CN116744909A - Metal organic frameworks deliver small and biological macromolecules for cancer immunotherapy - Google Patents

Metal organic frameworks deliver small and biological macromolecules for cancer immunotherapy Download PDF

Info

Publication number
CN116744909A
CN116744909A CN202180060168.XA CN202180060168A CN116744909A CN 116744909 A CN116744909 A CN 116744909A CN 202180060168 A CN202180060168 A CN 202180060168A CN 116744909 A CN116744909 A CN 116744909A
Authority
CN
China
Prior art keywords
mof
dbb
metal
optionally
dbp
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202180060168.XA
Other languages
Chinese (zh)
Inventor
林文斌
倪开元
罗韬堃
蓝光旭
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Chicago
Original Assignee
University of Chicago
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 University of Chicago filed Critical University of Chicago
Priority claimed from PCT/US2021/033886 external-priority patent/WO2021237209A1/en
Publication of CN116744909A publication Critical patent/CN116744909A/en
Pending legal-status Critical Current

Links

Abstract

Modified Metal Organic Frameworks (MOFs) are described that have surfaces that have the ability to coordinately bind or electrostatically interact with therapeutic agents such as nucleic acids and small molecules and proteins having phosphate or carboxylate groups. Also described is a method of providing a modified MOF comprising replacing a strongly coordinated metallo-oxygen cluster capping group with a weakly coordinated capping group and/or introducing an organic bridging ligand having an electron withdrawing group. Also described are MOFs having surface-attached therapeutic agents (e.g., immunotherapeutic agents) prepared from modified MOFs, and methods of treating cancer using MOFs, e.g., by radiotherapy-radiokinetic therapy (RT-RDT), with or without co-administration of another therapeutic agent, such as a chemotherapeutic agent or an immunomodulatory agent. Thus, the methods described may involve cancer immunotherapy and in situ cancer vaccination.

