CN109481696B - Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof - Google Patents

Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof Download PDF

Info

Publication number
CN109481696B
CN109481696B CN201811575728.1A CN201811575728A CN109481696B CN 109481696 B CN109481696 B CN 109481696B CN 201811575728 A CN201811575728 A CN 201811575728A CN 109481696 B CN109481696 B CN 109481696B
Authority
CN
China
Prior art keywords
cage
chemotherapy
nps
nanoparticles
supramolecular
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.)
Active
Application number
CN201811575728.1A
Other languages
Chinese (zh)
Other versions
CN109481696A (en
Inventor
王明
蒋鑫
于浩
曾云婷
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.)
Jilin University
Original Assignee
Jilin University
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 Jilin University filed Critical Jilin University
Priority to CN201811575728.1A priority Critical patent/CN109481696B/en
Publication of CN109481696A publication Critical patent/CN109481696A/en
Application granted granted Critical
Publication of CN109481696B publication Critical patent/CN109481696B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6949Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0057Photodynamic therapy with a photosensitizer, i.e. agent able to produce reactive oxygen species upon exposure to light or radiation, e.g. UV or visible light; photocleavage of nucleic acids with an agent
    • A61K41/0071PDT with porphyrins having exactly 20 ring atoms, i.e. based on the non-expanded tetrapyrrolic ring system, e.g. bacteriochlorin, chlorin-e6, or phthalocyanines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0057Photodynamic therapy with a photosensitizer, i.e. agent able to produce reactive oxygen species upon exposure to light or radiation, e.g. UV or visible light; photocleavage of nucleic acids with an agent
    • A61K41/0076PDT with expanded (metallo)porphyrins, i.e. having more than 20 ring atoms, e.g. texaphyrins, sapphyrins, hexaphyrins, pentaphyrins, porphocyanines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The present invention provides a method for preparing supramolecular cage-loaded nanoparticles for use in photodynamic therapy and chemotherapy of cancer, comprising: s1: preparing a supermolecular cage required by the nano-particles; s2: introducing mPEG-b-PEBP, RGD-PEG-b-PEBP and a supramolecular cage to form a nano particle, wherein the supramolecular cage comprises a tetrapyridine porphyrin derivative or a tetrapyridine metalloporphyrin derivative and a Pt (II) receptor. The nano particles formed with the supramolecular cage can stably and durably exist in the circulation in a living body, have a targeting effect on cancer tissues, and facilitate the transportation of medicaments and the treatment of cancers.

