Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
ACS Nano ; 18(23): 14877-14892, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38809421

ABSTRACT

Effective anticancer immunity depends on properly activating multiple stepwise events in the cancer-immunity cycle. An immunologically "cold" tumor microenvironment (TME) engenders immune evasion and refractoriness to conventional checkpoint blockade immunotherapy. Here, we combine nanoparticle formulations and an in situ formed hydrogel scaffold to treat accessible tumors locally and to stimulate systemic immunity against metastatic tumor lesions. The nanoparticles encapsulate poly(ε-caprolactone)-derived cytotoxic chemotherapy and adjuvant of Toll-like receptor 7/8 through a reactive oxygen species (ROS)-cleavable linker that can be self-activated by the coassembled neighboring photosensitizer following near-infrared (NIR) laser irradiation. Further development results in syringeable, NIR light-responsive, and immunogenic hydrogel (iGEL) that can be implanted peritumorally and deposited into the tumor surgical bed. Upon NIR laser irradiation, the generated ROS induces iGEL degradation and bond cleavage in the polymer-drug conjugates, triggering the immunogenic cell death cascade in cancer cells and spontaneously releasing encapsulated agents to rewire the cancer-immunity cycle. Notably, upon application in multiple preclinical models of melanoma and triple-negative breast cancer, which are aggressive and refractory to conventional immunotherapy, iGEL induces durable remission of established tumors, extends postsurgical tumor-free survival, and inhibits metastatic burden. The result of this study is a locally administrable immunogenic hydrogel for triggering host systemic immunity to improve immunotherapeutic efficacy with minimal off-target side effects.


Subject(s)
Hydrogels , Infrared Rays , Animals , Mice , Hydrogels/chemistry , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Nanoparticles/chemistry , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Immunotherapy , Female , Polyesters/chemistry
2.
EBioMedicine ; 92: 104594, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37167784

ABSTRACT

BACKGROUND: Long-term treatment with immunosuppressants is necessary to attenuate allograft rejection following organ transplantation (OT). Consequently, the overall survival of OT recipients with malignancies has been substantially compromised by tumour recurrence. Rapamycin (RAPA) is a clinically approved immunosuppressive agent with antitumour activity that is considered beneficial in preventing posttransplant tumour recurrence. However, the clinical outcome of RAPA is impeded by acquired drug resistance and its poor oral bioavailability. METHODS: A nanotherapeutic strategy was developed by supramolecular assembly of RAPA into a polymer cytotoxic 7-ethyl-10-hydroxycamptothecin (SN38) prodrug nanoparticle (termed SRNP) for simultaneous codelivery of cytotoxic/immunosuppressive agents. Cell-based experiments were used to evaluate the cytotoxicity of SRNPs against hepatocellular carcinoma (HCC). The therapeutic efficacy of SRNPs was evaluated in multiple preclinical models including an orthotopic HCC mouse model, an orthotopic liver transplantation (OLT) rat model and a clinically relevant cancer-transplant model to examine its antitumour and immunosuppressive activity. FINDINGS: The combination of SN38 with RAPA resulted in synergetic effects against HCC cells and alleviated RAPA resistance by abrogating Akt/mTOR signalling activation. SRNPs exhibited potent antitumour efficiency in the orthotopic HCC model while substantially prolonging the survival of allografts in the OLT model. In the cancer-transplant model that simultaneously bears tumour xenografts and skin allografts, SRNPs not only effectively inhibited tumour growth but also attenuated allograft damage. INTERPRETATION: The nanotherapy presented here had enhanced efficacy against tumours and maintained satisfactory immunosuppressive activity and thus has great potential to improve the survival outcomes of patients with a high risk of tumour recurrence following OT. FUNDING: This work was supported by the National Natural Science Foundation of China (32171368 and 31671019), the Zhejiang Provincial Natural Science Foundation of China (LZ21H180001), the Zhejiang Province Preeminence Youth Fund (LR19H160002), and the Jinan Provincial Laboratory Research Project of Microecological Biomedicine (JNL-2022039c).