Description

Metal organic frameworks deliver small and biological macromolecules for cancer immunotherapy
Citation of related applications
The presently disclosed subject matter claims to be entitled to U.S. provisional patent application Ser. No. 63/028,891 filed on day 22 of 5 in 2020; and U.S. provisional patent application Ser. No. 63/045,499, filed on 6/29 of 2020, the disclosures of each of which are incorporated herein by reference.
Government rights and interests
The invention is completed under government funding of grant item number CA253655 granted by national institutes of health and grant item number PC170934P2 granted by national defense. The government has certain rights in this invention.
Technical Field
The presently disclosed subject matter provides compositions and methods for treating cancer. In particular, the presently disclosed subject matter relates to Metal Organic Framework (MOF) nanomaterials having modified surfaces for enhancing binding to various small molecules, peptides, proteins, and nucleic acid therapeutics. The presently disclosed subject matter also relates to MOFs with therapeutic agent surface modifications and their use in the treatment of cancer by, for example, activating an anti-tumor immune system.
Abbreviations (abbreviations)
C = degrees celsius
Percent =percent
μg = microgram
Mu l = microliter
μmol = micromolar
μΜ = micromolar concentration
AFM = atomic force microscope
Apc=antigen presenting cell
APF = aminophenyl fluorescein
bpy=2, 2' -bipyridine
CBI = checkpoint blocking immunotherapy
cGAMP = cyclic guanosine-phosphate adenosine)
CLSM = confocal laser scanning microscope
cm = cm
CRT = calreticulin
CTL = cytotoxic T lymphocytes
Cu=copper
DAMPs = risk related molecular pattern
Dbb=4, 4 '-bis (4-benzoic acid) -2,2' -bipyridine
Dbp=5, 15-bis (p-toluic acid) porphyrin
DC = dendritic cell
dF(CF 3 ) ppy=2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine
DMF = dimethylformamide
DMSO = dimethyl sulfoxide
eV = electron volts
g=g
Gy=gray
h=h
Hf=hafnium
IC 50 =50% inhibitory concentration
ICD = immunogenic cell death
ICP-ms=inductively coupled plasma-mass spectrometry
Ifn=interferon
IL = interleukin
Imd=imiquimod
Ir=iridium
kg=kg
kvp=peak kilovolts
ma=milliamp
mg = mg
min = min
mL = milliliter
mm = millimeter
mM = millimolar concentration
mmol = millimoles
Mof=metal organic framework
Mol=metal organic layer
MOP = metal organic nanoplate
MUC-1 = mucin-1
mv=millivolt
ng=nanogram
NIR = near infrared
nm=nm
nMOF=nano metal organic frameworks
Nmr=nuclear magnetic resonance
ODN = oligodeoxynucleotide
PAMPs = pathogen associated molecular pattern
PBS = phosphate buffered saline
PD-1 = apoptosis-1
PD-l1=apoptosis-ligand 1
PDT = photodynamic therapy
ppy=2-phenyl-pyridine
PS = photosensitizer
Pt=platinum
PXRD = powder X-ray diffraction
RDT = radiation power therapy
REF 10 Radiation enhancement factor at 10% cell survival
ROS = reactive oxygen species
RT = radiation therapy
Ru=ruthenium
SBU = secondary building block
s=second
Sosg=singlet oxygen fluorescent probe (singlet oxygen sensor green)
STING = interferon gene stimulators
Tem=transmission electron microscope
TFA = trifluoroacetic acid
Tbp=5, 10,15, 20-tetra (p-benzoic acid) porphyrin
TMS = trimethylsilyl group
TGI = tumor growth inhibition
TLR9 = toll-like receptor 9
Tme=tumor microenvironment
UV = ultraviolet light
wt = weight
Z=atomic number
Background
Cancer immunotherapy utilizes the patient's own immune system to recognize and treat cancer. Cancer immunotherapy drugs use various strategies including checkpoint blockade, chimeric antigen receptor T cells, vaccination, and the like. For example, checkpoint Blocking Immunotherapy (CBI) uses agents that target T lymphocyte regulatory pathways, such as the programmed death-1/programmed death ligand 1 (PD-1/PD-L1) axis. CBI has shown clinical response in some solid tumors. However, many patients do not respond to current cancer immunotherapy.
Thus, there remains a need for other compositions and methods of treating cancer, including those that are capable of providing immunotherapy of cancer.
Disclosure of Invention
This disclosure lists several embodiments of the presently disclosed subject matter, and in many cases, variations and permutations of these embodiments. This disclosure is merely illustrative of the many varied embodiments. References to one or more representative features of a given embodiment are also exemplary. Such embodiments may or may not generally have the features described; likewise, these features may be applicable to other embodiments of the presently disclosed subject matter, whether or not listed in this disclosure. To avoid undue repetition, this disclosure does not list or suggest all possible combinations of such features.
In some embodiments, the presently disclosed subject matter provides a Metal Organic Framework (MOF) having a surface modified to coordinately or electrostatically bind one or more therapeutic agents of interest, wherein the MOF comprises: (a) A plurality of metallo-oxygen cluster Secondary Building Units (SBUs), wherein the metallo-oxygen cluster SBUs each comprise one or more first metal ions and one or more anions, wherein the one or more anions each coordinate with one or more of the one or more first metal ions; and (b) a plurality of organic bridging ligands linking the plurality of SBUs together to form a two-dimensional or three-dimensional matrix; wherein (i) each of the plurality of SBUs at the MOF surface comprises a weakly coordinating anion as an SBU capping group anion, or (ii) the plurality of organic bridging ligands comprises an organic bridging ligand comprising an electron withdrawing group or ligand, a positive charge, or a combination thereof, optionally wherein the plurality of organic bridging ligands comprises a ligand comprising a nitrogen donor group coordinately bound to a second metal ion, wherein the second metal ion is further coordinated to at least one second metal ligand comprising one or more electron withdrawing groups; wherein the surface of the MOF has enhanced ability to coordinately or electrostatically bind to one or more therapeutic agents of interest.
In some embodiments, the one or more first metal ions comprise at least one metal ion that absorbs ionizing radiation (optionally X-rays), and/or wherein the metal is selected from the group consisting of Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi; further optionally wherein the first metal ion is a Hf ion. In some embodiments, each of the plurality of SBUs on the MOF surface comprises a weakly coordinating anion as a capping group, optionally wherein the weakly coordinating anion is selected from trifluoroacetate and trifluoromethanesulfonate. In some embodiments, the plurality of organic bridging ligands comprises a porphyrin substituted with at least two carboxylate groups, optionally wherein the plurality of organic bridging ligands comprises 5, 15-bis (p-benzoic acid) porphyrin (DBP).
In some embodiments, the MOF further comprises a small molecule therapeutic that is sequestered in pores and/or cavities of a two-dimensional or three-dimensional network, optionally wherein the small molecule therapeutic is a chemotherapeutic, a small molecule inhibitor, and/or a small molecule immunomodulator. In some embodiments, the MOF comprises a chemotherapeutic agent sequestered in pores and/or cavities of a two-or three-dimensional network, optionally wherein the chemotherapeutic agent is selected from cisplatin, carboplatin, paclitaxel, SN-35, and etoposide. In some embodiments, the MOF comprises a small molecule inhibitor sequestered in pores and/or cavities of a two-dimensional or three-dimensional network, optionally wherein the small molecule inhibitor is selected from the group consisting of PLK1 inhibitor, wnt inhibitor, bcl-2 inhibitor, PD-L1 inhibitor, ENPP1 inhibitor, and IDO inhibitor. In some embodiments, the MOF comprises a small molecule immunomodulator sequestered in pores and/or cavities of a two-dimensional or three-dimensional network. In some embodiments, the small molecule immunomodulator is Imiquimod (IMD).
In some embodiments, the plurality of organic bridging ligands comprises an organic bridging ligand comprising a nitrogen donor group, wherein the nitrogen donor group coordinates to a second metal ion, and wherein the second metal cation is further coordinated to at least one second metal ligand comprising one or more electron withdrawing groups, optionally wherein the one or more electron withdrawing groups are selected from halo and perhaloalkyl. In some embodiments, the organic bridging ligand comprising a nitrogen donor group is 4,4 '-bis (p-benzoic acid) -2,2' -bipyridine (DBB). In some embodiments, the second metal ion is an iridium (Ir) ion or a ruthenium (Ru) ion, and/or wherein the second metal cation is coordinated to two second metal ligands, wherein one or both of the second metal ligands comprises one or more electron withdrawing groups. In some embodiments, one or both of the second metal ligands is 2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine (dF (CF) 3 ) ppy). In some embodiments, the MOF has a Zeta (Zeta) -potential value of at least about 5 millivolts (mV), optionally wherein the MOF has a Zeta-potential value of at least about 30mV.
In some embodiments, the MOF comprises a three-dimensional network, wherein the three-dimensional network is provided in the form of nanoparticles.
In some embodiments, the presently disclosed subject matter provides a MOF for delivering one or more therapeutic agents of interest, wherein the MOF comprises: (a) A plurality of metal oxide clusters SBU, wherein each of said metal oxide clusters SBU contains one or more first metal ions and one or more anions, wherein each of said anions coordinates to one or more of said one or more first metal ions; (b) A plurality of organic bridging ligands linking together the plurality of SBUs to form a two-dimensional or three-dimensional matrix; and (c) one or more therapeutic agents of interest bound to the MOF surface by coordinative binding or electrostatic interaction, optionally wherein one or more drugs of interest are coordinately bound to the metal ion of one or more of the plurality of SBUs on the MOF surface. In some embodiments, the first metal ion is a metal ion that absorbs ionizing radiation (optionally X-rays), and/or wherein the first ion is a metal ion selected from Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi; further optionally wherein the first metal ion is a Hf ion.
In some embodiments, each of the one or more therapeutic agents of interest is selected from the group consisting of a nucleic acid, a small molecule comprising phosphate or carboxylate groups, and/or a large molecule comprising surface accessible phosphate or carboxylate groups. In some embodiments, the one or more therapeutic agents of interest comprise a macromolecule comprising a surface accessible phosphate or carboxylate group, and wherein the macromolecule is a protein, optionally wherein the protein is an antibody. In some embodiments, the protein is selected from the group consisting of an anti-CD 37 antibody, an anti-CD 44 antibody, an anti-CD 47 antibody, an anti-CD 73 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-LAG 3 antibody, and an anti-CTLA-4 antibody. In some embodiments, the one or more therapeutic agents of interest comprise a nucleic acid, and wherein the nucleic acid is selected from the group consisting of miRNA, mRNA, siRNA, cpG ODN, and a cyclic dinucleotide, optionally wherein the nucleic acid is a cyclic dinucleotide and the cyclic dinucleotide is a STING agonist, further optionally wherein the STING agonist is c-di-AMP or cGAMP.
In some embodiments, the MOF comprises one or more additional therapeutic agents sequestered in pores or cavities of a two-dimensional or three-dimensional network; optionally wherein the MOF comprises from about 1wt% to about 50wt% of the one or more additional therapeutic agents.
In some embodiments, the plurality of SBUs comprises Hf oxygen clusters, wherein the plurality of organic bridging ligands comprises DBPs, and wherein the one or more therapeutic agents of interest are bound to the surface of the MOF by coordination bonds to Hf ions of the surface-accessible SBUs. In some embodiments, the one or more therapeutic agents of interest include one or more antibodies. In some embodiments, the one or more therapeutic agents comprise an anti-CD 47 antibody. In some embodiments, the MOF further comprises an IMD sequestered in a pore or cavity of a two-dimensional or three-dimensional network.
In some embodiments, the MOF is a three-dimensional network and is provided as a nanoparticle. In some embodiments, the MOF comprises from about 1wt% to about 50wt% IMD or anti-CD 47 antibody; optionally wherein the MOF comprises about 9wt% IMD and about 7.5wt% anti-CD 47 antibody.
In some embodiments, the plurality of SBUs comprises Hf oxygen clusters, wherein the plurality of organic bridging ligands comprises DBBs coordinated to Ir ions, wherein the Ir ions are further coordinated to two (dF (CF 3 ) ppy); and wherein the one or more therapeutic agents of interest bind to the surface of the MOF via electrostatic interactions. In some embodiments, the one or more therapeutic agents of interest comprise a nucleic acid. In some embodiments, the nucleic acid is a STING agonist or a CpG Oligodeoxynucleotide (ODN), optionally wherein the nucleic acid is a CpG ODN.
In some embodiments, the MOF comprises from about 1wt% to about 50wt% of one or more therapeutic agents of interest, optionally wherein the one or more therapeutic agents of interest comprise antibodies.
In some embodiments, the presently disclosed subject matter provides a method of treating cancer in need thereof, the method comprising: (a) administering a MOF to a subject, wherein the MOF comprises: (i) A plurality of metal oxide clusters SBU, wherein each of said metal oxide clusters SBU comprises one or more first metal ions and one or more anions, wherein each of said anions coordinates to one or more of said one or more first metal ions; (ii) A plurality of organic bridging ligands linking together the plurality of SBUs to form a two-dimensional or three-dimensional matrix; and (iii) one or more therapeutic agents of interest bound to the MOF surface by coordinative binding or electrostatic interaction, optionally wherein the one or more therapeutic agents of interest coordinately bind to metal ions of one or more of the plurality of SBUs on the MOF surface; and (b) exposing at least a portion of the subject to ionizing radiation energy, optionally X-rays. In some embodiments, the method further comprises administering to the subject an additional therapeutic agent or treatment, optionally an immunotherapy and/or a cancer treatment selected from the group consisting of surgery, chemotherapy, toxin therapy, cryotherapy, and gene therapy.
In some embodiments, the additional therapeutic agent is an immunotherapeutic agent, optionally wherein the immunotherapeutic agent is an immune checkpoint inhibitor. In some embodiments, the immunotherapeutic agent is an anti-PD-1 or anti-PD-L1 antibody.
In some embodiments, the cancer is colorectal cancer, melanoma, head and neck cancer, brain cancer, breast cancer, liver cancer, cervical cancer, lung cancer, or pancreatic cancer. In some embodiments, administration of the MOF provides an extended release profile for one or more therapeutic agents of interest, optionally wherein the release rate is adjustable, and/or wherein the MOF provides sustained release of one or more therapeutic agents of interest over a period of hours or days. In some embodiments, administration of the MOF reduces a therapeutically effective dose of one or more therapeutic agents of interest.
In some embodiments, the presently disclosed subject matter provides methods of enhancing the interaction and/or binding of one or more therapeutic agents of interest to a MOF surface, the methods comprising modifying the surface of the MOF by (i) providing one or more surface accessible coordination sites coordinately bound to a weakly coordinating anion capable of being substituted with carboxylate or phosphate substituents of the therapeutic agent of interest, or (ii) providing a MOF comprising one or more electron withdrawing bridging ligands, one or more positively charged bridging ligands, or a combination thereof. In some embodiments, modifying the surface of the MOF comprises: (ia) providing a parent MOF comprising metallo-oxy SBUs linked together by an organic bridging ligand, wherein each of said SBUs comprises one or more metal ions and one or more anions, and wherein said MOF comprises a plurality of surface accessible metallo-oxy SBUs, wherein said one or more anions of each of said surface accessible metallo-oxy SBUs comprise a strongly coordinating anion that is an SBU capping group; optionally wherein the strongly coordinating anion comprises acetate or formate; and (ib) removing the strongly coordinating anion, wherein the removing comprises contacting the parent MOF with an agent selected from the group consisting of trimethylsilyl trifluoroacetate, trimethylsilyl triflate, and mineral acids having a pKa of less than about 3; whereby the strongly coordinating anion is replaced with a weakly coordinating anion, optionally wherein the strongly coordinating anion is selected from acetate or formate anions, optionally wherein the weakly coordinating ion is selected from trifluoroacetate or trifluoromethanesulfonate anions.
In some embodiments, providing a MOF comprising one or more bridging ligands comprising an electron withdrawing group, one or more bridging ligands comprising a positive charge, or a combination thereof comprises providing a MOF comprising metallo-oxygen cluster SBUs linked together by an organic bridging ligand, wherein each of the SBUs comprises one or more first metal ions and one or more anions coordinated to the one or more first metal ions, and wherein the organic bridging ligand comprises at least one organic bridging ligand comprising a non-SBU linked coordinated second metal ion, optionally wherein the electron withdrawing ligand is a halo-and/or perhaloalkyl-substituted bipyridine ligand. In some embodiments, providing the MOF comprises providing a MOF comprising a DBB bridging ligand, wherein the DBB bridging ligand coordinates to a first metal ion and a second metal ion of two different metal oxygen clusters SBU, and wherein the second metal is further coordinated to two halo-and/or perhaloalkyl-substituted pyridine ligands, optionally wherein the two halo-and/or perhaloalkyl-substituted pyridine ligands are each 2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine. In some embodiments, the second metal ion is Ir or Ru.
In some embodiments, the MOF comprises one or more SBUs comprising metal ions that absorb ionizing radiation (optionally X-rays) and/or wherein the metal ions are ions of an element selected from the group consisting of Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi; further optionally wherein the metal ion is a Hf ion. In some embodiments, the MOF has enhanced interaction and/or binding capacity for one or more therapeutic agents of interest as compared to a MOF without surface modification, wherein the one or more therapeutic agents of interest are selected from the group consisting of nucleic acids, small molecules, and/or macromolecules comprising surface accessible phosphate or carboxylate groups. In some embodiments, the protein is selected from the group consisting of an anti-CD 37 antibody, an anti-CD 44 antibody, an anti-CD 47 antibody, an anti-CD 73 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-LAG 3 antibody, and an anti-CTLA-4 antibody. In some embodiments, the nucleic acid is selected from the group consisting of miRNA, mRNA, siRNA, cpG ODN, and a cyclic dinucleotide, optionally wherein the cyclic dinucleotide is a STING agonist, further optionally wherein the STING agonist is c-di-AMP or cGAMP.
Accordingly, it is an object of the presently disclosed subject matter to provide MOFs suitable for binding and delivering one or more therapeutic agents of interest, methods of treating cancer using the MOFs, and methods of enhancing therapeutic agent interactions and/or binding to the surface of MOFs.
One object of the presently disclosed subject matter has been set forth hereinabove, and which is accomplished in whole or in part by the presently disclosed subject matter, other objects will become evident as the description proceeds when taken in connection with the accompanying drawings and the following best described embodiments.
Drawings
Fig. 1: a schematic showing repolarization and promotion of phagocytosis of M2 to M1 macrophages by Metal Organic Frameworks (MOFs) comprising hafnium (Hf) -oxygen clusters and 5, 15-bis (p-benzoic acid) porphyrin (DBP) bridging ligands, together with Imiquimod (IMD) chelated in the pores of the MOF and an antibody cluster (anti-cluster) of differentiated 47 (αcd 47) antibodies attached to the surface of the MOF (where the MOF is referred to as imd@hf-DBP/αcd 47) plus an X-ray radiation blocking "don't eat me" signal on tumor cells is shown. This macrophage therapy works synergistically with anti-apoptotic ligand 1 antibody (αpd-L1) Checkpoint Blocking Immunotherapy (CBI) to systematically eradicate tumors.
Fig. 2A-2C: FIG. 2A is a schematic diagram showing surface modification of metallo-oxygen clusters of hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) metallo-organic frameworks (MOFs) for differentiation 47 antibody (. Alpha.CD47) antibody cluster loading. The parent Hf-DBP with an acetate end group on the hafnium (Hf) oxygen cluster (left) is contacted with trimethylsilyl trifluoroacetate (TMS-TFA), exchanging the acetate end group with a trifluoroacetate end group (middle), then substituting with αcd 47. FIG. 2B is a graph showing the αCD47 loading efficiency of Hf-DBP (right) and TFA modified Hf-DBP (left). Fig. 2C is a graph showing imiquimod (IMD, square) and αcd47 (circle) release profile from Hf-DBP MOFs, where the IMD is sequestered in the MOF pores and αcd47 is attached to the surface (i.e., where the MOF is imd@hf-DBP/αcd 47).
Fig. 3A and 3B: transmission Electron Microscope (TEM) images of hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) Metal Organic Frameworks (MOFs). Fig. 3A is a large field-of-view TEM image. The scale bar in the lower left corner is 200 nanometers (nm). Fig. 3B is a high resolution TEM image (bottom left scale 20 nm) and a cropped Fast Fourier Transform (FFT) image (inset, bottom right).
Fig. 4A and 4B: transmission Electron Microscope (TEM) images of Trifluoroacetate (TFA) -modified hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) Metal Organic Frameworks (MOFs). Fig. 4A is a large field-of-view TEM image. The scale bar in the lower left corner is 200 nanometers (nm). Fig. 4B is a high resolution TEM image (bottom left scale 10 nm) and a cropped Fast Fourier Transform (FFT) image (inset, bottom right).
Fig. 5: a graph showing powder X-ray diffraction (PXRD) patterns of hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP, solid line) Metal Organic Frameworks (MOFs) and Trifluoroacetate (TFA) modified Hf-DBP-MOFs (dashed lines).
Fig. 6: schematic showing Imiquimod (IMD) supported in Trifluoroacetate (TFA) modified hafnium-5, 15-bis (p-benzoic) porphyrin (Hf-DBP) nanoplatelets. Hafnium-12 (Hf) 12 ) The Secondary Building Units (SBUs) are shown as polyhedrons and the DBP organic ligands are shown as rods. The TFA group and the trifluoromethyl moiety of the IMD are indicated by arrows.
Fig. 7A and 7B: IMD@Hf-DBP, i.e., transmission Electron Microscope (TEM) image of Imiquimod (IMD) supported hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) metal-organic framework (MOF). Fig. 7A is a large field-of-view TEM image. The scale bar in the lower left corner is 200 nanometers (nm). Fig. 7B is a high resolution TEM image (bottom left scale 50 nm) and a cropped Fast Fourier Transform (FFT) image (inset, bottom right).
Fig. 8A and 8B: transmission Electron Microscope (TEM) images of imd@hf-DBP/αcd47, i.e., imiquimod (IMD) -loaded antibody cluster with differentiated 47 antibodies (αcd 47) attached to hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) metal-organic frameworks (MOFs) of the surface. Fig. 8A is a large field-of-view TEM image. The scale bar in the lower left corner is 100 nanometers (nm). Fig. 8B is a high resolution TEM image (bottom left scale 50 nm) and a cropped Fast Fourier Transform (FFT) image (inset, bottom right).
Fig. 9: a graph showing the powder X-ray diffraction (PXRD) pattern of Imiquimod (IMD) -loaded hafnium-5, 15-bis (p-benzoate) porphyrin (Hf-DBP, solid line) metal-organic frameworks (MOFs), i.e., imd@hf-DBP (solid line) and Hf-DBP MOFs, i.e., imd@hf-DBP/αcd47 (dashed line), loaded with IMD and having an antibody cluster (αcd 47) of differentiated 47 antibodies attached to the surface of the MOFs.
Fig. 10: a graph showing the release of fluorescein-isothiocyanate (FITC) -labeled immunoglobulin G (IgG-FITC) from IgG-FITC-modified Imiquimod (IMD) -loaded hafnium-5, 15-bis (p-benzoic acid) porphyrin metal-organic framework surfaces (i.e., imd@hf-DBP/IgG FITC) in serum-containing Phosphate Buffered Saline (PBS) (percent versus time (in hours (h)). n=3.
Fig. 11: shows cellular uptake (in millimoles of hafnium (nmol of Hf)/10) of hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) Metal Organic Frameworks (MOFs) in mouse colon adenocarcinoma (CT 26) cells quantified by inductively coupled plasma-mass spectrometry (ICP-MS) 5 Individual cytometers). Cell uptake of Imiquimod (IMD) loaded Hf-DBP (IMD@Hf-DBP, dark shaded bar) and IMD loaded Hf-DBP (IMD@Hf DBP/αCD47, light grey shaded bar) with attachment surface of antibody cluster (αCD47) of differentiation 47 antibodies are shown. n=3.
Fig. 12: a graph showing the dark toxicity (no X-ray exposure) of Imiquimod (IMD) loaded hafnium-5, 15-bis (p-benzoate) porphyrin (Hf-DBP) Metal Organic Frameworks (MOFs) (i.e., imd@hf-DBP, circles) and imd@hf-DBP MOFs (imd@hf-DBP/αcd47, squares) with differentiated 47 antibody cluster attachment surfaces in mouse colon adenocarcinoma (CT 26) cells. Cell viability (expressed as percent (%)) was determined by the (3- (4, 5-dimethylthiazol-2-yl) -5- (3-carboxymethoxyphenyl) -2- (4-sulfophenyl) -2H-tetrazolium) (MTS) assay. n=6.
Fig. 13: representative gating strategies for macrophages and their subtypes M1 and M2. Histograms show the different expression levels of differentiation cluster 86 (CD 86) or differentiation cluster 206 (CD 206) in the M1 or M2 subtypes, respectively.
Fig. 14A-14D: FIG. 14A is a series of representative flow cytometry analyses of M2 macrophages repolarized co-cultured with Phosphate Buffered Saline (PBS), imiquimod (IMD), hafnium-5, 15-bis (p-benzoate) porphyrin (Hf-DBP) Metal Organic Frameworks (MOFs) or IMD-loaded Hf-DBP (IMD@Hf-DBP) MOFs and irradiated with X-rays at 0 (-) or 2 (+) Gray (Gy) doses. Macrophages were stained with phycoerythrin blue dye (PE-Cy 7) conjugated differentiation cluster 206 (CD 206) and Allophycocyanin (APC) conjugated differentiation cluster 86 (CD 86) antibodies. FIG. 14B is a series of representative flow cytometry analyses of carboxyfluorescein succinimidyl ester (CFSE) labeled CT26 cells by macrophages treated with PBS, an antibody cluster of differentiated 47 antibodies (αCD47), hf-DBP, or αCD47 surface modified Hf-DBP (Hf-DBP/αCD47) and irradiated with X-rays at 0 (-) or 2 (+) Gray (Gy) doses. CFSE-labeled mouse colon adenocarcinoma (CT 26) cells were gated by a Zhou Pigan chlorophyll protein cyanine dye 5.5 (PerCP-cy 5.5) -labeled macrophage population. FIG. 14C is a graph showing quantification of repolarization of macrophages as described in FIG. 14A. Fig. 14D is a graph showing the quantification of phagocytosis described in fig. 14B. n=3; p <0.05, < P <0.01, < P <0.005, compared to the control group.
Fig. 15A-15C: fig. 15A is a series of histograms showing an immunoassay in which macrophages in a mouse colon adenocarcinoma (CT 26) tumor repolarized using the following treatments: phosphate Buffered Saline (PBS) and X-ray free irradiation (PBS (-)); PBS + irradiation (PBS (+); imiquimod (IMD) and differentiated 47 antibodiesTogether with the irradiation (IMD/aCD 47 (+)); hafnium-5, 15-bis (p-benzoic acid) porphyrin metallo-organic framework (Hf-DBP) and no irradiation (imd@hf-DBP- αcd47 (-)) carried by IMD with αcd47 attached to the surface; hf-DBP and irradiation (Hf-DBP+); IMD loading Hf-DBP and irradiation (IMD@Hf-DBP (+)); hf-DBP with surface attached αCD47 plus illumination (Hf-DBP- αCD47 (+); or IMD@Hf-DBP- αCD47 plus radiation (IMD@Hf-DBP- αCD47+). FIG. 15B is a graph showing the Mean Fluorescence Intensity (MFI) of major histocompatibility complex class II (MHC-II) expression in the same tumor treatment group depicted in FIG. 15A. FIG. 15C is a graph showing the efficacy of the various treatments depicted in FIG. 15A (in cubic centimeters (cm) 3 ) Tumor volume in meters). n=5 or 6.
Fig. 16: representative gating strategies for differentiated cluster 45 positive (cd45+) cells, differentiated 11b cluster positive (cd11b+) cells, dendritic Cells (DCs), macrophages and subtypes M1 and M2.
Fig. 17A-17F: FIG. 17A is a graph showing the percentage of differentiation cluster 45 positive (CD45+) cells in colon adenocarcinoma (CT 26) tumor-bearing mice of mice treated as described in FIG. 15A. Fig. 17B is a graph showing the percentage of Dendritic Cells (DCs) in tumor-bearing mice of colon adenocarcinoma (CT 26) of mice treated as described in fig. 15A. Fig. 17C is a graph showing the percentage of macrophages in colon adenocarcinoma (CT 26) tumor-bearing mice of mice treated as described in fig. 15A. Fig. 17D is a graph showing the percentage of M1 macrophages in colon adenocarcinoma (CT 26) tumor-bearing mice of mice treated as described in fig. 15A. Fig. 17E is a graph showing the percentage of M2 macrophages in colon adenocarcinoma (CT 26) tumor-bearing mice of mice treated as described in fig. 15A. FIG. 17F is a graph showing the ratio of M1 to M2 macrophages in colon adenocarcinoma (CT 26) tumor-bearing mice treated as described in FIG. 15A. (+) and (-) refer to the presence and absence of X-ray irradiation, respectively. The center line, the frame boundary and the box must represent the average value, 25% -75% of the data range, and 1.5 times the outlier distance quartile range, respectively. t-test, P < 0.05, P < 0.01 and P < 0.001.
Fig. 18: resective tumor photographs of tumor-bearing mice treated with colon adenocarcinoma (CT 26) of mice: phosphate Buffered Saline (PBS) and X-ray free radiation (PBS (-)); PBS and illumination (PBS (+); a mixture of Imiquimod (IMD) and the antibody cluster of differentiated 47 antibodies (αcd47) and irradiation (IMD/αcd47+); hafnium-5, 15-bis (p-benzoic acid) porphyrin metallo-organic framework (Hf-DBP) and no irradiation (imd@hf-DBP- αcd47 (-)) carried by IMD with αcd47 attached to the surface; hf-DBP+ irradiation (Hf-DBP (+); hf-DBP and irradiance (IMD@Hf-DBP (+)) on IMD load; hf-DBP and irradiation of surface-attached αcd47 (Hf-DBP- αcd47 (+); or IMD@Hf-DBP- αCD47) and radiation (IMD@Hf-DBP- αCD47+). n=6.
Fig. 19: tumor weight plot (units: grams (g)) for each treatment group shown in fig. 18 in the colon adenocarcinoma tumor model of mice after sacrifice. N=6.
Fig. 20A-20F: fig. 20A is a plot of primary tumor growth in bilateral mouse colon adenocarcinoma (CT 26) tumor-bearing mice treated with: phosphate Buffered Saline (PBS) and no radiation (PBS (-)); x-ray irradiation and PBS (); anti-apoptotic ligand 1 antibody (αpd-L1) and irradiation (αpd-L1 (+); imiquimod (IMD) loaded hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) metal-organic frameworks (MOFs) and irradiation (imd@hf-DBP/αcd47 (+)) with anti-differentiation cluster 11b47 antibody (αcd47); the Hf-DBP MOF alpha CD47 antibody with alpha CD47 attached on the surface, irradiation and combined administration of alpha PD-L1 (Hf-DBP/alpha CD47 (+) +alpha PDL 1); IMD, αcd47, αpdl1 and irradiation (IMD/αcd47 (+) +αpdl1); surface-attached αCD47 IMD loaded Hf-DBP MOF, no irradiation and co-administration of αPDL1 (IMD@Hf-DBP/αCD47 (-) +αPD-L1); or surface-attached αCD47, with the application of Hf-DBP MOF, irradiation, and co-administration of αPDL1 (IMD@Hf-DBP/αCD47 (+) +αPD-L1). Fig. 20B is a graph of the growth of distant tumors in the mice depicted in fig. 20A. FIG. 20C is a graph of the results of an ELISPot assay to detect tumor-specific response-producing interferon-gamma (IFN-gamma) -producing T cells in spleen cells after six of the eight treatments described in FIG. 20A. The results of treatment with cells stimulated with no peptide (left) or with SPSYVYHQF (SEQ ID No: 4) (right) are shown. FIGS. 20D-20F are graphs of the percentages of tumor infiltrating differentiation cluster 8 positive (CD8+, FIG. 20D), differentiation cluster 4 positive (CD4+) T cells (FIG. 20E) and Natural (NK) cells (FIG. 20F) relative to the total number of tumor cells for six of the eight treatment groups depicted in FIG. 20A. n=5. P <0.05, P <0.01, P <0.005 relative to the control group.
FIG. 21 is a series of histograms showing that the differentiation cluster 45 was positive (CD 45 + ) Cell, T cell, differentiation 8 cluster positive (CD 8 + ) Representative gating strategies for T cells, B cells, natural Killer (NK) cells, dendritic cells, macrophages, and M1 and M2 subtypes.
Fig. 22A and 22B: the differentiation cluster 45 positive (CD 45) in primary tumors and distant tumors of treated mice colon adenocarcinoma (CT 26) bilateral tumor-bearing mice as described for six of the eight treatment groups in fig. 20A + ) A pair of percentile plots of cells (fig. 22A) and B cells (fig. 22B). (+) and (-) refer to irradiation and non-irradiation, respectively. The center line, frame boundary and box must represent the average value, data range 25% -75%, 1.5 times the outlier distance quartile range, respectively. t-test, P < 0.05, P < 0.01 and P < 0.001.
Fig. 23 is a schematic representation of the anti-tumor effect of in situ cancer vaccination by nanoscale metal organic frameworks (nMOFs) + Checkpoint Blocking Immunotherapy (CBI). (1) Hf-DBB F -ir@cpg (i.e. MOF with hafnium secondary building blocks; organic bridging ligands comprising 4,4 '-bis (4-benzoic acid) -2,2' -bipyridine (DBB) coordinately bound to Iridium (IR) which is also coordinately bound to two 2- (2, 4-difluorophenyl) -5-trifluoromethyl) pyridine ligands; and surface-bound CpG Oligodeoxynucleotides (ODNs)) are intratumorally administered into the primary tumor. (2) After X-ray activation, hf-DBB F Ir generates Reactive Oxygen Species (ROS) to induce immunogenic cell death, exposing tumor antigens and danger-related molecular patterns (DAMP), whereas CpG ODN acts as a pathogen-related molecular pattern, in the cation Hf-DBB F Delivery to antigen presenting cells with the aid of Ir. (3) DAMP and PAMP promote Dendritic Cell (DC) maturation. (4) Tumor antigens are presented by mature DCs to T cells in tumor draining lymph nodes. (5) T cells are expanded and primed towards distant tumors and primary tumors. (6) Systemic administration of immune checkpoint blocking inhibitors anti-apoptotic ligand 1 antibodies (αpd-L1) attenuate T cell depletion.
FIGS. 24A and 24B are a pair of schematic diagrams showing a composition consisting of hafnium tetrachloride (HfCl 4 ) And H 2 DBB-Ir-F (FIG. 24A) or H 2 DBB-Ir (FIG. 24B) SynthesisHf-DBB F -Ir (FIG. 24A) and Hf-DBB-Ir (Table 24B) Metal Organic Frameworks (MOFs).
Fig. 25A and 25B: FIG. 25A is a graph showing Hf-DBB freshly prepared or after 24 hours of incubation in 0.6mM Phosphate Buffered Saline (PBS) as compared to model metal organic frameworks (UiO) -69) F -powder X-ray diffraction Pattern (PXRD) of Ir and Hf-DBB-Ir. FIG. 25B shows Hf-DBB in ethanol (EtOH) F Number average diameters of Ir (112.2.+ -. 2.8 nanometers (nm), square) and Hf-DBB-Ir (113.9.+ -. 1.6nm, circle). n=3.
Fig. 26A-26E: FIG. 26A is a graph based on hafnium (Hf) -oxygen clusters and DBB, respectively F Controlled synthesis of Hf-DBB-Ir and Hf-DBB by-Ir or DBB-Ir ligand F -schematic diagram of Ir-nmos ofs. Under the irradiation of X-rays, the Hf oxygen cluster absorbs the X-rays and generates by radiolysis * OH and transferring energy to adjacent photoactive ligands to generate 1 O 2 And/or O 2- . FIG. 26B is a view of Hf-DBB-Ir (top) and Hf-DBB F A pair of Transmission Electron Microscope (TEM) images of Ir (bottom), scale bar = 100nm. FIGS. 26C-26E are diagrams showing detection of aminophenyl fluorescein (APF) after X-ray exposure using a fluorescence imaging system (FIG. 26C, n=6) * OH generation, detected by Singlet Oxygen Sensor Green (SOSG) using a fluorescence imaging device (fig. 26d, n=6) 1 O 2 Formation and use of 5-tert-butoxycarbonyl-5-methyl-1-pyrroline-N-oxide (BMPO) in DBB-Ir, DBB F -Ir, hf-DBB-Ir or Hf-DBB F O detected in Electron Paramagnetic Resonance (EPR) of Ir (FIG. 26E) 2- And (5) generating.
Fig. 27A-27D: hf-DBB F -Transmission Electron Microscope (TEM) images of Ir and Hf-DBB-Ir Metal Organic Frameworks (MOFs) with and without surface-attached CpG Oligodeoxynucleotides (ODNs). FIGS. 27A and 27B are, respectively, hf-DBB F Large area TEM images of Ir and Hf-DBB-Ir. The scale bar in the lower right hand corner of the two images represents 500 nanometers (nm). FIGS. 27C and 27D are Hf-DBB F TEM image of Ir@CpG. In fig. 27C, the scale bar at the lower right represents 100nm, and the scale bar at the lower right of fig. 27D represents 500nm.
Fig. 28A-28D: FIG. 28A is a view showing the Hf-DBB F -Ir and Hf-DBB-Ir Metal Organic Frameworks (MOFs) and ligandsBody DBB F UV-vis spectra (intensity in arbitrary units) versus wavelength in nanometers (nm) for Ir and DBB-Ir FIGS. 28B and 28C are Hf-DBB F Ir (FIG. 28B) and Hf-DBB-Ir (see FIG. 28C) are each independently of DBB F -graph of excitation (Ex) and emission (Em) spectra of Ir compared to DBB-Ir. FIG. 28D is a comparative Hf-DBB F -graphs of excitation (Ex) and emission (Em) spectra of Ir and Hf-DBB-Ir.
Fig. 29: mouse colon cancer (MC 38) cells were isolated from 20 micromolar (μM) Hf-DBB-Ir or Hf-DBB based on Hf F Hf-DBB-Ir and Hf-DBB after 1, 2, 4 or 8 hours of Ir incubation F Cell uptake profile of Ir, n=3. Hafnium (Hf) concentration (in millimoles (nmol)/10% of cells) was determined by inductively coupled plasma-mass spectrometry (ICP-MS).
Fig. 30A-30D: in vitro generation of danger-related molecular patterns (DAMP) and phagocytosis. Phosphate Buffered Saline (PBS), ligand (DBB-Ir or DBB) for colon cancer (MC 38) cells in mice F -Ir) or Metal Organic Framework (MOF) (Hf-DBB-Ir or Hf-DBB) F -Ir) for 4 hours, then irradiated with X-rays at a dose of 0 (-) or 2 (+) Gray (Gy). FIG. 30A is a chart for evaluating Hf-DBB-Ir or Hf-DBB on MC38 cells after X-ray irradiation F Ir-radiation-enhanced clones generated analytical test result plots, n=6. FIG. 30B is a chart showing the results of 2Gy of MC38 cells irradiated with X-rays with Hf-DBB-Ir or Hf-DBB F Graph of cytotoxicity of Ir, n=6. FIGS. 30C and 30D are graphs of phagocytosis of carboxyfluorescein succinimidyl ester (CFSE) labeled MC38 cells by Dendritic Cells (DCs). DCs co-cultured with MC38 cells untreated (-, fig. 30C) or (+fig. 30D) were stained with phycoerythrin-cyanin dye 5.5 (PE-cy 5.5) -conjugated cluster of differentiation 11C (CD 11C) antibody, n=3. Data are expressed as mean ± s.d. By t-test, P<0.05,**P<0.01 and P<0.001。
Fig. 31A and 31B: generation and phagocytosis of danger-related molecular patterns (DAMP). PBS, DBB-Ir and DBB for colon cancer (MC 38) cells of mice F -Ir, hf-DBB-Ir or Hf-DBB F Ir treatment for 4 hours, followed by irradiation with X-rays at a dose of 0 (-) or 2 (+) Gray (Gy) and co-culture with Dendritic Cells (DCs) for phagocytosis assay. FIG. 31A is a series ofA chart showing flow cytometry analysis of calreticulin exposure in treated MC38 cells. The light gray histogram (control) and the dark gray histogram show differences in Calreticulin (CRT) levels in the cells, respectively. FIG. 31B is a series of charts showing the phagocytosis of Carboxyl Fluorescein Succinimidyl Ester (CFSE) labeled MC38 cells by flow cytometry analysis of DC. DCs co-cultured with treated MC38 cells were stained with phycoerythrin-cyanin dye 5.5 (PE-cy 5.5) conjugated CD11c antibody. CD11c + CFSE + The biscationic population was gated as DC phagocytosed MC38 cells.
Fig. 32A-32I: pathogen-associated molecular pattern (PAMP) and Dendritic Cell (DC) activated in vitro delivery. FIG. 32A is a view showing Hf-DBB-Ir and Hf-DBB F -graph of Zeta potential (millivolts (mV)) of Ir Metal Organic Frameworks (MOFs). FIG. 32B is a graph of quantification of adsorbed CpG Oligodeoxynucleotides (ODNs) by DNA gel (inset) and quantification of unadsorbed CpG-ODNs by NanoDrop using free CpG ODNs as a control (right lane). FIG. 32C is a series of free CpG ODN, hf-DBB-Ir@CpG or Hf-DBB at a concentration of 20. Mu.M hafnium (Hf) and 0.1. Mu.g/mL CpG ODN as quantified by flow cytometry F -a series of charts and fluorescence micrographs of a DC uptake Fluorescein Isothiocyanate (FITC) labeled CpG ODN cultured at ir@cpg and observed under Confocal Laser Scanning Microscopy (CLSM). Scale bar = 4 micrometers (μm). FIGS. 32D-32F are graphs showing functional surface marker differentiation cluster 80 (CD 80, FIG. 32D), differentiation cluster 86 (CD 86, FIG. 32E) and major histocompatibility complex class II (MHC-II, FIG. 32F) quantified by flow cytometry. The legend in fig. 32D applies to all three of fig. 32D-32F. FIGS. 32G and 32H are schematic representations of free CpG ODN, hf-DBB-Ir@CpG or Hf-DBB F -quantitative plots of biomarker interferon alpha (IFN-alpha, fig. 32G) and interleukin-6 (IL-6, fig. 32H) by enzyme-linked immunosorbent assay (ELISA) with increasing CpG ODN concentration, n=6. FIG. 32I is a graph of expression levels of ovalbumin Kb binding protein (SIINFEKL (SEQ ID NO: 3)) co-cultured with MC 38-egg cells at a 1:3 ratio, n=6. The legend in fig. 32I also applies to fig. 32F and 32H.
Fig. 33A-33C: delivery of pathogen-associated molecular patterns (PAMPs) and Dendritic Cell (DC) maturation. FIGS. 33A and 33B are graphs showing the expression levels of interferon alpha (IFN-. Alpha., FIG. 33A) and interleukin-6 (IL-6, FIG. 33B) quantified by quantitative polymerase chain reaction (qPCR). n=3. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a housekeeping gene for comparison of gene expression. DC and use of free CpG Oligodeoxynucleotides (ODN), hf-DBB-Ir@CpG or Hf-DBB F The MC38 egg cells pretreated with Ir@CpG plus X-ray radiation at a dose of 2 Gray (Gy) were co-cultured at a ratio of 1:3. Fig. 33C is a graph showing quantitative analysis of Kb-ova expression levels of DCs, n=3. Data are expressed as mean ± s.d. (n=3). t-test, P<0.05 and P<0.001。
Fig. 34A-34E: toxicity and efficacy in vivo. PBS (-), PBS (+), hf-DBB for C57BL/6 mice with mouse colon adenocarcinoma (MC 38) F -Ir@CpG(-)、CpG(+)、Hf-DBB F -Ir (+), hf-DBB-Ir (+), and Hf-DBB F -ir@cpg (+) treatment, n=6. FIG. 34A is a series of pairs of dimensions of 100-150 cubic millimeters (mm) 3 ) Immunoassay of tumor microenvironment for 7-day and 14-day MC38 tumor models, n=6. Fig. 34B is a photograph, and fig. 34C and 34C are graphs of the weight (in grams (g)) of the MC38 tumor-bearing mice resected tumors. Pancreatic tumor cells (Panc 02) tumor-bearing C57BL/6 mice were treated with PBS (-), PBS (+), PBS- (+) and PBS- (-), hf-DBB F -Ir@CpG(-)、CpG(+)、Hf-DBB F -Ir (+) and Hf-DBB F -ir@cpg (+) treatment, n=6. FIG. 34D is a photograph and FIG. 34E is a plot of tumor weight (in g) resected by Panc 02-loaded mice.
Fig. 35A-35J: nanoscale metal organic frameworks (nMOFs) for in situ personalized cancer vaccination to improve the innate immunity of in vivo anti-cancer therapies. FIGS. 35A and 35B are FIGS. 35A and 35B show the use of PBS (-), PBS (+), cpG (+), hf-DBB F -Ir(+)、Hf-DBB F Ir@CpG (-) or Hf-DBB F Tumor growth charts of colon adenocarcinoma (MC 38) (fig. 35A) and pancreatic carcinoma (Panc 02) (fig. 35B) tumor-bearing mice of ir@cpg (+) treated mice. Hf-DBB-Ir (+) was used as a control for the MC38 model, n=6. FIG. 35C is a sample of interleukin-6 (IL-6) and interferon-alpha (IFN-alpha) tested by ELISA assay 48 hours after the first irradiation (ELISA) Graph of quantitative plasma concentration in nanograms per liter (ng/L). Fig. 35D and 35E are graphs of the percentage (%) of total cells in tumor Draining Lymph Nodes (DLN) resected from MC38 tumor-bearing mice by macrophages (fig. 35D) and dendritic cells (DC, fig. 35E) relative to tumor neutralization and 2 days post-treatment. FIG. 35F is a tumor infiltrating CD80 + MHCII + Graph of the percentage of DC in DLN from cells excised from mice bearing MC38 relative to tumor neutralization at day 2 post-treatment. Figures 35G and 35H are graphs (μg/L) of total immunoglobulin G (IgG, figure 35G) and total immunoglobulin M (IgM, figure 35H) measured in micrograms/liter in plasma 2 days and 12 days after the fu-irradiation, n=6. FIG. 35I is SIINFEKL (SEQ ID NO: 3) -H excised from MC 38-egg tumor bearing mice on day 6 post-treatment 2 K b+ Percentage graph of cells. The legend in FIG. 35D also applies to FIGS. 35C and 35E-35I. FIG. 35J is a chart of the method using Hf-DBB F -Ir (+) or Hf-DBB F MC 38-egg tumor bearing Rag2 treated with or without OT-I T cell metastasis studies by Ir@CpG (+) -/- Tumor growth curve of mice (in cubic centimeters (cm) 3 ) Meter) is provided. Data are expressed as mean ± standard deviation (n=6). t-test, n.s.P>0.05,*P<0.05,**P<0.01 and P<0.001. The center line, box boundary and box must represent the average value, data range 25% -75%, 1.5 times outlier distance quartile spacing, respectively.
Fig. 36: PBS (-), PBS (+), hf-DBB F -Ir@CpG(-)、CpG(+)、Hf-DBB F -Ir(+)、Hf-DBB-Ir(+)、Hf-DBB F Weight map of tumor draining lymph nodes (unit: milligrams (mg)) of an ir@cpg (+) treated mouse colon adenocarcinoma (MC 38) tumor-bearing C57BL/6 mouse, n=6. t-test, P<0.05,**P<0.01,***P<0.001。
Fig. 37A-37I: in situ cancer vaccination in conjunction with Checkpoint Blocking Immunotherapy (CBI) employs the far-reaching effect (abscopal effect) of enhancing adaptive immunity. FIGS. 37A-37C show the use of PBS (-), PBS (+), alpha PD-L1 (+), hf-DBB F -Ir@CpG(+)、Hf-DBB F -Ir@CpG (-) +αPD-L1 or Hf-DBB F Primary treatment of colon adenocarcinoma (MC 38) tumor-bearing mice of-ir@cpg (+) +αpd-L1 treated mice (fig. 37A) and tumor growth of distal untreated (fig. 37B) tumorsCurves and survival curves (fig. 37C). Treatment was started on day 14 after tumor inoculation, at which time the tumor volume reached 100-150mm 3 . PBS or Hf-DBB injection at i.t F The mice were X-rayed at a dose of 1 gray (Gy)/fraction for five consecutive days 12 hours after ir@cpg. Antibody (αpd-L1) was injected i.t. once every three days at a dose of 75 micrograms (μg)/mouse, n=6. The legend in fig. 37A also applies to fig. 37B. FIGS. 37D-37I are graphs showing tumor infiltration CD45 10 days after treatment + Cells (FIG. 37D), dendritic Cells (DC) (FIG. 37E), CD80 + MHCII + DC (FIG. 37F), natural Killer (NK) cells (FIG. 37G), CD4 + T cells (Table 37H) and CD8 + Percentage plot of T cells. Data are expressed as mean ± standard deviation (n=6). t-test, P < 0.05, P < 0.01 and P < 0.001. The center line, box boundary and box must represent the average value, data range 25% -75%, 1.5 times outlier distance quartile spacing, respectively. The legend in FIG. 37E also applies to FIGS. 37D and 37F-37I.
Fig. 38A-38D: immunoassay of a bilateral model. Colon adenocarcinoma (MC 38) tumor bearing mice were treated with PBS (-), PBS (+), alpha PD-L1 (+), hf-DBB F -Ir@CpG(+)、Hf-DBB F -Ir@CpG+αPD-L1 (-) and Hf-DBB F -ir@cpg+αpd-L1 (+) treatment. Fig. 38A and 38B are graphs showing the percentage (%) of neutrophils (fig. 38A) or macrophages (fig. 38B) relative to total cells infiltrated in tumors excised from double sided MC38 tumor-bearing mice 10 days after the first radiation treatment, n=6. The legend in fig. 38A also applies to fig. 38B. Fig. 38C is a photograph, and fig. 38D is the weight (in milligrams (mg)) of resected tumor draining lymph nodes of mice, n=6.
Fig. 39A-39J: in situ cancer vaccination + checkpoint blocking the specificity and immune memory effects of immunotherapy (CBI). Fig. 39A (left) is a representative image of a series of colonies (top: control, bottom: stimulated with KSPWFTTL (SEQ ID NO: 5)) and (right) a statistical analysis of ELISpot assays (control or stimulated with SEQ ID NO: 5) for detecting T cells producing tumor specific interferon gamma (IFN- γ), n=6. Fig. 39B is a schematic diagram of a bilateral model created by subcutaneously injecting MC38 and B16F10 or LL2 cells (s.c.) flanking, respectively, primary and distal tumors.FIGS. 39C-39F are diagrams showing the use of PBS (+), alpha PD-L1 (+), hf-DBB F -Ir@CpG (+) or Hf-DBB F Primary treatment MC38 (fig. 39C and 39E) and distal untreated (B16F 10 of fig. 39D and LL2 of fig. 39F) tumor growth charts on unmatched bilateral tumor models of ir@cpg (+) +αpd-L1 treatment, n=4. FIGS. 39G and 39H are diagrams showing the use of PBS (-) or Hf-DBB F Treatment of MC38 tumor-bearing Rag2 with-Ir@CpG (+) +αPD-L1 -/- Primary (fig. 39G) and distal (fig. 39H) tumor growth patterns on the mouse model, n=6. FIG. 39I is a graph of tumor growth following challenge with MC38 tumor cells and re-challenge with B16F10 cells in cured mice treated according to FIG. 37C. FIG. 39J is CD44 High height CD62L Low and low Percent (%) of cells relative to total spleen cells, n=6. The legend in fig. 39A also applies to fig. 39J.
Fig. 40: quantitative Fluorescein Isothiocyanate (FITC) signal profile of FITC-mucin 1 (MUC-1)/cell, n=3, was shown by Confocal Laser Scanning Microscopy (CLSM). CLSM indicated that FITC-MUC-1/Hf-DBP-Pt was effective in delivering MUC-1 peptide into HEK293T cells.
Fig. 41: the graph of the analytical test results of 3- (4, 5-dimethylthiazol-2-yl) -5- (3-carboxymethoxyphenyl) -2- (4-sulfo-phenyl) -2H-tetrazolium (MTS) shows that mucin-1 (MUC-1) peptide can act synergistically with the radiation-powered therapy (RT-RDT) by hafnium-5, 15-bis (p-benzoic acid) porphyrin (Hf-DBP) +platinum (Pt) (Hf-DBP-Pt) to better kill cancer cells (mouse colon adenocarcinoma (MC 38) cells) upon X-ray irradiation in vitro.