Description

Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof
Technical Field
The invention belongs to the technical field of medicines, relates to a cancer treatment medicine, and particularly relates to a medicine for photodynamic cancer treatment and chemotherapy and a preparation method thereof.
Background
Over the past decades, photodynamic therapy (PDT) has proven to be an attractive and promising clinical method for the treatment of lung, bladder, skin and esophageal cancer. PDT is the activation of photosensitizing drugs (PS) by light of the appropriate wavelength, generating Reactive Oxygen Species (ROS) that can damage cellular compartments including plasma, mitochondria, lysosomes and nuclear membranes, leading to irreversible damage to the desired cells to cause cell death. Compared with the traditional treatment means, PDT shows several obvious advantages, such as noninvasive treatment, controllable time, negligible drug resistance and low toxic and side effects. Porphyrins and their derivatives are of particular interest among the many photosensitizing drugs that may have PDT functionality. Therefore, researchers have endeavored to design and synthesize a series of novel porphyrin photosensitizers with improved PDT effects. For example, the research group of Lin (j.am. chem. soc.2014,136,16712) developed several novel NMOFs that combine structural order and porosity with specific photosensitizing drugs to achieve high PS loading. Research group of Tang (Angew. chem. int. Ed., 2018,57,4891)Several new MOFs, including cu (ii) and metalloporphyrin derivatives, were designed to reduce intracellular glutathione concentrations or modulate singlet oxygen based on hydrogen sulfide activation. The group of Yan (j.am. chem.soc.,2017,139,1921) utilizes nanoparticle-delivered delivery platforms based on simple peptide or amphiphilic amino acid self-assembly to improve the EPR effect of drugs. The panel of Spingler (angelw. chem. int. ed.,2014,53,6938) reported four porphyrin derivatives and demonstrated that porphyrins exhibit significant DNA cleavage phenomena and photo-cytotoxicity under light irradiation. However, most reported porphyrin derivatives resulted in severe pi-pi stacking due to large planar structures, with a substantial reduction1O2The PDT efficiency is suppressed. This accumulation, producing low singlet oxygen concentrations under hypoxic conditions, has become one of the common limitations limiting the ability of porphyrin derivatives and other photosensitizing drugs to perform PDT functions in potential clinical applications.
Disclosure of Invention
In order to solve the above problems and simultaneously impart better anticancer properties to porphyrin derivatives, a first aspect of the present invention provides a method for preparing supramolecular cage-supported nanoparticles for photodynamic therapy and chemotherapy of cancer, comprising:
s1: preparing a supermolecular cage required by the nano-particles;
s2: dissolving a supramolecular cage in a solution of acetone, amphiphilic polymer mPEG-b-PEBP and end group targeting polymer RGD-PEG-b-PEBP, dropwise adding the solution into Milli-Q water, violently stirring, spin-drying the acetone, and performing ultrasonic treatment for five minutes to obtain a well-dispersed nanoparticle suspension, thereby finally forming nanoparticles;
wherein, the supermolecule cage comprises a tetrapyridine porphyrin derivative or a tetrapyridine metalloporphyrin derivative and a Pt (II) receptor, and the structure of the supermolecule cage is (I):
Figure BDA0001916700850000021
Figure BDA0001916700850000031
wherein the content of the first and second substances,
Figure BDA0001916700850000032
is a tetrapyridylporphyrin derivative;
Figure BDA0001916700850000033
is a tetrapyridine metalloporphyrin derivative;
Figure BDA0001916700850000034
is a Pt (II) acceptor.
The supermolecule cage structure formed by coordination-driven self-assembly is adopted, and compared with the structures such as MOF and nano particles, the supermolecule cage structure has a very special and unique structure, is single in molecular weight distribution, and is easier to control and use. The relative position and the number of the design components of the molecular cage structure are strictly fixed, the Pt (II) receptor distributes tetrapyridine porphyrin on two sides of the tetrapyridine porphyrin so as to greatly reduce the self aggregation of porphyrin in molecules, and the pi-pi accumulation and aggregation of porphyrin between molecules are further avoided by forming nano particles, thereby producing the tetrapyridine porphyrin1O2The efficiency of the device is effectively improved. The tetrapyridyl porphyrin derivative or the tetrapyridyl metalloporphyrin derivative in the molecular cage serves as a PDT photosensitive drug and a donor, the Pt (II) receptor serves as a receptor for chemotherapy, and the introduction of the Pt (II) receptor can not only increase the capacity of the chemotherapy of the drug, but also realize higher synergistic anticancer efficiency with photodynamic therapy because the Pt (II) receptor enters an accurate supermolecular cage.
Preferably, the structure of the tetrapyridylporphyrin derivative is represented by formula (II):
Figure BDA0001916700850000035
wherein:
r1 is H, alkyl;
r2 is H, F, Cl, Br, I or alkyl.
More preferably, the tetrapyridylporphyrin derivative has the structure
Figure BDA0001916700850000041
(P)。
Preferably, the structure of the tetrapyridine metalloporphyrin derivative is shown as a formula (III):
Figure BDA0001916700850000042
wherein:
r1 is H, alkyl;
r2 is H, F, Cl, Br, I, alkyl;
m is Zn, Co, Mn, Ce, Fe, Mg, Hg, Ru, Cu, Zr, Rh, Pt, Sn, Tl, Al, Pt, Pd, Ir, Sb, V, Ti, Hf, Au, Cr, Ag, In, Tb, Gd, Er, Yb, Lu, Dy, Nd, Eu, Pr, Ho, Tm, La, Sm;
more preferably, the structure of the tetrapyridine metalloporphyrin derivative is
Figure BDA0001916700850000051
Preferably, the structure of the pt (ii) acceptor is any one of the following formulae (IV):
Figure BDA0001916700850000052
wherein:
r3 is H, alkyl, amino, aldehyde group, amide group, aryl;
x is
Figure BDA0001916700850000053
Y is O, S, N, Se or Te;
more preferably, the Pt (II) acceptor has a structure
Figure BDA0001916700850000061
Preferred carrier materials are mPEG-b-PEBP and RGD-PEG-b-PEBP, in a ratio of 5: 1 mass ratio and the supermolecular cage action to form the nano particles. Wherein the mPEG-b-PEBP amphiphilic polymer and the RGD-PEG-b-PEBP are amphiphilic polymers with targeting groups at the tail ends. The nano particles formed by the supermolecule cages can stably and durably exist in the circulation of organisms, have a targeting effect on cancer tissues, and facilitate the transportation of medicaments and the treatment of cancers.
