironment. The change in microenvironment alters the phenotype and metabolism of macrophages to induce a tumor-promoting reprogramming. Nutrient stress also provokes autophagy to guarantee cell survival or to induce cell death. Death of tumor cells is communicative system attracting macrophages and directing their phenotype. Depending on the mode of tumor cell death macrophage polarization ranges from pro-inflammatory activation to anti-inflammatory/immuno-suppressive activation.
Chronic inflammation contributes to cancer development. The presence and activation of chronic innate immune cell types (e.g., neutrophils, macrophages, and mast cells) promote cancer development. Thus it is clear that some subsets of chronically activated innate cells promote the growth and/or facilitate the survival of neoplastic cells. Depending on their polarization status, immune cells can exert either antitumor (e.g. T-helper 1 (Th1) vs Th17 subsets of CD4(+) T cells, type I) or protumor (e.g. vs type II NKT cells, M1 vs M2 macrophages, and N1 vs N2 neutrophils) functions. Chronically activated and polarized immune cells (e.g. M2 macrophages and N2 neutrophils) produce or carry a myriad of chemokines, cytokines, growth factors, and proteases leading to tissue remodeling, angiogenesis, cell proliferation, genomic instability, and expansion of neoplastic cells into ectopic tissue.
Macrophages are a type of white blood cell, of the immune system, that engulfs and digests cellular debris, foreign substances, microbes, cancer cells, and anything else that does not have the type of proteins specific to healthy body cells on its surface in a process called phagocytosis. Macrophages increase inflammation and stimulate the immune system. Macrophages also play an important anti-inflammatory role and can decrease immune reactions through the release of cytokines. Macrophages that encourage inflammation are called M1 macrophages, whereas macrophages that decrease inflammation and encourage tissue repair are called M2 macrophages. Macrophages are a dominating immune cell population in most solid tumors. Tumor-associated macrophages (TAMs) are a type of macrophage. TAMs are thought to acquire an M2 phenotype, contributing to tumor growth and progression. TAMs can regulate tumor progression. Therapeutic strategies to reduce the number of TAMs or to manipulate the TAM phenotype are of interest to cancer therapy.
Retinoid X receptors (RXR) are members in the superfamily of nuclear receptor and have essential nuclear and cytoplasmic functions as transcription factors. RXR is a type of nuclear receptor that is activated by 9-cis retinoic acid, which is contemplated to be of endogenous relevance. RXR is also activated by 9-cis-13,14-dihydro-retinoic acid, may be the major endogenous mammalian RXR-selective agonist. There are three retinoic X receptors (RXR): RXR-alpha (RXRA), RXR-beta (RXRB), and RXR-gamma (RXRG), encoded by the RXRA, RXRB, RXRG genes, respectively. RXR is a hetero-dimerization partner for the members of the subfamily II nuclear receptors which regulate the transcription of numerous target genes, following chemical activation. RXR can heterodimerize with subfamily I nuclear receptors including CAR, FXR, LXR, PPAR, PXR, RAR, TR, and VDR. The RXR heterodimer in the absence of ligand is bound to hormone response elements complexed with corepressor protein. Binding of agonist ligands to RXR results in dissociation of corepressor and recruitment of coactivator protein. This promotes transcription of the downstream target gene into mRNA and eventually protein. RXR can regulate macrophages in inflammatory and metabolic disorders and there is potential for direct modulation of RXR signaling to treat macrophage-related pathologies. The cell surface expression of RXRB is specific to TAMs and RXRB is responsible for the preferential binding of CRV and other contemplated molecules to tumor macrophages.
Molecules that Target TAMs
The peptide CRV (CRVLRSGSC) is a cyclic macrophage-targeting peptide with a disulfide bond between the terminal cysteine residues. CRV selectively homes to tumors and binds with TAMs within said tumors. CRV recognizes and binds to RXRB on the surface of TAMs within tumors. CRV only recognizes TAMs, but not macrophages in atherosclerotic plaques.
CRV can be linked to a therapeutic agent. The therapeutic agent can comprise antibodies (e.g. IgG, IgA, or IgM).