Fig. 42: tumor growth (in cubic millimeters (mm) of C57BL/6 mice treated with Phosphate Buffered Saline (PBS), mucin-1 peptide (MUC-1) and a metallo-organic framework with surface conjugated MUC-1 (i.e., hf-DBP-Pt/MUC-1) 3 ) Measured tumor) graph. All mice received 1 Gray (Gy) X-rays daily for 6 consecutive days, while MUC-1/Hf-DBP-Pt system was least toxic.
Fig. 43: cpG oligodeoxynucleotide loading percentile for different nmofs.
FIG. 44 treatment of colon adenocarcinoma in mice intratumorally with Phosphate Buffered Saline (PBS), cyclic guanosine monophosphate adenosine monophosphate (cGAMP) or a metallo-organic framework with surface conjugated cGAMP (cGAMP/Hf-DBP-Pt)(MC 38) tumors of tumor-bearing C57BL/6 mice (in cubic millimeters (mm) 3 ) Measured tumor) growth chart. All mice received 2 gray (Gy) X-rays daily for 5 consecutive days starting on day 7. As shown by the trend towards stable body weight, the cGAMP/Hf-DBP-Pt system has minimal systemic toxicity.
Fig. 45A and 45B: a pair of graphs of the adsorption percentage of cyclic guanosine monophosphate adenosine (cGAMP) (fig. 45A) and cGAMP release profile (fig. 45B) for cGAMP/nanoscale metal-organic layer (nMOL) in different buffers.
Fig. 46: graph of Isothermal Titration Calorimetry (ITC) fitting results of titration of cyclic guanosine monophosphate adenosine (cGAMP) into aqueous solution of nanoscale metal organic layer (nMOL).
Fig. 47: by THP1-DUAL TM A graph of Interferon Regulatory Factor (IRF) response measured by KO-MyD88 reporter cells (InvivoGen, san Diego, california, united States of America). Cyclic guanosine monophosphate adenosine monophosphate (cGAMP)/nanometal organic layer (nMOL) has a lower half maximum effective concentration (EC 50) and higher IRF response level than free 2',3' -cGAMP.
Fig. 48: quantification of fluorescent signal after intratumoral injection of cyclic guanosine monophosphate adenosine (cGAMP) -Cy5 or cGAMP-Cy 5/nanoscale metal organic layer (nMOL). The nMOL retains and protects the cGAMP for a much longer time than the free cGAMP.
Fig. 49A and 49B: tumor growth charts (in cubic centimeters (cm) of mice colon adenocarcinoma (MC 38) tumor-bearing C57BL/6 mice (FIG. 49A) and mice colorectal carcinoma (CT 26) -cancer-bearing BALB/C mice (FIG. 49B) treated with (+) or without (-) irradiation by Phosphate Buffered Saline (PBS), nanoscale metalorganic layers (nMOL) and cGAMP/nMOL 3 ) Tumor measured by a meter). All mice received 2 gray (Gy) X-rays daily for 6 consecutive days, starting on day 7. The cGAMP/nMOL system showed minimal toxicity.
Fig. 50A and 50B: tumor growth curves (in cubic centimetres) of double-sided murine colon adenocarcinoma (MC 38) tumor-bearing C57BL/6 mice treated with Phosphate Buffered Saline (PBS), anti-apoptotic ligand 1 antibody (. Alpha.PD-L1), nanoscale metalorganic layers with surface conjugated cyclic guanosine monophosphate adenosine (cGAMP/nMOL) and. Alpha.PD-L1+cGAMP/nMOL with irradiationRice (cm) 3 ) Tumor measured by a meter). Fig. 50A is a graph of tumor growth for a primary tumor. Fig. 50B is a graph of tumor growth for distant tumors. All mice received 2Gy X-rays daily for 6 consecutive days starting on day 7. The αPD-L1 was injected intraperitoneally on days 10 and 13.
Reference to an electronically submitted sequence Listing
The contents of the list of electronically submitted sequences in the ASCII text file filed with the application (title: 3072-19-PCT. ST25.Txt; size: 2KB; date of creation: 2021, month 5, 21) are incorporated herein by reference.
Detailed Description
The subject matter of the present disclosure will now be described more fully hereinafter with reference to the accompanying examples, in which representative implementations are shown. The subject matter of the present disclosure may, however, be embodied in different forms and should not be construed as limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the embodiments to those skilled in the art.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the disclosed subject matter, representative methods, devices, and materials are now described. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.
Throughout the specification and claims, a given formula or name shall encompass all optical and stereoisomers, as well as racemic mixtures of such isomers and mixtures.
I. Definition of the definition
Although the following terms are believed to be well understood by those of ordinary skill in the art, the following definitions are set forth to aid in explaining the subject matter of the present disclosure.
In accordance with long-term patent statutes, the terms "a," an, "and" the "when used in this disclosure, including the claims, refer to" one or more. Thus, for example, reference to "a metal ion" includes a plurality of such metal ions, and so forth.
Unless otherwise indicated, all numbers expressing dimensions, reaction conditions, and so forth, used in the specification and claims are to be understood as being modified in all instances by the term "about". Accordingly, unless indicated to the contrary, the numerical parameters set forth in this specification and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by the presently disclosed subject matter.
As used herein, the term "about," when referring to a value or amount of size (i.e., diameter), weight, concentration, or percentage, means that in one embodiment, 20% or 10% of the specified amount is contemplated, in another embodiment 5% of the specified amount is contemplated, in another embodiment 1% of the specified amount is contemplated, and in yet another embodiment 0.1% of the specified amount is contemplated, as such a change is suitable for practicing the disclosed methods.
As used herein, the term "and/or" when used in the context of a list of entities refers to entities that exist alone or in combination. Thus, for example, the phrase "A, B, C and/or D" includes A, B, C and D, respectively, but also includes any and all combinations and subcombinations of A, B, C and D.
The term "comprising" is synonymous with "including," "containing," or "characterized by," and is inclusive or open-ended, and does not exclude additional, unrecited elements or method steps. "comprising" is a term used in the claim language that means that the named element is present but that other elements can be added and still form a structure or method within the scope of the claims.
As used herein, the phrase "consisting of …" excludes any element, step or component not specified in the claims. When the phrase "consisting of …" appears in one clause of the claim text, rather than immediately after the preamble, it merely limits the elements specified in that clause; other elements as a whole are not excluded from the stated claims.
As used herein, the phrase "consisting essentially of …" will limit the scope of the claims to the materials or steps specified, plus those materials and steps that do not materially affect the basic and novel characteristics of the claimed subject matter.
With respect to the terms "comprising," "consisting of …," and "consisting essentially of …," when one of these three terms is used herein, the subject matter of this disclosure and claimed may include the use of either of the other two terms.
As used herein, the term "alkyl" may refer to C 1-20 Including linear (i.e., "straight"), branched or cyclic, saturated or at least partially and in some cases fully unsaturated (i.e., alkenyl and alkynyl) hydrocarbon chains, including, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, pentyl, hexyl, octyl, ethenyl, propenyl, butenyl, pentenyl, hexenyl, octenyl, butadienyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl and propadienyl. "branched" refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a straight alkyl chain. "lower alkyl" refers to an alkyl group having 1 to about 8 carbon atoms, e.g., 1, 2, 3, 4, 5, 6, 7, or 8 carbons (i.e., C 1-8 Alkyl). "higher alkyl" refers to an alkyl group having from about 10 to about 20 carbon atoms, for example, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbons. In certain embodiments, "alkyl" refers specifically to C 1-8 A linear alkyl group. In other embodiments, "alkyl" refers specifically to C 1-8 Branched alkyl groups.
The alkyl group may optionally be substituted with one or more alkyl substituents which may be the same or different ("substituted alkyl"). The term "alkyl substituent" includes, but is not limited to, alkyl, substituted alkyl, halo, arylamino, acyl, hydroxy, aryloxy, alkoxy, alkylthio, arylthio, aralkoxy, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl. In some embodiments, one or more oxygen, sulfur, or substituted or unsubstituted nitrogen atoms can optionally be inserted along the alkyl chain, wherein the nitrogen substituent is hydrogen, lower alkyl (also referred to herein as "alkylaminoalkyl"), or aryl.
Thus, as used herein, the term "substituted alkyl" includes alkyl groups as defined herein wherein one or more atoms or functional groups of the alkyl group are substituted with another atom or group, including, for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxy, hydroxy, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
The term "aryl" is used herein to refer to an aromatic substituent that may be a single aromatic ring or multiple aromatic rings fused together, covalently linked or linked to a common group (e.g., without limitation, a methylene or ethylene moiety). The common linking group may also be a carbonyl group, as in benzophenone, or oxygen, as in diphenyl ether, or nitrogen, as in diphenylamine. The term "aryl" specifically includes heterocyclic aromatic compounds. The aromatic ring may include phenyl, naphthyl, biphenyl, diphenyl ether, diphenylamine, benzophenone, and the like. In particular embodiments, the term "aryl" refers to cyclic aromatic compounds containing from about 5 to about 10 carbon atoms, for example, 5, 6, 7, 8, 9, or 10 carbons, and includes 5-and 6-membered hydrocarbons and heterocyclic aromatic rings.
The aryl group may be optionally substituted with one or more aryl substituents which may be the same or different ("substituted aryl") wherein "aryl substituent" includes alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, hydroxy, alkoxy, aryloxy, aralkoxy, carboxy, acyl, halo, nitro, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, acyloxy, amido, aralkoxy, carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, arylthio, alkylthio, alkylene, and-NR 'R ", wherein R' and R" can each be independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, and aralkyl.
Thus, as used herein, the term "substituted aryl" includes aryl groups as defined herein wherein one or more atoms or functional groups of the aryl group are substituted with another atom or functional group, including, for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxy, hydroxy, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
Specific examples of aryl groups include, but are not limited to, cyclopentadienyl, phenyl, furan, thiophene, pyrrole, pyran, pyridine, imidazole, benzimidazole, isothiazole, isoxazole, pyrazole, pyrazine, triazine, pyrimidine, quinoline, isoquinoline, indole, carbazole, and the like.
"heteroaryl" as used herein refers to an aryl group containing one or more non-carbon atoms (e.g., O, N, S, se, etc.) in the backbone of the ring structure. Nitrogen-containing heteroaryl moieties include, but are not limited to, pyridine, imidazole, benzimidazole, pyrazole, pyrazine, triazine, pyrimidine, and the like.
"aralkyl" refers to an-alkyl-aryl group, optionally wherein the alkyl and/or aryl moieties are substituted.
"alkylene" means a straight or branched chain divalent aliphatic hydrocarbon radical having from 1 to about 20 carbon atoms, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbons. The alkylene group may be linear, branched or cyclic. Alkylene groups may also be optionally unsaturated and/or substituted with one or more "alkyl substituents". One or more oxygen, sulfur, or substituted or unsubstituted nitrogen atoms (also referred to herein as "alkylaminoalkyl") can optionally be inserted along the alkylene group, wherein the nitrogen substituent is an alkyl group as previously described. Exemplary alkylene groups include methylene (-CH) 2 (-) -; ethylene (-CH) 2 -CH 2 (-) -; propylene (- (CH) 2 ) 3 (-) -; cyclohexylidene (-C) 6 H 10 -);-CH=CH-CH=CH-;-CH=CH-CH 2 -;-(CH 2 ) q -N(R)-(CH 2 ) R-, wherein q and R are each independently integers from 0 to about 20, e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and R is hydrogen or low A lower alkyl group; methylenedioxy (-O-CH) 2 -O-); and ethylenedioxy (-O- (CH) 2 ) 2 -O-). The alkylene group may have from about 2 to about 3 carbon atoms, and may further have 6-20 carbons.
The term "arylene" refers to a divalent aromatic group, for example, a divalent phenyl or naphthyl group. Arylene groups can be optionally substituted with one or more aryl substituents and/or include one or more heteroatoms.
The term "amino" refers to the group-N (R) 2 Wherein each R is independently H, alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, or substituted aralkyl. The terms "aminoalkyl" and "alkylamino" may refer to the group-N (R) 2 Wherein each R is H, alkyl or substituted alkyl, and wherein at least one R is alkyl or substituted alkyl. "arylamine" and "aminoaryl" refer to the groups-N (R) 2 Wherein each R is H, aryl or substituted aryl, and wherein at least one R is aryl or substituted aryl, such as aniline (i.e., -NHC) 6 H 5 )。
The term "sulfanyl" may refer to the group-SR, wherein R is selected from the group consisting of H, alkyl, substituted alkyl, aralkyl, substituted aralkyl, aryl, and substituted aryl. Similarly, the terms "thioarylalkyl" and "thioaryl" refer to the-SR group, wherein R is aralkyl and aryl, respectively.
The terms "halogen", "halide" or "halogen" as used herein refer to fluorine, chlorine, bromine and iodine groups.
"haloalkyl" refers to an alkyl group substituted with one or more halo groups. "perhaloalkyl" refers to an alkyl group in which all hydrogen atoms attached to a carbon atom are replaced with halo groups. An exemplary perhaloalkyl group is trifluoromethyl (i.e., -CF) 3 )。
The terms "hydroxy" and "hydroxyl" refer to the-OH group.
The term "mercapto" or "thiol" refers to a-SH group.
The terms "carboxylate" and "carboxylic acid" may refer to the groups-C (=o) O-and-C (=o) OH, respectively. The term "carboxyl" may also refer to a-C (=o) OH group. In some embodiments, "carboxylate" or "carboxyl" may refer to a-C (=o) O-or-C (=o) OH group. In some embodiments, when the term "carboxylate" is used to refer to the anion of the SBU, the term "carboxylate" may be used to refer to the anion HCO 2 - Thus, it can be synonymous with the term "formate".
The term "carbonyl" refers to a-C (=o) -R group, wherein R is H, alkyl, substituted alkyl, aralkyl, substituted aralkyl, aryl, or substituted aryl.
The term "phosphonate" refers to-P (=o) (OR) 2 A group, wherein each R can independently be H, alkyl, aralkyl, aryl, or a negative charge (i.e., wherein there is effectively no R group bound to an oxygen atom, resulting in an unshared pair of electrons on the oxygen atom). Thus, in other words, each R may be present or absent and is selected from H, alkyl, aralkyl, or aryl when present.
The term "phosphate" refers to-OP (=o) (OR') 2 A group wherein R' is H or a negative charge.
The term "lipid" may refer to a hydrophobic or amphiphilic small molecule such as, but not limited to, a fatty acid, a phospholipid, a glycerolipid, a glycerophospholipid, a sphingolipid, a glycolipid, or a polyketone.
The term "bound" or "bound" and variants thereof may refer to covalent or non-covalent binding (e.g., hydrogen bonding, ionic bonding, van der Waals interactions, etc.). In some cases, the term "bind" refers to binding by coordination bonds or electrostatic interactions.
The term "conjugation" may also refer to a binding process, such as covalent attachment or formation of a coordinative bond.
As used herein, the term "metal-organic framework" or "MOF" refers to a solid two-dimensional or three-dimensional network comprising both metal and organic components, wherein the organic components include at least one, and typically more than one, carbon atom. In some embodiments, the material is crystalline. In some embodiments, the material is amorphous. In some embodiments, the material is porous. In some embodiments, the metal-organic matrix material is a coordination polymer comprising repeat units of a coordination complex comprising a metal-based secondary building block (SBU), such as a metal ion or a metal complex, and a bridged multidentate (e.g., bidentate or tridentate) organic ligand. In some embodiments, the material comprises more than one type of SBU or metal ion. In some embodiments, the material may comprise more than one type of organic bridging ligand.
The term "nanoscale metal-organic framework" may refer to a nanoscale structure comprising, consisting essentially of, or consisting of MOFs.
The terms "nanoscale," "nanomaterial," and "nanoparticle" refer to structures having regions with at least one dimension (e.g., length, width, diameter, etc.) less than about 1000 nm. In some embodiments, the size is smaller (e.g., less than about 500nm, less than about 250nm, less than about 200nm, less than 150nm, less than 125nm, less than 100nm, less than 80nm, less than 70nm, less than 60nm, less than 50nm, less than 40nm, or even less than 30 nm). In some embodiments, the dimension is from about 30nm to about 250nm (e.g., about 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 nm).
In some embodiments, the nanoparticle is approximately spherical. When the nanoparticle is approximately spherical, the characteristic dimension may correspond to a sphere diameter. The nanomaterial/nanoparticle may be disk-shaped, plate-shaped (e.g., hexagonal plate-shaped), rectangular, polyhedral, rod-shaped, cubic, or irregularly shaped, in addition to spherical.
As used herein, the terms "nanoplate," "metal organic nanoplate," and "MOP" refer to a MOF having a plate or disc shape, i.e., wherein the MOF is substantially longer and wider than its thickness. In some embodiments, the MOP is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 25, or 50 times longer and/or wider than its thickness. In some embodiments, the MOP thickness is less than about 100nm, 50nm, or about 30nm. In some embodiments, the MOP has a thickness of about 3nm to about 30nm (e.g., about 5, 10, 15, 20, 25, or 30nm thick). In some embodiments, the MOP has a thickness of about 3nm to about 12nm (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12nm thick). In some embodiments, the MOP is about two, three, four, five, six, seven, eight, nine, or ten layers thick. In some embodiments, the MOP has a thickness of about two to about five layers, wherein each layer has a thickness of about SBU. In some embodiments, the MOP is crystalline. In some embodiments, the MOP is amorphous. In some embodiments, the MOP is porous. In some embodiments, strong coordination modifiers, such as monocarboxylic acids, such as acetic acid (AcOH), formic acid, benzoic acid, or trifluoroacetic acid (TFA), are used to control the nanoplate morphology of MOP and introduce defects (missing bridging ligands) to enhance ROS diffusion through MOP channels.
As used herein, the term "metal-organic layer" (or "MOL") refers to a solid, primarily a two-dimensional network, comprising a metal and an organic component, wherein the organic component comprises at least one, and typically more than one carbon atom. In some embodiments, the MOL is crystalline. In some embodiments, MOL is amorphous. In some embodiments, the MOL is porous.
In some embodiments, MOL is a coordination polymer comprising repeat units of a coordination complex comprising a metal-based Secondary Building Unit (SBU), such as a metal ion or a metal complex, and a bridged polydentate (e.g., bidentate or tridentate) organic ligand. In some embodiments, the bridging ligand is substantially planar. In some embodiments, a majority of the bridging ligands bind to at least three SBUs. In some embodiments, the material comprises more than one type of SBU or metal ion. In some embodiments, the material may comprise more than one type of bridging ligand.
In some embodiments, the MOL can be substantially a monolayer coordination complex network between the SBU and the bridging ligand, wherein the monolayer extends in the x-and y-planes, but has a thickness of only about one SBU.
In some embodiments, the MOL can be a substantially planar monolayer coordination complex network between the SBU and the bridging ligand, wherein substantially all of the bridging ligand is in the same plane. In some embodiments, more than 80%, 85%, 90%, or 95% of the bridging ligands are substantially in the same plane. In some embodiments, more than 95%, 96%, 97%, 98%, 99% or about 100% of the bridging ligands are in the same plane. Thus, while the MOL may extend in the x-and y-planes a distance that may comprise the lengths and/or diameters of the SBUs and bridging ligands, in some embodiments the MOL may have a thickness of only about one SBU. In some embodiments, the MOL has a thickness of about 3nm or less (e.g., about 3nm, 2, or about 1nm or less), and the MOL has a width, length, and/or diameter that is at least about 5 times, 10 times, 20 times, 30 times, 40 times, 50 times, 60 times, 70 times, 80 times, 90 times, or about 100 times or more the MOL thickness. In some embodiments, the MOL has a plate or disc shape. In some embodiments, a coordinating modifier, such as a monocarboxylic acid, such as acetic acid (AcOH), formic acid, benzoic acid, or trifluoroacetic acid (TFA), is used to control the nanoplate morphology of the MOL and/or introduce defects (missing bridging ligands), while enhancing ROS diffusion through the MOL channels or modulating other properties of the MOL.
The terms "metal-organic framework", "nanoscale metal-organic framework", "MOF", and/or "nMOF", as used herein, generally refer to a solid metal-organic matrix material particle, as compared to MOL and/or MOP, wherein the MOF has a length, width, thickness, and/or diameter that is greater than about 30 or 31nm (or greater than about 50nm or greater than about 100 nm), and/or wherein the MOF has a width, length, and/or diameter that is no greater than 5 times or more than the MOF thickness.
"coordination complex" refers to a compound in which there is a coordination bond between a metal ion and an electron pair donor, ligand or chelating group. Thus, a ligand or chelating group is typically an electron pair donor, a molecule or molecular ion having an unshared electron pair that can be utilized for the supply of metal ions.
The term "coordination bond" refers to an interaction between an electron pair donor and a coordination site on a metal ion, resulting in an attractive force between the electron pair donor and the metal ion. The use of this term is not intended to be limiting, as certain coordination bonds can also be classified as having more or less covalent properties (if not entirely covalent properties) based on the identity of the metal ion and electron pair donor.
As used herein, the term "ligand" generally refers to a substance, such as a molecule or ion, that interacts (e.g., binds) in some manner with another substance. More specifically, as used herein, a "ligand" may refer to a molecule or ion that binds a metal ion in solution to form a "coordination complex. See Martell,A.E.,and Hancock,R.D.Metal Complexes in Aqueous Solutions, plenum: new York (1996), incorporated herein by reference in its entirety. The terms "ligand" and "chelating group" may be used interchangeably. The term "bridging ligand" may refer to a group that binds more than one metal ion or complex, thereby providing a "bridge" between metal ions or complexes. The organic bridging ligands may have two or more groups separated by, for example, an alkylene or arylene group, having unshared electron pairs. Groups having unshared electron pairs include, but are not limited to, -CO 2 H、-NO 2 Amino, hydroxy, thio-, sulfanyl, -B (OH) 2 、-SO 3 H、PO 3 H. Phosphonates and heteroatoms in the heterocycle (e.g., nitrogen, oxygen or sulfur).
The term "coordination site" as used herein with respect to a ligand (e.g., a bridged ligand) refers to an unshared pair of electrons, a negative charge, or an atom or functional group that forms an unshared pair of electrons or a negative charge (e.g., by deprotonation at a particular pH).
"electrostatic binding" refers to the attractive force between two fully or partially ionized species of opposite charge.
The term "small molecule" refers to a non-polymeric natural or synthetic molecule. Small molecules typically have a molecular weight of about 900 daltons (Da) or less (e.g., about 800Da, about 750Da, about 700Da, about 650Da, about 600Da, or about 550Da or less).
The term "macromolecule" as used herein refers to a molecule of greater than about 900 Da. In some embodiments, the macromolecule is a polymer or biopolymer, e.g., a protein or nucleic acid.
The terms "polymer" and "polymeric" refer to chemical structures having repeating units (i.e., multiple replicators of a given chemical substructure). The polymer may be formed from a polymerizable monomer. A polymerizable monomer is a molecule that comprises one or more moieties that can react with moieties on other molecules of the polymerizable monomer to form a bond (e.g., a covalent bond or a coordination bond). In general, each polymerizable monomer molecule can be combined with two or more other molecules. In some cases, the polymerizable monomer is only bound to another molecule, forming the end of the polymeric material.
The polymer may be organic or inorganic, or a combination thereof. As used herein, the term "inorganic" refers to a compound or composition containing at least some atoms other than carbon, hydrogen, nitrogen, oxygen, sulfur, phosphorus, or one halogen. Thus, for example, the inorganic compound or composition may contain one or more silicon atoms and/or one or more metal atoms.
As used herein, "organic polymers" are those polymers that do not contain silicon oxide or metal atoms in their repeating units. Exemplary organic polymers include polyvinylpyrrolidone (pvon), polyesters, polyamides, polyethers, polydienes, and the like. Some organic polymers contain biodegradable linkers, such as esters or amides, that enable them to degrade over time under biological conditions.
The term "hydrophilic polymer" as used herein generally refers to hydrophilic organic polymers such as, but not limited to, polyvinylpyrrolidone (PVP), polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazole, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethyl acrylate, hydroxymethyl cellulose, hydroxyethyl imine (PEI), polyethylene glycol (i.e., PEG) or another hydrophilic poly (alkylene oxide), polyglycerol, and polyaspartic acid. The term "hydrophilic" refers to the ability of a molecule or chemical to interact with water. Thus, hydrophilic polymers are typically polar, or have groups that can hydrogen bond with water.
"polypeptide" and "peptide" refer to polymers composed of amino acid residues linked by peptide (amide) bonds, related natural structural variants, and synthetic non-natural analogs thereof. In some embodiments, "peptide" refers to a polymer consisting of 2-50 amino acid residues.
"synthetic peptide or polypeptide" refers to a non-natural peptide or polypeptide. Synthetic peptides or polypeptides may be synthesized, for example, using an automated polypeptide synthesizer. Various methods of solid phase peptide synthesis are known to those skilled in the art.
The term "protein" generally refers to a large polypeptide (e.g., >50 amino acid residues). The polypeptide sequences are described herein using conventional symbols: the left end of the polypeptide sequence is the amino terminus; the right end of the polypeptide sequence is the carboxy terminus.
The term "preventing" as used herein refers to preventing something from happening or taking precautions against what may or may happen. In the medical context, "prevention" generally refers to actions taken to reduce the chances of a disease or illness. It is noted that "prevention" is not necessarily absolute, but can therefore occur to some extent.
In some embodiments, a "prophylactic" or "preventative" treatment is a treatment of a subject that does not exhibit symptoms of a condition, disease, or disorder, or that exhibits only early symptoms. Thus, prophylactic or preventative treatment can be performed with reduced risk of pathological development associated with developing a condition, disease or disorder.
The term "photosensitizer" (PS) refers to a compound or moiety capable of being excited by light of a specific wavelength, typically visible or Near Infrared (NIR) light, and generating Reactive Oxygen Species (ROS). For example, in its excited state, a photosensitizer may undergo intersystem crossing and transfer energy to oxygen (O 2 ) (e.g., in PDT treated tissue) to produce ROS, such as singlet oxygen [ ] 1 O 2 )。
In some embodiments, the photosensitizer is a porphyrin, chlorophyll, dye, or derivative or analog thereof, such as porphyrin, chlorophyll, or a dye comprising one or more additional aryl or alkyl substituents, having one or more carbon-carbon double bonds substituted with carbon-carbon single bonds, and/or comprising a substituent (e.g., a substituted alkylene group) capable of being covalently substituted with a bond to an organic bridging ligand. In some embodiments, porphyrins, chlorine, bacterial chlorine, or porphyrinenes can be used. In some embodiments, the photosensitizer may have one or more functional groups, such as carboxylic acid, amine, or isothiocyanate, for example, for linking the photosensitizer to another molecule or moiety, such as an organic bridging ligand or SBU, and/or for providing additional one or more sites to enhance coordination or to coordinate additional one or more metals. In some embodiments, the photosensitizer is a porphyrin or a derivative or analog thereof. Exemplary porphyrins include, but are not limited to, hematoporphyrins, protoporphyrins, and Tetraphenylporphyrins (TPPs). Exemplary porphyrin derivatives include, but are not limited to, pyropheophorbide, bacteriochlorophyll, chlorophyll a, benzoporphyrin derivatives, tetrahydroxyphenyl chlorophyllins, purines, benzophenones, naphthochlorophyllins, vedins, rotins, oxo chlorophyllins, azachlorophyllins, bacteriochlorophyllins, tolylporphyrins, and benzobacteriochlorophyllins. Porphyrin analogs include, but are not limited to, extended porphyrin family members (e.g., texaphrins, saphyrins and hexaphyrins), porphyrin isomers (e.g., porphyrinenes, flipped porphyrins (inverted porphyrins), phthalocyanines, and naphthalocyanines), and TPPs substituted with one or more functional groups.
In some embodiments, the PS is a metal coordination complex comprising a metal (e.g., ru or Ir) and one or more nitrogen donor ligands (e.g., one or more nitrogen-containing aromatic groups). In some embodiments, the one or more nitrogen donor ligands are selected from the group including, but not limited to, bipyridine (bpy), phenanthroline, terpyridine, or phenylpyridine (ppy), each of which may be optionally substituted with one or more aryl substituents (e.g., on a carbon atom of the aryl group).
The term "cancer" as used herein refers to a disease caused by uncontrolled cell division and/or cell metastasis or the ability to establish new growth at other sites. The terms "malignant", "tumor" and "carcinoma" and variants thereof refer to a cancer cell or group of cancer cells.
Specific types of cancers include, but are not limited to, skin cancer (e.g., melanoma), connective tissue cancer (e.g., sarcoma), fat cancer, breast cancer, head and neck cancer, lung cancer (e.g., mesothelioma), stomach cancer, pancreatic cancer, ovarian cancer, cervical cancer, uterine cancer, anogenital cancer (e.g., testicular cancer), kidney cancer, bladder cancer, colorectal cancer (i.e., colon or rectal cancer), prostate cancer, central Nervous System (CNS) cancer, retinal cancer, blood cancer, neuroblastoma, multiple myeloma, and lymphoma (e.g., hodgkin's lymphoma and non-hodgkin's lymphoma).
The term "metastatic cancer" refers to cancer that spreads from an initial site (i.e., a primary site) within a patient.
The terms "anti-cancer drug," "chemotherapeutic drug," and "anti-cancer prodrug" refer to drugs (i.e., compounds) or prodrugs that are known or suspected to be capable of treating cancer (i.e., killing cancer cells, inhibiting proliferation of cancer cells, or treating symptoms associated with cancer). In some embodiments, the term "chemotherapy" as used herein refers to a non-PS molecule that is used to treat cancer and/or has cytotoxic capabilities. Such more traditional or conventional chemotherapeutic agents can be described by mechanism of action or class of compounds and can include, but are not limited to, alkylating agents (e.g., melphalan), anthracyclines (e.g., doxorubicin), cytoskeletal disrupting agents (e.g., paclitaxel), epothilones, histone deacetylase inhibitors (e.g., inhibitors of topoisomerase I or II (e.g., irinotecan or etoposide), kinase inhibitors (e.g., bortezomib), nucleotide analogs or precursors thereof (e.g., methotrexate), peptide antibiotics (e.g., bleomycin), platinum-based drugs (e.g., cisplatin or oxaliplatin), retinoids (e.g., retinoic acid), and vinca alkaloids (e.g., vinblastine).
General considerations
Cancer vaccines have been developed for decades to amplify tumor-specific T cell responses (Goldman and DeFrancesco, 2009). In particular, cancer vaccines based on tumor antigens have been widely studied clinically, resulting in approval by the U.S. food and drug administration (United States Food and Drug Administration) of prostate cancer vaccine Sipleucel T (Hu et al, 2018) based on prostatectonase. However, traditional approaches to cancer vaccine development face several obstacles, including tumor heterogeneity with different somatic mutations, and thus also different tumor antigens in patients (Tanyi et al,2018; sahin and tureci, 2018), failure to deliver peptide tumor antigens to lymph nodes due to rapid renal clearance and enzymatic degradation (Purcell et al,2007;Du et al,2018), failure to evade immune surveillance by Antigen Presenting Cells (APCs) (Liu et al, 2014) and tumor mechanisms such as the programmed death-1/programmed death ligand 1 (PD-1/PD-L1) axis (Joyce and Fearon, 2015) on tumor antigen internalization.
Personalized vaccines using neoantigens or autologous whole tumor lysates can overcome tumor heterogeneity (Kuai et al, 2017), but their production process is lengthy, complex and expensive (Scheetz et al, 2019). One promising strategy to improve personalized cancer vaccination is the in situ generation of tumor antigens using immunostimulating therapy, which can provide systemic anti-tumor immune responses in a personalized way and modulate the local tumor microenvironment to alleviate immunosuppression (Wang et al, 2018). For example, intratumoral injection of oncolytic viruses such as talimogene laherparepvec (T-VEC) produces direct cytotoxic effects on cancer cells and antigen presentation with DCs acting as an in situ cancer vaccine with reduced side effects (gillieet al, 2008). Non-viral therapies with potent anti-tumor effects, such as phototherapy (Castano et al, 2006), radiotherapy (RT) (Weichselbaum et al,2014;Chao et al,2018), and some chemotherapies, can also generate danger-related molecular patterns (DAMP) and tumor antigens (Kepp et al, 2019) by inducing immunogenic cell death.
Stimulation of DCs with immunoadjuvants such as interferon gene Stimulators (STING) agonists (Shae et al 2019;Luo et al,2017) or CpG Oligodeoxynucleotides (ODNs) further promotes antigen presentation and immune responses (Kliman, 2004). CpG is a short DNA strand widely explored as a vaccine adjuvant for toll-like receptor 9 (TLR 9) stimulation, DC maturation, antigen presentation and priming of tumor-specific Cytotoxic T Lymphocytes (CTLs) naturally occurring as microbial DNA, known as pathogen-associated molecular patterns (PAMPs) (Figdor et al, 2004). Furthermore, in the absence of immune adjuvants, antigen presentation of immature DCs induces tolerance, rather than stimulating an immune response (Lutz and schulter, 2002). In particular, C class CpG can enhance the production of type I Interferons (IFNs) to activate DCs and stimulate B cells, thereby up-regulating costimulatory molecules and secreting pro-inflammatory cytokines, providing excellent anti-cancer effects (Radovic-Moreno et al,2015;Brody et al,2010). However, like peptide vaccines, even topically administered CpG is susceptible to enzymatic degradation and, due to its anionic nature, cannot be efficiently internalized by APC (brown et al,2010;Rosi et al,2006).
In the context of antigen and adjuvant delivery, nanotechnology is at the front of the antitumor activity of mixed drug delivery and immunostimulation to elicit cancer vaccination (Jiang et al,2017;Nam et al,2019;Song et al,2017;Zhang et al,2019;Ma et al,2019,Lou et al,2009; and Wilson et al, 2019). In one aspect, the presently disclosed subject matter is based on a nanoscale metal-organic framework (nMOF) for personalized cancer vaccination by X-ray activated production of DAMP and tumor antigens with CpG effectively delivered as PAMP to APCs. However, cpG is but one exemplary agent that can be delivered by nMOF. Other immunotherapeutic agents including small molecule or macromolecular arrays can be delivered using nMOFs according to the presently disclosed subject matter, as described further below.
nMOF, assembled from tunable metal-oxygen clusters and functional organic ligands, has become a novel porous and crystalline molecular nanomaterial with interesting potential for biomedical applications (Furukawa et al,2013;Wang et al,2017). nMOF exhibits potent antitumor activity by generating highly cytotoxic and immunogenic Reactive Oxygen Species (ROS) under external light or X-ray irradiation (Lan et al,2019a;Ni et al,2018a). nMOF is also capable of converting X-ray energy directly into ROS (Ni et al,2018 b) by unique radiotherapy-radiodynamic therapy (RT-RDT). According to one aspect of the presently disclosed subject matter, cationic nMOFs are designed by molecular engineering, while DAMP and tumor antigens are released by X-ray activated RT-RDT, thereby delivering CpG through, for example, electrostatic interactions. The in situ vaccination provided by nMOF effectively expands cytotoxic T cells in tumor draining lymph nodes, while reactivating the adaptive immune system for tumor regression. See fig. 23. The local therapeutic effect of nMOF-based in situ vaccines was extended to distal tumors by combination therapy with anti-PD-L1 antibodies (αPD-L1), while providing a cure rate of 83.3% for MC38 colorectal cancer models.
Other exemplary nMOFs for treating cancer by activating the immune system are also described herein. For example, nMOF co-delivers (i) the Toll-like receptor 7/8 (TLR 7/8) agonist Imiquimod (IMD) as a pathogen-associated molecular pattern (PAMP), and (ii) the antibody cluster of differentiated 46 antibodies (αCD47) for macrophage modulation and reversal of immunosuppression, as described below. See fig. 1.IMD repolarizes immunosuppressive M2 macrophages to immunostimulatory M1 macrophages, while antibodies block CD47 tumor cell surface markers to promote phagocytosis. IMD may sequester in the pores of the nMOF core, while the surface of nMOF is modified to enhance its ability to bind to antibodies. The nMOF combined with IMD and αCD47 for intratumoral injection followed by low grade X-ray irradiation showed enhanced tumor regression relative to αCD47, IMD or bare nMOF treatment alone. When nMOF triggers RT-RDT, IMD and αCD47 in combination with an anti-PDL 1 immune checkpoint inhibitor, following X-ray exposure, co-regulate the immunosuppressive tumor microenvironment and active innate immunity to coordinate adaptive immunity, which can lead to complete eradication of primary and distal tumors in a bilateral colorectal cancer model. Thus, nMOF provides an effective platform for co-delivery of a variety of immunoadjuvants to induce systemic immune responses and provide improved anti-tumor efficacy.
III nanoscale Metal organic frameworks
In some embodiments, the presently disclosed subject matter relates to Metal Organic Frameworks (MOFs) (e.g., nanoscale MOFs (nMOFs), such as MOF nanoparticles or nanoscale Metal Organic Layers (MOLs)) that are modified to improve the surface absorption of therapeutic agents compared to parent, unmodified MOFs.
In some embodiments, the therapeutic agent targets the immune system. Such therapeutic agents include, but are not limited to, PAMPS, such as Toll-like receptor (TLR) agonists and RIG-I like receptor (RLR) agonists and STING agonists; oncolytic molecules, including small molecule oncolytic molecules targeting lysosomes, such as PV-10, mitochondrial targeting peptides, such as Lu Temi peptide (ruxoemide); cytokines including, but not limited to, interleukins (IL) (e.g., IL-2, IL-7, IL-12, IL-15 and other interleukins and natural or synthetic derivatives thereof), interferons (IFNs) (e.g., type I, II, III IFNs, including IFN- α, IFN- β, IFN- γ and other IFNs and natural or synthetic derivatives thereof), and Tumor Necrosis Factors (TNF) (e.g., TNF- α and natural or synthetic derivatives thereof); antibodies, including monoclonal antibodies (mabs) that target PD-1, PD-L1, CTLA, CD47, sirpa, CD28, OX40, CD127, and CD40, as well as bispecific monoclonal antibodies; and nucleic acids such as, but not limited to siRNA, miRNA, mRNA and non-coding RNAs (ncrnas). Exemplary TLR agonists and RLR agonists include TLR3 agonists such as poly (I: C), poly (a: U), double-stranded RNA (dsRNA) and natural or synthetic derivatives thereof; TLR4 agonists such as Lipopolysaccharide (LPS) and natural and synthetic derivatives thereof, such as monophosphoryl lipid a (MPLA), CRX-527, G100, etc.; TLR7/8 agonists, including imidazoquinoline compounds such as Imiquimod (IMD), jidiaquinimod, resiquimod, CL075, CL097, CL264, CL307; benzazepine analogs such as VTX-2337, TL8-506; single stranded RNA (ssRNA) and natural and synthetic derivatives thereof; TLR-9 agonists, such as CpG Oligonucleotides (ODNs), including class a, class B and class C CpG ODNs, double-stranded DNA (dsDNA), and natural and synthetic derivatives thereof; and RLR Ligands (RLRs) targeting RIG-I like receptors, such as dsRNA like 5' ppp-dsRNA;3 p-hairpin RNA; poly (dA: dT); poly (I: C); and natural and synthetic derivatives thereof. Exemplary STING agonists include, but are not limited to, cyclic Dinucleotides (CDNs), including natural CDNs, such as 2'3' -cGAMP, 3' -cGAPP, c-di-AMP, c-di-GMP; adenosine cyclophosphate-inosine phosphate (caimep); xanthine analogues such as DMXAA; and natural and synthetic derivatives thereof, such as CL656 and ADU-S100; and phosphodiesterase inhibitors, such as ENPP1/2 inhibitors and SVPD inhibitors.
IIIA modified metal organic frameworks
In some embodiments, the presently disclosed subject matter relates to modified Metal Organic Frameworks (MOFs), e.g., nmofs, that include metal-containing Secondary Building Units (SBUs) linked together by organic bridging ligands. For example, the organic bridging ligand may include at least two functional groups (e.g., carboxylate or phosphate) capable of forming a coordination bond with a metal ion, and the two SBU units can be linked together by forming a coordination bond with a metal ion on each of the two SBUs with one of the two functional groups. In some embodiments, the SBU is a metal oxide cluster. In some embodiments, the metal ions of the SBU are capable of absorbing X-rays. In a parent, unmodified MOF, the SBU may include a strongly coordinating end capping group such as an acetate or formate group that coordinates to the metal ion. Such strongly coordinating end capping groups may be removed by treatment with trimethylsilyl trifluoroacetate, trimethylsilyl triflate, or an inorganic acid of suitable acid strength to exchange the strongly coordinating end capping groups for weakly coordinating groups, such as trifluoroacetate or trifluoromethanesulfonate groups, in the modified MOF. The weakly coordinating groups can then be substituted with carboxylate groups of the protein or phosphate groups of carboxylate-containing small molecules or nucleic acids, thereby attaching the protein, peptide, small molecule or nucleic acid to the MOF surface by coordination to the metal ion. Alternatively, a combination of electron withdrawing bridging ligands can be used to increase the defects of the end capping groups and the cationic charge on the bridging ligands to increase the electrostatic interaction between the MOF and the macromolecule. Thus, the term "modified MOF" also refers to MOFs having electron withdrawing bridging ligands. Methods of preparing the parent MOF to be modified are described, for example, in U.S. patent No. 10206871, the disclosure of which is incorporated herein by reference in its entirety.
The modified MOFs can serve as a delivery platform for one or more MOF surface attachment therapeutics (e.g., one or more therapeutics targeted to the immune system). In some embodiments, the modified MOF can provide improved absorption of a surface-attached therapeutic agent. In addition, the pores/cavities of the MOF may be further loaded with small molecule chemotherapeutic agents such as, but not limited to, cisplatin, carboplatin, paclitaxel, SN-35, etoposide, and the like; or small molecule inhibitors such as, but not limited to, PLK1 inhibitors (TAK-960, ON01910, BI 2536, etc.), wnt inhibitors (CCT 036477, etc.), bcl-2 inhibitors, PD-L1 inhibitors, ENPP1 inhibitors or IDO inhibitors. Thus, the modified MOFs can act as multiple agent delivery platforms for combinations of agents with different solubilities.
In some embodiments, the presently disclosed subject matter provides nanoparticles (e.g., MOF nanoparticles (nmofs)) or nanoparticle formulations for treating cancer. The nanoparticle (e.g., nMOF) can comprise a metal-containing secondary building block (e.g., a metal-oxygen cluster, such as a Hf metal-oxygen cluster) and an organic bridging ligand. Attached to the surface of the nMOF may be one or more macromolecules, such as αCD47 or CpG ODN, and/or one or more small molecule therapeutic agents to activate the immune system against the tumor. Additional therapeutic agents (e.g., additional chemotherapeutic or immunotherapeutic agents), such as IMD, may be loaded in the pores and/or cavities of the MOF.
For example, as described in the examples below, exemplary IMD-nMOF-DBP-CD47 nanoparticles were prepared. In a carcinoma model of large intestine, intratumoral treatment with IMD-nMOF-DBP-CD47 followed by low-grade X-ray irradiation provides enhanced tumor regression compared to CD47, imiquimod or nMOF DBP alone. Thus, in some embodiments, the presently disclosed subject matter provides improved tumor treatment using lower doses of CD47 and imiquimod, which may be toxic when delivered at current systemic doses. In some embodiments, MOFs can deliver peptides (e.g., mucin-1 peptide) or STING activators/agonists, such as cyclic guanosine monophosphate (cGAMP) or CpG Oligodeoxynucleotides (ODN).
In some embodiments, the presently disclosed subject matter relates to MOFs (e.g., nmofs) as locally activatable immunotherapy to release risk-related molecular patterns (DAMP) and tumor antigens, and deliver pathogen-related molecular patterns for in situ personalized cancer vaccination. As described in the examples below, cationic nmofs, when activated by X-rays, can be effective to generate reactive oxygen species to release DAMP and tumor antigens, while at the same time delivering anionic CpG ODN as PAMPs to promote antigen presenting cell maturation.
In some embodiments, the presently disclosed subject matter provides MOFs (e.g., nmofs, such as MOF nanoparticles or MOL) having surfaces modified to coordinately or electrostatically bind one or more therapeutic agents of interest. In some embodiments, the MOF comprises: (a) A plurality of metal oxide cluster Secondary Building Units (SBUs), wherein the metal oxide clusters SBUs each comprise one or more first metal ions and one or more anions, wherein the one or more anions each coordinate with one or more of the one or more first metal ions; and (b) a plurality of organic bridging ligands linking the plurality of SBUs together to form a two-dimensional or three-dimensional matrix. In some embodiments, each of the plurality of SBUs at the surface of the MOF comprises a weakly coordinating anion as the SBU capping group anion. Alternatively, in some embodiments, the plurality of organic bridging ligands comprises an organic bridging ligand comprising an electron withdrawing group or ligand, a positive charge, or a combination thereof. In some embodiments, the plurality of organic bridging ligands comprises a ligand comprising a nitrogen donor group coordinately bound to a second metal ion, wherein the second metal ion is further coordinated to at least one second metal ligand comprising one or more electron withdrawing groups. For example, the modified MOFs of the present disclosure can have a surface that has enhanced (i.e., increased) ability to coordinately or electrostatically bind to one or more therapeutic agents of interest as compared to a MOF surface that does not contain weakly coordinated end capping groups or organic bridging ligands that contain electron withdrawing groups or ligands. For example, the MOF can have more sites on its surface that can bind through coordinate binding or electrostatic interactions with the therapeutic agent.