The invention also provides a supramolecular cage nanoparticle, which is obtained by the method.
The light absorption wavelength of the nano-particles is 600 nm-900 nm. Experiments prove that the photodynamic effect under the wavelength is the best.
The particle diameter of the nano particles is within 20-120 nm; preferably, the particle size is 30 to 90 nm. Experiments prove that the particle size of the particle accords with the endocytosis size of cells.
The invention also provides an injectable or oral composition, which comprises any one of the nanoparticles and a pharmaceutically acceptable carrier.
Use of said nanoparticles for the preparation of a medicament for use in a method of killing cancer cells using photodynamic therapy and chemotherapy.
A method of killing cancer cells using photodynamic therapy and chemotherapy, comprising contacting the cancer cells with nanoparticles and irradiating the nanoparticles with a therapeutically effective amount of light to induce the self-assembled supramolecular cage to emit singlet oxygen; meanwhile, the composition has chemotherapy ability. Because the photodynamic therapy has outstanding therapeutic effect on superficial cancer self-organization, the medicament has potential application to the treatment of breast cancer, lung cancer, bladder cancer, skin cancer, esophageal cancer and the like.
The self-assembled supramolecular cage with a novel structure designed by the invention has the following advantages. First, the Supramolecular Coordination Complex (SCC) has a very specific and unique structure, easier to control and use than many structures such as MOFs and nanoparticles. Secondly, in SCC, the relative positions and amounts of the components are strictly fixed, especially in a 3D cage structure, porphyrin derivatives can well avoid intramolecular pi-pi accumulation and aggregation, and nanoparticles can further avoidThe pi-pi accumulation and aggregation among porphyrin derivatives are generated1O2The efficiency of (2) will be further improved. Finally, the introduction of pt (ii) into the cage structure can increase the chemotherapeutic capacity in PDT function. Thus, nanoparticles based on tetrapyridine porphyrin derivatives or tetrapyridine metalloporphyrin derivatives and pt (ii) receptors entering precise supramolecular cages can achieve higher synergistic anticancer efficiency in combination with synergistic effects of PDT and chemotherapy. The introduction of mPEG-b-PEBP and RGD-PEG-b-PEBP, the nano particles formed by the mPEG-b-PEBP and the supermolecule cage can stably and durably exist in the circulation in the organism, and have a targeting effect on cancer tissues, thereby facilitating the transportation of medicines and the treatment of cancers.
Drawings
FIG. 1 electrospray ionization mass spectrometry (ESI-MS) of PDP and ZPDP;
FIG. 2 PDP and ZPDP structure correlation spectra (2D COSY);
FIG. 3 PDP NPs and ZPDP NPs Transmission Electron Microscopy (TEM) and Dynamic Laser Scattering (DLS) studies;
FIG. 4 investigation of singlet oxygen fluorescent probe (SOSG);
FIG. 5 in vitro cellular uptake and synergy cytotoxicity studies of PDP NPs;
FIG. 6 in vivo distribution and synergistic anti-cancer mechanism studies of PDPNPs in mice with 4T1 orthotopic breast cancer;
FIG. 7 is a study of cancer cell metastasis after treatment and tumor volume and lung surface coverage.
Detailed Description
The present invention is described in further detail below by way of examples, but the embodiments of the present invention are not limited thereto.
Example 1
Synthesis of PDP
Figure BDA0001916700850000081
16.2. mu. mol of 5,10,15, 20-tetrakis (3-pyridyl) porphyrin (P) and 32.4. mu. mol of 4,4' -bis (trans-bis (triethylphosphine) (trifluoromethane) platinium) benzophenone (DP) were weighed, dissolved in dimethyl sulfoxide, reacted at 80 ℃ for 12 hours, cooled to room temperature, added with excess diethyl ether to form a precipitate, filtered, washed with diethyl ether, and then dried in vacuo to give a dark brown solid (51.5mg, 96.4%).
1H NMR(ppm):9.60(m,8H,Py-Ha),9.33(d,8H,Py-Hb),8.94 (m,24H,Py-Hd,P-He),8.28(t,8H,Py-Hc),7.63(d,16H,Ph-H2), 7.20-7.30(m,16H,Ph-H1),-3.13(s,4H,P-Hf).
31P{1H}NMR:=12.64ppm
ESI-MS (FIG. 1 (A)): 6600Da
2D correlation Spectrum (2D COSY) (FIG. 2(A))
Example 2
ZPDP synthesis
Figure BDA0001916700850000091
14.7. mu. mol of zinc 5,10,15, 20-tetrakis (4-pyridyl) -21H, 23H-porphyrin (ZP) and 29.4. mu. mol of 4,4' -bis (trans-bis (triethylphosphine) (trifluoromethane) platinum) benzophenone (DP) were weighed, dissolved in dimethyl sulfoxide, reacted at 80 ℃ for 12 hours, cooled to room temperature, added with excess diethyl ether to form a precipitate, filtered, washed with diethyl ether, and then dried under vacuum to obtain a purple black solid (47.0mg, 95.1%).
1H NMR:9.60(s,8H,Py-Ha),9.29-9.30(d,8H,Py-Hb),8.84-8.87 (m,24H,Py-Hd,ZP-He),8.22-8.25(t,8H,Py-Hc),7.64(d,16H,Ph-H2), 7.21-7.32(m,16H,Ph-H1)
31P{1H}NMR:12.73ppm
ESI-MS (FIG. 1 (B)): 6732Da
2D correlation Spectrum (2D COSY) (FIG. 2(B))
Example 3
Preparation of supramolecular cage-loaded nanoparticles
Preparation of supermolecular cage loaded nano-particle PDP NPs and ZPDP NPs
5mL of an acetone solution containing 6.0mg of supramolecular cages (PNP or ZNP), mPEG-b-PEBP (25.0mg) and RGD-PEG-b-PEBP (5.0mg) was added dropwise to 20mL of Milli-Q water with vigorous stirring and dried in vacuo. After 5 minutes of sonication, a well-dispersed nanoparticle suspension was obtained.
The morphology and size of the nanoparticles were studied by Transmission Electron Microscopy (TEM) and Dynamic Laser Scattering (DLS), see fig. 3, and it can be seen that spherical PDP NPs with a diameter ranging from 30 to 90nm were observed in the dry state as shown in fig. 3A. Slightly larger hydrophilic diameters were recorded from DLS (fig. 3B) due to hydration of NP. An increase in diameter from 35.7nm to 61.8nm was observed after loading the PDP cage into the NP, indicating that the amphiphilic polymer successfully encapsulated the cage. In addition, the zeta potential of NP formed from PEBP-b-PEG-RGD increased from-46.9 mV to-5.4 mV after encapsulation of PDP by PE (FIG. 3C), indicating that charge-charge interaction is one of the major driving forces. In addition to hydrophobic interactions, the formation of self-assembly. The obtained PDP NPs can stably exist in Phosphate Buffered Saline (PBS) containing 10% fetal calf serum at 37 ℃ for 48 hours, and the PDP NPs are proved to have good colloidal stability in a biological environment.