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Liver Transplantation , Mice , Humans , Rats , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/etiology , Neoplasm Recurrence, Local/drug therapy , Liver Neoplasms/drug therapy , Immunosuppressive Agents/adverse effects , Sirolimus/pharmacology , Sirolimus/therapeutic use , Liver Transplantation/adverse effects , Antineoplastic Agents/therapeutic use
3.
Adv Sci (Weinh) ; 10(15): e2204890, 2023 05.
Article in English | MEDLINE | ID: mdl-37017572

ABSTRACT

Immune checkpoint blockade (ICB) therapies have had a tremendous impact on cancer therapy. However, most patients harbor a poorly immunogenic tumor microenvironment (TME), presenting overwhelming de novo refractoriness to ICB inhibitors. To address these challenges, combinatorial regimens that employ chemotherapies and immunostimulatory agents are urgently needed. Here, a combination chemoimmunotherapeutic nanosystem consisting of a polymeric monoconjugated gemcitabine (GEM) prodrug nanoparticle decorated with an anti-programmed cell death-ligand 1 (PD-L1) antibody (αPD-L1) on the surface and a stimulator of interferon genes (STING) agonist encapsulated inside is developed. Treatment with GEM nanoparticles upregulates PD-L1 expression in ICB-refractory tumors, resulting in augmented intratumor drug delivery in vivo and synergistic antitumor efficacy via activation of intratumor CD8+ T cell responses. Integration of a STING agonist into the αPD-L1-decorated GEM nanoparticles further improves response rates by transforming low-immunogenic tumors into inflamed tumors. Systemically administered triple-combination nanovesicles induce robust antitumor immunity, resulting in durable regression of established large tumors and a reduction in the metastatic burden, coincident with immunological memory against tumor rechallenge in multiple murine tumor models. These findings provide a design rationale for synchronizing STING agonists, PD-L1 antibodies, and chemotherapeutic prodrugs to generate a chemoimmunotherapeutic effect in treating ICB-nonresponsive tumors.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Mice , Animals , B7-H1 Antigen/metabolism , Neoplasms/drug therapy , CD8-Positive T-Lymphocytes , Immunotherapy/methods , Gemcitabine
4.
Proc Natl Acad Sci U S A ; 120(8): e2210385120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36787350

ABSTRACT

Immunotherapy holds great promise for the treatment of aggressive and metastatic cancers; however, currently available immunotherapeutics, such as immune checkpoint blockade, benefit only a small subset of patients. A photoactivatable toll-like receptor 7/8 (TLR7/8) nanoagonist (PNA) system that imparts near-infrared (NIR) light-induced immunogenic cell death (ICD) in dying tumor cells in synchrony with the spontaneous release of a potent immunoadjuvant is developed here. The PNA consists of polymer-derived proimmunoadjuvants ligated via a reactive oxygen species (ROS)-cleavable linker and polymer-derived photosensitizers, which are further encapsulated in amphiphilic matrices for systemic injection. In particular, conjugation of the TLR7/8 agonist resiquimod to biodegradable macromolecular moieties with different molecular weights enabled pharmacokinetic tuning of small-molecule agonists and optimized delivery efficiency in mice. Upon NIR photoirradiation, PNA effectively generated ROS not only to ablate tumors and induce the ICD cascade but also to trigger the on-demand release of TLR agonists. In several preclinical cancer models, intravenous PNA administration followed by NIR tumor irradiation resulted in remarkable tumor regression and suppressed postsurgical tumor recurrence and metastasis. Furthermore, this treatment profoundly shifted the tumor immune landscape to a tumoricidal one, eliciting robust tumor-specific T cell priming in vivo. This work highlights a simple and cost-effective approach to generate in situ cancer vaccines for synergistic photodynamic immunotherapy of metastatic cancers.


Subject(s)
Neoplasms , Toll-Like Receptor 7 , Animals , Mice , Toll-Like Receptor 7/agonists , Reactive Oxygen Species , Immunotherapy/methods , Neoplasms/therapy , Adjuvants, Immunologic , Polymers/chemistry , Vaccination , Cell Line, Tumor
5.
Bioact Mater ; 20: 449-462, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35765468