In some embodiments, the one or more first metal ions are metals that absorb ionizing radiation. In some embodiments, the ionizing radiation that is absorbable by the metal ion is X-rays. In some embodiments, one or more of the one or more first metal ions are ions of an element selected from the group consisting of, but not limited to: hf. Lanthanide metals (La, ce, pr, nd, pm, sm, eu, gd, tb, dy, ho, er, tm, yb or Lu), ba, ta, W, re, os, ir, pt, au, pb, and Bi. In some embodiments, the MOF is free of Bi and/or W ions. In some embodiments, the first metal ion is a Hf ion. In some embodiments, the plurality of SBUs are metal oxygen clusters, e.g., hf oxygen clusters. In some embodiments, the oxygen cluster comprises a compound selected from the group consisting of oxides (O 2- ) Hydroxide (OH) - )、S 2- 、SH - And formic acid radical (HCO) 2 - ) Is an anion of (a).
In some embodiments, each of the plurality of SBUs on the surface of the MOF comprises a weakly coordinating anion as a capping group. "blocking group" refers to a metal ligand that coordinates to only one metal ion in the SBU. In some embodiments, the capping group is a metal ligand in the MOF SBU located on the surface of the MOF (e.g., the upper or lower surface of the MOL or the outer surface of the MOF nanoparticle). In some embodiments, the weakly coordinating anion is selected from trifluoroacetate and trifluoromethanesulfonate. These weakly coordinating anions can be introduced by modifying a MOF that contains more strongly coordinating end-capping groups such as acetate, formate, or benzoate. In some embodiments, the MOF comprises Hf 12 Oxygen cluster or Hf 6 Oxygen clusters.
Each SBU is bound to at least one other SBU by coordination binding to the same bridging ligand. In other words, each SBU is bound via a coordination bond to at least one bridging ligand that is also coordinately bound to at least one other SBU.
Any suitable bridging ligand may be used. In some embodiments, each bridging ligand is an organic compound comprising multiple coordination sites. The coordination sites may each include a group capable of forming a coordination bond with a metal cation or a group capable of forming such a group. Thus, each coordination site may contain a non-shared electron pair, a negative charge, or an atom or functional group capable of forming a non-shared electron or a negative charge. Typical coordination sites include, but are not limited to, functional groups such as carboxylates and derivatives thereof (e.g., esters, amides, anhydrides), nitrogen-containing groups (e.g., amines, nitrogen-containing aromatic and non-aromatic heterocycles), alcohols, phenols, and other hydroxy-substituted aromatic groups; ethers, phosphonates, phosphates, thiols, and the like.
In some embodiments, each bridging ligand comprises 2-10 coordination sites (i.e., 2, 3, 4, 5, 6, 7, 8, 9, or 10 coordination sites). In some embodiments, each bridging ligand is capable of binding two or three SBUs. In some embodiments, each organic bridging ligand is a dicarboxylic acid salt or a tricarboxylic acid salt. For example, the organic bridging ligand may be a porphyrin, a smectite or a bacteriosmectite substituted with two carboxylate groups or two substituents each containing a carboxylate group. In some embodiments, two carboxylate groups can each form a coordinate bond with a metal ion of two separate SBUs, while a nitrogen atom of a porphyrin, chlorophyllin, or bacteriochlorophyllin is capable of forming a coordinate bond with another cation or cations (e.g., another metal cation).
In some embodiments, each organic bridging ligand comprises at least two groups, wherein each of the two groups is individually selected from the group consisting of carboxylate salts, aromatic or non-aromatic nitrogen-containing groups (e.g., pyridine, piperidine, indole, acridine, quinolone, pyrrole, pyrrolidine, imidazole, pyrimidine, pyridazine, pyrazine, triazole, and oxazole), phenol, acetylacetonate (acac), phosphonate, and phosphate. In some embodiments, at least one bridging ligand is a carboxylate-containing ligand, a pyridine-containing bridging ligand, a phenol-containing ligand, an acetylacetone-containing bridging ligand, a phosphonate-containing bridging ligand, or a phosphate-containing bridging ligand. In some embodiments, at least one bridging ligand comprises at least two carboxylate groups.
In some embodiments, the plurality of organic bridging ligands comprises a porphyrin substituted with at least two carboxylate groups, optionally wherein the plurality of organic bridging ligands comprises 5, 15-bis (p-benzoate) porphyrin (DBP).
In some embodiments, each bridging ligand is an organic compound comprising a plurality of coordination sites, wherein the plurality of coordination sites are substantially coplanar or capable of forming coplanar coordination bonds.
Exemplary organic bridging ligands include, but are not limited to,
/>
/>
/>
wherein X (if present) is selected from H, halo (e.g., cl, br or I), OH, SH, NH 2 Nitro (NO) 2 ) Alkyl, substituted alkyl (e.g., hydroxy-substituted alkyl, mercapto-substituted alkyl, or amino-substituted alkyl), and the like. In some embodiments, X comprises a covalently linked photosensitizer such as, but not limited to, a dye, porphyrin, chlorophyllin, bacteriochlorophyllin, porphyrinene, or chlorophyll, or a derivative or analog thereof. For example, X can be a porphyrin covalently linked to a bridging ligand through an alkylene linking moiety and an amide, ester, thiourea, disulfide, or ether linkage.
In some embodiments, the linking group of the organic bridging ligand comprises a nitrogen donor moiety. In some embodiments, the organic bridging ligand may comprise a nitrogen donor moiety selected from, but not limited to, bipyridine, phenylpyridine, phenanthroline, and terpyridine, which can be optionally substituted at one or more carbon atoms of the nitrogen donor moiety with one or more aryl substituents. In some embodiments, the at least one organic bridging ligand comprises a ligand selected from the group consisting of 4,4' -bis (4-benzoic acid) -2,2' -bipyridine (DBB), 4',6' -dibenzoic acid- [2,2' -bipyridine ] -4-carboxylate (BPY), and 4' - (4-carboxyphenyl) - [2,2':6', 2' -terpyridine ] -5,5 ' -dicarboxylate (TPY). In some embodiments, the at least one organic bridging ligand comprises DBB.
In some embodiments, the MOF further comprises a small molecule therapeutic (e.g., a small molecule chemotherapeutic) sequestered in the pores and/or cavities of the two-or three-dimensional MOF network. In some embodiments, the small molecule therapeutic is a chemotherapeutic drug, a small molecule inhibitor, and/or a small molecule immunomodulator. In some embodiments, the small molecule therapeutic is a small molecule chemotherapeutic such as, but not limited to, cisplatin, carboplatin, paclitaxel, SN-35, and etoposide. In some embodiments, the small molecule therapeutic is a small molecule inhibitor, such as, but not limited to, a marquee kinase 1 (PLK 1) inhibitor, a Wnt inhibitor, a B-cell lymphoma 2 protein (Bcl-2) inhibitor, a PD-L1 inhibitor, an exonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP 1) inhibitor, and an indoleamine 2, 3-dioxygenase (IDO) inhibitor. In some embodiments, the IDO inhibitor is selected from the group including, but not limited to, indomethacin (i.e., 1-methyl-D-tryptophan), BMS-986205, ai Kaduo stat (i.e., ICBN 24360), and 1-methyl-L-tryptophan. In some embodiments, the small molecule therapeutic is a small molecule immunomodulator. In some embodiments, the small molecule immunomodulator is Imiquimod (IMD).
In some embodiments, the plurality of organic bridging ligands comprises an organic bridging ligand comprising a nitrogen donor group, wherein the nitrogen donor group coordinates to a second metal ion, and wherein the second metal cation further coordinates to at least one second metal ligand comprising one or more electron withdrawing groups, optionally wherein the one or more electron withdrawing groupsIs a halo or haloalkyl (e.g., perhaloalkyl) group. For example, in some embodiments, the organic bridging ligand comprising a nitrogen donor group comprises a pyridine or bipyridine moiety. In some embodiments, the organic bridging ligand comprising a nitrogen donor group is 4,4 '-bis (p-benzoic acid) -2,2' -bipyridine (DBB). In some embodiments, the second metal ion is an iridium (Ir) or ruthenium (Ru) ion. In some embodiments, the second metal ion is an Ir ion. In some embodiments, the second metal ion coordinates to two second metal ligands, wherein one or both of the second metal ligands comprises one or more electron withdrawing groups, such as, but not limited to, halogen, carbonyl, sulfonyl, cyano, nitro, and haloalkyl (e.g., perhaloalkyl). In some embodiments, the haloalkyl is halogen substituted methyl. In some embodiments, the haloalkyl is perhaloalkyl (i.e., wherein all hydrogen atoms of the alkyl are substituted with halogen). In some embodiments, one or both of the second metal ligands is bipyridine (bpy) or phenylpyridine (ppy) substituted with one or more electron withdrawing groups. In some embodiments, the bpy or ppy is substituted with at least two groups selected from halo and haloalkyl. In some embodiments, the bpy or ppy is substituted with at least two or three groups selected from fluoro (F) and trifluoromethyl. In some embodiments, one or both of the second metal ligands is 2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine (dF (CF) 3 )ppy)。
In some embodiments, the MOF has a Zeta (Zeta) -potential value of at least about 5 millivolts (mV) (e.g., at least about 5mV, at least about 10mV, at least about 15mV, at least about 20mV, or at least about 25 mV). In some embodiments, the MOF has a zeta potential value of at least about 30mV.
In some embodiments, the MOF comprises a three-dimensional network. In some embodiments, the three-dimensional network is provided in the form of nanoparticles. In some embodiments, the MOF comprises a two-dimensional network. In some embodiments, the MOF comprises a nanoscale MOL.
IIIB Metal organic frameworks with surface-bound therapeutic Agents
In some embodiments, the presently disclosed subject matter provides MOFs (e.g., MOF nanoparticles or MOL) for delivering one or more therapeutic agents of interest. In some embodiments, one or more of the one or more therapeutic agents of interest are attached to the surface of the MOF by coordination bonds or electrostatic interactions. In some embodiments, the MOF comprises: (a) A plurality of metallo-oxo cluster Secondary Building Units (SBUs), wherein each of the metallo-oxo cluster SBUs comprises one or more first metal ions and one or more anions, wherein each of the anions coordinates to one or more first metal ions; (b) A plurality of organic bridging ligands linking together the plurality of SBUs to form a two-dimensional or three-dimensional matrix; and (c) one or more therapeutic agents of interest bound to the MOF surface by a coordination bond or electrostatic interaction, optionally wherein one or more therapeutic agents of interest are coordinately bound to the metal ions of one or more of the plurality of SBUs of the MOF surface.
In some embodiments, the first metal ion is a metal ion that absorbs ionizing radiation. The ionizing radiation absorbing metal includes high Z metals (i.e., elements having Z (i.e., atomic number or proton number) greater than 40). The ionizing radiation energy may include, for example, X-rays, gamma (gamma) -rays, beta (beta) -radiation, or proton radiation. In some embodiments, the first metal ion is an X-ray absorbing metal ion. In some embodiments, the first metal ion is an ion of a metal selected from Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi. In some embodiments, the first metal ion is a Hf ion.
Any suitable therapeutic agent of interest may be bound to the surface. In some embodiments, the one or more therapeutic agents of interest are selected from the group consisting of nucleic acids, small molecule therapeutic agents comprising phosphate or carboxylate groups, and macromolecules comprising surface accessible phosphate or carboxylate groups. In some embodiments, the therapeutic agent is a therapeutic agent that targets the immune system, such as one of the agents described above. In some embodiments, the macromolecule is a protein or peptide. In some embodiments, the protein is an antibody or antibody fragment (e.g., an antibody fragment comprising an antigen binding region). In some embodiments, the protein is selected from the group consisting of antibodies, such as, but not limited to, an anti-cluster 37 (CD 37) antibody, an anti-cluster 44 (CD 44) antibody, an anti-cluster 47 (CD 47) antibody, an anti-cluster 73 (CD 73) antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-lymphocyte activation gene 3 (LAG 3) antibody, and an anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) antibody. In some embodiments, the protein is an anti-PD-1 antibody or an anti-PD-L1 antibody.
In some embodiments, one of the one or more therapeutic agents of interest is a peptide. In some embodiments, the peptide is a peptide that targets annexin-1 (MUC-1) mucin. In some embodiments, the peptide has a cysteine-glutamine-cysteine (CQC) motif. In some embodiments, the peptide is a D-amino acid sequence. In some embodiments, the D-amino acid sequence comprises or consists of the amino acid sequence D-CQCRRKN (SEQ ID NO: 1). In some embodiments, the peptide is a transmembrane peptide. In some embodiments, the peptide comprises or consists of the amino acid sequence RRRRRRRRRCQCRRKN (SEQ ID NO: 2).
In some embodiments, one of the one or more therapeutic agents of interest comprises or consists of a nucleic acid. For example, the nucleic acid may be DNA, RNA, miRNA, mRNA, siRNA, ODN or a cyclic dinucleotide. In some embodiments, the ODN is a CpG ODN (i.e., a short single-stranded DNA comprising cytosine followed by guanine). In some embodiments, the cyclic dinucleotide is a STING agonist, such as, but not limited to, c-di-AMP or cGAMP.
In some embodiments, the MOF further comprises one or more additional therapeutic agents that sequester in the pores or cavities of the two-dimensional or three-dimensional network (e.g., in the pores within the core of the MOF nanoparticle). In some embodiments, the MOF comprises from about 1wt% to about 50wt% (e.g., about 1wt%, about 5wt%, about 10wt%, about 15wt%, about 20wt%, about 25wt%, about 30wt%, about 35wt%, about 40wt%, about 45wt%, or about 50 wt%) of the one or more additional therapeutic agents. In some embodiments, the one or more additional therapeutic agents may be selected from the group consisting of small molecule chemotherapeutic agents, small molecule inhibitors, and small molecule immunomodulators. The small molecule chemotherapeutic agents include, but are not limited to, cisplatin, carboplatin, paclitaxel, SN-35, and etoposide. In some embodiments, the small molecule inhibitor may be selected from the group consisting of a PLK1 inhibitor, a Wnt inhibitor, a Bcl-1 inhibitor, a PD-L1 inhibitor, an ENPP1 inhibitor, and an IDO inhibitor. In some embodiments, the additional therapeutic agent comprises a small molecule immunomodulator. In some embodiments, the small molecule immunomodulator is IMD.
In some embodiments, the plurality of SBUs comprises Hf oxygen clusters and the plurality of organic bridging ligands comprises DBPs. Thus, in some embodiments, the MOF comprises Hf-DBP nanoparticles or MOL, wherein one or more therapeutic agents of interest are bound to the surface of the MOF by coordination bonds to Hf ions of the surface accessible SBU. In some embodiments, the one or more therapeutic agents of interest include one or more antibodies. In some embodiments, the one or more therapeutic agents comprise an anti-CD 47 antibody. In some embodiments, the MOF further comprises an IMD sequestered in a pore or cavity of a two-dimensional or three-dimensional network of MOFs.
In some embodiments, the MOF is a three-dimensional network and is provided as a nanoparticle. In some embodiments, the MOF comprises from about 1wt% to about 50wt% IMD or anti-CD 47 antibody. In some embodiments, the MOF comprises about 1wt% to about 25wt% IDM or anti-CD 47 antibody. In some embodiments, the MOF comprises about 9wt% imd and about 7.5wt% anti-CD 47 antibody.
In some embodiments, the plurality of SBUs comprise Hf oxygen clusters and the plurality of organic bridging ligands comprise DBBs coordinated to a second metal ion (e.g., ir or Ru ions), wherein the second metal ion (e.g., ir or ruthenium ions) is further coordinated to two (dF (CF) 3 ) ppy) coordination; and wherein the one or more therapeutic agents of interest are bound to the surface of the MOF by electrostatic interactions. In some embodiments, the one or more therapeutic agents of interest comprise a nucleic acid. In some embodiments, the nucleic acid is a STING agonist orCpG ODN. In some embodiments, the nucleic acid is a CpG ODN.
In some embodiments, the MOF comprises from about 1wt% to about 50wt% of the one or more therapeutic agents of interest (e.g., about 1, about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, or about 50wt% of the one or more therapeutic agents of interest). In some embodiments, the MOF comprises from about 1wt% to about 25wt% of the one or more therapeutic agents of interest. In some embodiments, the one or more therapeutic agents of interest comprise antibodies.
In some embodiments, the plurality of SBUs comprise Hf oxygen clusters, the plurality of organic bridging ligands comprise DBPs, wherein the nitrogen atoms of the DBPs are coordinated to metal ions (e.g., pt ions), and the one or more therapeutic agents of interest are bound to the surface of the MOF. In some embodiments, the MOF comprises a nanoparticle. In some embodiments, the one or more agents of interest are selected from one or more MUC-1 peptides (i.e., one or more peptides that target MUC-1), cpG ODNs, and cGAMP. In some embodiments, the one or more SBUs comprise Hf oxygen clusters and the one or more organic bridging ligands comprise Ir (DBB) [ dF (CF) 3 )ppy] 2 + (i.e., DBB) F -Ir) and cGAMP binds to the surface of the MOF. In some embodiments, the MOF is MOL.
IIIC pharmaceutical preparation
In some embodiments, the presently disclosed subject matter provides a pharmaceutical composition or formulation comprising (i) a MOF as described above comprising one or more therapeutic agents of interest bound to the surface of the MOF, and (ii) a pharmaceutically acceptable carrier, e.g., a pharmaceutically acceptable carrier for humans. In some embodiments, the composition may further comprise other components such as, but not limited to, lipids, antioxidants, buffers, bacteriostats, bactericidal antibiotics, suspending agents, thickening agents, and solutes that render the composition isotonic with the body fluid of the subject to which the composition is to be administered. In some embodiments, the pharmaceutical composition or formulation further comprises other therapeutic agents, such as conventional chemotherapeutic agents or immunotherapeutic agents. For example, in some embodiments, the pharmaceutical composition or formulation further includes an antibody immunotherapeutic (e.g., an antibody immune checkpoint inhibitor, such as, but not limited to, an anti-PD-1/PD-L1 antibody, an anti-CTLA-4 antibody, an anti-OX 40 antibody (i.e., also known as the tumor necrosis factor receptor superfamily, member 4 (TNFRSF 4)), an anti-T cell immunoglobulin and mucin-containing domain 3 (TIM 3) antibody, an anti-LAG 3 antibody, and an anti-CD 47 antibody).
In some embodiments, the compositions of the presently disclosed subject matter comprise compositions comprising a pharmaceutically acceptable carrier. Any suitable pharmaceutical formulation may be used to prepare the composition for administration to a subject. In some embodiments, the composition and/or carrier may be pharmaceutically acceptable to humans.
For example, suitable formulations may include aqueous and non-aqueous sterile injection solutions, which may contain antioxidants, buffers, bacteriostats, bactericidal antibiotics, and solutes that render the formulation isotonic with the body fluids of the subject; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers (e.g., sealed ampoules and vials) and may be stored in a frozen or freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water for injection, immediately prior to use. Some exemplary ingredients are Sodium Dodecyl Sulfate (SDS), in one example at a concentration in the range of 0.1-10mg/mL, in another example at a concentration of about 2.0 mg/mL; and/or mannitol or another sugar, e.g., at a concentration in the range of 10-100mg/mL, in another example at a concentration of about 30 mg/mL; and/or Phosphate Buffered Saline (PBS).
It should be understood that the formulations of the presently disclosed subject matter may include other formulations conventional in the art, in addition to the ingredients specifically mentioned above, in view of the type of formulation. For example, sterile pyrogen-free aqueous and nonaqueous solutions may be used.
IV. method of treatment
In some embodiments, the presently disclosed subject matter provides a method of treating cancer in a subject in need thereof. In some embodiments, the method comprises administering to the subject a MOF as described above, wherein the MOF comprises a plurality of metal oxide clusters SBU, wherein each metal oxide cluster SBU comprises one or more first metal ions and one or more anions, wherein each of the anions coordinates with one or more of the first metal ions; a plurality of organic bridging ligands linked together by the plurality of SBUs to form a two-dimensional or three-dimensional network; and one or more therapeutic agents of interest bound to the MOF surface by coordination bonds or electrostatic interactions (e.g., metal ions of one or more SBUs bound to the MOF surface). In some embodiments, the method further comprises exposing the subject (i.e., at least a portion of the subject) to ionizing radiation energy (e.g., X-rays, gamma radiation, or proton radiation). For example, after waiting a period of time (e.g., minutes to hours) after administration of the MOF to allow the MOF to localize to a tumor site, the subject may be exposed to ionizing radiation energy. The time period may be adjusted according to the method of administering the MOF. In some embodiments, the ionizing radiation energy is X-rays. In some embodiments, the MOF is administered directly to a tumor or intravenously. In some embodiments, a portion of the anatomy of a subject suffering from cancer or the vicinity of a site affected by cancer is exposed to ionizing radiation.
The subject may be exposed to ionizing radiation energy in any suitable manner and/or using any suitable device, such as those currently used for delivering X-rays in a medical or veterinary environment. In some embodiments, the X-ray source and/or output may be refined to enhance disease treatment. For example, the X-rays may be generated using peak voltages, currents, and/or optionally selected filters to minimize patient DNA damage due to X-ray radiation and maximize scintillator absorption of X-rays.
In some embodiments, conventional techniques, intensity Modulated Radiation Therapy (IMRT), image Guided Radiation Therapy (IGRT) or Stereotactic Body Radiation Therapy (SBRT), are used, 60 Co radiation sources, implanted radiation seeds as used in brachytherapy, positive voltage or overvoltage X-raysA line illuminator, a high energy electron beam generated by a LINAC, or a proton source. In some embodiments, the irradiating may include generating X-rays using tungsten or another metal target, a cobalt-60 source (cobalt unit), a linear accelerator (linac), an Ir-192 source, and a cesium-137 source. In some embodiments, irradiating includes passing X-rays (e.g., X-rays generated using a tungsten target) or other ionizing radiation through a filter prior to irradiating the subject. In some embodiments, the filter may include an element having an atomic number of at least 20. In some embodiments, the filter comprises copper (Cu). In some embodiments, the filter may have a thickness of less than about 5 millimeters (mm). In some embodiments, the filter may have a thickness of less than about 4mm (e.g., less than about 3mm, less than about 1mm, less than about 0.5mm, less than about 0.4mm, less than about 0.3mm, less than about 0.2mm, or less than about 0.1 mm).
The X-rays may be generated using peak voltages, currents and/or optionally filters selected to minimize patient DNA damage due to X-ray radiation and maximize scintillator absorption of X-rays. In some embodiments, X-rays are generated using a peak voltage of less than about 230 kVp. In some implementations, the peak voltage is less than about 225kVp, less than about 200kVp, less than about 180kVp, less than about 160kVp, less than about 140kVp, less than about 120kVp, less than about 100kVp, or less than about 80kVp. In some embodiments, a peak voltage of about 120kVp is used to generate X-rays.
In some embodiments, the X-rays are generated by temporarily or permanently placing a radioactive source inside the subject. In some embodiments, the MOFs of the presently disclosed subject matter are injected with the implantation of a radioactive source.
In some embodiments of the presently disclosed subject matter, the X-ray (or other ionizing radiation energy) source can be refined to enhance the RT-RDT effect of the MOF, thereby enabling more efficient killing of cancer cells. In some embodiments, the X-ray irradiator may comprise a panoramic irradiator comprising at least one X-ray source within a shielded enclosure, the one or more sources each operable to emit an X-ray flux in an area equal to a surface area facing proximal the tumor. See U.S. patent application publication nos. 2010/0189222 and WO 2011/049743, each of which is incorporated herein by reference. X-ray generators based on tungsten target emission are suitable for this application. The output energy is typically in the range of 100-500 kV. In certain embodiments, the application relates to a removable attenuator or filter of at least one selected material comprising at least one metal having an atomic number > 20. Each attenuator may be a flat plate or a flat plate having a gradient thickness. See U.S. patent No. 7430282, incorporated by reference in its entirety. The attenuator may also be modulated with a periodically spaced grid/holes. The output X-ray energy may also be adjusted after the attenuator filters to maximize the energy absorption of the radiosensitizer/radioscintillator in the application. An X-ray bandpass filter having an X-ray refractive lens for refracting X-rays may also be used. See WO2008/102632, the entire contents of which are incorporated herein by reference.
In some embodiments, the method further comprises administering to the subject an additional therapeutic agent or treatment, such as an immunotherapeutic agent and/or cancer treatment. In some embodiments, the additional therapeutic agent or treatment is selected from the group consisting of surgery, chemotherapy, toxin therapy, cryotherapy, and gene therapy. The additional cancer treatment may be selected based on the treating cancer and/or other factors such as the patient's treatment history, overall health, etc., at the best discretion of the treating physician.
In some embodiments, the additional cancer treatment may include administration of a conventional chemotherapeutic agent, such as, but not limited to, a platinum-containing agent (e.g., cisplatin or oxaliplatin or a prodrug thereof), doxorubicin, daunorubicin, docetaxel, mitoxantrone, paclitaxel, digitalis, digoxin, and a hypusin (septacidin), or another conventional chemotherapeutic agent known in the art. The additional chemotherapeutic agent may be present in (e.g., encapsulated or coordinated to or covalently bound to) the MOF. In addition, the additional chemotherapeutic agent may be present in the same pharmaceutical composition or formulation as the MOF, or in a separate pharmaceutical composition or formulation, administered prior to, at the time of, or after administration of the pharmaceutical composition and formulation comprising the MOF and/or radiation.
In some embodiments, the additional cancer treatment may include administering to the patient a pharmaceutical formulation selected from the group consisting of a polymer micelle formulation, an asymmetric lipid bilayer, a liposome formulation, a dendrimer formulation, a polymer-based nanoparticle formulation, a silica-based nanoparticle formulation, a nanoscale coordination polymer formulation, a nanoscale metal-organic framework formulation, and an inorganic nanoparticle (gold, iron oxide nanoparticle, etc.) formulation. In some embodiments, the pharmaceutical formulation may be a formulation comprising a conventional chemotherapeutic agent.
The immunotherapeutic agent used in accordance with the presently disclosed subject matter may be any suitable immunotherapeutic agent known in the art. Immunotherapeutic agents suitable for use in the presently disclosed subject matter include, but are not limited to: PD-1, PD-L1, CTLA-4, IDO and CCR7 inhibitors, i.e., compositions that inhibit or alter the function, transcription stability, translation, modification, localization or secretion of polynucleotides or polypeptides encoding a target or target-related ligand, such as anti-target antibodies, small molecule antagonists of a target, peptides that block a target, blocking fusion proteins of a target, or small interfering ribonucleic acid (siRNA)/shRNA/microrna/pDNA of a target. Antibodies that may be used according to the presently disclosed subject matter include, but are not limited to: anti-CD 52 (Alemtuzumab), anti-CD 20 (ofatuzumab), anti-CD 20 (Rituximab), anti-CD 47 antibodies, anti-GD 2 antibodies, and the like. Conjugated monoclonal antibodies for use in accordance with the presently disclosed subject matter include, but are not limited to: radiolabeled antibodies (e.g., temozolomide (Ibritumomab tiuxetan) (Zevalin) and the like), chemically labeled antibodies (antibody-drug conjugates (ADCs)), (e.g., brinetuximab (Brentuximab vedotin) (Adcetris), adotrastuzumab (Ado-trastuzumab emtansine) (Kadcyla), diniinterleukin (denilukin diftitox) (Ontak) and the like). Cytokines used in accordance with the presently disclosed subject matter include, but are not limited to: interferons (i.e., IFN- α, INF- γ), interleukins (i.e., IL-2, IL-12), TNF- α, etc. Other immunotherapeutic agents used in accordance with the presently disclosed subject matter include, but are not limited to, polysaccharide-K, neoantigens, and the like.
In some embodiments, the immunotherapeutic agent may be selected from agonists of a DNA or RNA sensor, such as a RIG-I agonist (e.g., a compound described in U.S. patent No. 7271156, which is incorporated herein by reference in its entirety), a TLR3 agonist (e.g., polyinosinic acid: polycytidylic acid), a TLR 7 agonist (e.g., IMD), a TLR-9 agonist (e.g., cpG ODN), and a STING agonist (e.g., STING vax or ADU-S100). In some embodiments, the immunotherapeutic agent is selected from the group consisting of a PD-1 inhibitor (e.g., peberolizumab) or nivolumab (nivolumab)), a PD-L1 inhibitor (e.g., atizolizumab), avistuzumab (avelumab) or duvalumaab), a CTLA-4 inhibitor (e.g., ipilimumab (ipilimumaab)), an IDO inhibitor (e.g., indoximod (indoximod), BMS-986205, or Ai Kaduo stavta (epacoaddostat)), and a CCR7 inhibitor. In some embodiments, the immunotherapeutic agent is selected from the group consisting of, but not limited to, anti-PD-1/PD-L1 antibodies, anti-IDO inhibitors, anti-CTLA-4 antibodies, anti-OX 40 antibodies, anti-TIM 3 antibodies, anti-LAG 3 antibodies, siRNA targeted PD-1/PD-L2, siRNA targeted IDO, and siRNA targeted CC chemokine receptor 7 (CCR 7), as well as any other immunotherapeutic agent described elsewhere herein or known in the art.
Thus, in some embodiments, the presently disclosed subject matter provides a method of treating cancer in combination with X-ray induced RDT and immunotherapy. Accordingly, in some embodiments, the presently disclosed subject matter provides a method comprising the steps of: administering to a patient a MOF or MOL as described herein comprising one or more surface-bound therapeutic agents; and irradiating at least a portion (e.g., a fiftieth portion) of the patient with X-rays; and administering an immunotherapeutic agent to the patient. The immunotherapeutic agent may be administered simultaneously with or before or after the MOF and/or irradiation. In some embodiments, the immunotherapeutic agent is selected from the group consisting of, but not limited to: agonists of DNA or RNA sensors, such as RIG-1 agonists, toll-like receptor 3 (TLR 3) agonists (e.g., polyinosinic acid: polycytidylic acid), toll-like receptor 7 (TLR 7) agonists (e.g., IMD), toll-like receptor 9 (TLR 9) agonists (e.g., CPG ODNs), interferon gene (STING) agonists (e.g., STINGVAX or ADU-S100), or IDO inhibitors. In some embodiments, the IDO inhibitor is selected from the group including, but not limited to, indomethacin (i.e., 1-methyl-D-tryptophan), BMS-986205, ai Kaduo stat (i.e., ICBN 24360), and 1-methyl-L-tryptophan. In some embodiments, the immunotherapeutic agent is an immune checkpoint inhibitor. The immune checkpoint inhibitor may be an antibody, such as an anti-PD-1/PD-L1 antibody (i.e., an anti-PD-1 antibody or an anti-PD-L1 antibody), an anti-CTLA-4 antibody, an anti-OX 40 antibody, an anti-TIM 3 antibody, an anti-LAG 3 antibody, or an anti-CD 47 antibody. In some embodiments, the immunotherapeutic agent is an anti-PD-L1 antibody.
In some embodiments, the cancer is selected from the group consisting of a head tumor, a neck tumor, a breast cancer, a gynecological tumor, a brain tumor, a colorectal cancer, a lung cancer, a mesothelioma, a soft tissue sarcoma, a skin cancer, a connective tissue cancer, a fat cancer, a gastric cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a central nervous system cancer, a retinal cancer, a blood cancer, a neuroblastoma, a multiple myeloma, a lymphatic cancer, and a pancreatic cancer. In some embodiments, the disease is selected from colorectal cancer, melanoma, head and neck cancer, brain cancer, breast cancer, liver cancer, lung cancer, and pancreatic cancer. In some embodiments, the disease is selected from colorectal cancer, melanoma, lung cancer, and pancreatic cancer. In some embodiments, the disease is a metastatic cancer.
In some embodiments, the use of MOFs of the present disclosure provides an extended release profile for one or more therapeutic agents of interest (e.g., as compared to administration of free, non-MOF-bound therapeutic agents of interest). In some embodiments, the release rate is adjustable. In some embodiments, the MOF provides sustained release of one or more therapeutic agents of interest over a period of hours or days. In some embodiments, administration of the MOF reduces the therapeutically effective dose of the one or more therapeutic agents of interest (e.g., independent of the MOF as compared to administration of the same therapeutic agent of interest in free form). In some embodiments, the sustained release of the therapeutic agent may be modulated from 4 hours to 2 weeks. In some embodiments, this can be achieved by adjusting the number and charge number of metal open coordination sites on the SBU and the charge number and electron density of the organic ligand and by selecting a suitable charge, coordination strength, and multivalent therapeutic agent.
V. subject
The methods and compositions disclosed herein can be used on samples in vitro (e.g., on isolated cells or tissues) or in vivo (i.e., in vivo, such as a patient) in a subject. In some embodiments, the subject or patient is a human subject, but it should be understood that the principles of the presently disclosed subject matter indicate that the presently disclosed subject matter is effective with respect to all vertebrate substances, including mammals, which are intended to be included in the terms "subject" and "patient". Furthermore, mammals should be understood to include any mammalian species, particularly agricultural and domestic mammalian species, for which it is desirable to use the compositions and methods disclosed herein.
Thus, the methods of the presently disclosed subject matter are particularly useful in warm-blooded vertebrates. Thus, the presently disclosed subject matter relates to mammals and birds. More specifically, the methods and compositions provided are applicable to mammals such as humans, as well as those mammals of great importance (e.g., northeast tigers), mammals of economic importance (farm animals raised for human consumption), and/or mammals of social importance, e.g., carnivores other than humans (e.g., cats and dogs), pigs (swine, pigs, and wild boars), rodents (e.g., mice, hamsters, guinea pigs, etc.), ruminants (e.g., cows, cattle, sheep, giraffes, deer, goats, bison, and camels), and horses. Also provided are treatments for birds, including treatment of endangered birds raised in zoos or as pets (e.g., parrots) and poultry, particularly poultry, for example, poultry such as turkeys, chickens, ducks, geese, guinea fowl, and the like, as they are also of economic importance to humans. Thus, treatment of livestock is also provided, including, but not limited to, domesticated pigs (pigs and castors), ruminants, horses, poultry, and the like.
VI administration of
Suitable methods for administering the compositions of the presently disclosed subject matter include, but are not limited to, intravenous and intratumoral injection, oral, subcutaneous administration, intraperitoneal injection, intracranial injection, and rectal administration. Alternatively, the composition may be deposited in any other manner, for example, by spraying the composition within the pulmonary passages, at the site where treatment is desired. The particular mode of administration of the compositions of the presently disclosed subject matter depends on a variety of factors, including the distribution and abundance of the cells to be treated and the mechanism by which the composition is metabolized or cleared from its site of administration. For example, relatively superficial tumors may be injected intratumorally. In contrast, internal tumors can be treated after intravenous injection.
In some embodiments, the method of administration includes features that localized delivery or accumulation at the site to be treated. In some embodiments, the composition is delivered intratumorally. In some embodiments, selective delivery of the composition to the subject is achieved by intravenous injection of the composition, followed by ionizing radiation treatment (e.g., X-ray irradiation) of the subject.
For delivery of the composition to the pulmonary pathways, the compositions of the presently disclosed subject matter may be formulated as an aerosol or a coarse spray. Methods of preparation and administration of aerosol or spray formulations can be found, for example, in U.S. patent No. 5858784;6,013,638;6,022,737; and 6,136,295.
VII dosage of
An effective dose of the composition of the presently disclosed subject matter is administered to a subject. An "effective amount" refers to an amount of a composition sufficient to produce a detectable treatment. The actual dosage levels of the components of the compositions of the presently disclosed subject matter can be varied so as to administer the composition in an amount effective to achieve a desired effect on a particular subject and/or target. The selected dosage level may depend on the activity (e.g., RT-RDT activity or MOF and/or MOL loading) and route of administration of the composition.
After reviewing the presently disclosed subject matter disclosed herein, one of ordinary skill in the art would consider the specific formulation, method of administration used with the composition, and nature of the target to be treated, and can tailor the dosage of each subject. Such adjustments or variations and the evaluation of when and how such adjustments or variations are made are well known to those of ordinary skill in the art.
Modification method of metal organic framework
In some embodiments, the presently disclosed subject matter provides a method of enhancing the interaction and/or binding of one or more therapeutic agents of interest with the surface of a MOF. In some embodiments, the method comprises modifying the surface of a MOF (i.e., a MOF comprising a plurality of metal oxide clusters SBUs and a plurality of organic bridging ligands linking the plurality of SBUs together) by: (i) Providing one or more surface accessible coordination sites that coordinate to a weakly coordinating anion that may be substituted with carboxylate or phosphate substituents of a therapeutic agent of interest, or (ii) providing a MOF comprising one or more electron withdrawing bridging ligands, one or more positively charged bridging ligands, or a combination thereof.
In some embodiments, a method comprises (ia) providing a parent MOF comprising metal oxygen cluster SBUs linked together by an organic bridging ligand, wherein each of said SBUs comprises one or more metal ions and one or more anions, and wherein said MOF comprises a plurality of surface accessible metal oxygen cluster SBUs, wherein said one or more anions of each of said surface accessible metal oxygen cluster SBUs comprise a strongly coordinating anion that is an SBU capping group; and (ib) replacing the strongly coordinating anion with a weakly coordinating anion by contacting the parent MOF with a suitable reagent to remove the strongly coordinating anion. In some embodiments, the strongly coordinating anion is an acetate or formate anion. In some embodiments, the agent is selected from the group consisting of trimethylsilyl trifluoroacetate (TMS-TFA), trimethylsilyl triflate, and mineral acids having a pKa of less than about 3. In some embodiments, the weakly coordinating anion is a trifluoroacetate anion or a trifluoromethanesulfonate anion.
In some embodiments, the method comprises providing a MOF comprising one or more bridging ligands comprising an electron withdrawing group, one or more bridging ligands comprising a positive charge, or a combination thereof. In some embodiments, the electron withdrawing group is selected from halo, carbonyl, sulfonyl, cyano, nitro, and haloalkyl (e.g., perhaloalkyl). In some embodiments, the electron withdrawing group is fluoro or trifluoromethyl. In some embodiments, the method comprises providing a MOF comprising metal oxygen cluster SBUs linked together via organic bridging ligands, wherein each of the SBUs comprises one or more first metal ions (e.g., hf) and one or more anions coordinated to the one or more first metal ions, and wherein the organic bridging ligands comprise at least one organic bridging ligand comprising a coordinated non-SBU linked second metal ion (e.g., ir, ru or Pt), wherein the second metal ions are further coordinated to one or more electron withdrawing ligands, optionally wherein the electron withdrawing ligands are halo-and/or perhaloalkyl-substituted bipyridine or phenylpyridine ligands. In some embodiments, the electron withdrawing ligand is a halogenated and/or perhalogenated alkyl substituted bipyridine ligand. In some embodiments, providing the MOF comprises providing a MOF comprising a bis (4-benzoic acid) -2,2' -bipyridine (DBB) bridged ligand, wherein the DBB bridged ligand coordinates to a first metal ion and a second metal ion of two different metal oxide clusters SBU, and wherein the second metal cation is further coordinated to two halo-and/or perhaloalkyl-substituted pyridine ligands, each of which is 2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine in some embodiments. In some embodiments, the second metal ion is Ir. In some embodiments, the MOF comprises one or more SBUs comprising metal ions that absorb ionizing radiation. In some embodiments, the ionizing radiation is X-rays. In some embodiments, the metal ion is an ion of an element selected from Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi. In some embodiments, the metal ion is not a Bi ion or a W ion. In some embodiments, the metal ion is a Hf ion.
In some embodiments, the MOF has enhanced interaction and/or binding capacity for one or more therapeutic agents of interest as compared to the MOF without surface modification. In some embodiments, the one or more therapeutic agents of interest are selected from nucleic acids, small molecules, and/or macromolecules comprising surface accessible phosphate or carboxylate groups. In some embodiments, the protein is selected from the group comprising an anti-CD 37 antibody, an anti-CD 44 antibody, an anti-CD 47 antibody, an anti-CD 73 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-LAG 3 antibody, and an anti-CTLA-4 antibody. In some embodiments, the nucleic acid is selected from the group consisting of miRNA, mRNA, siRNA, cpG ODN, and a cyclic dinucleotide. In some embodiments, the cyclic dinucleotide is a STING agonist. In some embodiments, the STING agonist is c-di-AMP or cGAMP.
Examples
The following examples are included to provide guidance to those of ordinary skill in the art in practicing representative embodiments of the presently disclosed subject matter. In light of the present disclosure and the level of ordinary skill in the art, it will be appreciated by those skilled in the art that the following embodiments are intended to be exemplary only and that many variations, modifications, and alternatives may be employed without departing from the scope of the presently disclosed subject matter.
Example 1
Materials and methods of examples 1-8
Unless otherwise indicated, all chemicals were purchased from Sigma-Aldrich (st.louis, missouri, united States of America) and Fisher (Thermo Fisher Scientific, waltham, massachusetts, united States of America) and were used without further purification. Imiquimod (powder, 95%) was purchased from Cayman Chemical (Ann Arbor, michigan, united States of America). InVivoMAb anti-mouse CD47 polyclonal antibodies (. Alpha.CD47, MIAP 301) were purchased from Bio X Cell (Lebanon, new Hampshire, united States of America). By using the product under the trade name Avance II TM A commercially available 500MHz NMR spectrometer (Bruker, billerica, massachusetts, united States of America) uses a default Bruker QNP probe 19 F{ 1 H } (Bruker, billerica, massachusetts, united States) and referenced in DMSO-D 6 Incompletely deuterated 1 H resonance and Nuclear Magnetic Resonance (NMR) spectra were collected. Transmission Electron Microscopy (TEM) using a TECNAI Spirit TEM (FEI Company, hillsboro, oregon, united States of America). Ultraviolet-visible (UV-vis) absorption spectra were collected using a UV-2600UV-vis spectrophotometer (Shimadzu, kyoto, japan). Particle size was collected by Dynamic Light Scattering (DLS) and zeta potential was measured by electrophoresis using Nano-Series Zeta Sizer (Malvern Panalytic, westborough, massachusetts, united States). Using Cu K α Radiation sourcePowder X-ray diffraction (PXRD) data was collected on a D8 vector diffractometer (Bruker, billerica, massachusetts, united States) and processed using PowderX software. Inductively coupled plasma-mass spectrometry (ICP-MS) data were obtained using 7700x ICP-MS (Agilent Technologies, santa Clara, california, united States of America) and analyzed using ICP-MS mass Hunter B01.03 edition. Dilution of sample with 2% HNO 3 In a matrix and use 115 The In internal standard was analyzed with respect to a 10 point standard curve In the range of 0.1ppb to 500 ppb. The correlation for all the analyses of interest was greater than 0.999. Data acquisition was performed in spectroscopic mode, while five replicates were performed per sample, each replicate being scanned 100 times. LC-MS data was obtained with 1290UHPLC (C 18 Reverse) was collected on an Agilent 6540Q-Tof MS-MS spectrometer (Agilent Technologies, santa Clara, california, united States of America). Mobile phase 35% MeOH/65% H 2 O, the linear range of Imiquimod (IMD) calibration curve was 0-200ng/mL using an injection volume of 10. Mu.L. Thermogravimetric analysis (TGA) was performed in air using a platinum-disc-equipped Shimadzu TGA-50 (Shimadzu, kyoto, japan) and heating at a rate of 1 ℃/min. Flow cytometer data were collected on LSR Fortessa 4-15 (BD Biosciences, san Jose, california, united States) and analyzed by FlowJo software (Tree Star, ashland, oregon, united States). Confocal laser scanning microscopy images were collected on an Olympus FV1000 and analyzed using imageJ software (NIH, bethesda, maryland, united States of America). The concentration of αCD47 is determined by the trade name NANODROP TM 8000(Thermo Fisher Scientific,Waltham, massachusetts, united States) or under the trade name PIERCE TM BCA Protein Assay Kit (Thermo Fisher Scientific, waltham, massachusetts, united States of America). ELISPot analysis test under the trade name ELISPot READY-SET-GO-! TM (eBioscience, san Diego, california, united States of America) commercially available mouse IFN-gamma assay.
Mouse colon adenocarcinoma cells CT26 were obtained from American type culture Collection (American Type Culture Collection) (ATCC, manassas, virginia, united States of America) and contained 5% CO at 37 ℃C 2 In a moist environment, the cells were cultured in RPMI 1640 medium (GE Healthcare, chicago, illinois, united States of America) containing 10% fetal bovine serum, 100U/mL penicillin G sodium and 100. Mu.G/mL streptomycin sulfate. BALB/c mice (6-8 weeks) were obtained from Harlan Envigo Laboratories, inc (Indianapolis, indiana, united States of America).
In vitro assays were performed using a RT250 positive voltage X-ray model (Philips, andover, massachusetts, united States of America) with a 250kVp, 15mA fixed setup and a built-in 1mm Cu filter. In vivo studies were performed using an X-RAD 225 image guided bioirradiator (Precision X-ray inc., north Branford, connecticut, united States of America). The instrument was set to 225kVp and 13mA and a 0.3mm flat copper filter was installed before the 25mm collimator.