Example 4
Singlet oxygen fluorescent probe (SOSG) for quantification of laser irradiation (638nm, 0.5W/cm)2) Reactive Oxygen Species (ROS) generation of the lower nanomaterial. As shown in FIG. 4A, the fluorescence intensities of P and ZP without forming a supramolecular cage structure are similar and are about 2500, and after forming the supramolecular cage structure, the fluorescence intensity of ZP NPs at 532nm is observed to be about 10000, and the intensity is increased by 4 times, which is mainly because the special supramolecular cage structure enables the porphyrin derivative to well avoid pi-pi accumulation and aggregation, and the porphyrin derivative is produced1O2The efficiency of (2) will be improved.
However, the fluorescence intensity at 532nm was observed to be lower for both NPs and ZP NPs than for the supramolecular cage-loaded nanoparticles in example 3, and the quenching of ROS was mainly due to their poor solubility and small amount of pi-pi stacking interactions. In sharp contrast, the SOSG fluorescence showed a 4-fold enhancement with an intensity close to 40000 after the introduction of cage structures (PDP NPs) under the same conditions, confirming their high photosensitivity effect (fig. 4A). The ROS produced by PDP NPs is high in yield, due to the supramolecular cage structure, effectively prevents aggregation of P molecules, while heavy atoms (Pt) are incorporated into the cage, facilitating the conversion of molecular oxygen into singlet oxygen. It is worth to be noted that the generation of ROS is low under the laser irradiation of this band because the introduction of Zn ions in ZPDNPs causes the Q-band to move (FIG. 4B). Since PDP NPs have a larger absorption wavelength and a better penetration ability into cell tissues, they are preferable as drugs for in vitro and in vivo treatment
Example 5
Cytotoxicity test
αvβ3Integrin receptor is one of the most extensively studied targets for tumor cell overexpression, and has been selected in this study against mouse triple negative breast cancer cells (4T 1). Cyclo (Arg-Gly-Asp-D-Phe-Lys) (cRGD) has been selected as a specific targeting moiety because it can selectively bind α with high affinityvβ3Receptor-mediated cellular uptake PDPNPs were quantitatively confirmed by flow cytometry and inductively coupled plasma mass spectrometry (ICP-MS), with faster and higher cellular uptake of PDP NPs demonstrating cRGD by αvβ3Integrin overexpresses 4T1 cells instead of the counterpart without cRGD (fig. 5A, 5B).
Chemotherapy and PDT were placed in a single experiment, and PDP NPs were effective at eliciting cytotoxicity upon irradiation, as quantified using the MTT assay (fig. 5C). Half maximal inhibitory concentrations (IC50) for DP and DP + L were determined to be 7.85 ± 0.8 μ M and 8.15 ± 0.7 μ M, higher than commercially available cisplatin (IC50 ═ 4.39 ± 0.7 μ M, data not shown). This may be due to the instability of DP in aqueous environments, resulting in reduced toxicity. The results also demonstrate that laser irradiation does not enhance the toxicity of DP. In the absence of light irradiation, the IC50 value for the PDP NPs was reduced to 3.56. + -. 0.6. mu.M. The reason is that DP is stable in supramolecular cages and can induce cytotoxicity after cellular uptake. Light irradiation (638nm, 2 min, 0.2W/cm)2) Determination of the IC50 value for the P NPs 0.51. + -. 0.12. mu.M without laser irradiation, P NPs showed no significant cytotoxicity. The most exciting results observed from the PDP NPs + L group showed post-irradiation IC50 values of 87.4 + -8.7 nM, well below chemotherapy and PDT alone. The enhanced cytotoxicity of PDP NPs + L results from the excellent synergistic effects of chemotherapy and PDT, with a Combination Index (CI) well below 1(CI ═ 0.17). The Phototoxicity Index (PI) calculated as the ratio of IC50 values in the dark and exposed to light irradiation is as high as 40.7. The high PI of PDPNPs indicates that they are excellent photosensitizing drugs because they have low toxicity in the dark but high cytotoxicity upon light irradiation, which is very important for PDT. Notably, the molar ratio of DP and P NPs is 2: the mixture of 1 showed no significant improvement in cytotoxicity (IC50 ═ 0.49. + -. 0.08. mu.M based on the molar amount of Pt) upon irradiation, underscoring that the formation of supramolecular cages plays a significant role in synergistic photochemical therapy.
The Annexin V-FITC/PI assay was used to differentiate between viable (FITC-/PI-), early apoptotic (FITC +/PI-), late apoptotic (FITC-/PI +) and necrotic (FITC +/PI +) cell counts (FIG. 5D). For cells treated with PDP NPs in the dark, the populations of early apoptotic, late apoptotic and necrotic cells were 0.80%, 23.95% and 8.43%, respectively, indicating that chemotherapy alone had a relatively low ability to induce apoptosis. After PDT (P NPs + L) treatment, apoptotic late cells increased to 61.1% and necrotic cells remained 6.81%. Exciting, the marked increase in necrotic cells to 42.71% upon combined chemotherapy confirmed the potential use of PDP NPs in cancer co-therapy.
Fluorescein diacetate/propidium iodide (FDA/PI) co-staining was used to distinguish live from dead cells under the fluorescence image. As shown in fig. 5E, negligible cell death was induced due to very low concentrations, light, DP and PDP NPs. A significant increase in cell death was observed for cells incubated with PDP NPs following exposure compared to these controls. The percentage of dead cells rapidly increased with the increase of the laser irradiation time, confirming that the PDP NPs have an excellent PDT effect.
Example 6
In vivo experiments
PDP NPs have suitabilityAnd can effectively accumulate at the tumor site, resulting in better therapeutic efficacy and lower side effects. The long circulation of PDP NPs in the bloodstream is essential for successful targeted delivery and effective treatment, and is achieved by "masking" them by surface grafting a "protective" PEG shell, thereby preventing clearance by the reticuloendothelial system (RES). To investigate their pharmacokinetics (pKa), PDP NPs and free DP were injected intravenously (iv) into mice at a dose of 2mg/kg platinum. Various time points after injection of the blood sample were collected. By quantifying blood platinum concentrations using ICP-MS, the blood circulation half-life of PDP NPs was calculated to be 2.39 ± 0.4h, which is 4.5 times DP (fig. 6A). Approximately 11.4% of the Injected Dose (ID) of PDP NPs remained in the plasma at 24 hours. Although DP was almost completely eliminated from the bloodstream 8 hours after injection. The area under the curve (AUC) of the PDP NPs increased significantly to 149. mu.gmL-1h, specific DP (7.86. mu.gmL)-1h) 18.9 times greater, indicating a greatly extended cycle time due to the EPR effect and active localization.