ABSTRACT

The recent remarkable success and safety of mRNA lipid nanoparticle technology for producing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines has stimulated intensive efforts to expand nanoparticle strategies to treat various diseases. Numerous synthetic nanoparticles have been developed for pharmaceutical delivery and cancer treatment. However, only a limited number of nanotherapies have enter clinical trials or are clinically approved. Systemically administered nanotherapies are likely to be sequestered by host mononuclear phagocyte system (MPS), resulting in suboptimal pharmacokinetics and insufficient drug concentrations in tumors. Bioinspired drug-delivery formulations have emerged as an alternative approach to evade the MPS and show potential to improve drug therapeutic efficacy. Here we developed a biodegradable polymer-conjugated camptothecin prodrug encapsulated in the plasma membrane of lipopolysaccharide-stimulated macrophages. Polymer conjugation revived the parent camptothecin agent (e.g., 7-ethyl-10-hydroxy-camptothecin), enabling lipid nanoparticle encapsulation. Furthermore, macrophage membrane cloaking transformed the nonadhesive lipid nanoparticles into bioadhesive nanocamptothecin, increasing the cellular uptake and tumor-tropic effects of this biomimetic therapy. When tested in a preclinical murine model of breast cancer, macrophage-camouflaged nanocamptothecin exhibited a higher level of tumor accumulation than uncoated nanoparticles. Furthermore, intravenous administration of the therapy effectively suppressed tumor growth and the metastatic burden without causing systematic toxicity. Our study describes a combinatorial strategy that uses polymeric prodrug design and cell membrane cloaking to achieve therapeutics with high efficacy and low toxicity. This approach might also be generally applicable to formulate other therapeutic candidates that are not compatible or miscible with biomimetic delivery carriers.

6.
J Adv Res ; 47: 93-103, 2023 05.
Article in English | MEDLINE | ID: mdl-35931324

ABSTRACT

INTRODUCTION: Organosilica nanoparticles (ONs), which are a new type of photoluminescent nanomaterial (PM) with excellent biocompatibility, have caught more attention in recent years. However, their applications are significantly impeded by the complicated preparation process, poor photostability, and especially aggregation-induced quenching. OBJECTIVES: The present study was aimed to design and prepare solid-state fluorescent ONs to avoid aggregation-induced quenching effect. In addition, the uses of ONs for fingerprint detection, white light-emitting diodes (WLEDs) and lysosome-targetable cellular imaging were demonstrated. METHODS: Here, for the first time, we designed and prepared novel solid-state fluorescent ultrasmall ONs with orange-emitting photoluminescence via a one-step hydrothermal method. RESULTS: The prepared solid-state fluorescent ONs could be successfully employed in fingerprint detection, WLEDs fabrication and cellular imaging. Intriguingly, the ultrasmall ONs specifically localized to lysosomes rather than other subcellular organelles across distinct cell lines, including cancer cells and noncancerous cells. CONCLUSION: Collectively, these data showed that the new ONs presented in this study could be ideal candidates for PMs in biological and photoelectric applications.


Subject(s)
Nanoparticles , Fluorescence , Cell Line , Fluorescent Dyes , Lysosomes
7.
Int J Pharm ; 607: 121017, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34416334

ABSTRACT

Drug resistance remains a major challenge in achieving cures in cancer patients. Cabazitaxel has shown the ability to overcome drug resistance induced by paclitaxel and docetaxel; however, substantially high toxicity has been observed in patients receiving this agent, which compromises its efficacy. We have previously demonstrated that a polymeric platform (termed cabazitaxel-NPs) encapsulating the oligolactide-cabazitaxel conjugate exhibits desired antitumor efficacy and improved in vivo tolerability. However, we found that upon cabazitaxel treatment, cancer cells adapted to activate Akt signaling, which potentially discounts the drug efficacy. We therefore hypothesized that combing cabazitaxel nanotherapeutics with a pan-Akt inhibitor MK-2206 would synergistically sensitize the resistant cancer. In this study, we confirmed that nanoparticle formulation reduced the systemic toxicity, with higher tolerance than solution-based free cabazitaxel agent in animals. Interestingly, the activation of Akt signaling in the resistant cancer was reversed by the addition of MK-2206. In particular, the collaboration of these two ingredients was demonstrated to maximize the efficacy in vitro and in a xenograft model bearing paclitaxel-resistant tumors. Mechanistically, Akt inhibition increased the microtubule-stabilizing effect of cabazitaxel nanomedicine. Collectively, this report introduced a binary platform composed of cytotoxic nanotherapeutics and inhibitors with certain targets to combat multidrug resistance, and such a combined regimen has the potential for the clinical treatment of patients with resistant cancer.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Humans , Nanomedicine , Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt , Taxoids/pharmacology
8.
Theranostics ; 11(12): 5713-5727, 2021.
Article in English | MEDLINE | ID: mdl-33897877