Example 2
Synthesis and characterization of Hf-DBP and TFA modified Hf-DBP
Hf-DBP surface modification with TFA: synthesis of 5, 15-bis (p-benzoic acid) porphyrin (H) according to previous reports (Lu et al, J.am.chem.Soc.,2014, 136 (48), 16712-16715) 2 DBP) and Hf-DBP. The suspension of Hf-DBP in EtOH was washed sequentially with acetonitrile and benzene by sonication and centrifugation to reduce the polarity of the solvent. Using N for Hf-DBP in benzene 2 Deaeration and transfer to a glove box for surface modification reactions. Into a 1-dram vial with a stirring bar, 1mL of a suspension of Hf-DBP in benzene (2 mM) was addedTrimethylsilyl trifluoroacetate (TFA-TMS) 10 times and sealing the vial. The reaction mixture was stirred for 12 hours to obtain TFA-modified Hf-DBP. Outside the glove box, CH is used in sequence 3 The suspension was washed with CN and EtOH and stored in EtOH for further use.
Digestion and NMR analysis of TFA modified Hf-DBP: about 1.0mg of Hf-DBP or TFA modified Hf-DBP was dried under vacuum and resuspended in 500mL DMSO-D 6 And 50mL D 3 PO 4 In the mixture. The mixture was sonicated for 15 minutes and an additional 50mL D was added 2 O. The resulting mixture is sonicated to provide for 1 H and 19 homogeneous solution for F NMR analysis. Digestion of Hf-DBP 1 H NMR(500MHz,DMSO-D 6 Ppm): δ=10.65 (s, 2H), 9.67 (d, 4H), 9.03 (d, 4H), 8.40 (m, 8H). Digested Hf-DBP 1 H NMR(500MHz,DMSO-D 6 Ppm): δ=10.65 (s, 2H), 9.67 (d, 4H), 9.03 (d, 4H), 8.40 (m, 8H). Digested TFA modified Hf-DBP 1 H NMR(500MHz,DMSO-D 6 Ppm): δ=10.65 (s, 2H), 9.67 (d, 4H), 9.03 (d, 4H), 8.40 (m, 8H) (DBP aryl H), δ=1.89(s) (HOAc alkyl H). Digestion of TFA-modified Hf-DBP 19 F NMR(471MHz,DMSO-D 6 Ppm): delta= -74.25(s). In Hf-DBP, the surface modifier is HOAc, in 1 Strong signal δ=1.89 (s, 1.2H) is given in the H NMR spectrum. In Hf-DBP TFA, δ=1.89 (s, 0.11H) showed a significantly reduced signal, indicating that the surface conditioner OAc is mostly @>90%) was exchanged to TFA. Delta = -74.25(s) 19 F NMR also confirmed the presence of TFA on the MOF surface.
Example 3
Synthesis and characterization of IMD@Hf-DBP
Synthesis of IMD@Hf-DBP: in a 20mL glass vial, 10mg IMD and 5mL of 2mM TFA modified Hf-DBP in EtOH were added. The mixture was sonicated at 40-50℃for 1 hour. The reaction was stirred at room temperature for a further 12 hours, then excess imiquimod was removed by centrifugation and ultrasonic washing with DMSO: etoh=1:1 and EtOH in sequence. IMD@Hf-DBP supernatant was checked by UV-Vis detection to confirm the absence of free imiquimod.
Digestion of IMD@Hf-DBP and UV-Vis analysis: to determine the components and estimate the load percentage of the IMD, 5mL of 2mM IMD@Hf-DBP was added to 900mL of DMSO and 100mL of H 3 PO 4 Is a kind of medium. The mixture was sonicated for 2 hours and then analyzed by UV-Vis spectroscopy.
Digestion and Nuclear magnetic resonance analysis of IMD@Hf-DBP: about 1.0mg of IMD@Hf-DBP was dried under vacuum and resuspended in 500. Mu.L of DMSO-D 6 And 50mL D 3 PO 4 Is a kind of medium. The mixture was sonicated for 15 minutes and an additional 50mL D was added 2 O. The mixture was vortexed to provide a homogeneous solution. Sample passage 1 H and 19 f NMR was used for analysis. 1 H NMR(500MHz,DMSO-D 6 Ppm): δ=10.65 (s, 2H), 9.67 (d, 4H), 9.03 (d, 4H), 8.40 (m, 8H) (DBP aryl H), δ=1.89(s) (HOAc alkyl H), δ=8.41 (s, 1H), 8.14 (d, 1H). 19 F NMR(471MHz,DMSO-D 6 Ppm): delta= -74.25(s) (TFA F). The weight percent of loaded IMD was 8.8% calculated by integration of δ=0.90.
TGA analysis of imd@hf-DBP: about 2mg of IMD@Hf-DBP was dried under vacuum and used for TGA analysis. The theoretical weight loss was 62.0% and the experimental weight loss was 66.2%. The percentage load calculated by TGA was 11.1%.
Example 4
Synthesis and characterization of IMD@Hf-DBP/αCD47
Synthesis of IMD@Hf-DBP/αCD47: IMD@Hf-DBP was resuspended in 1mL PBS at an equivalent Hf concentration of 2mM in a 1.5mL Eppendorf tube. 750mg of CD47 (8.8 mg/mL,85.2mL in PBS) was added to the tube and vortexed for 15 seconds. αcd47 loading was verified by measuring nanodrops of supernatant IgG concentration after centrifugation.
Particle size and zeta potential of Hf-DBP, TFA modified Hf-DBP, IMD@Hf-DBP and IMD@Hf-DBP/alpha CD47 are determined by the reaction of the polymer with purified water (MILI-Water, millipore Sigma, burlington, massachusetts, united States of America). The results are summarized in table 1 below.
TABLE 1 particle size, PDI and zeta potential
Particles Number average size PDI Zeta potential
Hf-DBP 130.8±7.1nm 0.099 -23.3±1.0mV
TFA modified Hf-DBP 134.2±12.2nm 0.250 -19.5±0.7mV
IMD@Hf-DBP 161.2±14.3nm 0.131 -16.1±0.7mV
IMD@Hf-DBP/αCD47 212.9±4.2nm 0.163 -4.7±0.5mV
IMD@Hf-DBP/αCD47 release profile:
quantification method of αcd 47: imd@hf-DBP/oc CD47 was resuspended in 27mL PBS at an equivalent Hf concentration of 0.5mM and aliquoted into 27 Eppendorf tubes (n=3 for each time point). Each tube was transferred to an incubator at 37℃and measurements were made at 0 hours, 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 24 hours, 36 hours and 48 hours. At each time point, three Eppendorf tubes were removed and spun at 12000 rpm. The supernatants were then collected for LC-MS analysis of imiquimod and BCA assay of αcd 47.
Method for quantifying IMD: standard curves for imiquimod were prepared by dissolving pure imiquimod in PBS (1 ppm stock and gradient dilutions) and ranged linearly from 10ppb to 500ppb. The gradient elution of LC-MS was set as: 1) 0-3 min, 100% H 2 O; 2) 3-8 min, 65% H 2 O and 35% MeOH; 3) 8-10 minutes 100% methanol. The flow rate was 0.5mL/min, the injection volume was 10mL, and the sample was diluted 10-fold for LC-MS analysis.
Release profile of IgG-FITC in serum-containing PBS: IMD@Hf-DBP/IgG-FITC was prepared as in IMD@Hf-DBP/. Alpha.CD47 and resuspended in 27mL PBS containing 10% FBS at 0.1mM equivalent Hf concentration and aliquoted into 27Eppendorf tubes (N=3 at each time point). Each tube was transferred to an incubator at 37℃and measurements were made at 0 hours, 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 24 hours, 36 hours and 48 hours. At each time point, three Eppendorf tubes were removed and spun at 12000 rpm. The supernatant was then collected and subjected to fluorescence detection by a microplate reader. The percent release was calculated based on the relative fluorescence fitted into the IgG-FITC standard curve. IgG is released relatively rapidly within the first about 10 hours and then begins to stabilize. The percent release after 12 hours was about 50%, and after 48 hours was about 60%
Example 5
In vitro studies of IMD@Hf-DBP and IMD@Hf-DBP/αCD47
Cellular uptake of IMD@Hf-DBP and IMD@Hf-DBP/αCD47: cellular uptake of IMD@Hf-DBP and IMD@Hf-DBP/αCD47 in RPMI medium at 2.5X10 5 The density of/mL was evaluated on CT26 cells in 6-well plates. IMD@Hf-DBP and IMD@Hf-DBP/. Alpha.CD47 were added to each well in medium at 20. Mu.M equivalent Hf concentration and the well plate was shaken at 150rpm for 1min. The cells were then returned to the 37℃incubator and incubated for 1, 2, 4 and 8 hours. At each time point, the medium was removed, the cells were washed three times with 2mL DPBS, trypsinized, collected by centrifugation at 3000rpm and counted with a hemocytometer. Using 1mL of 99% concentrated HNO in a 1.5mL ep tube 3 (67% -70% trace metal grade) and 1% HF digest the cell particles for 48 hours, with strong vortexing and sonication every 12 hours. Then, the Hf concentration was determined by ICP-MS.
Cytotoxicity of IMD@Hf-DBP and IMD@Hf-DBP/αCD47: dark toxicity of IMD@Hf-DBP and IMD@Hf-DBP/. Alpha.CD47 was evaluated against CT26 cells or HEK293T cells using the 3- (4, 5-dimethylthiazol-2-yl) -5- (3-carboxymethoxyphenyl) -2- (4-sulfo-phenyl) -2H-tetrazolium (MTS) assay (Promega Corporation, madison, wisconsin, united States of America,1/10 diluted in DMEM). First, each cell was seeded on a 96-well plate at a density of 15000 cells/mL using 100 μl RPMI/DEM medium/well and further cultured overnight. IMD@Hf-DBP and IMD@Hf-DBP/. Alpha.CD47 were added to each well at equivalent Hf concentrations of 0, 0.5, 1, 2, 5, 10, 20, 50, 100. Mu.M and incubated for a further 72 hours before cell viability was determined by MTS assay. In the absence of X-ray exposure, IMD@Hf-DBP/αCD47 was slightly toxic to CT26 colon cancer cells (see FIG. 12), but not HEK-293T cells.
Macrophage activation: BALb/c-bone marrow derived monocytes were harvested, cultured and activated. For classical activated M1 macrophages, bone marrow cells were cultured for 48 hours with fresh Dulbecco Modified Eagle Medium (DMEM) medium supplemented with 20% v/v fetal bovine serum, 100ng/mL lipopolysaccharide and 25ng/mL IFN-gamma. Adherent cells were harvested for the following study. For M2 macrophages, 100ng/mL mouse granulocyte-macrophage colony stimulating granulocyte factor and 20ng/mL IL-4 were added. Cell at 37℃、5% CO 2 And (5) culturing. The culture medium is replaced every 2-3 days, and the cells are cultured for 6-8 days.
Macrophage repolarization: CT26 cells were cultured overnight in 6-well plates and incubated with PBS, IMD, hf-DBP or IMD@Hf-DBP for 4 hours at an equivalent dose of 20. Mu.M, followed by irradiation with X-rays at a dose of 0 or 2 Gy. Differentiated M2 macrophages were added and co-cultured with treated CT26 cells at 37℃for 12 hours. Cells were then collected, washed twice with cold PBS, blocked with anti-CD 16/32, while reducing non-specific binding to FcR, stained with CD86 (GL 1), CD206 (C068C 2), and analyzed by flow cytometry.
Calreticulin translocation: the immunogenic cell death induced by RT-RDT treatment was studied by detecting exposure to cell surface Calreticulin (CRT). CT26 cells were cultured overnight in 6-well plates and incubated with PBS, aCD 47, hf-DBP or Hf-DBP/aCD 47 for 4 hours at an equivalent dose of 20 μM, followed by X-ray irradiation at a dose of 0 or 2 Gy. Each cell was then cultured in an incubator for another 4 hours to induce CRT translocation, then fixed with AlexaFluor 488-CRT antibody (Enzo Life Science, farm dale, new York, united States of America) and DAPI and stained on a coverslip for confocal imaging or flow cytometry analysis.
Phagocytosis: will be 5X 10 5 CFSE-labeled CT26 cells were cultured overnight in 6-well plates and incubated with PBS, αCD47, hf-DBP or Hf-DBP- αCD47 for 4 hours at an equivalent dose of 20 μM, followed by X-ray irradiation at a dose of 0 or 2 Gy. 1.5X10 are added 6 PerCP-Cy5.5-labeled macrophages and co-cultured with treated CT26 cells at 37℃for 4 hours. The cells were then collected, washed twice with cold PBS, imaged by CLSM or analyzed by flow cytometry.
Example 6
In vivo studies of IMD@Hf-DBP and IMD@Hf-DBP/αCD47
Performance profiling of innate immunity: tumors were harvested, treated with 1mg/mL collagenase I (Gibco Laboratories, gaithersburg, maryland, united States of America) in a 37℃water bath for 1 hour, andthe rubber end of the syringe was used for grinding. Cells were filtered through a nylon mesh filter of 70 μm in size and washed with PBS. The single cell suspension was incubated with anti-CD 16/32 (clone 93;eBiosciences,San Diego,California,United States of America) to reduce non-specific binding to FcR. Cells were further stained with the following fluorochrome conjugated antibodies: CD45 (30-F11), CD3e (145-2C 11), CD11b (M1/70), F4/80 (BM 8), CD86 (GL 1), CD206 (C068C 2), MHC-II (AF 6-120) and yellow fluorescence (all from eBioscience, san Diego, california, united States of America). Under the trade name lsrforterssa TM Flow cytometers (BD Biosciences, san Jose, california, united States) are sold for cell collection and data analysis is performed using FlowJo software (Tree Star, ashland, oregon, united States).
Immunofluorescence assay test: the tumor sections were air dried for at least 1 hour and then fixed in acetone at 20 ℃ for 10 minutes. After blocking with 20% donkey serum, each section was incubated overnight at 4 ℃ with each primary antibody against CD47 (MIAP 301, bioleged, san Diego, california, united States) or F4/80 (BM 8), CD86 (GL 1), CD206 (C068C 2), all from eBiosciences (San Diego, california, united States of America), followed by incubation with dye conjugated secondary antibodies at room temperature for 1 hour. After 10 minutes of re-staining with DAPI, the sections were washed twice with PBS and observed under CLSM.
Antitumor effect: to evaluate Hf-DBP regulated macrophage therapy, 2X 10 on day 0 6 The individual CT26 cells were injected into the right subcutaneous tissue of Balb/c mice as a single tumor CT26 model. When the tumor volume reaches 100-150mm 3 In the case of Hf-DBP, IMD@Hf-DBP, hf-DBP/αCD47 or IMD@Hf-DBP/αCD47, intratumoral injection was performed at an equivalent Hf dose of 0.1. Mu. Mol or equivalent amount of IMD or αCD47. Mice that were not treated at all served as controls. Mice were anesthetized with 2.5% (v/v) isoflurane 12 hours after injection and tumors were irradiated with 2Gy doses of X-rays for 2 consecutive days. Tumor size was measured daily with calipers and tumor volume was measured as (wide 2 X length)/2. The body weight of each group was monitored daily. Mice were sacrificed on day 25 and resected tumors were photographed and weighed. Swelling of the heartTumor sections were stained with hematoxylin-eosin (H)&E) And immunofluorescence analysis.
Example 7
IMD@Hf-DBP and IMD@Hf-DBP/alpha-CD 47 combined checkpoint blocking immunotherapy
Abscopal effect: to evaluate nMOF-mediated macrophage therapy in combination with checkpoint blocking immunotherapy, one was performed by administering 2X 10 on day 0 6 And 1X 10 6 The individual cells were injected into the right and left subcutaneous tissues of Balb/c mice, respectively, to simulate primary and distal tumors, creating a bilateral syngeneic CT26 model. When the primary tumor volume reaches 100-150mm 3 At this point, 0.2. Mu. Mol equivalent dose of IMD/. Alpha.CD47, hf-DBP/. Alpha.CD47, IMD@Hf-DBP or IMD@Hf-DBP/. Alpha.CD47 or PBS was injected intratumorally. Mice were anesthetized with 2% (v/v) isoflurane 12 hours after injection, and primary tumors were irradiated once with 2Gy of X-rays for 2 consecutive days. anti-PD-L1 antibody was intraperitoneally injected at a dose of 75 μg/dose every three days for a total of 2 injections. Untreated mice served as controls. Mice were sacrificed on day 25. Tumors and major organs were harvested for immunoassay.
ELISPpot test: by ELISPot assay (mouse IFN-. Gamma.ELISPot READY-SET-GO! TM The method comprises the steps of carrying out a first treatment on the surface of the Cat.No. 88-7384-88; eBioscience, san Diego, california, united States of America) in vivo. Millipore Multiscreen HTS-IP plates (Millipore Sigma, burlington, massachusetts, united States of America) were coated overnight at 4℃with anti-mouse IFN-gamma capture antibody. Single cell suspensions of splenocytes were obtained from CT26 tumor bearing mice treated with PBS (-), PBS (+), alpha PD-L1 (+), IMD@Hf-DBP/alpha CD47 (-) +aPD-L1, IMD@Hf-DBP/alpha CD47 (-) +alpha PD-L1, and at 2X 10 5 Individual cells/wells were seeded onto antibody coated plates. Stimulation (10 mg/mL; purity) with or without SPSYVYHQF (SEQ ID NO: 4)>95%; PEPTIDE 2.0,Chantilly,Virginia,United States of America) was cultured at 37 ℃ for 42 hours, and then the suspension was discarded. Plates were then incubated with biotin-conjugated anti-IFN-gamma detection antibody for 2 hours at room temperature and then used at room temperatureAvidin (Avidin) -HRP was incubated for 2 hours. 3-amino-9-ethylcarbazole substrate solution (Sigma, st. Louis, missouri, united States of America, cat. AEC 101) was added for cytokine spot detection.
Performance analysis of adaptive immunity: tumors were harvested, treated with 1mg/mL collagenase I (Gibco Laboratories, gaithersburg, maryland, united States of America) for 1 hour in a 37℃water bath, and ground with the rubber end of a syringe. Cells were filtered through a 70 μm nylon mesh filter and washed with PBS. The single cell suspension was incubated with anti-CD 16/32 (clone 93;eBiosciences,San Diego,California,United States of America) to reduce non-specific binding to FcR. The cells were further stained with the following fluorochrome-conjugated antibodies: CD45 (30-F11), CD3e (145-2C 11), CD4 (GK 1.5), CD8 (53-6.7), B220 (RA 3-6B 2), nkp (29A1.4) and yellow fluorescent staining solutions (all from eBioscience, san Diego, california, united States of America). By using the products under the trade name LSRFORTESSA TM Cell collection was performed using a commercially available flow cytometer (BD Biosciences, san Jose, california, united States) and data analysis was performed using FlowJo software (Tree Star, ashland, oregon, united States).
Example 8
Discussion of examples 1-7
As a major defense of the immune system, macrophages engulf the neoplastic cancer cells before they maintain host homeostasis (alarena et al, 2008). However, the ability of macrophages to target cancer cells is limited by the overexpression of the "don't eat me" CD47 checkpoint signaling molecule on the tumor cell surface that evades immune surveillance (Jaiswal et al, 2009) and promotes immunosuppressive Tumor Microenvironment (TME), whereas the host is anti-inflammatory (pro-tumor) M2 macrophages (Chao et al, 2010). Immunostimulatory therapies have been explored to remodel TME and reverse immunosuppression with the goal of activating an anti-tumor immune response (Lou et al,2019;Chen et al,2015;Louttit et al,2019; and Nam et al, 2019).
One such treatment is IMB, a hydrophobic small molecule drug, which can repolarize innate immunity by activating toll-like receptor-7 (TLR-7) pathways (Rodell et al,2018; and O' Neill et al, 2013). TLR-7 agonists such as IMD can repolarize M2 macrophages to pro-inflammatory (anti-tumor) M1 macrophages, which aid in phagocytosis, inflammation, and antigen presentation. Furthermore, macrophage checkpoint inhibition was proposed as an anti-tumor therapy by promoting phagocytosis of dying tumor cells and releasing tumor antigens (Chen et al,2019a; and Liu et al, 2015). In fact, blocking the CD-47 signaling pathway with anti-CD 47 antibodies (αCD 47) is under clinical investigation (Chen et al,2019b;Kojima et al,2016; and Feng et al, 2019). However, systemic toxic IMD and aCD 47 therapies are limited by inadequate anti-tumor efficacy and adverse side effects even by intratumoral injection (Liu et al,2015;Kamath et al,2018; and Xiong et al, 2011).
The presently disclosed subject matter describes using nMOF to deliver IMD and αCD47 to tumor cells to enhance the radiotherapy-radiodynamic therapy (RT-RDT) effects of nMOF and low dose X-ray irradiation. In particular, nMOFs with heavy metal secondary building blocks (SBUs) and photoactive ligands can mediate RT-RDT by enhancing X-ray energy deposition and generating a variety of Reactive Oxygen Species (ROS).
IMD@Hf-DBP/αCD47 was synthesized by sequential Hf-DBP-nMOF surface modification, IMD loading and αCD47 adsorption. IMD@Hf-DBP/αCD47 plus X-ray radiation causes a strong RT-RDT effect, resulting in ICD of tumor cells and enhances the immunomodulatory effects of TLR-7 agonists and CD-47 blockers. See fig. 1. Further combination with anti-PD-L1 immune Checkpoint Blockade (CBI) completely rooted out primary and distant tumors on a bilateral colorectal tumor model.
The synthesis of Hf-DBP nMOF containing acetate (OAc) end-capping groups as previously described (Lu et al, 2014) shows empirical formula Hf 123 -O) 83 -OH) 82 -OH) 6 (AcO) 3.5 (DBP) 6.8 -(OH) 0.9 (OH 2 ) 0.9 . Hf-DBP nano-plate by stacking Hf in hcp-like mode 12 SBU and DBP ligand. Hf on surface 12 SBU is blocked by OAc groups (see FIG. 2A), while it isH 2 The zeta potential in O is-23.3.+ -. 1.0mV, which prevents the loading of αCD47 on the surface of the Hf-DBP nano-plate. See fig. 2B. Hf-DBP was treated with trimethylsilyl trifluoroacetate (TMS-TFA) by 1 H and 19 f NMR spectroscopy, TFA group substituted>90% of OAc groups, while TFA-modified Hf-DBP is provided. TEM imaging and PXRD studies show that the morphology and crystallinity of the surface-modified Hf-DBP remain unchanged. See fig. 3A, 3B, 4A, 4B and 5. The weakly coordinating TFA groups may be substituted with carboxylate groups in the protein or phosphate groups on the nucleic acid. Hf-DBP-TFA showed almost complete adsorption of αCD47 (97.2%) by BCA assay. See fig. 2B.
IMD@Hf-DBP was obtained by ultrasonic treatment in ethanol with the loading of IMD into the pores of TFA-modified Hf-DBP (see FIG. 6), by 1 H NMR and UV-vis spectra and thermogravimetric analysis determined that IMD loading was-9 wt%. αCD47 was added to the PBS suspension of IMD@Hf-DBP under vortexing to provide IMD@Hf-DBP/αCD47 with an αCD47 loading of 7.5wt%. Based on TEM images and PXRD images, the IMD@Hf-DBP and the IMD@Hf-DBP/αCD47 maintain the morphology and crystallinity of the Hf-DBP. See fig. 7A, 7B, 8A, 8B and 9. See also table 1 above.
IMD at 37℃in PBS and αCD47 release profiles were determined using liquid chromatography-mass spectrometry (LC-MS) and bicinchoninic acid (BCA) assays, respectively. See fig. 2C. About 8% of the IMD is released slowly over 48 hours, which is desirable to maintain a high local IMD concentration while continuing repolarization of M2 to M1 macrophages. About 30% and 60% of the αcd47 was released in PBS or 10% (v/v) FBS-containing PBS, respectively, within 48 hours, and substitution was readily achieved by phosphate complexation to Hf12 SBU. See fig. 10.IMD@Hf-DBP/αCD47 showed high cellular uptake. See fig. 11.
Studies were conducted to determine if released IMD and αcd47 could target macrophages and tumor cells, respectively, to exert an in vitro antitumor effect. First, bone marrow-derived monocytes are harvested and differentiated into M1 or M2 macrophages. From F4/80 + Cells were gated and M1 or M2 cells displayed CD86, respectively + CD206 - Or CD86 - CD206 + Phenotype. See fig. 13. A mouseColorectal cancer CT26 cells were treated with IMD@Hf-DBP, hf-DBP or IMD followed by 2Gy X-ray irradiation (hereinafter +X-ray irradiation). M2 macrophages were then co-cultured with treated CT26 cells for 24 hours and immunostained for flow cytometry analysis. As shown in FIG. 14A, CT26 cells treated with IMD@Hf-DBP (+) induced stronger macrophage repolarization with an M1/M2 ratio of 382 compared to the other groups (0.01-1.15). Interestingly, hf-DBP (+) induced a higher M1 population than IMD (+) (see FIG. 14C), reflecting the intrinsic nature of RT-RDT to repolarize macrophages from M2 to M1 phenotype.
CT26 cells were then treated with Hf-DBP/αCD47, hf-DBP or αCD47 to assess phagocytosis. In the Hf-DBP (+) and Hf-DBP/. Alpha.CD47 (+) groups, calreticulin was demonstrated to translocate to the cell surface as a "eat me" signal. M1 macrophages were co-cultured with treated CT26 cells and a significant increase in phagocytosis of CFSE-labeled CT26 cells was observed in the Hf-DBP/αCD47 (+) group by PerCP-Cy5.5 conjugated F4/80-labeled M1 macrophages, indicating that exposure of the "eat me" signal (calreticulin) and blocking of the "do not eat me" signal (CD 47) on the tumor cell surface promoted enhanced phagocytosis. See fig. 14B and 14D. Similar results were observed under CLSM, where the Hf-DBP-. Alpha.CD47 (+) group showed that most CT26 cells were phagocytosed by PerCP-Cy5.5-labeled M1 macrophages.
IMD@Hf-DBP/αCD47 plus X-rays were tested to determine if TME could be remodeled by modulating anti-tumor immune activation of macrophages. PBS, IMD/αCD47, hf-DBP/αCD47 or IMD@Hf-DBP/αCD47 was intratumorally injected into CT26 tumor-bearing BALB/c followed by 2 Gy/X-ray exposures for 2 consecutive days. At 48 hours after the first irradiation, tumor section slides were immunostained to detect CD47 blockage. Although IMD/αCD47 (+) slightly reduced the CD47 fluorescence signal compared to the PBS control, hf-DBP/αCD47 (+) and IMD@Hf-DBP/αCD47 (+) strongly blocked CD47, indicating a higher blocking effect of Hf-DBP-delivered αCD47. Interestingly, hf-DBP (+) treatment also reduced CD47. Tumor-infiltrating innate immune cells were analyzed 48 hours after the first irradiation. See fig. 16.IMD@Hf-DBP/αCD47 (+) treatment statistically increased tumor-infiltrating leukocytes, dendritic cells and macrophages (see FIGS. 15A and 17A-17C), indicating enhanced infiltration of antigen presenting cells of inflammatory TME. In addition, IMD@Hf-DBP/αCD47 (+) treatment significantly increased M1 macrophages and decreased M2 macrophages, suggesting in vivo macrophage repolarization by synergistic nMOF-mediated RT-RDT, TLR-7 agonists and CD-47 blockade. See fig. 15A and 17D-7F. Interestingly, a significant increase in the M2 phenotype was observed in the PBS (+) group, indicating that low doses of X-rays alone induced a more immunosuppressive TME. See fig. 17E. In addition, a significant increase in the total number of macrophages (see FIG. 17C) and M1 phenotype (see FIG. 17D) was found in the Hf-DBP/αCD47 (+) or IMD@Hf-DBP (+) groups, respectively, indicating that nMOF delivery would enhance macrophage modulating effects of IMD and αCD47. Macrophage repolarization was confirmed by CLSM imaging of immunostained tumor section slides. Furthermore, M1 macrophages significantly increased MHC-II expression in both the IMD@Hf-DBP (+) and IMD@Hf-DBP/αCD47 (+) groups (see FIG. 15B), indicating enhanced function in antigen presentation.
The anti-tumor efficacy of IMD@Hf-DBP/αCD47 against the CT26 tumor model was evaluated. When the tumor reaches 150mm 3 At this time, the IMD/αCD47, IMD@Hf-DBP, hf-DBP/αCD47 or IMD@Hf-DBP/α0CD47 was injected intratumorally and then irradiated with 2 Gy/fraction of X-rays for 2 days continuously. These treatments showed no systemic toxicity, as evidenced by stable body weight. IMD@Hf-DBP (+) and Hf-DBP/. Alpha.1CD47 (+) showed strong tumor inhibition, with Tumor Growth Inhibition (TGI) indices of 83.3% and 89.3%, respectively, on day 25 compared to PBS (-) control. IMD@Hf-DBP/αCD47 (+) effectively eradicated the tumor with a TGI of 98.2% and a cure rate of 50%. See fig. 15C. See also table 2 below. Hf-DBP (+) and IMD@Hf-DBP/. Alpha.CD47 (-) showed moderate efficacy with TGI values of 52.8% and 33.8%, respectively, but IMD/. Alpha.CD47 (+) showed minimal effect with a TGI of 2.1%. These results support that nMOF delivered IMDs and αCD47 would enhance antitumor efficacy. On day 25, the average weight of resected tumors for the PBS (-), PBS (+), IMD/αCD47 (+), IMD@Hf-DBP/αCD47 (-), hf-DBP (+), IMD@Hf-DBP (+), hf-DBP/αCD47 (+), and IMD@Hf Hf-DBP/αCD47 (+), groups were 2.34+ -0.73, 2.44+ -0.86, 2.18+ -0.60, 1.38+ -0.15, 0.87+ -0.26, 0.58+ -0.21, 0.41+ -0, respectively. 08 and 0.12 + -0.15 g. See fig. 18 and 19.H&E staining showed severe necrosis of IMD@Hf-DBP/αCD47 (+) treated tumor sections, whereby IMD@Hf-DBP/αCD47 (+) treatment repolarizes macrophages, remodelled the intratumoral immunity, providing excellent antitumor effect.
Table 2. Student T-test analysis and p-value of tumor volume of single CT26 tumor bearing mice after treatment endpoint.
Synergy between macrophage therapy and CBI was investigated. imd@hf-DBP/αcd47 (+) or each control was injected into primary tumors of a bilateral CT26 tumor model, followed by two intraperitoneal injections of αpdl1 and two X-ray exposures at 2 Gy/fraction. As shown in fig. 20A, αpdl1 (+) treatment did not show significant differences from the PBS (+/-) control group. IMD@Hf-DBP/αCD47 (+) treatment effectively inhibited the primary tumor, but had no effect on the distant tumor. In contrast, IMD@Hf-DBP/αCD47 (+) +αPDL1 treatment completely eradicates the primary and distant tumors. IMD@Hf-DBP/αCD47 (+) or IMD@Hf-DBP/αCD47 (-) +aPDL1 only moderately controls tumor growth. See fig. 20B. See also table 3 below. Hf-DBP mediated synergism of RT-RDT, IMD reversal of macrophage-related immunosuppression and macrophage checkpoint blockade of αcd47 thus constitute an immunocaloric bed of potent CBI, leading to excellent distal effect (abscopal effect).
Table 3 student T-test analysis and p-value of tumor volumes of bilateral CT26 tumor-bearing mice after treatment endpoint (= < 0.0001).
Anti-tumor immunity induced by IMD@Hf-DBP/αCD47 (+) +αPDL1 was probed. First, an ELISpot test was performed to determine specific anti-tumor immunity by detecting IFN- γ producing cytotoxic T cells in spleen cells 12 days after the first irradiation. Cytotoxic T cell epitopes expressed by CT26 tumor cellsUpon stimulation with AH1 (SPSYVYHQF) (SEQ ID NO: 4), cytotoxic T cells constitute counted IFN-gamma spots. 8.0.+ -. 9.3/10 compared to PBS (-) 6 The number of cells, IMD@Hf-DBP/. Alpha.CD47 (+) (61.6.+ -. 28.6/10) 6 Individual cells) and IMD@Hf-DBP/αCD47 (+) +αPD-L1 (213.8.+ -. 126.9/10) 6 Individual cells) a significant increase in antigen-specific IFN- γ producing T cells was observed in the group, indicating that strong tumor-specific adaptive immunity was induced. See fig. 20C. Flow cytometry analysis (see FIG. 21) showed tumor infiltrating leukocytes (see FIG. 22A), CD8+ T cells (see FIG. 20D) and CD4 in primary and distal tumors of the IMD@Hf-DBP/αCD47 (+) +αPD-L1 group + T cells (see fig. 20E) were significantly increased. Interestingly, natural killer cells (NK, see fig. 20F) and B cells (see fig. 22B) were also significantly increased in both primary and distant tumors. These results demonstrate that imd@hf-DBP/αcd447 (+) mediated local macrophage therapy and systemic CBI enhance infiltration of effector T cells, B cells and NK cells, activating anti-tumor immunity and producing strong distal effects.
In summary, the presently disclosed subject matter provides a new strategy to modify the surface of nMOFs for biomacromolecule delivery. The activation of the immunomodulation and innate immunity by macrophages with the loading of IMD in the pores and αcd47 on the surface of the modified Hf-DBP results in excellent antitumor effects. Hf-DBP mediated RT-RDT and slow released IMD repolarize immunosuppressive M2 macrophages to immunostimulatory M1 macrophages, while simultaneously released αCD47 blocks the "do-it-yourself" signal on tumor cells to enhance phagocytosis. When combined with αpd-L1, imd@hf-DBP/αcd47 (+) mediated macrophage therapy was extended to systematically eradicate the tumor on a bilateral CT26 tumor model. Thus, macrophage-mediated "trinity" therapy appears to be a strategy to modulate the synergistic combination of TME and immunotherapy.
Example 9
Materials and methods of examples 10-16
Cell lines and animals: mouse colorectal cancer cell line MC38, lewis lung cancer cell line LL2 and melanoma cell line B16F10 were purchased from American dictionaryType culture collection (American Type Culture Collection) (Rockville, maryland, united States of America). The mouse pancreatic cancer cell line Panc02 was provided by Hans Schreiber doctor friendly, university of Chicago pathology (Department of Pathology, university of Chicago) (Chicago, illinois, united States of America). MC 38-egg cell line [ OVA (257-264) -ZGREEN ]Produced by transfection of MC38 cells with LZRS-based retrovirus. All cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) medium (GE Healthcare, chicago, illinois, united States of America) supplemented with 10% FBS, 100U/mL penicillin G sodium and 100. Mu.G/mL streptomycin sulfate. The cells contained 5% CO at 37 ℃C 2 Is cultured in a moist environment. Mycoplasma is passed under the trade name MYCALERT before use TM A commercial kit (Lonza Walkersville, inc., walkersville, maryland, united States of America) was used for detection, and C57BL/6 mice (6-8 weeks) were obtained from Harlan Envigo Laboratories, inc. (Indianapolis, indiana, united States of America).
H 2 DBB-Ir-F and H 2 Synthesis of DBB-Ir-Ir (DBB): the synthesis of [ dF (CF) as shown in scheme 1 is reported below in accordance with the literature (Zhu et al, 2018) 3 )ppy] 2 + [H 2 DBB F -Ir, dbb=4, 4 '-bis (4-benzoic acid) -2,2' -bipyridine; dF (CF) 3 ) ppy=2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine]。 1 H NMR(500MHz,DMSO-d 6 ): delta 9.08 (d, 2H), 8.76 (d, 2H), 8.49 (d, 2H), 8.44 (d, 2H), 8.15 (s, 2H), 8.02 (d, 4H), 7.82 (s, 2H), 7.62 (d, 4H), 7.12 (t, 2H) and 5.91 (d, 2H). Ir (H) 2 DBB)(ppy) 2 + (H 2 DBB-Ir, dbb=4, 4 '-bis (4-benzoic acid) -2,2' -bipyridine; ppy=2-phenylpyridine) the synthesis was performed as reported in literature (Lan et al,2019 b) as shown in scheme 1 below. 1 H NMR(500MHz,DMSO-d 6 ):δ9.05(d,2H),8.67(d,2H),8.28(d,2H),8.07(s,2H),7.96(m,8H),7.89(d,2H),7.52(d,4H),7.17(t,2H),7.09(t,2H),6.98(t,2H),6.33(d,2H)。
Scheme 1.DBB F Synthetic routes to Ir and DBB-Ir
Hf-DBB F -synthesis of Ir and Hf-DBB-Ir: into a 4mL glass vial was added 0.5mL HfCl 4 Solution (2.0 mg/mL in DMF), 0.5mL H 2 DBB F Ir solution (4.0 mg/mL in DMF), 2.6. Mu.L TFA and 2. Mu.L water. The reaction mixture was kept in an oven at 70 ℃ for 24 hours. The yellow precipitate was collected by centrifugation and washed with DMF and ethanol. Hf yield, determined based on ICP-MS, was 61%. Into a 4mL glass vial was added 0.5mL HfCl 4 Solution (1.6 mg/mL in DMF), 0.5mL H 2 DBB-Ir solution (6.4 mg/mL in DMF) and 100. Mu.L of AcOH. The reaction mixture was kept in an oven at 70 ℃ for 72 hours. The orange precipitate was collected by centrifugation and washed with DMF and ethanol. The yield of Hf, as determined on the basis of ICP-MS, was 52%.
Hf-DBB F Digestion of Ir and Hf-DBB-Ir: drying under vacuum 1.0mg of Hf-DBB F -Ir. The resulting solid was purified in 500. Mu.L DMSO-d 6 And 50 mu L D 3 PO 4 Is digested and sonicated for 10 minutes. The mixture was then added to 50 mu L D 2 In O and pass through 1 H NMR analysis. Digested Hf-DBB F Ir shows a reaction with H 2 DBB F All signals corresponding to Ir without any other aromatic signals, which confirms Hf-DBB F The DBB is only present in Ir F -Ir ligands. 1.0mg of Hf-DBB-Ir was dried under vacuum. The resulting solid was purified in 500. Mu.L DMSO-d 6 And 50 mu L D 3 PO 4 Is digested and sonicated for 10 minutes. The mixture was then added to 50 mu L D 2 In O and pass through 1 H NMR analysis. The digested Hf-DBB-Ir showed a reaction with H 2 All signals corresponding to DBB-Ir, without any other aromatic signals, confirm that only DBB-Ir ligands are present in Hf-DBB-Ir.
APF test · OH production: aminophenyl luciferins (APF, thermo Fisher Scientific, waltham, massachusetts, united States of America) and · the reaction of OH is carried out,while generating bright green fluorescence (excitation/emission maximum 490/515 nm). H was performed in the presence of 5. Mu.M APF 2 DBB F -Ir、H 2 DBB-Ir、Hf-DBB F Ir and Hf-DBB-Ir are suspended in water at an equivalent concentration of 20. Mu.M. As a blank, 5 μm APF aqueous solution was used. mu.L of each suspension was added to a 96-well plate and then irradiated with 0, 1, 2, 3, 5 or 10Gy of X-rays (RT 250X-ray generator (Andof Philips, mass.) 250kVp,15mA,1mm Cu filter). Fluorescent signals were immediately collected using an IVIS200 imaging system (Xenogen, hopkitton, massachusetts, united States).
Production of 1O2 monomeric oxygen sensor Green (SOSG, thermo Fisher Scientific, waltham, massachusetts, united States of America) Using SOSG assay 1 O 2 The reaction produced bright green fluorescence (excitation/emission maximum 504/525 nm). H was performed in the presence of 12.5. Mu.M SOSG 2 DBB F -Ir、H 2 DBB-Ir、Hf-DBB F Ir and Hf-DBB-Ir are suspended in water at an equivalent concentration of 20. Mu.M. An aqueous solution of 12.5. Mu.M SOSG was used as a blank. mu.L of each suspension was added to a 96-well plate and then irradiated with 0, 1, 2, 3, 5 or 10Gy of X-rays (RT 250X-ray generator (Philips, andover, massachusetts, united States of America), 250KVp,15mA,1mm Cu filter). Fluorescent signals were immediately collected using an IVIS200 imaging system (Xenogen, hopkitton, massachusetts, united States).
O for BMPO test 2 - Generating: 5-tert-Butoxycarbonyl-5-methyl-1-pyrroline N-oxide (BMPO) is a nitrone spin-trap (spin trap) which can be used with O 2 - (BNPO-O 2 - ) Forming recognizable adducts with long half-lives (t 1/2 =23 min). H was performed in the presence of 25mM BMPO 2 DBB F -Ir、H 2 DBB-Ir、Hf-DBB F Ir and Hf-DBB-Ir are suspended in benzene at an equivalent concentration of 200. Mu.M. A25 mM BMPO in benzene was used as a blank. 1mL of each suspension was added to a 4mL vial, and then irradiated with 5Gy X-rays (RT 250X-ray generator (Philips, andover, massachusetts, united States of America),250kVp,15mA,1mm Cu filter). The Electron Paramagnetic Resonance (EPR) signal was immediately acquired by X-Band ELEXSYS-II 500EPR (Bruker, billerica, massachusetts, united States).
DNA double strand breaks: by detecting phosphorylated gamma-H 2 AX detects DNA double strand breaks. MC38 cells were plated in 6-well plates at 5X 10 5 Well incubated overnight with PBS, H 2 DBB-Ir、H 2 DBB F -Ir, hf-DBB-Ir or Hf-DBB F Ir was grown at an equivalent concentration of 20. Mu.M and then irradiated with 0 and 2Gy (RT 250X-ray generator (Philips, andover, massachusetts, united States of America), 250kVp, 15mA, 1mm Cu filter). Cells were stained with hcsdn injury kit (Life Technologies, carlsbad, california, united States of America) 2 hours after irradiation and flow cytometry analysis was performed with 1:500 dilutions.
Clonogen assay MC38 cells were cultured overnight in 6-well plates and incubated with the particles at 20. Mu.M Hf concentration for 4 hours, then irradiated with 0, 1, 2, 4, 8 and 16Gy (RT 250X-ray generator (Philips, andover, massachusetts, united States of America), 250KVP, 15mA, 1mm Cu filter). The irradiated cells were immediately trypsinized and counted. 200-2000 cells were seeded in 6-well plates and cultured with 2mL of medium for 14 days to form visible colonies, which were counted to determine viability. Once colony formation is observed, the medium is discarded. The plate was washed twice with PBS and then with 500. Mu.L of 0.5% w/v crystal violet in 50% methanol/H 2 O staining. The wells were rinsed three times with water and colonies were counted manually. Radiation Enhancement Factor (REF) at 10% survival dose 10 ) The ratio of the equivalent radiation dose required to obtain 10% survival relative to the experimental group was calculated for the PBS control group.
Cytotoxicity test: 3- (4, 5-dimethylthiazol-2-yl) -5- (3-carboxymethoxy-phenyl) -2- (4-sulfophenyl) -2H-tetrazolium (MTS) assay (Promega, madison, wisconsin, united States of America) was used to evaluate cytotoxicity of X-ray irradiation. MC38 cells at 1X 10 4 The wells were seeded on 96-well plates and further incubated for 12 hours. 0, 1, 2, 5,10. Equivalent ligand doses of 20, 50 and 100. Mu.M were added to PBS, H 2 DBB-Ir、H 2 DBB F -Ir, hf-DBB-Ir or Hf-DBB F Ir and incubated for 4 hours. The cells were then irradiated with X-rays at a dose of 2Gy (RT 250X-ray generator (Philips, andover, massachusetts, united States), 250KVp,15mA,1mm Cu filter). The cells were further cultured for 72 hours and then cell viability was determined by the MTS assay.
Live cell/dead cell analysis: the live/dead cell assay was evaluated with the cell permeation dye calcein AM and Propidium Iodide (PI) kit. MC38 cells were plated in 6-well plates at 5X 10 5 Culture overnight in wells and irradiation with PBS, H at an equivalent concentration of 20. Mu.M by 0 or 2Gy (RT 250X-ray generator (Philips, andover, massachusetts, united States of America), 250kVp, 15mA, 1mm Cu filter) 2 DBB-Ir、H 2 DBB F -Ir, hf-DBB-Ir or Hf-DBB F Ir is incubated for 4 hours. Cells were then gently washed with PBS and the trade name Fluoview was used TM Commercial confocal microscopy of FV1000 (Olympus, tokyo, japan) observed live cells stained with calcein AM (green) and dead cells stained with PI (red) under confocal microscopy.
Apoptosis/necrosis: the cell death assay was evaluated using an apoptotic cell death kit. MC38 cells were plated in 6-well plates at 5X 10 5 Culture in wells overnight and use PBS, H 2 DBB-Ir、H 2 DBB F -Ir, hf-DBB-Ir or Hf-DBB F Ir was incubated at an equivalent concentration of 20. Mu.M for 4 hours and then irradiated with 0 or 2Gy (RT 250X-ray generator (Philips, andover, massachusetts, united States of America), 250kVp, 15mA, 1mm Cu filter). After 24 hours, cells were stained according to AlexaFluor 488Annexin V/dead apoptosis kit (Life Technologies, carlsbad, california, united States of America), imaged under CLSM and used under the trade name LSRFORTESSA TM 4-15 (BD Biosciences, san Jose, california, united States of America) were quantified by a commercially available flow cytometer.
Immunogenic cell death: immunogenic cell death was examined by Calcium Reticulin (CRT) exposure. M is MC38 cells were plated in 6 well plates at 5X 10 5 Culture in wells overnight and use PBS, H 2 DBB-Ir、H 2 DBB F -Ir, hf-DBB-Ir or Hf-DBB F Ir was grown at an equivalent concentration of 20. Mu.M and then irradiated with 0 and 2Gy (RT 250X-ray generator (Philips, andover, massachusetts, united States of America), 250kVp, 15mA, 1mm Cu filter). Cells were then gently washed with PBS and stained with AlexaFluor 488-CRT antibody (Enzo Life Sciences, farmingdale, new York, united States of America) in a 1:100 dilution for flow cytometry analysis.
Phagocytosis: c57BL/C bone marrow derived monocytes are harvested, cultured and activated. Mouse granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-4 were supplied at 1% final concentration for 168 hours and non-adherent cells were harvested as immature DCs for the following study. Cells were incubated at 37℃with 5% CO 2 And (5) culturing. The medium was changed every 2-3 days and each cell was used after 6-8 days of culture. Will be 5X 10 5 MC38 cells of each CFSE-tag (Life Technologies, carlsbad, california, united States of America) were cultured overnight in 6-well plates and dosed with H at an equivalent dose of 20. Mu.M 2 DBB-Ir、H 2 DBB F -Ir, hf-DBB-Ir and Hf-DBB F Ir was incubated for 4 hours, followed by irradiation with 0 or 2Gy (RT 250X-ray generator (Philips, andover, massachusetts, united States of America), 250kVp, 15mA, 1mm Cu filter). Adding 1×10 6 The PE-Cy5.5 labeled DCs were co-cultured with treated MC38 cells at 37℃for 4 hours. Cells were then collected, washed twice with cold PBS, imaged by CLSM or analyzed by flow cytometry.
Immunofluorescent staining: tumors and lymph nodes were collected and then frozen. Tissue sections with a thickness of 5 μm were prepared using a CM1950 cryostat (Leica Camera, wetzler, germany). The sections were air dried for at least 1 hour and then fixed in acetone at 20 ℃ for 20 minutes. After blocking with 20% donkey serum, these sections were incubated overnight at 4℃with respective primary antibodies of CD11b (53-6.7), F4/80 (H57-597), CD11b (53-6.7), MHC-II (53-6.7), CD86 (53-67), CD206 (53-6.7) and CD 8. Alpha. (53-6.7), followed by incubation with dye-conjugated secondary antibodies at room temperature for 1 hour. After 10 minutes of staining with DAPI, each section was then washed twice with PBS and observed with an SP8 light confocal microscope (Leica Camera, wetzler, germany).
In situ inoculation of homologous gene model: collaborative tumor models MC38, MC 38-egg cells and Panc02 were constructed to evaluate the in vivo anticancer effects of nMOF-mediated in situ vaccination. For a single tumor model, 5×10 will be 5 Individual MC38 cells, 1X 10 6 Individual MC 38-egg cells or 1X 10 6 Each Panc02 cell was inoculated subcutaneously to the right of the C57BL/6 mice. When the tumor volume reaches 100-150mm 3 At this time, mice were intratumorally injected with nMOF at a 0.2. Mu. Mol Hf dose, cpG or PBS at a 1. Mu.g dose. 12 hours after injection, mice were anesthetized with 2% (v/v) isoflurane and tumors were irradiated with 1Gy of X-rays/fraction (225kVp,13mA,0.3mm Cu filter) for a total of 5 fractions daily. For a bilateral tumor model, 5×10 will be 5 The MC38 cells were inoculated subcutaneously on the right as primary tumor, whereas 2X 10 cells were inoculated 5 Individual MC38 cells, 2X 10 5 Individual B16F10 cells or 5×10 cells 5 The individual LL2 cells were inoculated subcutaneously on the left as distal tumors of C57BL/6 mice. Alpha PD-L1 (clone number: 10F.9G2, catalog number: BE0101, bioXcell, lebanon, new Hampshire, united States of America) was provided at a dose of 75 μg/dose per three days of intraperitoneal injection. Tumor size was measured daily with calipers, where tumor volume was equal to (width 2 X length)/2.
ELISpot test. By an ELISPot test (mouse IFN-. Gamma.test under the trade name ELISPot READY-SET-GO! TM Are commercially available; accession numbers 88-7384-88; eBioscience, san Diego, california, united States of America) in vivo. Millipore Multiscreen HTS-IP plates (Millipore Sigma, burlington, massachusetts, united States) were coated overnight at 4℃with anti-mouse IFN-gamma capture antibody. Single cell suspensions of spleen cells were obtained from MC38 tumor bearing mice and were used at a rate of 2X 10 5 The individual cell/well concentrations were seeded onto antibody coated plates. Cells were incubated with the peptide sequence (KSPWFTTL) (SEQ ID NO: 5) or without the peptide sequence at 37℃for 42 hours and then discarded. The well plate was then conjugated with biotin against IFNThe gamma detection antibody was incubated at room temperature for 2 hours, followed by incubation with avidin-HRP for 2 hours at room temperature. 3-amino-9-ethylcarbazole substrate solution (Sigma, st. Louis, missouri, united States of America, cat. AEC 101) was added for cytokine spot detection. The spots were commercially available under the trade name Immundospot TM Imaging and quantification was performed by the analyzer (Cellular Technology Ltd, shaker Heights, ohio, united States of America).
Lymphocyte performance analysis. Tumors and lymph nodes were harvested and treated with 1mg/mL collagenase I (Gibco Laboratories, gaithersburg, maryland, united States of America) for 1 hour at 37 ℃. Cells were filtered through a nylon mesh filter of size 40 μm and washed with PBS. Tumor draining lymph nodes were collected and directly grinded by cell filters. The single cell suspension was incubated with anti-CD 16/32 (clone 93) to reduce non-specific binding to FcR. Cells were further stained with the following fluorochrome conjugated antibodies: CD45 (30-F11), CD3 ε (145-2C 11), CD4 (GK 1.5), CD8α (53-6.7), nkp (29A1.4), F4/80 (BM 8), CD11b (M1/70), gr-1 (RB 6-8C 5), MHC-II (AF 6-120), CD80 (16-10A 1), CD86 (GL 1), CD206 (C068C 2), CD44 (IM 7), CD62L (MEL-14), H-2K b SIINFEKL (SEQ ID NO: 3) (25-D1.16), PI, and CD45 of yellow fluorescent reactive dyes (CD 206 and CD62L from BD Bioscience (San Jose, california, united States of America), CD206 and CD62L from bioleged (San Diego, california, united States of America), others from eBioscience (San Diego, california, united States of America)). Antibodies were used at a dilution of 1:200. Representative gating strategies for different immune cells are shown in supplementary figures 31-32. Under the trade name lsrforterssa TM 4-15 commercially available flow cytometry (BD Biosciences, san Jose, california, united States) was used for cell collection and data analysis was performed using FlowJo software (Tree Star, ashland, oregon, united States).
Secondary OT-I T cell transfer: will be 2X 10 6 Subcutaneous injection of MC 38-egg cells into C57BL/6Rag2 -/- Right side of the mice. After 14 days, mice were dosed with 0.2. Mu. Mol Hf-DBB F Intratumoral injection of Ir with or without 1 μg dose of CpG followed by 1Gy X-ray/partial irradiationThe injection is 5 parts per day. 2 days after the first irradiation, spleen and lymph nodes were isolated from OT-I mice and mouse CD8 was used + CD8 pair T cell isolation kit (Miltenyi Biotec, bergisch Gladbach, germany) + T lymphocytes were negatively classified, 1X 10 6 CD8 + Intravenous injection of T cells in Rag2 with MC 38-egg -/- Is a human subject. Tumor size was measured daily with calipers and tumor volume was equal to (width 2 X length)/2.
And (5) carrying out statistical analysis. The group size (n.gtoreq.5) was chosen as appropriate for statistical ANOVA analysis to ensure efficacy studies. Student's t-test was used to determine if the differences between groups were similar. Statistical analysis was performed using an originPro (originLab Corp., northampton, massachusetts, united States of America). Statistical significance was calculated using a two-tailed student t-test, defined as P <0.05, P <0.01, P <0.001. Animal experiments were not performed in blind mode and are expressed as mean ± standard deviation. Immunoassays were performed in blind mode and are expressed as median ± SD.
Example 10
F Synthesis and characterization of Hf-DBB-Ir and Hf-DBB-Ir-NMOF
According to one aspect of the presently disclosed subject matter, a novel strategy for personalized cancer vaccination using nanoscale metal-organic frameworks (nMOFs) through X-ray activated DAMP and tumor antigen generation and efficient delivery of CpG as PAMP to APCs is described. Cationic nMOF is designed by molecular engineering, whereas DAMP and tumor antigens are released by X-ray activated RT-RDT and CpG is delivered by electrostatic interactions. In situ vaccination provided by nMOF effectively expands cytotoxic T cells in tumor draining lymph nodes, while reactivating the adaptive immune system for tumor regression. See fig. 23. The local therapeutic effect of nMOF-based in situ vaccines was extended to distal tumors (αPD-L1) by combination therapy with anti-PD-L1 antibodies, while providing a cure rate of 83.3% on MC38 colorectal cancer model.
More specifically, to generate DAMP and tumor antigens via RT-RDT and deliver PAMPs with high CpG loading rates, provision is made forTwo positively charged nMOFs were counted: hf-DBB F -Ir and Hf-DBB-Ir, each with a high Z metal Hf 6 Two-level building block (SBU) and photosensitized DBB F -Ir and DBB-Ir ligands. See scheme 1 above and fig. 24A and 24B. Hf-DBB F -Ir and Hf-DBB-IrnMOF have the formula Hf 63 -O) 43 -OH) 4 L 6 UiO-like structure of (1), wherein l=dbb F -Ir or DBB-Ir. See fig. 25A. As shown by Transmission Electron Microscope (TEM) imaging (see FIGS. 26B and 27A-27D) and Dynamic Light Scattering (DLS) measurements, hf-DBB F Ir and Hf-DBB-Ir exhibit a spherical to octahedral morphology with diameters of 100 nm. See fig. 25B. Confirmation of Hf-DBB by UV-Vis absorption Spectrum and luminescence Spectrum F Photosensitive Properties of Ir and Hf-DBB-Ir, wherein Hf-DBB F Ir and Hf-DBB-Ir show the same properties as DBB, respectively F Light absorption and luminescence similar to those of Ir and DBB-Ir. See fig. 28A-28D.