The distribution of PDP NPs in the organism was quantitatively evaluated by ICP-MS analysis of platinum content in different organs (FIG. 6B). The biodistribution evaluation showed that PDP NPs were efficiently accumulated in tumors, with approximately 2.24. + -. 0.31. mu.g/g tissue concentration, significantly higher than DP (0.39. + -. 0.05. mu.g/g tissue concentration) 24 hours after administration. Due to RES capture and metabolism, fairly high concentrations of PDP NPs can be observed in the liver. DP showed a significantly different distribution in major organs compared to PDP NPs mainly treated by the kidney. These findings clearly demonstrate that PDP NPs are easier to use for tumor uptake than small molecule drugs and are beneficial for increasing their anti-tumor efficacy while reducing adverse effects on normal tissues.
In vivo synergistic photochemotherapy
Based on excellent in vitro combinatorial cytotoxicity, biostable cycling and reasonable biodistribution of PDP NPs, a single treatment evaluation of the in vivo anti-tumor efficacy of PDP NPs was performed on highly invasive 4T1 orthotopic breast cancer bearing mice. Tumor mice were randomly divided into six groups, and when the tumor volume reached 130mm3On the left and right, respectively administering physiological saline, cisplatin, DP and PNPs +L, PDP NPs or PDP NPs + L (n ═ 6). Selecting the material with relatively low power density (0.5W/cm) at 24 hours of intravenous injection2) And optimized irradiation duration (6 minutes) for light-based treatment (laser alone, no significant skin burn). The tumors of mice dosed with saline grew rapidly (fig. 6C), increasing the tumor volume by 13.0-fold within 21 days compared to its original volume. Cisplatin, DP or PDP NPs slightly reduced tumor growth due to the limited efficacy of chemotherapy alone, which was not effective enough to inhibit tumor growth. Although PDT significantly reduced tumor size within the first 6 days using NPs and laser irradiation, tumor growth then resumed rapidly because the ROS production of the NPs was relatively low and virtually impossible to eliminate all cancer cells. It is worth noting that PDP NPs + L showed the highest antitumor efficiency in these groups and almost completely eradicated tumors without recurrence during the experiment (5 out of 6 mice total). Tumors were excised 21 days post treatment and tumor weights were evaluated (fig. 6D). The tumor growth inhibition ratio of the light-irradiated group treated with PDP NPs was 98.4%, while the tumor growth inhibition ratios of PDP NPs, DP, P NPs + L and cisplatin were 51.5%, 37.3%, 48.3%, 60.5% and 31.2%, respectively. These results clearly demonstrate the synergistic anti-tumor efficacy between PDP NPs mediated chemotherapy and laser irradiation activated PDT to completely ablate the tumor without recurrence after a single treatment.
Immunohistochemical analysis highly supported the results discussed above regarding tumor inhibition (fig. 6E). Hematoxylin and eosin (H & E) staining showed that tumors receiving chemotherapy (DP, cisplatin and PDP NPs) or PDT (P NPs + L) showed different degrees of tumor regression compared to the saline treated group, indicating that these administrations had different degrees of anti-tumor effect. After a minimal treatment with PDP NPs + L, we did not find tumor cells, indicating successful destruction of the tumor. Ki67 positive immunohistochemical staining further showed minimal proliferation in the tumor area from the light chemotherapy group (PDP NPs + L).
Lung tissue was excised and observed after sacrifice (fig. 7A), and the number of metastatic tumor nodules and percent tumor coverage on the lung surface were further analyzed to assess the anti-metastatic effect of these treatments. Mean metastatic tumor nodule counts for mice treated with saline, PDP NPs, DP, P NPs + L and cisplatin, respectively, were 5.67,2.17,3.67,1.17 and 4.2 (fig. 7B). The percentage of tumor coverage on lung surfaces was calculated as 9.33,2.23,4.37,0.83 and 5.51% for mice treated with saline, PDP NPs, DP, P NPs + L and cisplatin, respectively (fig. 7C), demonstrating that limited anti-metastatic effects could be achieved by chemotherapy or PDT alone. It is worth noting that only one tumor nodule was visible from the six lungs of the mice receiving the combination treatment, and the average tumor coverage over the lung surface was only 0.07% of the age of the lungs, indicating that photochemotherapy had excellent anti-metastatic efficacy.
The systemic toxicity of nanomedicine was carefully assessed using body weight size and survival as indications. For DP and cisplatin administration, the body weight of mice decreased within the first week due to their systemic toxicity and associated side effects (fig. 6G). Histological analysis provided insight into DP or cisplatin-induced systemic toxicity. Some degree of lung and liver damage was observed in DP mice in the cisplatin group. However, no significant changes in body weight and histology were observed in mice receiving other treatments, which means that PDP NPs had minimal systemic toxicity to mice after a single injection. Median survival rates were calculated for mice treated with saline, PDP NPs, DP, P NPs + L and cisplatin for 36, 49, 42, 54 and 42 days, respectively, while phototherapy greatly extended survival time of mice. There was only one death over 75 days (fig. 6F). These results indicate that the combination of chemotherapy and PDT in tumor treatment effectively extends their life span without significant side effects.
The above embodiments are preferred embodiments of the present invention, but the present invention is not limited to the embodiments, and any other changes, modifications, combinations, substitutions and simplifications which do not depart from the spirit and principle of the present invention should be construed as equivalents and are included in the scope of the present invention.