CRV can be modified to produce a related peptide which is contemplated to also act as a TAM molecule. Examples of such a modification include one or more substitutions, deletions, or additions of amino acids. Conservative substitutions include amino acid substitutions that substitute a given amino acid with another amino acid of similar characteristics and further include, among the aliphatic amino acids interchange of alanine, valine, leucine, and isoleucine; interchange of the hydroxyl residues serine and threonine, exchange of the acidic residues aspartate and glutamate, substitution between the amide residues asparagine and glutamine, exchange of the basic residues lysine and arginine, and replacements among the aromatic residues phenylalanine and tyrosine. In some embodiments CRV is modified with one or more amino acid conservative substitutions. In other embodiments, CRV is modified with one amino acid conservative substitution.
Other TAM binding molecules which can act as a cell surface RXRB binding molecule to selectively bind TAMs comprise antibodies (e.g. IgG, IgA, or IgM), antigen binding fragments, peptides, ligands, non-IG domains, or small molecules. Said TAM binding molecules may bind RXRB at different locations on the RXRB molecule. If the TAM binding molecule is an antibody, it can be a single domain antibody, chimeric antibody, humanized antibody, human antibody, and/or monoclonal antibody. If the TAM binding molecule is an antigen binding fragment, it can be a Fab, Fab′, Fab′-SH, Fv, scFv, F(ab′)2, or a diabody. If the TAM binding molecule is a peptide, it can be cyclic. If the TAM binding molecule is cyclic, it can be CRV or another peptide.
Modifications to the TAM binding molecules are contemplated. These modifications include conjugation to an additional moiety. This moiety can be a therapeutic agent or a diagnostic agent to be used clinically or for research purposes.
These modifications can further constitute a delivery system, which can deliver the TAM binding molecules in a targeted fashion to the tumor or tumor microenvironment. Benefits of this delivery system can include reduction in the frequency of the dosages taken by a patient, a uniform effect of the drug, reduction of drug side-effects, and reduction of fluctuation in circulating drug levels.
Said moiety or TAM binding molecule or both can be held in a delivery agent. Said delivery agent can be a liposome, microsphere, nanoparticle, microemulsion, microcapsule, polymer matrix, hydrogel, or viral vector.
Liposomes are non-toxic, non-hemolytic, and non-immunogenic even upon repeated injections; they are biocompatible and biodegradable and can be designed to avoid clearance mechanisms (reticuloendothelial system (RES), renal clearance, chemical or enzymatic inactivation, or other undesirable effects. Lipid-based, ligand-coated nanocarriers can store their payload in a hydrophobic shell or a hydrophilic interior depending on the nature of the agent being carried.
Microspheres can encapsulate many types of drugs including small molecules, proteins, and nucleic acids and are easily administered through a syringe needle. Microspheres are generally biocompatible, can provide high bioavailability, and are capable of sustained release for long time periods. Disadvantages of microspheres include difficulty of large-scale manufacturing, inactivation of drug during fabrication, and poor control of drug release rates.
Nanoparticle-based drug delivery systems are contemplated to be a useful method for delivering agents. Some benefits of using nanoparticles include controllable release of the payload into the cytoplasm and circumvention of tumor drug resistance.
Microemulsions are isotropic, thermodynamically stable transparent (or translucent) systems of oil, water and surfactant, frequently in combination with a cosurfactant with a droplet size usually in the range of 20-200 nm. Said microemulsions can be classified as oil-in-water (o/w), water-in-oil (w/o) or bicontinuous systems depending on their structure and are characterized by ultra-low interfacial tension between oil and water phases. Said systems are currently of great technological and scientific interest to the researchers because of their potential to incorporate a wide range of drug molecules (hydrophilic and hydrophobic) due to the presence of both lipophilic and hydrophilic domains. These adaptable delivery systems provide protection against oxidation, enzymatic hydrolysis and improve the solubilization of lipophilic drugs and hence enhance their bioavailability. Microemulsions are suitable for oral and intravenous delivery systems and for sustained and targeted delivery (e.g. through ophthalmic, dental, pulmonary, vaginal and topical routes). Microemulsions have been used to improve the oral bioavailability of various poorly soluble drugs. In some instances, microemulsions are employed for challenging tasks such as carrying chemotherapeutic agents to neoplastic cells and oral delivery of insulin.