Example 11
Reactive oxygen species, DAMP and in vitro immunogenicity
Detection of active oxygen species: without being bound by any one theory, it is believed that photosensitizing Hf-DBB F Ir and Hf-DBB-Ir can generate various Reactive Oxygen Species (ROS) under X-ray irradiation, including: by Hf 6 Hydroxyl radical generated by water radiation decomposition of SBU · OH) and singlet oxygen generated by excitation of a photoactive ligand 1 O 2 ) And superoxide anion (O) 2 - ). See fig. 26A. Quantification by APF and SOSG assays, hf-DBB compared to its ligand control F Ir plus X-ray irradiation [ denoted Hf-DBB ] F -Ir(+)]And Hf-DBB-Ir (+) both showed · OH and 1 O 2 a significant enhancement is generated. See fig. 26C and 26D. However, through BMPO test, only Hf-DBB F Ir (+) shows an effective O 2 - Generation due to DBB F -Ir has a higher reduction potential than DBB-Ir. See fig. 26E.
In vitro generation of active oxygen: evaluation of MC38 cell pairs Hf-DBB-Ir and Hf-DBB F Uptake of Ir.Inductively coupled plasma mass spectrometry (ICP-MS) studies showed that both nmos ofs reached similar intracellular Hf levels after 4 hours of culture. See fig. 29. Then, SOSG and superoxide test kit are used to detect the presence of the target in vitro 1 O 2 And O 2 - And (5) generating. Hf-DBB-Ir (+) and Hf-DBB F Ir (+) both induced intense green fluorescence, indicating significant 1 O 2 And (5) generating. However, only Hf-DBB F Ir (+) shows a strong red fluorescence, which indicates that it is passed through Hf-DBB F the-Ir mediated RT-RDT process produces O 2 - . See fig. 30A. To confirm the RT effect, OH-induced DNA Double Strand Breaks (DSBs) were quantified by flow cytometry analysis of phosphorylated γ -H2AX in cells treated with PBS, ligand or nMOFs with or without X-rays. Interestingly, 2 hours after irradiation, with Hf-DBB F A significantly higher red gamma-H2 AX fluorescence than Hf-DBB-Ir (+) was observed in the Ir (+) treated group, probably due to the O-by the superoxide dismutase 2 - Conversion to · OH. No fluorescence was observed in the group without irradiation with X-rays or without culture with nMOF.
Release and in vitro immunogenicity of DAMP: to verify Hf-DBB F The hypothesis that the large amount of ROS produced by Ir is more effective in damaging cancer cells than other treatments, hf-DBB was evaluated by clonogenic assays F Ir mediated cell damage and DAMP production, indicating Hf-DBB F the-Ir (+) is slightly better than the Hf-DBB-Ir (+), REF 10 The values were 1.75 and 1.68, respectively. See fig. 30B. MTS test further showed that Hf-DBB F -Ir (+) exhibits higher cytotoxicity than Hf-DBB-Ir (+) at 2Gy, IC 50 The values were 4.28.+ -. 1.15. Mu.M and 7.85.+ -. 2.41. Mu.M, respectively. See fig. 30C. Hf-DBB was also observed by live/dead cell imaging and apoptotic cell quantification by CLSM and flow cytometry F -higher levels of cell death by Ir (+). See fig. 30D. These results indicate that Hf-DBB F Ir (+) has a stronger cell killing effect through the RT-RDT process.
Then, the tumor cells and their apoptotic fragments are subjected to death by detecting Immunogenic Cell Death (ICD) and APC of the tumor cells Is studied for nMOF mediated DAMP production of RT-RDT. During ICD, calreticulin (CRT) is transferred to the cell membrane as a "eat me" signal recognized by macrophages and DCs to engulf dying tumor cells and their apoptotic fragments. Quantitative reveal of flow cytometry, hf-DBB F Ir (+) treated cells showed higher CRT fluorescence, indicating Hf-DBB F Ir (-) induces stronger ICD, with higher cytotoxicity. See fig. 31A.
To evaluate the effect of nMOF-mediated RT-RDT on antigen processing and immune activation of APCs, bone marrow cell differentiated DCs were compared to the use of PBS, DBB-Ir, DBB F Ir, hf-DBB-Ir or Hf-DBF-Ir and CFSE-labeled MC38 cells with or without X-ray irradiation treatment were co-cultured. Flow cytometry showed that Hf-DBB compared to the other treatment groups F The Ir (+) treatment induced a significant increase in the population of PE-cy5.5 conjugated CD11 c-labeled DCs with phagocytosed CFSE-labeled MC38 cells, indicating cationic nMOF-mediated immune stimulation enhancement. See fig. 31B. CLSM imaging confirmed more CD11c + Hf-DBB phagocytosed by DC F -Ir (+) treated cfse+mc38 cells.
In vitro delivery of PAMPs: without being bound by any one theory, it is reasonably deduced that Hf-DBB F DBB in Ir F Fluorination of the ligand may introduce an electron withdrawing effect, increasing the surface charge, thereby delivering CpG more efficiently. Hf-DBB F The zeta potential values exhibited by the-Ir and the Hf-DBB-Ir are 31.6+ -1.2 mV and 23.8+ -0.8 mV, respectively, confirming that the Hf-DBB F Ir has a stronger cationic backbone for the electrostatic attraction of anionic CpG. See fig. 32A. 1mg of CpG was added to Hf-DBB at a Hf concentration of 10mM F Incubation in 20mL of PBS solution of-Ir or Hf-DBB-Ir for 10 min. After centrifugation, DNA gel electrophoresis showed 82.7% adsorption of CpG to Hf-DBB F On Ir, 46.5% of CpG was adsorbed by Hf-DBB-Ir, and 8.6% and 43.8% of CpG remained in the corresponding supernatant as quantified by NanoDrop spectrophotometry. See fig. 32B. The DCs were then checked for CpG internalization. Flow cytometry and CLSM imaging showed that in their free CpG, hf-DBB-Ir@CpG or Hf-DBB with FITC label F After cultivation of-Ir@CpG, hf-DBB F The highest amount of CpG delivered to DC by Ir. See the figure32C. These results confirm that Hf-DBB F The superior ability of Ir to deliver CpG as PAMP to APC.
In vitro DC maturation: to evaluate the effect of CpG delivery on DC maturation, cpG, hf-DBB-Ir@CpG or Hf-DBB was used F -ir@cpg bone marrow derived DCs were incubated at CpG concentrations of 0, 62.5, 125, 250, 500 and 1000ng/mL for 60 hours. Cells were harvested and stained to detect DC maturation markers, including MHC-II and costimulatory molecules CD80 and CD86. Supernatants were also collected and analyzed for the presence of cytokines interferon alpha (IFN-alpha) and interleukin-6 (IL-6). Hf-DBB-Ir@CpG and Hf-DBB F Both Ir@CpG are effective in promoting DC maturation with increased MFI signal for CD80 (see FIG. 32D), CD86 (see FIG. 32E) and MHC-II compared to free anionic CpG. See fig. 32F. Hf-DBB F Ir@CpG is more potent than Hf-DBB-Ir@CpG in that it delivers CpG, and thus appears to be more excellent in terms of upregulation of CD80, CD86 and MHC-II signals. Only cationic nMOF-delivered CpG showed elevated IFN- α levels while free CpG had no effect at all. See fig. 32G. Furthermore, DCs treated with free CpG secreted IL-6 only at high CpG concentrations, whereas DCs treated with nMOFs/CpG also secreted IL-6 at low CpG concentrations. See fig. 32H. qPCR of IL-6 and IFN-alpha expression confirmed Hf-DBB F Ir more effectively delivers CpG as PAMP to activate DCs. See fig. 33A and 33B. To directly prove Hf-DBB F DC enhanced antigen presentation properties following stimulation with Ir@CpG with CpG, hf-DBB-Ir@CpG or Hf-DBB F Ir@CpG stimulated ovalbumin antigen for DC culture (OVA, cell line expressed as MC 38-egg cell) at a 3:1 ratio. By detection of H-2K on DC surfaces b Expression of SIINFEKL (SEQ ID NO: 3) complex (Kb-egg cells) detects uptake and presentation of tumor antigens. Hf-DBB F Ir@CpG is more excellent than Hf-DBB-Ir@CpG and free CpG in promoting antigen uptake and presentation of DC (see FIGS. 32I and 33C), probably due to the more efficient delivery of PAMP and antigen presentation.
Example 12
In situ cancer vaccine
X-ray induced carcinoma in situ vaccine: research of Hf-DBB F Ir@CpG (+) as in situ cancer vaccineIs a local anticancer effect. Two weeks total 4 doses of intravenous 2 μmol DBB F -Ir or Hf-DBB F Ir was not toxic to the C57BL/6 mice, as judged by stable body weight gain. Next, the injection was performed by injecting 5X 10 into the right side 5 MC38 cells A T cell depleted mouse colorectal cancer model MC38 was constructed on C57BL/6C mice. In previous studies, by inoculation with 2X 10 6 Individual MC38 cells established subcutaneous MC38 tumors and reached 100-150mm on day 7 before treatment began 3 Is of a size of (a) and (b). In contrast, MC38 tumors grew to 100-150mm within 14 days when seeded with fewer cells 3 And showed more immunosuppressive tumor microenvironment than the 7 day model. See fig. 34A. PBS, hf-DBB-Ir, hf-DBB F -Ir or Hf-DBB F Ir@CpG was injected intratumorally at a Hf dose of 0.2. Mu. Mol and/or at a CpG dose of 1. Mu.g. After 12 hours, the tumor was irradiated with 1Gy of X-rays (225 kVp,13mA,1 Gy) and then irradiated with 4 more X-rays (1 Gy) per day. Hf-DBB F the-Ir (+) is superior to the Hf-DBB-Ir (+) and the tumor growth inhibition index (TGI) is 81.9% and 64.7% respectively, indicating that Hf-DBB F Ir-mediated RT-RDT would release DAMP more effectively in vivo. Hf-DBB F Ir@CpG (+) showed a ratio of CpG (+) (99.6% and 34.8% TGI, respectively) or Hf-DBB on day 31 F -Ir (+) stronger tumor regression, indicating synergy of nMOF-mediated RT-RDT released DAMP and cationic nMOF delivered PAMP. See fig. 35A. See also table 4 below. The optical image on day 31 and the average weight of resected tumor confirm the anticancer effect. See fig. 34B and 34C. Terminal deoxynucleotidyl transferase dUTP notch end markers (TUNEL) and H&Immunofluorescence of E-staining showed that Hf-DBB was used F -ir@cpg (+) treatment, significant apoptosis of tumor cells, no systemic toxicity was observed for all treatment groups. Antitumor activity was evaluated on a mouse pancreatic cancer model Panc02 on C57BL/6C mice with high radio-resistance and low immunogenicity. Hf-DBB F Ir@CpG (+) provided superior tumor growth inhibition over the other groups (see FIGS. 34D, 34E, 35B and Table 4), indicating use of Hf-DBB F Ir@CpG (+) holds promise as a cancer vaccine in situ for a broad spectrum of cancers with different immunogenicity.
TABLE 4 tumor growth inhibition index (TGI) of MC38 and Panc02 tumor models with different treatments
Natural immunization after in situ cancer vaccination: plasma IL-6 and IFN-gamma concentrations were tested by ELISA and gene expression in tumors and tumor Draining Lymph Nodes (DLNs) was determined by qPCR 24 hours after treatment to evaluate the innate immune response. Hf-DBB F -Ir@CpG (+) treatment versus CpG (+) or Hf-DBB F Ir (+) treatment showed significant increases in plasma and intratumoral IL-6 and IFN- α levels. See fig. 35C. Furthermore, flow cytometry and CLSM studies showed that Hf-DBB F In the ir@cpg (+) treated group, tumor and DLN infiltrated APCs were significantly increased, including macrophages (see fig. 35D) and DCs (see fig. 35E), indicating the synergistic effect of nMOF-mediated RT-RDT and cationic nMOF delivered PAMP released DAMP and tumor antigen. From Hf-DBB F The accelerated DC maturation by Ir@CpG (+) is further demonstrated by increased expression of MHC-II and costimulatory CD80 molecules. See fig. 35F. Hf-DBB F The elevation of total IgG (see fig. 35G) and IgM (see fig. 35H) in plasma 2 and 12 days after ir@cpg (+) treatment is indicative of an effective promotion of B cell mediated humoral immunity. Since IgM can bind and activate the complement system, promoting opsonization and degradation of antigens and antigen presentation by phagocytes, an increase in plasma IgG and IgM levels suggests that B cells play an important role in promoting antigen presentation after in situ vaccination. Kb-ova complex (from CD45 + Cell-gated SIINFEKL (SEQ ID NO: 3) -H2Kb expression in Hf-DBB F The significant up-regulation of-Ir@CpG (+) after treatment on the MC38-ova model confirms the antigen presentation process. See fig. 35I. Hf-DBB F the-Ir@CpG (+) group also showed an increase in DLN (see FIG. 36), indicating T cell expansion in DLN. CLSM increased Ki67 expression in DLN corroborating Hf-DBB F T cell expansion in DLN after ir@cpg (+) treatment. Finally, in immunodeficiency Rag2 -/- Construction of MC 38-ovarian tumor in mice followed by Hf-DBB F -Ir (+) or Hf-DBB F Secondary transfer of-Ir@CpG (+) plus OT-I T cellsAnd (5) transferring to treat. By Hf-DBB F Mice treated with-Ir@CpG (+) plus OT-I T cell transfer showed a higher ratio than Hf-DBB alone F Ir (+) + plus OT-I T lymphocyte metastasis or Hf-DBB F More potent tumor suppression by Ir@CpG (+) (see FIG. 35J), which corroborates Hf-DBB as an in situ cancer vaccine F Effective antigen presentation process following ir@cpg (+) treatment. Interestingly, hf-DBB F Following ir@cpg (+) treatment, repolarization of macrophages was observed with increasing ratio of pro-inflammatory M1 subtype to anti-inflammatory (pro-tumor) M2 subtype.
Example 13
Far end effect
Then, MC38 double-sided model was constructed to evaluate Hf-DBB F -systemic anti-cancer efficacy of ir@cpg (+) in combination with anti-PD-L1 (αpd-L1) antibodies. Hf-DBB was used 14 days after tumor inoculation F Ir@CpG was intratumorally injected into the primary tumor at a dose of 0.2. Mu. Mol Hf and 1. Mu.g CpG, and X-ray irradiation was performed at a dose of 1 Gy/fraction every day from day 15 for a total of 5 fractions. Every three days, 75 μg of alpha PD-L1 was given by intraperitoneal injection for a total of 3 doses. Hf-DBB without alpha PD-L1 F Ir@cpg (+) almost exclusively of the primary tumor, but only moderately delayed progression of distant tumors. In sharp contrast, hf-DBB-Ir-F@CpG (+) and αPD-L1 significantly regressed primary and distant tumors with a cure rate of 83.3%. This result indicates that Hf-DBB F Strong synergy between ir@cpg (+) -based in situ cancer vaccination and CBI. See fig. 37A-37C.
Example 14
Adaptive immunity
The infiltrating leukocytes of the primary and distal tumors were analyzed 10 days after irradiation. Hf-DBB F the-ir@cpg+αpd-L1 (+) treated group showed a significant increase in tumor infiltration of cd45+ leukocytes (see fig. 37D), DCs (see fig. 37E), macrophages (see fig. 38A and 38B) and cd8+ T cells (see fig. 38F) in primary and distal tumors, which means an enhancement of the innate immune response following in situ vaccination. Specifically, under use of Hf-DBB F after-Ir@CpG (+) +αPD-L1 treatmentThe percentages of natural killer cells (NK cells, see fig. 37G), cd4+ T cells (see fig. 37H) and cd8+ T cells (see fig. 37I) increased significantly from 0.06±0.05% and 0.13±0.19%,0.05±0.04% and 0.03±0.02%, and 0.41±0.30% and 0.36±0.27% to 0.52±0.42% and 1.21±0.89%,0.25±0.23% and 1.15±1.14%, and 1.46±0.59% and 1.43±0.55%, respectively, in the total number of primary and distal tumor cells in the PBS (-) group. Both flow cytometry and CLSM showed effector T cell infiltration. DLN on both sides was collected, weighed and immunostained to detect T cell expansion, indicating Hf-DBB F Treatment with-Ir@CpG+αPD-L1 (+) promotes T cell expansion on bilateral DLNs. See fig. 38C and 38D. These results indicate that Hf-DBB F Not only does Ir@CpG (+) +αPD-L1 induce an innate immune response, but also enhances adaptive immunity of the treated local and untreated distal tumors.
Example 15
Induction and long-term immunization
Specificity of the induced immunity: the presence of tumor antigen specific cytotoxic T cells was determined using an IFN-gamma enzyme linked immunosorbent assay (ELISpot). Spleen cells were harvested in MC38 tumor-bearing mice 10 days after the first irradiation and stimulated with peptide sequence KSPWFTTL (SEQ ID NO: 5) for 42 hours. IFN-gamma spot forming cells were counted with an Immunospot (Immunospot) reader. In the case of using Hf-DBB F -Ir@CpG (+) and Hf-DBB F Antigen-specific IFN-gamma production of T cell number/10 in tumor-bearing mice treated with-Ir@CpG (+) +αPD-L1 6 The significant increase in splenocytes (60.2.+ -. 39.6 and 139.0.+ -. 52.4, respectively, and 16.4.+ -. 5.9 for PBS (-), see FIG. 39A) indicates Hf-DBB F -Ir@CpG (+) and Hf-DBB F both-Ir@CpG (+) +αPD-L1 are effective in generating tumor-specific T cell responses. To further investigate specific anti-tumor immunity, hf-DBB was used for MC38 primary tumor F -Ir@CpG (+) or Hf-DBB F Treatment with Ir@CpG (+) +αPD-L1 was performed to see if the treatment could regress unmatched syngeneic tumors of the distal flank. As shown in FIG. 39B, MC38 was used as the primary tumor treatment, while the syngeneic tumor cell lines B16F10 and LL2 were implanted simultaneously as far-end untreated A tumor to be treated. Hf-DBB F -Ir@CpG (+) and Hf-DBB F Both-ir@cpg (+) +αpd-L1 treatments were effective in regressing primary MC38 tumors, but had no effect on distant B16F10 or LL2 tumors. See fig. 39C-39F. These experiments show that, in the case of using Hf-DBB F After in situ vaccination of-Ir@CpG (+) +αPD-L1 treatment, the newly expanded T cells have tumor specificity and individuality.
Long-term antitumor immunity: the role of cytotoxic T cells in effective distal effects is further demonstrated by the following evidence: hf-DBB F Treatment of Rag2 in the absence of mature T cells and B cells with-Ir@CpG (+) +αPD-L1 -/- C57BL/6 mice MC38 bilateral subcutaneous models lack efficacy. By Hf-DBB F The primary tumor treated with-Ir@CpG (+) +αPD-L1 was initially inhibited (see FIG. 39G), but increased rapidly after the end of the X-ray irradiation. No distal effect was observed for distal tumors. See fig. 39H. This result demonstrates that both distal effects and local tumor regression/eradication require the presence of tumor-specific adaptive immunity. Finally, tumor re-challenge studies were performed to confirm long-term immune memory effects. For use with Hf-DBB F Mice cured after treatment with-Ir@CpG (+) +αPD-L1 will be 5X 10 5 The MC38 cells were seeded on the left side of the opposite side 30 days after tumor elimination, while these cured mice remained tumor-free after the first challenge, showing a powerful anti-tumor immune memory effect. 2 months after the first challenge, 2X 10 flank inoculation on the right side 6 B16F10 cells, while the cured mice constructed tumors similar to the original mice, suggesting tumor specificity of the immune memory effect. See fig. 39I. Memory effector cells (CD 3 epsilon) + CD8α + CD44 High height CD62L Low and low Phenotype) was also expressed in spleen cells after combination therapy. As shown in FIG. 39J, in Hf-DBB F A significant increase in memory effector cells was observed in the spleen after the end of the-ir@cpg (+) +αpd-L1 treatment.
Example 16
Discussion of examples 9-15
Advanced tumors avoid epidemic surveillance by inactivating, deregulating and hijacking the host immune system (Mahoney et al,2015;Dunn et al,2002). To address this problem, anti-PD- (L) 1CBI has become the standard of treatment for some cancers by targeting T cell inhibition checkpoint signaling pathways to provide durable anti-cancer efficacy with low side effects (Brahmer et al 2012; erico, 2015). However, immune checkpoint inhibition only elicits a persistent response in a few cancer patients, due to the reliance on the immunogenic tumor microenvironment, the so-called "hot" tumor. For patients with relatively "cold" tumors, e.g., low tumor mutation burden, low PD-L1 expression levels, and/or low abundance of pre-existing T cells, an immune-assisted therapy of "cold" tumor-to-heat "is actively examined in combination with checkpoint inhibitor therapy to overcome immune tolerance and enhance anti-tumor immunity of the host system.
In accordance with the presently disclosed subject matter, it is proposed that "cold" tumors can be effectively re-stimulated into an immunogenic hotbed by local therapy that produces innate immunity through exposure to tumor antigens. Furthermore, the two Pattern Recognition Receptor (PRR) pathways (Kawai and Akira,2010;Gong et al,2019), cGAS STING induced by DAMP after RT injury (Deng et al, 2014) and TLR pathways induced by PAMPs such as CpG (Weiner et al, 1997) were independently operated (Emming and Schroder, 2019), suggesting that they may be activated simultaneously to achieve additive or synergistic effects on immune stimulation. Porous nMOFs constructed from Hf-oxygen SBU and photoactive ligands can enhance the radiotherapeutic effects of ionizing radiation by enhancing X-ray energy deposition, facilitated ROS diffusion, and unique RT-RDT modes of action (Lan et al,2018;Ni et al,2019;Lu et al,2019). The presently disclosed subject matter provides new cationic Hf-based nMOF, hf-DBB for non-viral in situ vaccination by mediating efficient RT-RDT to generate immunogenic tumor antigens and DAMP and delivering anionic CpG as PAMP F -Ir. It is believed that the Hf-DBB of the present disclosure F Ir@cpg (+) would provide a first treatment in local tumors with synergistic DAMPs and PAMPs packaged in cancer-in-situ vaccines while simultaneously engaging lymphoid organs in antigen presentation, while synergistic CBI to induce CTL infiltration in distant tumors. Furthermore, by Hf-DBB F -Ir@CpG (+) +αPD-L1 was achieved on a relatively immunosuppressive 14 day model of MC38 colorectal cancerThe 83.3% cure rate indicates the potential use of an nMOF-based in situ vaccine for immunizing "cold" tumors.
Compared to traditional cancer vaccines, nMOF provides in situ cancer vaccines with several potential advantages. First, nMOF provides in situ vaccines that are personalized by the release of large amounts of ROS from the tumor, and can overcome the tumor heterogeneity problems faced by traditionally manufactured peptide vaccines. Second, cationic nMOF can capture DAMP and tumor antigens from dying cancer cells by electrostatic interactions (Min et al, 2017), and has a viroid size distribution that can be recognized and absorbed by APCs for efficient antigen presentation to stimulate a strong cytotoxic T cell response. Third, cationic nMOF delivers and protects anionic CpG from TLR stimulation and enzymatic degradation by downstream immune processes. Fourth, the tumor antigen released by the nMOF-mediated RT-RDT process and the DAMP act synergistically with the CpG-based PAMP delivered by the cationic nMOF to stimulate DC maturation to promote antigen presentation and adaptive immunity. Fifth, nMOF-based vaccines are activated by X-rays to release DAMP and tumor antigens having relatively non-toxic components, and thus are not expected to produce side effects. In addition, systemic administration of αpd-L1 would block the immunosuppressive co-suppression marker PD-L1 to enhance antigen presentation and reduce T cell depletion. nMOF-mediated in situ cancer vaccines in combination with CBI provide tumor-specific long-term anti-tumor immunity.
In summary, the presently disclosed subject matter provides a new nMOF by rational fluorination of photoactive ligands to efficiently generate ROS through RT-RDT and modulate nMOF surface charge to payload CpG. Intratumoral administration of Hf-DBB F In situ release of DAMP and use of Hf-DBB after Ir and X-ray irradiation F The tumor antigen delivered by Ir and CpG act synergistically to act as an effective personalized cancer vaccine to activate APCs and expand cytotoxic T cells in the tumor draining lymph nodes to reactivate the adaptive immune system for localized tumor regression. When combined with immune checkpoint inhibitor therapy, the innate and adaptive immunity of the nMOF-based cancer vaccine is further enhanced, resulting in excellent anti-tumor effects with tumor-specific long-term immune memory effects. This combination therapy via systemic anti-swelling by reactivating CTLTumor immunity extends the local therapeutic effects of carcinoma-in-place vaccines to distant tumors. This study paves the way for introducing the concept of an nMOF-based personalized vaccine into human trials for the treatment of advanced cancers.
Example 17
nMOF and peptide
Hf-DBP-Pt-nMOF having acetate (OAc) end-capping group was synthesized in a similar manner to Hf-DBP. Like Hf-DBP, hf-DBP-Pt is formed by linking Hf12-SBU with DBP-Pt-ligand in hcp-like stacking mode. Hf on surface 12 SBU is also blocked by OAc groups, at H 2 The zeta potential in O was-22.5.+ -. 0.5mV. Treatment of Hf-DBP-Pt with trimethylsilyl trifluoroacetate (TMS-TFA) to obtain TFA-modified Hf-DBP-Pt by substituting the OAc group with the TFA group, this was achieved by 1 H and 19 f NMR spectrum determination. TEM imaging and PXRD studies showed that the surface modified Hf-DBP-Pt maintained morphology and crystallinity. The weakly coordinating TFA groups may be substituted with carboxylate groups in the protein or phosphate groups on the nucleic acid.
MUC-1 peptide is a short peptide (d-CQCRRKN) (SEQ ID NO: 1) targeting membrane MUC-1 mucin (CQC motif) that can induce apoptosis and initiate host anticancer immune responses. Therapeutic peptides face significant challenges such as low cellular uptake and low stability in vivo. Membrane penetrating peptide GO-203 (RRRRRRRRRCQCRRKN) (SEQ ID NO: 2) was developed to target MUC-1 mucin but the clinical efficacy was not significant. By nMOF conjugation with MUC-1 peptide, it was found that Hf-DBP-Pt-TFA could not only deliver MUC-1 more effectively in vitro, but that MUC-1 peptide could also act synergistically with RT-RDT, exhibiting better anticancer effect on MC38 subcutaneous model with C57 BL/6.
MUC-1/Hf-DBP-Pt was prepared by mixing 1mM TFA-modified Hf-DBP-Pt with 2mM MUC-1 peptide in aqueous solution. The suspension was vortexed every 5 minutes for 15 minutes to obtain MUC-1/Hf-DBP-Pt. TEM imaging and PXRD studies showed that morphology and crystallinity remained after modulator exchange.
Cellular uptake: HEK293T cells at 2X 10 5 The density of individual cells/mL was inoculated on 6-well plates and culturedAnd (5) culturing overnight. 10. Mu.M TFA-modified Hf-DBP-Pt and 20. Mu.M FITC-MUC-1 were mixed in water, vortexed, and the mixture was allowed to stand for 15 minutes. Then 40. Mu.L of the mixture was added to 2mL of medium and FITC-MUC-1 or MUC-1/Hf-DBP-Pt (without fluorescent label) was added to the control wells at the same concentration. After 4 hours, cells were washed and fixed with 4% PFA and observed under confocal laser scanning microscopy. The green channel shows that FITC-MUC-1 is more effective in delivering through Hf-DBP-Pt. See fig. 40.
Cytotoxicity: MC38 cells were seeded into 96-well plates at a density of 1500 cells/well and cultured overnight. 1000. Mu.M TFA modified Hf-DBP-Pt and 2000. Mu.M MUC-1 were mixed in water, vortexed, and the mixture was allowed to stand for 15 minutes. The mixture was then added to each well at different concentrations. Three well plates were selected for 4Gy X-ray irradiation 4 hours after drug addition. MTS assay was performed 3 days later, while MUC-1/Hf-DBP-Pt showed a synergistic effect of MUC-1 and RT-RDT upon X-ray irradiation. See fig. 41.
Therapeutic effects in vivo: subcutaneously vaccinating 6-8 week old C57BL/6 mice with 2X 10 6 And MC38 cells. In the case of tumor reaching 100mm 3 In this case, MUC-1/Hf-DBP-Pt (0.4. Mu. Mol/0.2. Mu. Mol) was injected intratumorally. After 8 hours, the mice were anesthetized and subjected to a first 1Gy X-ray irradiation. Then, in the following consecutive 5 days, the mice were irradiated with 1Gy X-ray at a dose of 6Gy in total. Tumor volumes and body weights were measured daily. MUC-1/Hf-DBP-Pt showed synergistic effects of RT-RDT and MUC-1. See fig. 42. Stable body weight trends indicate minimal toxicity and good biocompatibility of the system.
Example 18
nMOF and CpG ODN
CpG adsorption of different nmofs: cpG ODN 2395 (3. Mu.g) and different nMOFs (Hf-DBP, hf-DBP-TFA or Hf-DBP-Pt-TFA; 0.1. Mu. Mol) were mixed in water to prepare CpG/nMOFs in separate 1.5mL ep tubes. The mixture was allowed to stand for 15 minutes and the mixture was centrifuged at 14500rpm for 15 minutes. The DNA concentration of the supernatant was determined by NanoDrop. The CpG loading of each nMOF was then calculated. Hf-TBP and Hf-TBP-Pt may adsorb 80% CpG, while Hf-DBP-TFA, hf-DBP-Pt and Hf-DBP-Pt-TFA may adsorb >90% CpG. See fig. 43. However, in this case, hf-DBP without TFA-modification is not able to adsorb CpG.
Example 19
nMOF and STING agonists
Preparation of cGAMP/Hf-DBP-Pt: 0.2. Mu. Mol of TFA-modified Hf-DBP-Pt and 5. Mu.g of 2',3' -cGAMP were mixed in 30. Mu.L of aqueous suspension, vortexed every 5 minutes for 15 minutes.
Therapeutic effect in vivo: subcutaneous inoculation of 6-8 week old C57BL/6 mice with 2X 10 6 And MC38 cells. When the tumor reaches 100mm at 7 th day 3 In the case of intratumoral injection of PBS, cGAMP or cGAMP/Hf-DBP-Pt (5. Mu.g/0.2. Mu. Mol). After 8 hours, the mice were anesthetized and irradiated with 2Gy of X-rays. The mice were further irradiated with 2Gy X-rays for 4 days in succession with a total dose of 10Gy X-rays. Tumor volumes and body weights were measured daily. As shown in FIG. 44, cGAMP/Hf-DBP-Pt showed synergistic effects of RT-RDT and STING agonists. The stable body weight trend shows that the system has the lowest toxicity and good biocompatibility.
Hf 12 -Ir nMOL preparation: MOL is a sub-class of MOFs having a single layer thickness. Hf 12-IrnMOL was synthesized as follows: 500 μl HfC l4 Solution [2.0mg/mL in N, N-Dimethylformamide (DMF)]、500μL H 2 DBB-Ir-F solution (4.0 mg/mL in DMF), 2. Mu.L trifluoroacetic acid (TFA) and 5. Mu.L water were added to a 1-dram vial. The mixture was sonicated and heated in an oven at 80 ℃ for 1 day. The yellow suspension was collected by centrifugation and washed with DMF and ethanol. Final product Hf 12 Ir nMOL dispersed in ethanol for characterization and further use.
Hf 12 IrnMOL contains Hf 12 Two-stage building blocks (SBU) and Ir (DBB) [ dF (CF) 3 )ppy] 2 + A photoactive ligand. PXRD pattern verifies Hf 12 Ir nmos is a crystalline material. TEM and AFM reveal Hf 12 The morphology of IrnMOL, which is a thickness<2nm and a diameter of about 100-200 nm.
Preparation of cGAMP/nMOL: to prepare cGAMP/nMOL, hf is first applied 12 IrnMOL was dispersed in 100. Mu.L of nuclease-free water at an equivalent Hf concentration of 2 mM. 1 μg of 2'3' -cGAMP was then added to the nMOL suspension. The mixture was vortexed three times every 5 minutes to obtain cGAMP/nMOL nanoconjugates. The concentration of Hf was detected by inductively coupled plasma-mass spectrometry (ICP-MS) using Agilent 7700x ICP-MS (Agilent Technologies, santa Clara, california, united States of America) and analyzed using ICP-MS MassHunter version B01.03 (Agilent Technologies, santa Clara, california, united States of America). With 1% HF acid solution in concentrated HNO 3 The samples were digested in (trace metal grade) for 2 days and then treated with final concentration of 2% HNO 3 And diluting the matrix. Using Cu ka radiation sourceThe crystallinity of both nanoparticles was checked by powder X-ray diffraction (PXRD) on a Bruker D8 vent diffractometer (Bruker, billerica, massachusetts, united States). The size and zeta potential were measured by Malvern Nano Series ZetaSizer (Malvern Panalytic, malvern, united Kingdom). The topography was observed by Transmission Electron Microscopy (TEM) on TECNAI Spirit TEM (FEI Company, hillsboro, oregon, united States of America) and Atomic Force Microscopy (AFM) on a Bruker V/Multimode 8 instrument (Bruker, billerica, united Kings of America). AFM, TEM imaging and PXRD studies showed that morphology and crystallinity remained after cGAMP conjugation.
cGAMP load efficiency and cGAMP/nMOL release profile: the concentration of 2'3' -cGAMP was quantified by LC-MS on an Agilent 6540Q-Tof MS-MS (5 μm Agilent C18 reverse phase column) with 1290UHPLC (Agilent Technologies, santa Clara, california, united States of America). A standard curve of 2'3' -cGAMP was prepared by dissolving the lyophilized 2'3' -cGAMP powder in nuclease free water to provide 1000ppm stock solution. A gradient dilution was prepared with a linearity in the range of 50ppb to 20 ppm. The elution of LC-MS was set as: 0-5min,95% H 2 O5% MeOH. The flow rate was 0.5mL/min and the injection volume was 20. Mu.L. To determine that 2',3' -cGAMP is found in Hf 12 Load efficiency on IrnMOL, GAMP/nMOL was newly prepared as described above, and collected by centrifugation at 14000gThe supernatant was collected. The concentration of 2',3' -cGAMP in the supernatant was quantified by LC-MS, n=3. For the release profile, volumes of 1 XPBS, 0.1 XPBS, and FBS (100. Mu.L/tube) were redispersed in 1.5mL Eppendorf tubes (3 replicates per time point), respectively. Eppendorf tubes were transferred to a 37℃heating block, and supernatants (80. Mu.L/tube) were collected at 0h, 1h, 2h, 4h, 8h, 12h, 24h, 36h, and 48h by centrifugation at 14000 g. Supernatants from the 1 XPBS and 0.1PBS groups were analyzed by LC-MS. The supernatant of the FBS group was mixed with 320. Mu.L of methanol/tube, sonicated for 1 minute to give a white precipitate, centrifuged again at 14000g, and the supernatant was analyzed by LC-MS. As shown in fig. 45A, TFA modified nMOL had an adsorption efficiency of nearly 100%, which verifies the phosphate group on cGAMP and Hf on nMOL 12 Conjugation between SBUs. The rapid release in 1 XPBS is due to phosphate exchange on the SBU. However, in 0.1×pbs and FBS, which mimic the actual phosphate concentration in biomedical applications, cGAMP release is significantly slower. See fig. 45B.
Isothermal titration calorimetry: measurement and analysis of 2',3' -cGAMP and Hf on a MicroCal ittc 200 system (Malvern Instruments, malvern, united Kingdom) equipped with reference and sample wells (v=40 μl) 12 Interactions between Ir nMOL. All titrations were performed using a 40 μl syringe at 298.15K with a stirring rate of 250 rpm. Titration of 75. Mu.M Hf with 235. Mu.M cGAMP in water 12 -an aqueous Ir nMOL solution. Data analysis was performed using MicroCal ittc 200 software (Malvern Instruments, malvern, uk) and all data were fitted to an independent single-site model. Moderate binding interactions (k=3.80×10 3 M -1 ) Dynamic binding between cGAMP and nMOL was verified. See fig. 46.
In vitro STING activation: under the trade name THP1-DUAL TM KO-MyD88 cells (InvivoGen, san Diego, california, united States of America) commercially available reporter cells were used for quantification of STING activation of free 2',3' -cGAMP and cGAMP/nMOL in vitro. The cell is 10 5 Individual cells/mL (n=6) were seeded in 96-well plates, added to up to 10 μm of 2'3' -cGAMP and cGAMP/nMOL and cultured for 24 hours. Stimulation of the Interferon Regulatory Factor (IRF) pathway by a commercial product Named QUANTI-LUC TM (InvivoGen, san Diego, california, united States of America) commercially available under the trade name SYNERGY TM HTX commercially available plate readers (BioTek, winioski, vermont, united States of America) were quantified according to the vendor protocol. As shown in FIG. 47, cGAMP/nMOL activates the EC of STING pathway in vitro 50 Much lower%<1/6), which suggests the potential of cGAMP/nMOL as a promising nano STING agonist.
In vivo imaging of cGAMP retention in tumors: a subcutaneous MC38 tumor-bearing C57BL/6 mouse model was constructed as described in example 17 above. When the tumor reaches 150mm 3 At this time, 20. Mu.L of cGAMP-Cy5/nMOL (2. Mu.g/0.5. Mu. Mol Hf) and cGAMP-Cy5 (2. Mu.g) were injected into the mice. Mice were anesthetized with 2% (v/v) isoflurane/oxygen and imaged by IVIS Spectrum 200 (Xenogen, hopkitton, massachusetts, united States; ex.640nm/em.680 nm) 10 minutes, 20 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 24 hours, 48 hours and 96 hours after injection. These images are produced by the process under the trade name LIVING4.7.2 (Perkinelmer, waltham, massachusetts, united States of America). After 8 hours, cGAMP-Cy5/nMOL has a fluorescence signal retention at least one order of magnitude higher than free cGAMP. See fig. 48. Free cGAMP diffuses and rapidly disappears in vivo, but cGAMP/nMOL slowly releases cGAMP and has long-term STING activation.
In vivo antitumor effect: the anti-tumor efficacy of cGAMP/nMOL was evaluated on subcutaneous CT26 tumor-bearing BALB/C and MC38 tumor-bearing C57BL/6 mouse models. For a single tumor model, 2×10 will be 6 The individual CT26 cells or MC38 cells were subcutaneously injected in the right flank of BALB/C or C57BL/6 mice, respectively (both n=6). When the tumor reaches 75-100mm on day 7 3 At this time, 20. Mu.L of Hf was injected into the mice 12 IrnMOL (0.5. Mu. Mol Hf), cGAMP/nMOL (2. Mu.g/0.5. Mu. Mol Hg), cGAMP (2. Mu.g) or PBS. After 8 hours, the mice were anesthetized with 2.5% (v/v) isoflurane/oxygen and the tumors were irradiated with 2Gy X-rays/fraction for 6 consecutive days. Closely monitoring swelling in miceTumor volume, body weight, and health status. As shown in fig. 49A and 49B, the synergistic effect between RT-RDT and STING activation achieved local tumor control at 1/5 of the typical dose of cGAMP (10 μg).
Distal effect: for the bilateral MC38 tumor model, 2X 10 6 The MC38 cells were subcutaneously injected into the right flank of C57BL/6 mice and 1X 10 cells were used 6 The MC38 cells were injected on the left flank (n=6). When the primary tumor (right side) reached 100-125mm on day 7 3 At this time, 20. Mu.L of cGAMP/nMOL (2. Mu.g/0.5. Mu. Mol Hf) or PBS was injected into the primary tumor. Mice received the same X-ray treatment procedure as the single tumor model described above. Mice of the group αpd-L1 or cGAMP/nmol+αpd-L1 were intraperitoneally injected with 2×75 μg/mouse αpd-L1 antibody on day 3 and day 6 after the first X-ray treatment. Checkpoint blocking immunotherapy of αpd-L1 enhances cGAMP/nMOL, better disease control for local and distal sites. See fig. 50A and 50B. PD-L1 blockade extends the local synergy between RT-RDT and STING to systemic anti-cancer immune responses. The combination therapy of cGAMP/nmol+αpd-L1 provides: 1) Inhibition of cancer proliferation by enhanced radiosensitization; 2) Exposed tumor antigen of RT-RDT; 3) Maturation and activation of APC and T cells by STING agonists; 4) PD-1/PD-L1 checkpoint blocking effects of CBI. These four aspects are coordinated to integrate into a 2D nano-platform, ultimately achieving favorable immune responses and therapeutic results.
Reference to the literature
All references, including but not limited to, all patents, patent applications and publications thereof, scientific journal articles and database entries, cited herein are hereby incorporated by reference in their entirety as if fully set forth herein, provide a background to, or teach, the methods, techniques and/or compositions used herein.
Allavena,P.;Sica,A.;Garlanda,C.;Mantovani,A.,The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance.Immunol.Rev.2008,222(1),155-161.
An,J.;Geib,S.J.;Rosi,N.L.,Cation-triggered drug release from a porous zinc-adeninate metal-organic framework.J.Am.Chem.Soc.2009,131(24),8376-8377.
Brahmer,J.R.,et al.,Safety and activity of anti-PD-L1 antibody in patients with advanced cancer.New England Journal of Medicine 2012,366,2455-2465.
Brody,J.D.,et al.,In situ vaccination with a TLR9 agonist induces systemic lymphoma regression:a phase I/II study.Journal of clinical oncology 2010,28,4324.
Castano,A.P.,Mroz,P.,Hamblin,M.R.,Photodynamic therapy and anti-tumour immunity.Nature Reviews Cancer 2006,6,535.
Chao,M.P.;Jaiswal,S.;Weissman-Tsukamoto,R.;Alizadeh,A.A.;Gentles,A.J.;Volkmer,J.;Weiskopf,K.;Willingham,S.B.;Raveh,T.;Park,C.Y.,Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47.Sci.Transl.Med.2010,2(63),63ra94-63ra94.
Chao,Y.,et al.,Combined local immunostimulatory radioisotope therapy and systemic immune checkpoint blockade imparts potent antitumour responses.Nature Biomedical Engineering 2018,2,611.
Chen,H.;Wang,G.D.;Chuang,Y.-J.;Zhen,Z.;Chen,X.;Biddinger,P.;Hao,Z.;Liu,F.;Shen,B.;Pan,Z.,Nanoscintillator-mediated X-ray inducible photodynamic therapy for in vivo cancer treatment.Nano Lett.2015,15(4),2249-2256.
Chen,Q.;Wang,C.;Zhang,X.;Chen,G.;Hu,Q.;Li,H.;Wang,J.;Wen,D.;Zhang,Y.;Lu,Y.,In situ sprayed bioresponsive immunotherapeutic gel for post-surgical cancer treatment.Nat.Nanotechnol.2019a,14(1),89-97.
Chen,Q.;Chen,G.;Chen,J.;Shen,J.;Zhang,X.;Wang,J.;Chan,A.;Gu,Z.,Bioresponsive Protein Complex of aPD1 and aCD47 Antibodies for Enhanced Immunotherapy.Nano Lett.2019b,19(8),4879-4889.
Deng,L.,et al.,STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors.Immunity 2014,41,843-852.
Du,B.,Yu,M.,Zheng,J.,Transport and interactions of nanoparticles in the kidneys.Nature Reviews Materials 2018,1.
Dunn,G.P.,Bruce,A.T.,Ikeda,H.,Old,L.J.,R.D.Schreiber,R.D.,Cancer immunoediting:from immunosurveillance to tumor escape.Nature immunology 2002,3,991-998.
Emming,S.,Schroder,K.,Tiered DNA sensors for escalating responses.Science 2019,365,1375-1376.
Errico,A.,Immunotherapy:PD-1-PD-L1 axis:efficient checkpoint blockade against cancer.Nature reviews Clinical oncology 2015,12,63-63.
Feng,M.;Jiang,W.;Kim,B.Y.;Zhang,C.C.;Fu,Y.-X.;Weissman,I.L.,Phagocytosis checkpoints as new targets for cancer immunotherapy.Nat.Rev.Cancer 2019,19(10),568-586.
Figdor,C.G.,de Vries,I.J.M.,Lesterhuis,W.J.,Melief,C.J.,Dendritic cell immunotherapy:mapping the way.Nature medicine 2004,10,475.
Furukawa,H.;Cordova,K.E.;O’Keeffe,M.;Yaghi,O.M.,The chemistry and applications of metal-organic frameworks.Science 2013,341(6149),1230444.
Gilliet,M.,Cao,W.,Liu,Y.-J.,Plasmacytoid dendritic cells:sensing nucleic acids in viral infection and autoimmune diseases.Nature Reviews Immunology 2008,8,594.
Goldman,B.;L.DeFrancesco,L.,The cancer vaccine roller coaster.Nature biotechnology 2009,27,129.
Gong,T.,Liu,L.,Jiang,W.,Zhou,R.,DAMP-sensing receptors in sterile inflammation and inflammatory diseases.Nature Reviews Immunology,2019,1-18.
Hu,Z.,Ott,P.A.,Wu,C.J.,Towards personalized,tumour-specific,therapeutic vaccines for cancer.Nature Reviews Immunology 2018,18,168.
Jaiswal,S.;Jamieson,C.H.;Pang,W.W.;Park,C.Y.;Chao,M.P.;Majeti,R.;Traver,D.;van Rooijen,N.;Weissman,I.L.,CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis.Cell 2009,138(2),271-285.
Jiang,W.,et al.,Designing nanomedicine for immuno-oncology.Nature Biomedical Engineering 2017,1,0029.
Joyce,J.A.,Fearon,D.T.,T cell exclusion,immune privilege,and the tumor microenvironment.Science 2015,348,74-80.
Kamath,P.;Darwin,E.;Arora,H.;Nouri,K.,A Review on Imiquimod Therapy and Discussion on Optimal Management of Basal Cell Carcinomas.Clin.Drug Investig.2018,38(10),883-899.
Kawai,T.,S.Akira,S.,The role of pattern-recognition receptors in innate immunity:update on Toll-like receptors.Nature immunology 2010,11,373.
Kepp,O.,Marabelle,A.,Zitvogel,L.,Kroemer,G.,Oncolysis without viruses—inducing systemic anticancer immune responses with local therapies.Nature Reviews Clinical Oncology 2019,1-16.
Klinman,D.M.,Immunotherapeutic uses of CpG oligodeoxynucleotides.Nature Reviews Immunology 2004,4,249.
Kojima,Y.;Volkmer,J.-P.;McKenna,K.;Civelek,M.;Lusis,A.J.;Miller,C.L.;Direnzo,D.;Nanda,V.;Ye,J.;Connolly,A.J.,CD47-blocking antibodies restore phagocytosis and prevent atherosclerosis.Nature 2016,536(7614),86-90.
Kuai,R.,Ochyl,L.J.,Bahjat,K.S.,Schwendeman,A.,Moon,J.J.,Designer vaccine nanodiscs for personalized cancer immunotherapy.Nature materials 2017,16,489.
Lan,G.,Ni,K.,Veroneau,S.S.,Song,Y.,Lin,W.,Nanoscale Metal-Organic Layers for Radiotherapy-Radiodynamic Therapy.Journal of the American Chemical Society 2018,140,16971-16975.
Lan,G.,Ni,K.,Lin,W.,Nanoscale metal-organic frameworks for phototherapy of cancer.Coordination chemistry reviews 2019a,379,65-81.
Lan,G.,et al.,Nanoscale Metal-Organic Framework Hierarchically Combines High-Z Components for Multifarious Radio-Enhancement.J.Am.Chem.Soc.2019b,141,6859-6863.
Liu,H.,et al.,Structure-based programming of lymph-node targeting in molecular vaccines.Nature 2014,507,519.
Liu,X.;Pu,Y.;Cron,K.;Deng,L.;Kline,J.;Frazier,W.A.;Xu,H.;Peng,H.;Fu,Y.-X.;Xu,M.M.,CD47 blockade triggers T cell-mediated destruction of immunogenic tumors.Nat.Med.2015,21(10),1209.
Lou,J.;Zhang,L.;Zheng,G.,Advancing cancer immunotherapies with nanotechnology.Adv.Ther.2019,2(4),1800128.
Louttit,C.;Park,K.S.;Moon,J.J.,Bioinspired nucleic acid structures for immune modulation.Biomater.2019,119287.
Lu,K.;He,C.;Lin,W.,Nanoscale Metal-Organic Framework for Highly Effective Photodynamic Therapy of Resistant Head and Neck Cancer.J.Am.Chem.Soc.2014,136(48),16712-16715.
Lu,K.;He,C.;Guo,N.;Chan,C.;Ni,K.;Lan,G.;Tang,H.;Pelizzari,C.;Fu,Y.-X.;Spiotto,M.T.,Low-dose X-ray radiotherapy-radiodynamic therapyvia nanoscale metal-organic frameworks enhances checkpoint blockadeimmunotherapy.Nat.Biomed.Eng.2018,2(8),600.
Luo,M.,et al.,A STING-activating nanovaccine for cancerimmunotherapy.Nature nanotechnology 2017,12,648.
Lutz,M.B.,Schuler,G.,Immature,semi-mature and fully maturedendritic cells:which signals induce tolerance or immunityTrends inimmunology 2002,23,445-449.
Ma,L.,et al.,Enhanced CAR-T cell activity against solid tumors byvaccine boosting through the chimeric receptor.Science 2019,365,162-168.
Mahoney,K.M.,Rennert,P.D.,Freeman,G.J.,Combination cancerimmunotherapy and new immunomodulatory targets.Nature reviews Drugdiscovery 2015,14,561-584.
Min,Y.,et al.,Antigen-capturing nanoparticles improve the abscopaleffect and cancer immunotherapy.Nature nanotechnology 2017,12,877.
Morris,W.;Briley,W.E.;Auyeung,E.;Cabezas,M.D.;Mirkin,C.A.,Nucleic Acid-Metal Organic Framework(MOF)Nanoparticle Conjugates.J.Am.Chem.Soc.2014,136(20),7261-7264.
Nam,J.;Son,S.;Park,K.S.;Zou,W.;Shea,L.D.;Moon,J.J.,Cancernanomedicine for combination cancer immunotherapy.Nat.Rev.Mater.2019,4(6),398-414.
Ni,K.,et al.,Nanoscale metal-organic frameworks enhanceradiotherapy to potentiate checkpoint blockade immunotherapy.Naturecommunications 2018a,9,2351.
Ni,K.,et al.,Nanoscale metal-organic frameworks formitochondria-targeted radiotherapy radiodynamic therapy.Naturecommunications 2018b,9,4321.
Ni,K.;Lan,G.;Chan,C.;Duan,X.;Guo,N.;Veroneau,S.S.;Weichselbaum,R.R.;Lin,W.,Ultrathin Metal-Organic-Layer MediatedRadiotherapy-Radiodynamic Therapy.Matter 2019,1(5),1331-1353.
O'neill,L.A.;Golenbock,D.;Bowie,A.G.,The history of Toll-likereceptors—redefining innate immunity.Nat.Rev.Immunol.2013,13(6),453-460.
Purcell,A.W.,McCluskey,J.,Rossjohn,J.,More than one reason torethink the use of peptides in vaccine design.Nature reviews Drug discovery2007,6,404.
Radovic-Moreno,A.F.,et al.,Immunomodulatory spherical nucleic acids.Proceedings of the National Academy of Sciences,U.S.A.2015,112,3892-3897.
Rodell,C.B.;Arlauckas,S.P.;Cuccarese,M.F.;Garris,C.S.;Li,R.;Ahmed,M.S.;Kohler,R.H.;Pittet,M.J.;Weissleder,R.,TLR7/8-agonist-loaded nanoparticles promote the polarization oftumour-associated macrophages to enhance cancer immunotherapy.Nat.Biomed.Eng.2018,2(8),578.
Rosi,N.L.,et al.,Oligonucleotide-modified gold nanoparticles forintracellular gene regulation.Science 2006,312,1027-1030.
Sahin,U.,Türeci,Personalized vaccines for cancer immunotherapy.Science 2018,359,1355-1360.
Scheetz,L.,et al.,Engineering patient-specific cancer immunotherapies.Nature biomedical engineering 2019,3,768-782.
Shae,D.,et al.,Endosomolytic polymersomes increase the activity ofcyclic dinucleotide STING agonists to enhance cancer immunotherapy.Naturenanotechnology 2019,14,269.
Song,W.,Musetti,S.N.,Huang,L.,Nanomaterials for cancerimmunotherapy.Biomaterials 2017,148,16-30.
Tanyi,J.L.,et al.,Personalized cancer vaccine effectively mobilizesantitumor T cell immunity in ovarian cancer.Science translational medicine2018,10,eaao5931.
Wang,S.,et al.,General and direct method for preparingoligonucleotide-functionalized metal-organic framework Nanoparticles.Journal of the American Chemical Society 2017,139,9827-9830.
Wang,H.,Mooney D.J.,Biomaterial-assisted targeted modulation ofimmune cells in cancer treatment.Nature Materials 2018,17,761-772.
Weichselbaum,R.R.,Liang,H.,Deng,L.,Fu,Y.-X.,Radiotherapy and immunotherapy:a beneficial liaisonNature reviews Clinical oncology 2017,14,365.
Weiner,G.J.,Liu,H.-M.,Wooldridge,J.E.,Dahle,C.E.,Krieg,A.M.,Immunostimulatory oligodeoxynucleotides containing the CpG motif are effective as immune adjuvants in tumor antigen immunization.Proceedings of the National Academy of Sciences 1997,94,10833-10837.
Wilson,D.S.,et al.,Antigens reversibly conjugated to a polymeric glyco-adjuvant induce protective humoral and cellular immunity.Nature materials 2019,18,175.
Wu,M.X.;Yang,Y.W.,Metal-organic framework(MOF)-based drug/cargo delivery and cancer therapy.Adv.Mater.2017,29(23),1606134.
Xiong,Z.;Ohlfest,J.R.,Topical Imiquimod has Therapeutic and Immunomodulatory Effects Against Intracranial Tumors.J.Immunother.2011,34(3).
Zhang,Y.-N.,et al.,Nanoparticle size influences antigen retention and presentation in lymph node follicles for humoral immunity.Nano letters 2019,19,7226-7235.
Zhu,Y.-Y.,et al.,Merging Photoredox and Organometallic Catalysts in aMetal-Organic Framework Significantly Boosts Photocatalytic Activities.Angew.Chem.2018,130,14286-14290.
It will be understood that various details of the disclosed subject matter may be changed without departing from the scope of the disclosed subject matter. Furthermore, the foregoing description is for the purpose of illustration only, and not for the purpose of limitation.
Sequence listing
<110> university of Chicago
Lin Wen
Ni Kai yuan
Luo Tao
Lan Guangxu
<120> delivery of small and biological macromolecules from metal organic frameworks for cancer immunotherapy
<130> 3072-19 PCT
<150> 63/028,891
<151> 2020-05-22
<150> 63/045,499
<151> 2020-06-29
<160> 5
<170> PatentIn version 3.3
<210> 1
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic sequence
<400> 1
Cys Gln Cys Arg Arg Lys Asn
1 5
<210> 2
<211> 16
<212> PRT
<213> artificial sequence
<220>
<223> synthetic sequence
<400> 2
Arg Arg Arg Arg Arg Arg Arg Arg Arg Cys Gln Cys Arg Arg Lys Asn
1 5 10 15
<210> 3
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic sequence
<400> 3
Ser Ile Ile Asn Phe Glu Lys Leu
1 5
<210> 4
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic sequence
<400> 4
Ser Pro Ser Tyr Val Tyr His Gln Phe
1 5
<210> 5
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic sequence
<400> 5
Lys Ser Pro Trp Phe Thr Thr Leu
1 5