Claims (7)

1. A method for preparing supramolecular cage-loaded nanoparticles, comprising: the nanoparticles are used for photodynamic therapy and chemotherapy of cancer, the method comprising:
s1: preparing a supermolecular cage required by the nano-particles;
s2: dropwise adding the supermolecule cage and the solution of mPEG-b-PEBP and RGD-PEG-b-PEBP into Milli-Q water, violently stirring, drying in vacuum, and carrying out ultrasonic treatment for five minutes to obtain a well-dispersed nano-particle suspension, and finally forming nano-particles;
wherein the supramolecular cage comprises a tetrapyridine porphyrin derivative or a tetrapyridine metalloporphyrin derivative and a Pt (II) receptor, and the structure of the supramolecular cage is shown as a formula (I):
Figure FDA0002593788760000011
wherein the content of the first and second substances,
Figure FDA0002593788760000012
is a tetrapyridylporphyrin derivative, the structure of which is shown in formula (II):
Figure FDA0002593788760000013
wherein:
r1 is H, alkyl;
r2 is H, F, Cl, Br, I, alkyl;
Figure FDA0002593788760000021
is a tetrapyridine metalloporphyrin derivative, and the structure of the tetrapyridine metalloporphyrin derivative is shown as a formula (III):
Figure FDA0002593788760000022
wherein:
r1 is H, alkyl;
r2 is H, F, Cl, Br, I, alkyl;
m is Zn, Co, Mn, Ce, Fe, Mg, Hg, Ru, Cu, Zr, Rh, Pt, Sn, Tl, Al, Pt, Pd, Ir, Sb, V, Ti, Hf, Au, Cr, Ag, In, Tb, Gd, Er, Yb, Lu, Dy, Nd, Eu, Pr, Ho, Tm, La, Sm;
Figure FDA0002593788760000023
is a Pt (II) receptor having the structure of formula (IV):
Figure FDA0002593788760000024
2. the method for preparing supramolecular cage-loaded nanoparticles as claimed in claim 1, wherein the mass ratio of mPEG-b-PEBP to RGD-PEG-b-PEBP as the carrier material is 4-6: 1.
3. A nanoparticle comprising a supramolecular cage, comprising: the nanoparticle is obtained by the method for preparing the supermolecular cage loaded nanoparticle according to any one of claims 1 to 2.
4. The nanoparticle of claim 3, wherein: the particle diameter of the particles is 20-120 nanometers.
5. An injectable or orally administrable composition comprising the nanoparticle of claim 3 and a pharmaceutically acceptable carrier.
6. Use of nanoparticles according to claim 3 for the preparation of a medicament for use in a method of killing cancer cells using photodynamic therapy and chemotherapy.
7. Use of nanoparticles according to claim 3 for the preparation of a medicament for use in a method of killing cancer cells using photodynamic therapy and chemotherapy, comprising contacting said cancer cells with nanoparticles and irradiating the nanoparticles with a therapeutically effective amount of light to induce the production of singlet oxygen by said self-assembled supramolecular cage; meanwhile, the composition has chemotherapy ability.
CN201811575728.1A 2018-12-22 2018-12-22 Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof Active CN109481696B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811575728.1A CN109481696B (en) 2018-12-22 2018-12-22 Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811575728.1A CN109481696B (en) 2018-12-22 2018-12-22 Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof

Publications (2)

Publication Number Publication Date
CN109481696A CN109481696A (en) 2019-03-19
CN109481696B true CN109481696B (en) 2020-10-02

Family

ID=65711369

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811575728.1A Active CN109481696B (en) 2018-12-22 2018-12-22 Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof

Country Status (1)

Country Link
CN (1) CN109481696B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110548141A (en) * 2019-09-04 2019-12-10 广东医科大学 Preparation method and application of titanyl porphyrin complex-folic acid liposome nanoparticles
CN111154115B (en) * 2020-01-03 2021-09-21 大连理工大学 Preparation method and application of binuclear Ir (III) metal-organic supermolecular cage-like compound
CN115054689B (en) * 2022-06-13 2023-06-30 哈尔滨工程大学 Preparation method of platinum-tin bimetallic nano particles with near infrared light enhanced catalytic performance

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1316558A1 (en) * 2000-09-08 2003-06-04 San-Ei Gen F.F.I., Inc. Tetraphenylbacteriochlorin derivatives and compositions containing the same
CN101102766A (en) * 2004-11-16 2008-01-09 科英布拉大学 Novel derivatives of porphyrin, particularly chlorins and/orbacteriochlorins, and uses thereof in photodynamic therapy
CN101857597A (en) * 2010-05-27 2010-10-13 中国医学科学院生物医学工程研究所 Diethylene triamine pentaacetic acid or ethylene diamine tetraacetic acid or amine triacetic acid modified porphyrin, preparation method and application thereof
CN102408452A (en) * 2011-12-13 2012-04-11 中山大学 Tetrapyridylporphine bridged crossed tetra-palladium complexes, and preparation method and antitumor activity thereof
WO2013142886A1 (en) * 2012-03-30 2013-10-03 Joanneum Research Forschungsgesellschaft Mbh Opto-chemical sensor
CN103923125A (en) * 2014-03-31 2014-07-16 哈尔滨工业大学 Water-soluble porphyrin photosensitizer and preparation method thereof
CN106267203A (en) * 2016-09-18 2017-01-04 河南大学 A kind of porphyrin/SiO being applied to optical dynamic therapy2the controllable method for preparing of assemble nanometer composite altogether

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1316558A1 (en) * 2000-09-08 2003-06-04 San-Ei Gen F.F.I., Inc. Tetraphenylbacteriochlorin derivatives and compositions containing the same
CN101102766A (en) * 2004-11-16 2008-01-09 科英布拉大学 Novel derivatives of porphyrin, particularly chlorins and/orbacteriochlorins, and uses thereof in photodynamic therapy
CN101857597A (en) * 2010-05-27 2010-10-13 中国医学科学院生物医学工程研究所 Diethylene triamine pentaacetic acid or ethylene diamine tetraacetic acid or amine triacetic acid modified porphyrin, preparation method and application thereof
CN102408452A (en) * 2011-12-13 2012-04-11 中山大学 Tetrapyridylporphine bridged crossed tetra-palladium complexes, and preparation method and antitumor activity thereof
WO2013142886A1 (en) * 2012-03-30 2013-10-03 Joanneum Research Forschungsgesellschaft Mbh Opto-chemical sensor
CN103923125A (en) * 2014-03-31 2014-07-16 哈尔滨工业大学 Water-soluble porphyrin photosensitizer and preparation method thereof
CN106267203A (en) * 2016-09-18 2017-01-04 河南大学 A kind of porphyrin/SiO being applied to optical dynamic therapy2the controllable method for preparing of assemble nanometer composite altogether

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
Combinatorial photochemotherapy on liver cancer stem cells with organoplatinum(II) metallacage-based nanoparticles;Beng Yang et al;《J. Mater. Chem. B》;20190829(第7期);第6476-6487页 *
Electrochemical and Electrocatalytic Properties of Tetra(pyridyl 4",3")porphin Series Derivatives;N. M. Berezina et al;《Russian Journal of Electrochemistry》;20111231;第47卷(第1期);第46-51页 *
Improving Paclitaxel Delivery: In Vitro and In Vivo Characterization of PEGylated Polyphosphoester-Based Nanocarriers;Fuwu Zhang et al;《JACS》;20150128(第137期);第2056-2066页 *
Investigation of excited singlet state absorption and intersystem crossing mechanism of isomeric meso-tetra(pyridyl)porphyrins containing peripheral polypyridyl platinum(II) complexes;Leandro H.Z. Cocca et al;《Chemical Physics Letters》;20180727(第708期);第1-10页 *
Molecular Architecture via Coordination: Self-Assembly of Nanoscale Hexagonal Metallodendrimers with Designed Building Blocks;Hai-Bo Yang et al;《JACS》;20060718(第128期);第10014-10015页 *
Protein adsorption is required for stealth effect of poly(ethylene glycol)- and poly(phosphoester)-coated nanocarriers;Susanne Schottler et al;《NATURE NANOTECHNOLOGY》;20160215;第1-6页 *
Self-Assembly of Chiral Metallacycles and Metallacages from a Directionally Adaptable BINOL-Derived Donor;Yang Ye, et al;《JACS》;20150902(第137期);第11896-11899页 *
Self-Assembly of Porphyrin-Containing Metalla-Assemblies and Cancer Photodynamic Therapy;Xin Jiang, et al;《Inorg. Chem.》;20200121;第1-9页 *
Self-Assembly of Supramolecular Fractals from Generation 1 to 5;Lei Wang et al;《JACS》;20181005(第140期);第14087-14096页 *
Synthesis and photophysical studies of self-assembled multicomponent supramolecular coordination prisms bearing porphyrin faces;Yanhui Shi et al;《PNAS》;20140514;第111卷(第26期);第9390-9395页 *
金属铂(II)-卟啉二聚体配合物的分子内π-π相互作用;苏祎伟,等;《无机化学学报》;20131231;第29卷(第12期);第2695-2703页 *

Also Published As

Publication number Publication date
CN109481696A (en) 2019-03-19

Similar Documents

Publication Publication Date Title
Yu et al. Recent advances in delivery of photosensitive metal-based drugs
Li et al. Nanoscale metal‐organic frameworks: synthesis, biocompatibility, imaging applications, and thermal and dynamic therapy of tumors
Zheng et al. The recent progress on metal–organic frameworks for phototherapy
Ding et al. Nanoscale MOFs: From synthesis to drug delivery and theranostics applications
Rojas et al. Metal-organic frameworks: A novel platform for combined advanced therapies
Hu et al. Potentiating photodynamic therapy of ICG-loaded nanoparticles by depleting GSH with PEITC
Yang et al. Mesoporous silica nanorods intrinsically doped with photosensitizers as a multifunctional drug carrier for combination therapy of cancer
CN109678905B (en) Coordination-driven self-assembled supramolecular cage, preparation method and application thereof
Yan et al. Versatile nanoplatforms with enhanced photodynamic therapy: designs and applications
Chen et al. Current and promising applications of Hf (IV)-based MOFs in clinical cancer therapy
Bhana et al. Photosensitizer-loaded gold nanorods for near infrared photodynamic and photothermal cancer therapy
CN109481696B (en) Medicine for cancer photodynamic therapy and chemotherapy and preparation method thereof
Hang et al. Controllable photodynamic performance via an acidic microenvironment based on two-dimensional metal-organic frameworks for photodynamic therapy
Ding et al. Photo-enhanced chemotherapy performance in bladder cancer treatment via albumin coated AIE aggregates
Valenzuela et al. Strategies and applications of covalent organic frameworks as promising nanoplatforms in cancer therapy
Bao et al. Nanoscale metal–organic framework composites for phototherapy and synergistic therapy of cancer
Nosrati et al. Magnetite and bismuth sulfide Janus heterostructures as radiosensitizers for in vivo enhanced radiotherapy in breast cancer
Yang et al. Core-shell structured nanoparticles for photodynamic therapy-based cancer treatment and related imaging
Feng et al. Recent insight into functional crystalline porous frameworks for cancer photodynamic therapy
Liu et al. Recent development of amorphous metal coordination polymers for cancer therapy
Gandosio et al. Recent approaches towards the development of ru (II) polypyridyl complexes for anticancer photodynamic therapy
Yin et al. Hypoxia-alleviated sonodynamic therapy based on a hybrid protein oxygen carrier to enhance tumor inhibition
Zhong et al. Recent Progress in Carrier‐Free Nanomedicine for Tumor Phototherapy
Chen et al. Platinum (IV) Complex-Loaded nanoparticles with photosensitive activity for cancer therapy
Hu et al. Application of nanosonosensitizer materials in cancer sono-dynamic therapy

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
GR01 Patent grant
GR01 Patent grant