Microcapsules are particles with a diameter of 1-1000 μm, irrespective of the precise interior or exterior structure, which can be used for agent delivery. Microcapsules offer various significant advantages as drug delivery systems (e.g. an effective protection of the encapsulated active agent against (e.g. enzymatic) degradation, the possibility to accurately control the release rate of the incorporated drug over periods of hours to months, an easy administration (compared to alternative parenteral controlled release dosage forms, such as macro-sized implants), and desired, pre-programmed drug release profiles can be provided which match the therapeutic needs of the patient.
Agents can be embedded into a polymeric matric or co-crystallized with a polymeric template. Polymers are a drug delivery technology which provide controlled release of therapeutic agents in constant doses over long periods, cyclic dosage, and tunable release of both hydrophilic and hydrophobic drugs. Polymer Matrices may be tailored for specific cargo and engineered to exert distinct biological functions.
Hydrogels are three-dimensional, cross-linked networks of water-soluble polymers. Hydrogels can be made from virtually any water-soluble polymer, encompassing a wide range of chemical compositions and bulk physical properties. Furthermore, hydrogels can be formulated in a variety of physical forms, including slabs, microparticles, nanoparticles, coatings, and films. As a result, hydrogels are commonly used in clinical practice and experimental medicine for a wide range of applications, including tissue engineering and regenerative medicine, diagnostics, cellular immobilization, separation of biomolecules or cells, and barrier materials to regulate biological adhesions
Virus delivery vectors are a type of nanomaterial which can be a drug delivery material. A successful vector must be able to effectively carry and subsequently deliver a drug cargo to a specific target.
Said conjugation can be achieved directly or indirectly, and with or without a linker molecule. This linker molecule can be a pH sensitive linker, a disulfide linker, a peptide linker, a beta-glucoronide linker, a redox responsive linker, a hydrazone linker, a hydrophilic linker, an azo linker, or another type of linker. Said linker can respond to a stimulus to initiate drug release. Said stimulus can be internal or external. Said stimulus can be local. Said stimulus can be pH, enzyme, light, heat, or another stimulus.
If the TAM binding molecule is conjugated to a moiety which is a therapeutic agent, said therapeutic agent can be a cytotoxic agent (e.g. ribosome inactivating protein, histone deacetylase inhibitor, tubulin inhibitor, alkylating agent, antibiotic, antineoplastic agent, antiproliferative agent, antimetabolite, topoisomerase I or II inhibitor, hormonal agonist or antagonist, immunomodulator, DNA minor groove binder or radioactive agent), a chemotherapeutic agent (e.g. alkylating agent, anthracycline, taxane, epothilone, topoisomerase I or II inhibitor, histone deacetylase inhibitor, kinase inhibitor, nucleotide analog, peptide antibiotic, platinum based agent, retinoid, vinca alkaloid or derivative, or other chemotherapeutic agent), a protein, a peptide, an antibody, a growth inhibitory agent, a nucleic acid, or an anti-hormonal agent.
If the TAM binding molecule is conjugated to a moiety which is a diagnostic agent, said diagnostic agent can be a label, and said label can be a fluorescent label, a chromogenic label, or a radiolabel. Said label is contemplated to be used for diagnostic imaging (e.g. PET, or MRI imaging or an imaging protocol incorporating PET or MRI imaging).
RAW (tumor-derived mouse macrophage cell line RAW264.7), J774 (tumor-derived mouse macrophage cell line J774A. 1), THP-1 differentiated macrophages (human macrophages differentiated from the human monocytic cell line THP-1) and 4T1 (mouse breast cancer cell line) cells were cultured in DMEM supplemented with 10% FCS. 1×106 cells were incubated with FAM-CRV or the control peptide FAM-GGS (10 μM) in 300 μL of complete growth medium in an Eppendorf tube. After incubation at 4° C. for 1 h, the peptide-containing medium was removed by centrifugation and the cells were washed with PBS two times. 100 μL PFA (4% buffer) was then added to the cells for fixation and flow cytometry data were acquired on FACSCanto (BD Biosciences, San Jose). Experiment was repeated three times on different days.
FAM-CRV showed a much higher binding to RAW, J774, and THP-1 differentiated macrophages than did the control peptide FAM-GGS (
To investigate if CRV can bind to macrophages in vivo, FAM-CRV was intravenously injected into tumor bearing mice. To this end, 4T1 and MCF10CA1a human breast cancer cells and KRAS-Ink mouse PDAC cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin. Py8119 cells were cultured in Ham's F12K medium containing 5% FCS, 2.5 μg/mL amphotericin B, 50 μg/mL gentamycin, and MITO+. The human cell lines were authenticated by the DNA Analysis Core Facility at the Sanford Burnham Prebys Medical Discovery Institute (La Jolla, Calif.) and the KRAS-Ink cell line was authenticated by DDC Medical (Fairfield, Ohio). All cells were tested negative for mycoplasma contamination. To produce 4T1 tumors, 1×106 tumor cells (suspended in 100 μL of PBS) were orthotopically injected into the mammary fat pad of normal BALB/c mice. To produce MCF10CA1a tumors, 2×106 tumor cells (suspended in 100 μL of PBS) were injected into the mammary fat pads of female BALB/c athymic nude mice. To produce Py8119 tumors, 1×106 tumor cells (suspended in 100 μL of PBS) were orthotopically injected into the mammary fat pads of C57BL6 mice. To produce KRAS-Ink PDAC tumors, 1×106 cells (suspended in 100 μL of PBS) were injected into female BALB/c mice. To produce H1975 tumors, 1×106 cells (suspended in 100 μL of PBS) were injected into (What kind of mouse? Where was the injection?) All animal experimentation received approval from the Animal Research Committee of Sanford Burnham Prebys Medical Discovery Institute.
Biodistribution of fluorescein-conjugated peptides (FAM-CRV, FAM-GGS, or FAM-ARA) was examined after intravenous injection of 100 μL peptide solution (1 mg/mL PBS) into the tail vein of a mouse. The peptide was allowed to circulate for 1 h and transcardial perfusion was performed with PBS. Tissues were collected, fixed with 4% formaldehyde buffer solution and then soaked in 30% sucrose in PBS overnight.
The tumor models tested in our study were summarized in Table 1. FAM-CRV homing was found positive in orthotopic 4T1 breast cancer, orthotopic MCF10CA1a breast cancer, subcutaneous kras-INK pancreatic cancer, subcutaneous KPC pancreatic cancer, and subcutaneous H1975 lung cancer. Compared to other organs, the tumors displayed a strong fluorescent signal under UV illumination.
Representative images of FAM-CRV 1-h homing in the 4T1 breast cancer mouse model is shown in
FAM-CRV was intravenously injected into tumor bearing mice. To this end, 4T1 human breast cancer cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin. The human cell line was authenticated by the DNA Analysis Core Facility at the Sanford Burnham Prebys Medical Discovery Institute (La Jolla, Calif.) and tested negative for mycoplasma contamination. To produce 4T1 tumors, 1×106 tumor cells (suspended in 100 μL of PBS) were orthotopically injected into the mammary fat pad of normal BALB/c mice. All animal experimentation received approval from the Animal Research Committee of Sanford Burnham Prebys Medical Discovery Institute.
Biodistribution of fluorescein-conjugated peptides (FAM-CRV, FAM-GGS, or FAM-ARA) was examined after intravenous injection of 100 μL peptide solution (1 mg/mL PBS) into the tail vein of a mouse. The peptide was allowed to circulate for 1 h and transcardial perfusion was performed with PBS. Tissues were collected, fixed with 4% formaldehyde buffer solution and then soaked in 30% sucrose in PBS overnight. Tissues were finally frozen in OCT embedding medium (Tissue-Tek), and sliced for immunofluorescence staining.
Tissue sections were blocked in 1% bovine serum albumin with 0.1% Triton X-100 for 1 h, and incubated with appropriate primary antibodies and second antibodies. Blood vessels were visualized by staining tissue sections with monoclonal antibodies against CD-31. The primary antibody was rat anti-mouse CD31 (BD Biosciences). The secondary antibody was 594 donkey anti-rat IgG were from Invitrogen. After washing with PBS, sections were mounted in DAPI-containing mounting medium (Vector Laboratories, Burlingame, Calif.) and examined under a Zeiss LSM 710 NLO confocal microscope.
Immunofluorescence staining was performed for FAM-CRV and CD31 on all tissue sections. A representative image from 4T1 breast cancer model is shown in
To determine whether CRV targeted to macrophages or other cell types in the tumor, the tissue sections of 4T1 tumor-bearing mice were also stained for macrophages markers including CD11b, F4/80, and CD68 To this end, 4T1 human breast cancer cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin. The human cell line was authenticated by the DNA Analysis Core Facility at the Sanford Burnham Prebys Medical Discovery Institute (La Jolla, Calif.) and tested negative for mycoplasma contamination. To produce 4T1 tumors, 1×106 tumor cells (suspended in 100 μL of PBS) were orthotopically injected into the mammary fat pad of normal BALB/c mice. All animal experimentation received approval from the Animal Research Committee of Sanford Burnham Prebys Medical Discovery Institute.
Biodistribution of fluorescein-conjugated peptides (FAM-CRV, FAM-GGS, or FAM-ARA) was examined after intravenous injection of 100 μL peptide solution (1 mg/mL PBS) into the tail vein of a mouse. The peptide was allowed to circulate for 1 h and transcardial perfusion was performed with PBS. Tissues were collected, fixed with 4% formaldehyde buffer solution and then soaked in 30% sucrose in PBS overnight. Tissues were finally frozen in OCT embedding medium (Tissue-Tek), and sliced for immunofluorescence staining.
Tissue sections were blocked in 1% bovine serum albumin with 0.1% Triton X-100 for 1 h, and incubated with appropriate primary antibodies and second antibodies. The primary antibodies were rat anti-mouse CD11b (BD Biosciences), rat anti-mouse F4/80 monoclonal (BD Biosciences), and rabbit anti-fluorescein/Oregon Green (Invitrogen) polyclonal antibodies. The secondary antibodies, Alexa Fluor 488 goat anti-rabbit IgG and 594 donkey anti-rat IgG were from Invitrogen. After washing with PBS, sections were mounted in DAPI-containing mounting medium (Vector Laboratories, Burlingame, Calif.) and examined under a Zeiss LSM 710 NLO confocal microscope.
Mice with tumor size about 8 mm were euthanized under deep anesthesia (Avertin, lack of response to a toe pinch) by cervical dislocation. Target (tumor) and control (e.g., liver, spleen) tissues were collected and further dissociated to single cells. Tumor cells were dissociated using MACS tumor dissociation kit. Cells were then incubated with fluorescent-labeled CRV or other cell markers (CD11b, CD68, F4/80, FAB, EpCAM, CD31) at 4° C. for 1 h. Positive cells were quantified on BD LSRFORTESSA and data were analyzed using FCS Express Version 3 (De Novo Software).
FAM-CRV signals strongly overlapped with those macrophages markers as shown in
We further carried out flow cytometry analysis of dissociated 4T1 tumor cells after FAM-CRV in vivo homing (15, or 60 min post intravenous administration). To this end, FAM-CRV was intravenously injected into tumor bearing mice To this end, 4T1 human breast cancer cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin. The human cell line was authenticated by the DNA Analysis Core Facility at the Sanford Burnham Prebys Medical Discovery Institute (La Jolla, Calif.) and tested negative for mycoplasma contamination. To produce 4T1 tumors, 1×106 tumor cells (suspended in 100 μL of PBS) were orthotopically injected into the mammary fat pad of normal BALB/c mice. All animal experimentation received approval from the Animal Research Committee of Sanford Burnham Prebys Medical Discovery Institute.
Fluorescein-conjugated peptides (100 μL, 1 mg/mL PBS) were intravenously injected into the tail vein of tumor-bearing mice. The peptide was allowed to circulate for 1 h. Mice were then euthanized under deep anesthesia (Avertin, lack of response to a toe pinch) by cervical dislocation. Tumors were collected and further dissociated to single cells, and incubated with different cell markers (CD11b, CD68, F4/80, FAB, EpCAM, CD31) at 4° C. for 1 h. Positive cells were quantified on BD LSRFORTESSA and data were analyzed using FCS Express Version 3 (De Novo Software).
The tumor cells from animals with both circulation lengths exhibited higher FAM-CRV signal compared to the non-injected control animals (
As macrophages reside in other disease sites, such as atherosclerotic plaques, it is interesting to know whether CRV recognizes other pathological tissues containing macrophages. Therefore, in vivo homing of FAM-CRV was tested in ApoE −/− mice bearing atherosclerotic plaques in the aorta. Unlike LyP1, which homes to and bind to the macrophages both in tumors and plaques through the p32 receptor, FAM-CRV did not home to aortic plaques upon systemic administration (
The difference between CRV and LyP-1 suggests that CRV probably binds to a different receptor for TAM recognition. Therefore, the CRV receptor on macrophages was identified. To reduce the complexity, the membrane fraction of RAW cells was isolated, and performed affinity chromatography was performed to isolate putative CRV receptors. To this end, CRV was immobilized on a column. Lysates of 4T1 tumor tissue and membrane proteins of RAW cells (isolated with the Mem-PER Plus kit from Thermo Fisher) were utilized as two independent sources of putative receptors. After washing with buffer and the control peptide GGS, the putative receptor was eluted with an excess of free CRV peptide. Mass spectrometry analysis then identified the retinoid X receptor beta (RXRB) as a receptor candidate.
To confirm RXRB binds CRV, the binding of FAM-CRV to human recombinant RXRB protein immobilized on a plate was tested. Human RXRB recombinant protein (50 μL, 5 μg/mL) was immobilized in high binding 96-well plate at 4° C. overnight. Bovine serum albumin was used as the control protein. The protein solution was removed and washed one time with PBS. 1% BSA in PBS solution was then added to the wells and incubated at room temperature for 1 h to block the rest available binding sites. After one wash with PBS, RXRB and BSA were incubated with FAM-CRV (100 μL, 1 μM) at room temperature for 1 h. The wells were washed three times with PBS and the fluorescence was measured using a multiplate reader.
Compared to the control peptide FAM-GGS, there was significantly higher binding of CRV peptide to RXRB (
In addition, RXRB immunohistochemical (TIC) staining on 4T1 tumor tissues was performed. To this end, 4T1 human breast cancer cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 g/mL streptomycin. The human cell line was authenticated by the DNA Analysis Core Facility at the Sanford Burnham Prebys Medical Discovery Institute (La Jolla, Calif.) and tested negative for mycoplasma contamination. To produce 4T1 tumors, 1×106 tumor cells (suspended in 100 μL of PBS) were orthotopically injected into the mammary fat pad of normal BALB/c mice. All animal experimentation received approval from the Animal Research Committee of Sanford Burnham Prebys Medical Discovery Institute.
Tissue sections were blocked in 1% bovine serum albumin with 0.1% Triton X-100 for 1 h, and incubated with appropriate primary antibodies and second antibodies. The primary antibodies were rat anti-mouse CD11b (BD Biosciences), rat anti-mouse F4/80 monoclonal (BD Biosciences), and rabbit anti-fluorescein/Oregon Green (Invitrogen) polyclonal antibodies. The secondary antibodies, Alexa Fluor 488 goat anti-rabbit IgG and 594 donkey anti-rat IgG were from Invitrogen. After washing with PBS, sections were mounted in DAPI-containing mounting medium (Vector Laboratories, Burlingame, Calif.) and examined under a Zeiss LSM 710 NLO confocal microscope.
This experiment showed that RXRB co-localized with FAM-CRV as well as the macrophage markers (CD11b, F4/80, and CD68) (
To further investigate the correlation between CRV binding to macrophages and the surface expression of RXRB, ex vivo binding of CRV and RXRB antibody to dissociated tumor cells from 4T1 tumor models was performed. Mice with tumor size about 8 mm were euthanized under deep anesthesia (Avertin, lack of response to a toe pinch) by cervical dislocation. Target (tumor) and control (e.g., liver, spleen) tissues were collected and further dissociated to single cells. Tumor cells were dissociated using MACS tumor dissociation kit. Cells were then incubated with fluorescent-labeled CRV or other cell markers (CD11b, F4/80) at 4° C. for 1 h. Positive cells were quantified on BD LSRFORTESSA and data were analyzed using FCS Express Version 3 (De Novo Software).
Flow cytometry analyses showed that about 15% cells from the tumor were RXRB positive and FAM-CRV primarily bound to cells with high RXRB surface expression (
The good correlation of RXRB and CRV lead us to the interest to investigate the surface expression of RXRB in different tissues, which could potentially explain the biodistribution of CRV in vivo. RXRB antibody (or Rabbit IgG as control) and FAM-CRV was both intravenously injected (RXRB antibody: 4 h circulation; FAM-CRV: 1 h circulation) to the same mice, and the presentation of RXRB was assessed after transcardial perfusion. Tissue sections were blocked in 1% bovine serum albumin with 0.1% Triton X-100 for 1 h, and incubated with appropriate primary antibodies and second antibodies. Blood vessels were visualized by staining tissue sections with monoclonal antibodies against CD-31. The primary antibodies were rat anti-mouse CD31 (BD Biosciences), rat anti-mouse CD11b (BD Biosciences), rat anti-mouse F4/80 monoclonal (BD Biosciences), and rabbit anti-fluorescein/Oregon Green (Invitrogen) polyclonal antibodies. The secondary antibodies, Alexa Fluor 488 goat anti-rabbit IgG and 594 donkey anti-rat IgG were from Invitrogen. After washing with PBS, sections were mounted in DAPI-containing mounting medium (Vector Laboratories, Burlingame, Calif.) and examined under a Zeiss LSM 710 NLO confocal microscope.
Immunofluorescence staining of the tissue sections showed that major organs had different levels of RXRB expression. RXRB can be found in tumor, liver, spleen and kidney, but not in heart and lung, while the IgG control staining was not observable in any organs. FAM-CRV signal can only be identified in tumor and kidney after its 1 h circulation. In the tumor, where RXRB staining was the highest among all the organs, RXRB showed nice colocalization with FAM-CRV and macrophage markers, best with CD11b, followed by CD68, and then F4/80 (
50 μg rabbit anti-mouse RXRB antibody (GeneTex, diluted in PBS, total volume was 100 μL) was intravenously injected to each mouse (n=3). 50 μg rabbit IgG per mouse was injected to the control group (n=3). Three hours later, FAM-CRV (100 μg in 100 μL) were injected to all animals. After 1 h circulation for FAM-CRV, animals were sacrificed with transcardial perfusion. Tissues were collected, fixed and sliced as described above.
When RXRB antibody was intravenously injected alone to 4T1 mice (also 4 h circulation), the apparent distribution of RXRB was the same as with FAM-CRV co-injection, indicating that the FAM-CRV presence did not affect the RXRB recognition by the antibody. In other words, RXRB antibody did not block CRV binding to RXRB in vivo, nor vice versa. This indicated that FAM-CRV bound to a different binding site as the RXRB antibody, which was in agreement as what was observed in the in vitro binding studies. These in vivo homing results demonstrated that RXRB is a good surface marker with its high specific expression on tumor macrophages. It also showed that RXRB antibody can preferentially home to tumors upon intravenous injection, penetrate the blood vessels and bind to macrophages in the extravascular regions, and function as a targeting agent.
To investigate the translational potential of the CRV-targeting approach, CRV-mediated tumor homing of nanoparticles was evaluated. There has been extensive works showing porous silicon nanoparticles (pSiNP) as therapeutic carriers with controlled loading and release of various types of drugs (24, 25). Their high drug loading efficiency and time-gated imaging property make them particularly desirable as drug carriers.
As a proof of concept study, CRV-conjugated porous silicon nanoparticles (CRV-pSiNPs) were loaded with a red fluorescence probe, SR101 and the nanoparticles showed tumor homing effect in 4T1 tumor-bearing mice. 100-200 uL of a solution that contains CRV peptide, CRV-pSiNP, or control non-specific peptide was injected into the tail vein of tumor mice with a tumor size of about 8 mm. After 5 min, to up to 24 hours of circulation, the mice will be anesthetized and imaged with an imaging equipment (e.g., Xenogen IVIS, Lightools' light table, multiphoton LSM). When available as with Xenogen IVIS, the mice will be anesthetized with isoflurane during the imaging procedures (Induction: Flow rate 0.8-1.5 L/min, isoflurane vaporizer 2-3%, Maintenance: Flow rate 0.4-0.8 L/min, isoflurane vaporizer 2-3%). The mice will be then perfused with PBS. The tumors and control organs will be collected and used for histological characterization and immunohistochemical assessment.
The ex vivo IVIS imaging showed that there was higher SR101 intensity in the tumor of animals that received CRV-pSiNP compare to the control NP (
While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.
This application claims the benefit of U.S. Provisional Patent Application No. 62/717,656 filed on Aug. 10, 2018, which is incorporated herein by reference in its entirety.
This invention was made with government support under R21 EB022652 awarded by the National Institutes of Health (NIH). The government has certain rights in the invention.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2019/045676 | 8/8/2019 | WO | 00 |
Number | Date | Country | |
---|---|---|---|
62717656 | Aug 2018 | US |