Claims (48)

1. A Metal Organic Framework (MOF) having a surface modified to coordinately or electrostatically bind to one or more therapeutic agents of interest, wherein the MOF comprises:
(a) A plurality of metallo-oxygen cluster Secondary Building Units (SBUs), wherein each of the metallo-oxygen cluster SBUs comprises one or more first metal ions and one or more anions, wherein each of the one or more anions coordinates to one or more of the one or more first metal ions; and
(b) A plurality of organic bridging ligands linking together the plurality of SBUs to form a two-dimensional or three-dimensional matrix;
wherein (i) each of the plurality of SBUs at the MOF surface comprises a weakly coordinating anion as an SBU capping group anion, or (ii) the plurality of organic bridging ligands comprises an organic bridging ligand comprising an electron withdrawing group or ligand, a positive charge, or a combination thereof, optionally wherein the plurality of organic bridging ligands comprises a ligand comprising a nitrogen donor group coordinately bound to a second metal ion, wherein the second metal ion is further coordinated to at least one second metal ligand comprising one or more electron withdrawing groups; wherein the surface of the MOF has enhanced ability to coordinately or electrostatically bind to one or more therapeutic agents of interest.
2. The MOF of claim 1, wherein the one or more first metal ions comprise ions of at least one metal that absorbs ionizing radiation, optionally X-rays, and/or wherein the metal is selected from the group consisting of Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi; further optionally wherein the first metal ion is a Hf ion.
3. The MOF of claim 1 or claim 2, wherein each of the plurality of SBUs of the MOF surface comprises a weakly coordinating anion as a capping group, optionally wherein the weakly coordinating anion is selected from the group consisting of trifluoroacetate and trifluoromethanesulfonate.
4. The MOF of claim 3, wherein the plurality of organic bridging ligands comprises a porphyrin substituted with at least two carboxylate groups, optionally wherein the plurality of organic bridging ligands comprises 5, 15-bis (p-benzoate) porphyrin (DBP).
5. The MOF of claim 3 or 4, wherein the MOF further comprises a small molecule therapeutic sequestered in pores and/or cavities of a two-or three-dimensional network, optionally wherein the small molecule therapeutic is a chemotherapeutic, a small molecule inhibitor, and/or a small molecule immunomodulator.
6. The MOF of claim 5, wherein the MOF comprises a chemotherapeutic agent sequestered in pores and/or cavities of the two-or three-dimensional network, optionally wherein the chemotherapeutic agent is selected from cisplatin, carboplatin, paclitaxel, SN-35, and etoposide.
7. The MOF of claim 5, wherein the MOF comprises a small molecule inhibitor sequestered in pores and/or cavities of the two-or three-dimensional network, optionally wherein the small molecule inhibitor is selected from the group consisting of PLK1 inhibitor, wnt inhibitor, bcl-2 inhibitor, PD-L1 inhibitor, ENPP1 inhibitor, and IDO inhibitor.
8. The MOF of claim 5, wherein the MOF comprises a small molecule immunomodulator sequestered in pores and/or cavities of the two-dimensional or three-dimensional network.
9. The MOF of claim 8, wherein the small molecule immunomodulator is Imiquimod (IMD).
10. The MOF of claim 1 or claim 2, wherein the plurality of organic bridging ligands comprises an organic bridging ligand comprising a nitrogen donor group, wherein the nitrogen donor group coordinates to a second metal ion, and wherein the second metal ion is further coordinated to at least one second metal ligand comprising one or more electron withdrawing groups, optionally wherein the one or more electron withdrawing groups are selected from halo groups and perhaloalkyl groups.
11. The MOF of claim 10, wherein the organic bridging ligand containing a nitrogen donor group is 4,4 '-bis (p-benzoic acid) -2,2' -bipyridine (DBB).
12. The MOF of claim 10 or 11, wherein the second metal ion is an iridium (Ir) ion or a ruthenium (Ru) ion, and/or wherein the first metal ion is coordinated to two second metal ligands, wherein one or both of the second metal ligands comprises one or more electron withdrawing groups.
13. The MOF of claim 12, wherein one or both of the second metal ligands is 2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine (dF (CF) 3 )ppy)。
14. The MOF of any one of claims 10-13, wherein the MOF has a zeta (zeta) potential value of at least about 5 millivolts (mV), optionally wherein the MOF has a zeta potential value of at least about 30 mV.
15. The MOF of any one of claims 1-14, wherein the MOF comprises a three-dimensional network, wherein the three-dimensional network is provided in the form of nanoparticles.
16. A Metal Organic Framework (MOF) for delivering one or more therapeutic agents of interest, wherein the MOF comprises
(a) A plurality of metallo-oxygen cluster Secondary Building Units (SBUs), wherein each of the metallo-oxygen cluster SBUs comprises one or more first metal ions and one or more anions, wherein each of the anions coordinates with one or more of the one or more first metal ions;
(b) A plurality of organic bridging ligands linking together the plurality of SBUs to form a two-dimensional or three-dimensional matrix; and
(c) One or more therapeutic agents of interest bound to the surface of the MOF by coordination bonds or electrostatic interactions, optionally wherein one or more therapeutic agents of interest are coordinately bound to metal ions of one or more of the plurality of SBUs at the surface of the MOF.
17. The MOF of claim 16, wherein the first metal ion is an ion of a metal that absorbs ionizing radiation, optionally X-rays, and/or wherein the first metal ion is an ion of a metal selected from Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi; further optionally wherein the first metal ion is a Hf ion.
18. The MOF of claim 16 or 17, wherein each of the one or more therapeutic agents of interest is selected from the group consisting of a nucleic acid, a small molecule comprising phosphate or carboxylate groups, and/or a large molecule comprising surface accessible phosphate or carboxylate groups.
19. The MOF of claim 18, wherein the one or more therapeutic agents of interest comprise a macromolecule comprising a surface accessible phosphate or carboxylate group, and wherein the macromolecule is a protein, optionally wherein the protein is an antibody.
20. The MOF of claim 19, wherein the protein is selected from the group consisting of an anti-CD 37 antibody, an anti-CD 44 antibody, an anti-CD 47 antibody, an anti-CD 73 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-LAG 3 antibody, and an anti-CTLA-4 antibody.
21. A MOF according to claim 18, wherein the one or more therapeutic agents of interest comprise a nucleic acid, and wherein the nucleic acid is selected from the group consisting of miRNA, mRNA, siRNA, cpG ODN, and a cyclic dinucleotide, optionally the nucleic acid is a cyclic dinucleotide and the cyclic dinucleotide is a STING agonist, further optionally wherein the STING agonist is c-di-AMP or cGAMP.
22. The MOF of any one of claims 16-21, wherein the MOF comprises one or more additional therapeutic agents sequestered in pores or cavities of the two-dimensional or three-dimensional network; optionally wherein the MOF comprises from about 1wt% to about 50wt% of the one or more additional therapeutic agents.
23. The MOF of claim 16, wherein the plurality of SBUs comprise Hf oxygen clusters, wherein the plurality of organic bridging ligands comprise DBPs, and wherein the one or more therapeutic agents of interest are bound to the surface of the MOF via coordination bonds to Hf ions of surface-accessible SBUs.
24. The MOF of claim 23, wherein the one or more therapeutic agents of interest comprise one or more antibodies.
25. The MOF of claim 24, wherein the one or more therapeutic agents comprise an anti-CD 47 antibody.
26. The MOF of claim 25, wherein the MOF further comprises an IMD sequestered in a pore or cavity of the two-dimensional or three-dimensional network.
27. The MOF of claim 26, wherein the MOF is a three-dimensional network and is provided as a nanoparticle.
28. The MOF of claim 27, wherein the MOF comprises about 1wt% to about 50wt% IMD or anti-CD 47 antibody; optionally wherein the MOF comprises about 9wt% IMD and about 7.5wt% anti-CD 47 antibody.
29. The MOF of claim 16, wherein the plurality of SBUs comprise Hf oxygen clusters, wherein the plurality of organic bridging ligands comprise DBBs coordinated to Ir ions, wherein the Ir ions are further coordinated to two (dF (CF 3 ) ppy) coordination; and wherein the one or more therapeutic agents of interest bind to the surface of the MOF by electrostatic interactions.
30. The MOF of claim 29, wherein the one or more therapeutic agents of interest comprise a nucleic acid.
31. The MOF of claim 30, wherein the nucleic acid is a STING agonist or CpG Oligodeoxynucleotide (ODN), optionally wherein the nucleic acid is a CpG ODN.
32. The MOF of any one of claims 16-31, wherein the MOF comprises about 1wt% to about 50wt% of the one or more therapeutic agents of interest, optionally wherein the one or more therapeutic agents of interest comprise antibodies.
33. A method of treating cancer in a subject in need thereof, the method comprising:
(a) Administering to the subject the MOF of any one of claims 16-32; and
(b) Exposing at least a portion of the subject to ionizing radiation energy, optionally X-rays.
34. The method of claim 33, wherein the method further comprises administering to the subject an additional therapeutic agent or treatment, optionally an immunotherapeutic agent and/or a cancer treatment selected from the group consisting of surgery, chemotherapy, toxin therapy, cryotherapy, and gene therapy.
35. The method of claim 34, wherein the additional therapeutic agent is an immunotherapeutic agent, optionally wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
36. The method of claim 35, wherein the immunotherapeutic agent is an anti-PD-1 or anti-PD-L1 antibody.
37. The method of any one of claims 33-36, wherein the cancer is colorectal cancer, melanoma, head and neck cancer, brain cancer, breast cancer, liver cancer, cervical cancer, lung cancer, or pancreatic cancer.
38. The method of any one of claims 33-37, wherein administration of the MOF provides an extended release profile for one or more of the one or more therapeutic agents of interest, optionally wherein the release rate is adjustable and/or wherein the MOF provides sustained release of one or more therapeutic agents of interest over a period of hours or days.
39. The method of any one of claims 33-38, wherein administration of the MOF reduces a therapeutically effective dose of the one or more therapeutic agents of interest.
40. A method of enhancing the interaction and/or binding of one or more therapeutic agents of interest with a surface of a Metal Organic Framework (MOF), the method comprising modifying the surface of the MOF by: (i) Providing one or more accessible coordination sites on the surface to which the weakly coordinating anion is coordinately bound, which weakly coordinating anion may be substituted with carboxylate or phosphate substituents of the therapeutic agent of interest; or (ii) providing a MOF comprising one or more electron withdrawing bridging ligands, one or more bridging ligands comprising a positive charge; or a combination thereof.
41. The method of claim 40, wherein modifying the surface of the MOF comprises:
(ia) providing a parent MOF comprising metallo-oxy-cluster SBUs linked together by an organic bridging ligand, wherein each of said SBUs contains one or more metal ions and one or more anions, and wherein said MOF comprises a plurality of surface-accessible metallo-oxy-cluster SBUs, wherein said one or more anions of each of said surface-accessible metallo-oxy-cluster SBUs comprise a strongly coordinating anion that is an SBU capping group; optionally wherein the strongly coordinating anion comprises acetate or formate; and
(ib) removing the strongly coordinating anion, wherein the removing comprises contacting the parent MOF with an agent selected from the group consisting of trimethylsilyl trifluoroacetate, trimethylsilyl triflate, and mineral acids having a pKa of less than about 3; whereby the strongly coordinating anion is substituted with a weakly coordinating anion, optionally wherein the strongly coordinating anion is selected from acetate or formate anions, optionally wherein the weakly coordinating ion is selected from trifluoroacetate or trifluoromethanesulfonate anions.
42. The method of claim 40, wherein providing a MOF comprising one or more bridging ligands comprising electron withdrawing groups, one or more bridging ligands comprising positive charges, or a combination thereof comprises providing a MOF comprising metal oxygen clusters SBUs linked together by organic bridging ligands, wherein each of the SBUs comprises one or more first metal ions and one or more anions coordinated to the one or more first metal ions, and wherein the organic bridging ligands comprise at least one organic bridging ligand comprising coordinated non-SBU linked second metal ions, wherein the second metal ions are further coordinated to one or more electron withdrawing ligands, optionally wherein the electron withdrawing ligands are halogen and/or perhaloalkyl substituted bipyridine ligands.
43. The method of claim 42, wherein providing the MOF comprises providing a polymer comprising a bis (4-benzoic acid) -2,2' -bipyridine (DBB) bridged ligand, wherein the DBB bridged ligand coordinates a first metal ion and a second metal ion of two different metal oxide clusters SBU, and wherein the second metal ion is further coordinated to two halogen and/or perhaloalkyl substituted pyridine ligands, optionally wherein the two halogen and/or perhaloalkyl substituted pyridine ligands are each 2- (2, 4-difluorophenyl) -5- (trifluoromethyl) pyridine.
44. The method of claim 42 or 43, wherein the second metal ion is iridium (Ir) or ruthenium (Ru).
45. The method of any of claims 40-44, wherein the MOF comprises one or more SBUs comprising ions of a metal that absorbs ionizing radiation, optionally X-rays, and/or wherein the metal ions are ions of an element selected from the group consisting of Hf, lanthanide metals, ba, ta, W, re, os, ir, pt, au, pb, and Bi; further optionally wherein the metal ion is a Hf ion.
46. The method of any one of claims 40-45, wherein the MOF has enhanced interaction and/or binding capacity for one or more therapeutic agents of interest as compared to an unsurface modified MOF, wherein the one or more therapeutic agents of interest are selected from nucleic acids, small molecules, and/or macromolecules comprising surface accessible phosphate or carboxylate groups.
47. The method of claim 46, wherein the protein is selected from the group consisting of an anti-CD 37 antibody, an anti-CD 44 antibody, an anti-CD 47 antibody, an anti-CD 73 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-LAG 3 antibody, and an anti-CTLA-4 antibody.
48. The method of claim 46, wherein the nucleic acid is selected from the group consisting of miRNA, mRNA, siRNA, cpG ODN and a cyclic dinucleotide, optionally wherein the cyclic dinucleotide is a STING agonist, further optionally wherein the STING agonist is c-di-AMP or cGAMP.
CN202180060168.XA 2020-05-22 2021-05-24 Metal organic frameworks deliver small and biological macromolecules for cancer immunotherapy Pending CN116744909A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/028,891 2020-05-22
US202063045499P 2020-06-29 2020-06-29
US63/045,499 2020-06-29
PCT/US2021/033886 WO2021237209A1 (en) 2020-05-22 2021-05-24 Metal-organic frameworks deliver small molecules and biomacromolecules for cancer immunotherapy