ABSTRACT

New strategies to fabricate nanomedicines with high translational capacity are urgently desired. Herein, a new class of self-assembled drug cocktails that addresses the multiple challenges of manufacturing clinically useful cancer nanomedicines was reported. Methods: With the aid of a molecular targeted agent, dasatinib (DAS), cytotoxic cabazitaxel (CTX) forms nanoassemblies (CD NAs) through one-pot process, with nearly quantitative entrapment efficiency and ultrahigh drug loading of up to 100%. Results: Surprisingly, self-assembled CD NAs show aggregation-induced emission, enabling particle trafficking and drug release in living cells. In preclinical models of human cancer, including a patient-derived melanoma xenograft, CD NAs demonstrated striking therapeutic synergy to produce a durable recession in tumor growth. Impressively, CD NAs alleviated the toxicity of the parent CTX agent and showed negligible immunotoxicity in animals. Conclusions: Overall, this approach does not require any carrier matrices, offering a scalable and cost-effective methodology to create a new generation of nanomedicines for the safe and efficient delivery of drug combinations.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib/pharmacology , Drug Carriers/chemistry , Drug Delivery Systems/methods , Drug Liberation/drug effects , Humans , Mice , Nanomedicine/methods , Taxoids/pharmacology
9.
Theranostics ; 11(9): 4137-4154, 2021.
Article in English | MEDLINE | ID: mdl-33754053

ABSTRACT

Rationale: Prodrug strategies that render the drug temporarily inactive through a cleavable linkage are able to modulate the physicochemical properties of drugs for adaptive nanoparticle (NP) formulation. Here we used cabazitaxel as a model compound to test the validity of our "balancing NP stability and specific drug activation" strategy. Methods: Cabazitaxel is conjugated to hydrophobic polylactide fragments with varying chain lengths via a self-immolation linkage, yielding polymeric prodrugs that can be reactivated by reductive agents in cells. Following a nanoprecipitation protocol, cabazitaxel prodrugs can be stably entrapped in amphiphilic polyethylene-block-polylactide matrices to form core-shell nanotherapies with augmented colloidal stability. Results: Upon cellular uptake followed by intracellular reduction, the NPs spontaneously release chemically unmodified cabazitaxel and exert high cytotoxicity. Studies with near-infrared dye-labeled NPs demonstrate that the nanodelivery of the prodrugs extends their systemic circulation, accompanied with increased drug concentrations at target tumor sites. In preclinical mouse xenograft models, including two paclitaxel-resistant xenograft models, the nanotherapy shows a remarkably higher efficacy in tumor suppression and an improved safety profile than free cabazitaxel. Conclusion: Collectively, our approach enables more effective and less toxic delivery of the cabazitaxel drug, which could be a new generalizable strategy for re-engineering other toxic and water-insoluble therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Nanoparticles/administration & dosage , Neoplasms/drug therapy , A549 Cells , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Carriers/chemistry , Drug Delivery Systems/methods , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacology , Polyesters/chemistry , Polymers/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Taxoids/chemistry , Taxoids/pharmacology
10.
Int J Pharm ; 599: 120399, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33647408

ABSTRACT

The global burden of colorectal cancer (CRC), the third most commonly diagnosed malignancy, continues to rise. Therefore, more effective and less toxic therapies are needed for CRC. CPT-11 (also called irinotecan), the standard-of-care treatment for CRC, has only had limited effects on survival outcomes. In vivo, CPT-11 must be converted to an active metabolite, SN38, to exert antitumor activity in the presence of carboxylesterases, but the conversion rate is extremely low (usually less than 8%). To fully harness the active SN38 compound, we showed here that esterification of SN38 using α-linolenic acid (LNA) generated a prodrug (termed LSN38), which can be formulated in pharmaceutically acceptable surfactants, such as polysorbate 80. Upon blending with an aqueous ethanolic solution, the mixture of LSN38/polysorbate 80 formed self-emulsifying nanomicelles (termed LSN38 NMs), enabling systemic injection. Unlike the insufficient release of active SN38 from CPT-11, drug activation from the LSN38 prodrug was quantitative and relied on esterase, which is abundant in cancerous cells. Pharmacokinetics studies revealed that polysorbate 80-based nanomicelles stably constrained the prodrug in the reservoir and prolonged blood circulation compared to CPT-11. Furthermore, LSN38 NMs showed superior therapeutic efficacy against a colorectal xenograft-bearing mouse model that failed to be treated with clinically approved CPT-11. Overall, these studies highlight the feasibility of converting a chemotherapeutic agent that is not miscible or compatible with pharmaceutical surfactants into an injectable self-emulsifying formulation. This approach could be applied to rescue other drugs or drug candidates that are abandoned in the preclinical stages due to pharmaceutical challenges.


Subject(s)
Antineoplastic Agents, Phytogenic , Colorectal Neoplasms , Prodrugs , Animals , Camptothecin , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Drug Repositioning , Esterases , Mice
11.
Chem Commun (Camb) ; 57(18): 2261-2264, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33532809

ABSTRACT

Using hydrophobic cabazitaxel as a target anticancer drug, we show that the conjugation of oligo(ethylene glycol)-oligolactide (OEG-OLA) via a self-immolative linkage induces the self-assembly of the resulting prodrug into injectable nanoparticles. The nanoparticles release chemically unmodified cabazitaxel after endocytosis in cancer cells. With the optimal conjugate, the nanotherapy not only potently induces tumor regression but also has a higher safety margin in animals than the free drug administered in its clinical formulation. Our studies highlight the design rationale that attaching a short amphiphilic oligomer to a toxic drug can convert it to a self-deliverable and safe nanotherapy.


Subject(s)
Drug Delivery Systems/methods , Melanoma/drug therapy , Nanoparticles/administration & dosage , Prodrugs/pharmacology , Taxoids/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Hydrophobic and Hydrophilic Interactions , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Nude , Nanoparticles/chemistry , Prodrugs/chemistry , Taxoids/chemistry , Xenograft Model Antitumor Assays
12.
Biomaterials ; 270: 120705, 2021 03.
Article in English | MEDLINE | ID: mdl-33581609

ABSTRACT

Colorectal cancer (CRC) is one of the most common and lethal human cancers, and the clinical outcomes remain unsatisfactory because of the lack of effective and safe therapeutic regimens. Here, we describe a practical and potent delivery approach for the human topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN38) against CRC. Injectable SN38-loaded nanoparticles are obtained through covalent ligation of the SN38 agent with oligo-ε-caprolactone (oligoCL) to form oligoCL-SN38 conjugates via an esterase-activatable linkage followed by encapsulation of these prodrugs in exogenous polymer matrices. Prodrug nanoparticles with adaptive features are sufficiently stable during blood circulation, while active drugs can be released in response to intracellular esterase. The administration of nanoparticle drugs results in durable tumor recession, and the efficacy is superior to that of the current standard-of-care therapy, CPT-11, in multiple mouse models of CRC, one of which is a chemically induced orthotopic CRC. Elucidation of the mechanism underlying these differing efficacies shows that nanoparticle delivery produces a substantial increase in the intratumoral concentration of the therapeutic agent relative to CPT-11, which contributes to improved antitumor efficacy. Finally, these nanoparticle drugs are potentially less toxic in animals than CPT-11, as evidenced by the low incidence of bloody diarrhea and attenuated colonic damage. Overall, these results demonstrate that precisely engineered therapeutic nanoparticles are capable of enhancing efficacy, addressing the risk of tumor recurrence, and increasing drug tolerance, thus deserving further investigation.


Subject(s)
Colorectal Neoplasms , Nanoparticles , Prodrugs , Animals , Camptothecin , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Esterases , Irinotecan
13.
Nanoscale Horiz ; 6(1): 33-42, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33210687

ABSTRACT

Targeted therapy using small molecular inhibitors has been developed to rewire key signaling pathways in tumor cells, but these inhibitors have had mixed success in the clinic due to their poor pharmaceutical properties and suboptimal intratumoral concentrations. Here, we developed a "self-assembling natural molecular inhibitor" strategy to test the efficacy and feasibility of the water-insoluble agent dasatinib (DAS), a tyrosine kinase inhibitor, for cancer therapy. By exploiting a facile reprecipitation protocol, the DAS inhibitor self-assembled into soluble supramolecular nanoparticles (termed sDNPs) in aqueous solution, without an exogenous excipient. This strategy is applicable for generating systemically injectable and colloid-stable therapeutic nanoparticles of hydrophobic small-molecule inhibitors. Concurrently, during this process, we observed aggregation-induced emission (AIE) of fluorescence for this self-assembled DAS, which makes sDNPs suitable for bioimaging and tracing of cellular trafficking. Notably, in an orthotopic model of breast cancer, administration of sDNPs induced a durable inhibition of primary tumors and reduced the metastatic tumor burden, significantly surpassing the effects of the free DAS inhibitor after oral delivery. In addition, low toxicity was observed for this platform, with effective avoidance of immunotoxicity. To the best of our knowledge, our studies provide the first successful demonstration of self-assembling natural molecular inhibitors with AIE and highlight the feasibility of this approach for the preparation of therapeutic nanoparticles for highly lethal human cancers and many other diseases.


Subject(s)
Breast Neoplasms/drug therapy , Dasatinib/administration & dosage , Drug Delivery Systems , Nanoparticles/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Dasatinib/therapeutic use , Female , Humans , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use
14.
J Control Release ; 328: 237-250, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32866593

ABSTRACT

Organ transplantation remains the gold standard therapeutic option for patients with end-stage organ failure. However, there have been few improvements in the management of post-transplant immunosuppression. As the long-term use of immunosuppressive agents (ISAs) may result in off-target systemic toxicity and complications, minimizing the ISA dosage while preserving the pharmacological efficacy could be a promising solution to address these challenges. Here, we present the design and application of self-assembled prodrug nanoparticles based on chemically derived mycophenolate mofetil, which further provide a hydrophobic core to noncovalently encapsulate additional ISAs such as tacrolimus. The resulting immunosuppressant cocktail nanoparticles are further refined by PEGylation with amphiphilic polymers to form colloidally stable self-assembled immunosuppressant cocktails (SAICs) that are suitable for preclinical studies. In a rat model of allogeneic orthotopic liver transplantation (OLT), administration of SAICs markedly extends graft/recipient survival, retards weight loss and attenuates allograft damage. Furthermore, SAICs significantly abolish intragraft inflammatory cell infiltration and proinflammatory cytokine profiles as well as improve liver graft function. This study demonstrates the superiority of SAICs over traditional ISAs in the treatment of allograft rejection and may support the emerging application of the SAIC platform in clinical settings.


Subject(s)
Hematopoietic Stem Cell Transplantation , Liver Transplantation , Animals , Graft Rejection/prevention & control , Graft Survival , Humans , Immunosuppressive Agents , Mycophenolic Acid , Rats , Tacrolimus
15.
Acta Biomater ; 113: 464-477, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32652227

ABSTRACT

Although some formats of nanomedicines are now available for clinical use, the translation of new nanoparticles to the clinic remains a considerable challenge. Here, we describe a simple yet cost-effective strategy that converts a toxic drug, cabazitaxel, into a safe and effective nanomedicine. The strategy involves the ligation of drug molecules via a self-immolating spacer, followed by dimerization-induced self-assembly to assemble stable nanoparticles. Self-assembled cabazitaxel dimers could be further refined by PEGylation with amphiphilic polymers suitable for preclinical studies. This protocol enables the formation of systemically injectable nanoparticles (termed SNPs) with nearly quantitative entrapment efficiencies and exceptionally high drug loading (> 86%). In healthy mice, PEGylated SNPs show a favorable safety profile, with reduced systemic toxicity and negligible immunotoxicity. In two separate mouse xenograft models of cancer, administration of SNPs produces efficient antitumor activity with durable tumor suppression during therapeutic studies. Overall, this methodology opens up a practical and expedient route for the fabrication of clinically useful nanomedicines, transforming a hydrophobic and highly toxic drug into a systemic self-deliverable nanotherapy. STATEMENT OF SIGNIFICANCE: Despite the great progress in cancer nanomedicines, clinical translation of nanomedicines still remains a considerable challenge. In this study, we designed a self-assembling nanoplatform based on cabazitaxel dimer reversibly ligated via a bioactivatable linker. This approach enabled the generation of systemically injectable nanomedicines with quantitative entrapment efficiencies and exceptionally high drug loading (> 86%), which greatly obviates concerns about excipient-associated side effects. Self-assembled dimeric cabazitaxel exhibited a higher safety profile than free cabazitaxel and negligible immunotoxicity in animals. This is a practical and expedient example how the chemical ligation of a hydrophobic and highly toxic anticancer drug can be leveraged to create a self-assembling delivery nanotherapy which preserves inherent pharmacologic efficacy while reduces in vivo systemic and immune toxicity.


Subject(s)
Nanoparticles , Prodrugs , Animals , Dimerization , Drug Delivery Systems , Mice , Oxidation-Reduction , Prodrugs/pharmacology , Therapeutic Index
16.
J Control Release ; 324: 289-302, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32442582

ABSTRACT

Despite the progress made with the recent clinical use of the anticancer compound cabazitaxel, the efficacy in patients remains unsatisfactory, largely due to the high in vivo toxicity of the agent. Therefore, strategies that achieve favorable outcomes and good safety profiles will greatly expand the repertoire of this potent agent. Here, we propose a combinatorial strategy to reform the cabazitaxel agent and the use of sequential supramolecular nanoassembly with liposomal compositions to assemble a prodrug-formulated liposome, termed lipoprodrug, for safe and effective drug delivery. Reconstructing cabazitaxel with a polyunsaturated fatty acid (i.e., docosahexaenoic acid) via a hydrolyzable ester bond confers the generated prodrug with the ability to be readily integrated into the lipid bilayer of liposomes for systemic administration. The resulting lipoprodrug scaffold showed significantly sustained drug release profiles and improved pharmacokinetics in rats as well as a reduction in systemic toxicity in vivo. Notably, the lipoprodrug outperformed free cabazitaxel in terms of in vivo therapeutic efficacy in multiple separate tumor xenograft-bearing mouse models, one of which was a patient-derived xenograft model. Surprisingly, the lipoprodrug was able to reduce tumor invasiveness and reprogram the tumor immunosuppressive microenvironment by proinflammatory macrophage polarization. Our findings validate this lipoprodrug approach as a simple yet effective strategy for transforming the highly toxic cabazitaxel agent into an efficacious nanomedicine with excellent in vivo tolerability. This approach could also be applied to rescue other drugs or drug candidates that have failed in clinical trials due to poor pharmacokinetic properties or unacceptable toxicity in patients.


Subject(s)
Antineoplastic Agents , Melanoma , Prodrugs , Animals , Heterografts , Humans , Liposomes , Mice , Nanomedicine , Rats , Tumor Microenvironment , Xenograft Model Antitumor Assays
17.
ACS Appl Mater Interfaces ; 12(1): 1707-1720, 2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31816241

ABSTRACT

Combinatorial regimens that rationally pair molecular inhibitors with standard cytotoxic chemotherapeutics are used to improve therapeutic outcomes. Simultaneously engineering these therapies within a single nanocarrier that spans cytotoxic, antiangiogenic, and anti-invasive mechanisms and that enables the delivery of unique drug combinations remains a technical challenge. In this study, we developed a simple and broadly applicable strategy in which ultrastable cytotoxic nanoparticles with an established excellent antitumor efficacy and π-rich inner core structure supramolecularly stabilized the antiangiogenic molecular inhibitor apatinib to create a synergistic drug delivery system (termed sTKI-pSN38). This small-sized nanoparticle accomplished the sequential release of both encapsulated drugs to exert antimetastatic, antivascular, and cytotoxic activities simultaneously. In xenograft models of hepatocellular carcinoma, a single intravenous administration of sTKI-pSN38 elicited robust and durable tumor reduction and suppressed metastasis to lymph nodes. Interestingly, sTKI-pSN38 treatment alleviated intratumoral hypoxia, which could contribute to impaired tumor metastasis and reduced drug resistance. Collectively, this nanotherapeutic platform offers a new strategy for cancer therapy by simply engineering a drug cocktail in conventional nanoparticles and by enabling the spatiotemporal modulation of drug release to enhance the synergy of the combined drugs.


Subject(s)
Cytotoxins/pharmacology , Drug Delivery Systems , Nanoparticles/chemistry , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cytotoxins/chemistry , Drug Liberation , Drug Synergism , Humans , Neoplasms/pathology
18.
Mol Cancer Ther ; 19(3): 822-834, 2020 03.
Article in English | MEDLINE | ID: mdl-31848296

ABSTRACT

Taxane-based chemotherapeutics are clinically available as frontline treatment regimens for cervical cancer. However, drug resistance and life-threatening toxicity impair the clinical efficacy of taxanes, so more effective and less toxic therapeutic modalities are urgently needed. Cabazitaxel has attracted increasing interest due to its potential to circumvent the drug resistance by taxanes. We previously showed that tethering docosahexaenoic acid (DHA) to cabazitaxel enabled the prodrug to self-assemble into nanoparticles in water. Despite this encouraging finding, the DHA-cabazitaxel conjugate formulation requires further optimization to enhance nanoparticle retention and tumor delivery. We here integrated this conjugate into amphiphilic poly(ethylene glycol)-block-poly(D,L-lactic acid) copolymers to assemble dCTX NPs The nanoparticle abrogated P-glycoprotein-mediated resistance in cancer cells. In a docetaxel-resistant cervical tumor xenograft-bearing mouse model, the efficacy was augmented by the nanotherapy when compared with solution-based free drugs (i.e., docetaxel and cabazitaxel). Dose intensification of dCTX NPs markedly suppressed the tumor growth in this model. Detailed studies revealed that systemic toxicity was alleviated, and MTD of dCTX NPs was at least 3 times higher than that of free cabazitaxel in animals, which may enable dose increases for clinical studies. In conclusion, the new formulation addresses essential requirements in terms of the stability, safety, and translational capacity for initiating early-phase clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Docetaxel/pharmacology , Drug Resistance, Neoplasm/drug effects , Nanoparticles/administration & dosage , Prodrugs/pharmacology , Taxoids/pharmacology , Uterine Cervical Neoplasms/drug therapy , Animals , Apoptosis , Cell Proliferation , Drug Delivery Systems , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Int J Pharm ; 574: 118879, 2020 Jan 25.
Article in English | MEDLINE | ID: mdl-31770581

ABSTRACT

Traditional approaches used for transforming hydrophobic anticancer drugs into therapeutically available nanoparticles heavily rely on the noncovalent formulation of drugs within amphiphilic copolymers. However, these nanotherapies have not yet shown the expected favorable clinical outcomes in cancer patients, presumably due to their insufficient stability. To solve this dilemma, we conceive a new class of nanotherapies assembled with polymeric prodrugs that maintain pharmacological activity while substantially alleviate the drug toxicity in animals. By exploiting methoxypoly(ethylene glycol)-block-poly(D, L-lactic acid) (mPEG-PLA) as a promoiety, cabazitaxel is tethered to the terminus of the PLA fragment via a hydrolysable ester linkage. These conjugates recapitulate the self-assembly to produce colloidal stable nanotherapies. In a xenograft model of prostate cancer, this nanotherapy shows a durable inhibition of tumor progression upon the administration of a tolerable dose. Our results suggest that a hydrophobic and highly toxic drug can be rationally converted into a pharmacologically efficient and self-deliverable nanotherapy.


Subject(s)
Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Taxoids/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Carriers/chemistry , Drug Delivery Systems/methods , Humans , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Polymers/chemistry , Prodrugs/chemistry , Prostatic Neoplasms/drug therapy , Taxoids/pharmacology
20.
ACS Appl Mater Interfaces ; 12(3): 3327-3340, 2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31872760

ABSTRACT

Effective new therapies for pancreatic ductal adenocarcinoma (PDAC) are desperately needed as the prognosis of PDAC patients is dismal and treatment remains a major challenge. Gemcitabine (GEM) is commonly used to treat PDAC; however, the clinical use of GEM has been greatly compromised by its low delivery efficacy and drug resistance. Here, we describe a very simple yet cost-effective approach that synergistically combines drug reconstitution, supramolecular nanoassembly, and tumor-specific targeting to address the multiple challenges posed by the delivery of the chemotherapeutic drug GEM. Using our developed PUFAylation technology, the GEM prodrug was able to spontaneously self-assemble into colloidal stable nanoparticles with sub-100 nm size on covalent attachment of hydrophobic linoleic acid via amide linkage. The prodrug nanoassemblies could be further refined by PEGylation and PDAC-specific peptide ligand for preclinical studies. In vitro cell-based assays showed that not only were GEM nanoparticles superior to free GEM but also the decoration with PDAC-homing peptide facilitated the intracellular uptake of nanoparticles and thereby augmented the cytotoxic activity. In two separate xenograft models of human PDAC, one of which was a patient-derived xenograft model, the administration of targeted nanoparticles resulted in marked inhibition of tumor progression as well as alleviated systemic toxicity. Together, these data unequivocally confirm that the hydrophilic and rapidly metabolized drug GEM can be feasibly transformed into a pharmacologically efficient nanomedicine through exploiting the PUFAylation technology. This strategy could also potentially be applied to rescue many other therapeutics that show unfavorable outcomes in the preclinical studies because of pharmacologic obstacles.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Nanoparticles/administration & dosage , Pancreatic Neoplasms/drug therapy , Prodrugs/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Prodrugs/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...