Publications (1)

Publication Number Publication Date
CN116744909A true CN116744909A (en) 2023-09-12

Family

ID=87901624

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180060168.XA Pending CN116744909A (en) 2020-05-22 2021-05-24 Metal organic frameworks deliver small and biological macromolecules for cancer immunotherapy

Country Status (1)

Country Link
CN (1) CN116744909A (en)

Similar Documents

Publication Publication Date Title
US20230293698A1 (en) Metal-organic frameworks deliver small molecules and biomacromolecules for cancer immunotherapy
CN111194232B (en) Nanoscale metal-organic layer and metal-organic nanosheet
US10780045B2 (en) Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, chemotherapy, immunotherapy, and any combination thereof
US10806694B2 (en) Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
JP7090034B2 (en) Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy and any combination thereof
Le et al. Nano delivery systems and cancer immunotherapy
US20200261403A1 (en) Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
Guan et al. A novel prodrug and its nanoformulation suppress cancer stem cells by inducing immunogenic cell death and inhibiting indoleamine 2, 3-dioxygenase
Escriche‐Navarro et al. Mesoporous silica materials as an emerging tool for cancer immunotherapy
US20210353670A1 (en) Nanoparticles for use as a therapeutic vaccine
Huang et al. Mn-based cGAS-STING activation for tumor therapy
Ma et al. Rationally integrating peptide-induced targeting and multimodal therapies in a dual-shell theranostic platform for orthotopic metastatic spinal tumors
Dongye et al. Toll-like receptor 9 agonists and combination therapies: strategies to modulate the tumour immune microenvironment for systemic anti-tumour immunity
Yang et al. Cancer immunotherapy elicited by immunogenic cell death based on smart nanomaterials
Qu et al. Drug-drug conjugates self-assembled nanomedicines triggered photo−/immuno-therapy for synergistic cancer treatments
CN116744909A (en) Metal organic frameworks deliver small and biological macromolecules for cancer immunotherapy
CA3225062A1 (en) Nanoconstructs and nanoparticle-mediated delivery of immunogenic cell death inducers for enhancing cancer immunotherapy
EA040587B1 (en) METHOD FOR MALIGNANT NEOPLASMS TREATMENT

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination