Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Bioeng Transl Med ; 8(2): e10429, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36925689

ABSTRACT

The majority of patients with high grade serous ovarian cancer (HGSOC) develop recurrent disease and chemotherapy resistance. To identify drug combinations that would be effective in treatment of chemotherapy resistant disease, we examined the efficacy of drug combinations that target the three antiapoptotic proteins most commonly expressed in HGSOC-BCL2, BCL-XL, and MCL1. Co-inhibition of BCL2 and BCL-XL (ABT-263) with inhibition of MCL1 (S63845) induces potent synergistic cytotoxicity in multiple HGSOC models. Since this drug combination is predicted to be toxic to patients due to the known clinical morbidities of each drug, we developed layer-by-layer nanoparticles (LbL NPs) that co-encapsulate these inhibitors in order to target HGSOC tumor cells and reduce systemic toxicities. We show that the LbL NPs can be designed to have high association with specific ovarian tumor cell types targeted in these studies, thus enabling a more selective uptake when delivered via intraperitoneal injection. Treatment with these LbL NPs displayed better potency than free drugs in vitro and resulted in near-complete elimination of solid tumor metastases of ovarian cancer xenografts. Thus, these results support the exploration of LbL NPs as a strategy to deliver potent drug combinations to recurrent HGSOC. While these findings are described for co-encapsulation of a BCL2/XL and a MCL1 inhibitor, the modular nature of LbL assembly provides flexibility in the range of therapies that can be incorporated, making LbL NPs an adaptable vehicle for delivery of additional combinations of pathway inhibitors and other oncology drugs.

2.
Acta Pharm Sin B ; 12(3): 1148-1162, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35530140

ABSTRACT

Combination of passive targeting with active targeting is a promising approach to improve the therapeutic efficacy of nanotherapy. However, most reported polymeric systems have sizes above 100 nm, which limits effective extravasation into tumors that are poorly vascularized and have dense stroma. This will, in turn, limit the overall effectiveness of the subsequent uptake by tumor cells via active targeting. In this study, we combined the passive targeting via ultra-small-sized gemcitabine (GEM)-based nanoparticles (NPs) with the active targeting provided by folic acid (FA) conjugation for enhanced dual targeted delivery to tumor cells and tumor-associated macrophages (TAMs). We developed an FA-modified prodrug carrier based on GEM (PGEM) to load doxorubicin (DOX), for co-delivery of GEM and DOX to tumors. The co-delivery system showed small particle size of ∼10 nm in diameter. The ligand-free and FA-targeted micelles showed comparable drug loading efficiency and a sustained DOX release profile. The FA-conjugated micelles effectively increased DOX uptake in cultured KB cancer cells that express a high level of folate receptor (FR), but no obvious increase was observed in 4T1.2 breast cancer cells that have a low-level expression of FR. Interestingly, in vivo, systemic delivery of FA-PGEM/DOX led to enhanced accumulation of the NPs in tumor and drastic reduction of tumor growth in a murine 4T1.2 breast cancer model. Mechanistic study showed that 4T1.2 tumor grown in mice expressed a significantly higher level of FOLR2, which was selectively expressed on TAMs. Thus, targeting of TAM may also contribute to the improved in vivo targeted delivery and therapeutic efficacy.

3.
Sci Adv ; 7(50): eabj4226, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34890233

ABSTRACT

In this study, we report a novel role of metabotropic glutamate receptor 4 (GRM4) in suppressing antitumor immunity. We revealed in three murine syngeneic tumor models (B16, MC38, and 3LL) that either genetic knockout (Grm4−/−) or pharmacological inhibition led to significant delay in tumor growth. Mechanistically, perturbation of GRM4 resulted in a strong antitumor immunity by promoting natural killer (NK), CD4+, and CD8+ T cells toward an activated, proliferative, and functional phenotype. Single-cell RNA sequencing and T cell receptor profiling further defined the clonal expansion and immune landscape changes in CD8+ T cells. We further showed that Grm4−/− intrinsically activated interferon-γ production in CD8+ T cells through cyclic adenosine 3',5'-monophosphate (cAMP)/cAMP response element binding protein­mediated pathway. Our study appears to be of clinical significance as a signature of NKhigh-GRM4low and CD8high-GRM4low correlated with improved survival in patients with melanoma. Targeting GRM4 represents a new approach for cancer immunotherapy.

4.
Molecules ; 26(5)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33668746

ABSTRACT

Cancer immunotherapies have generated some miracles in the clinic by orchestrating our immune system to combat cancer cells. However, the safety and efficacy concerns of the systemic delivery of these immunostimulatory agents has limited their application. Nanomedicine-based delivery strategies (e.g., liposomes, polymeric nanoparticles, silico, etc.) play an essential role in improving cancer immunotherapies, either by enhancing the anti-tumor immune response, or reducing their systemic adverse effects. The versatility of working with biocompatible polymers helps these polymeric nanoparticles stand out as a key carrier to improve bioavailability and achieve specific delivery at the site of action. This review provides a summary of the latest advancements in the use of polymeric micelles for cancer immunotherapy, including their application in delivering immunological checkpoint inhibitors, immunostimulatory molecules, engineered T cells, and cancer vaccines.


Subject(s)
Immunotherapy , Neoplasms/therapy , Polymers/chemistry , Animals , Drug Delivery Systems , Humans , Micelles , Nanomedicine , Nanoparticles/chemistry , Neoplasms/immunology
5.
J Leukoc Biol ; 109(2): 425-436, 2021 02.
Article in English | MEDLINE | ID: mdl-33259068

ABSTRACT

In the past decade, clinical and laboratory studies have led to important new insights into the biology of leukemia and its treatment. This review describes the progress of leukemia research in the United States in recent years. Whereas the traditional method of treatment is chemotherapy, it is nonselective and could induce systemic toxicities. Thus, in parallel with research on new chemotherapies, great emphasis has been placed on developing immunotherapies. Here, we will review the current immunotherapies available in research and development that overcome current challenges, specifically looking in the field of chimeric antigen receptor T-cell (CAR-T) therapies, checkpoint inhibitors, and antibody-drug conjugates. With about 100 clinical trials for CAR-T therapies and 30 in checkpoint inhibitors for leukemia treatment, scientists are trying to make these technologies cheaper, faster, and more feasible. Further describing the delivery of these therapeutics, we look at the current progress, clinical, and preclinical status of nano-based medicines such as liposomes, polymeric micelles, and metal nanoparticles. Taking advantage of their physicochemical and biologic properties, nanoparticles have been shown to increase the efficacy of commonly administered chemotherapies with reduced adverse effects.


Subject(s)
Biomedical Research/trends , Immunotherapy , Leukemia/therapy , Nanomedicine , Clinical Trials as Topic , Combined Modality Therapy , Humans
6.
Acta Biomater ; 90: 300-313, 2019 05.
Article in English | MEDLINE | ID: mdl-30930305

ABSTRACT

Immunotherapy based on checkpoint blockade has been regarded as one of the most promising approaches towards many types of cancers. However, low response rate hinders its application due to insufficient tumor immunogenicity and immunosuppressive tumor microenvironment. To achieve an overall enhanced therapeutic outcome, we developed a dual-functional immuno-stimulatory polymeric prodrug carrier modified with pendent indoximod, an indoleamine 2,3-dioxygenase (IDO) inhibitor that can be used to reverse immune suppression, for co-delivery of Doxorubicin (Dox), a hydrophobic anticancer agent that can promote immunogenic cell death (ICD) and elicit antitumor immunity. The resulted carrier denoted as POEG-b-PVBIND, consisting of poly (oligo (ethylene glycol) methacrylate) (POEG) hydrophilic blocks and indoximod conjugated hydrophobic blocks, is rationally designed to improve immunotherapy by synergistically modulating the tumor microenvironment (TME). Our data showed that Dox-triggered ICD promoted intra-tumoral infiltration of CD8+ T cells and IFN-γ-production by CD8+ T cells. Meanwhile, cleaved indoximod significantly increased CD8+ T cell infiltration while reducing the immunosuppressive T regulatory cells (Tregs). More importantly, Dox/POEG-b-PVBIND micelles led to significantly improved tumor regression in an orthotopic murine breast cancer model compared to both Dox-loaded POEG-b-PVB micelles (a control inert carrier) and POEG-b-PVBIND micelles alone, confirming combination effect of indoximod and Dox in improving the overall antitumor activity. STATEMENT OF SIGNIFICANCE: Indoleamine 2,3-dioxygenase (IDO) is an enzyme that can induce immune suppressive microenvironment in tumors. As a well-studied IDO inhibitor, indoximod (IND) represents a promising agent for cancer immunotherapy and could be particularly useful in combination with other chemotherapeutic agents. However, three major problems hinder its application: (1) IND is barely soluble in water; (2) IND delivery efficiency is limited (3) simultaneous delivery of two agents into tumor site is still challenging. Currently, most reports largely focus on improving the pharmacokinetic profile of IND alone via different formulations such as IND prodrug and IND nanocrystal. However, there is limited information about IND based co-delivery systems, especially for delivering hydrophobic chemotherapeutic agents. Here, we developed a new dual-functional polymeric prodrug carrier modified with a number of pendent IND units (denoted as POEG-b-PVBIND). POEG-b-PVBIND shows immunostimulatory and antitumor activities by itself. More importantly, POEG-b-PVBIND polymer is able to self-assemble into nano-sized micelles that are highly effective in formulating and codelivering other hydrophobic agents including doxorubicin (Dox), sunitinib (Sun), and daunorubicin (Dau), which can elicit antitumor immunity via promoting immunogenic cell death (ICD). We have shown that our new combination therapy led to a significantly improved antitumor activity in an aggressive murine breast cancer model (4T1.2).


Subject(s)
Antibiotics, Antineoplastic , Drug Carriers , Immunotherapy , Mammary Neoplasms, Experimental , Prodrugs , Tryptophan/analogs & derivatives , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Female , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Tryptophan/chemistry , Tryptophan/pharmacokinetics , Tryptophan/pharmacology
7.
Eur J Med Chem ; 141: 417-426, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29032034

ABSTRACT

A family of 12 members of Naphthalene-2-ol-indolin-2-one-thiocarbamides (5a-l) with pharmacological potentials of cardiovascular modulator were efficiently synthesized and evaluated. These compounds show inhibitory activity on angiotensin-converting enzyme (ACE), which is a principal constituent of the renin-angiotensin system and causative source for hypertension (HTN) (elevated blood pressure) and congestive heart failure (CHF), a parameter that was tested in this report. Prior to this, to get more insight into the binding mode and inhibition of human ACE C-domain (PDB ID: 2XY9) and N-domain (PDB ID: 3NXQ) compounds 5a-l was docked into the active site of them. The established inhibitory constant (Ki) (range 40-500 nM) and least binding affinities (-18.52 to -30.57 kcal/mol) indicated the therapeutic selectivity of compounds 5a-l towards ACE C-domain inhibition over ACE N-domain. The cytotoxicity effect of most potent compounds among 5a-l were tested in normal breast cells and MCF-7 cell lines. Simultaneously, H2O2 induced antioxidant and DNA damage assessment was executed. Eventually, a thrombolytic activity followed by a human red blood cell (HRBC) membrane stabilization study to ensure the relaxation of blood and stabilization of RBC was executed. Structure-Activity Relationship (SAR) study discloses the potential of 5c, 5h, and 5k as cardiovascular protective therapeutic agents among 5a-l.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cardiovascular Diseases/drug therapy , Indoles/pharmacology , Thrombolytic Therapy , Angiotensin-Converting Enzyme Inhibitors/chemical synthesis , Angiotensin-Converting Enzyme Inhibitors/chemistry , Cardiovascular Diseases/metabolism , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Heart Failure/drug therapy , Heart Failure/metabolism , Humans , Hypertension/drug therapy , Hypertension/metabolism , Indoles/chemical synthesis , Indoles/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
8.
Mater Sci Eng C Mater Biol Appl ; 79: 950-957, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28629100

ABSTRACT

The major purpose of cardiac tissue engineering is to engineer cells on scaffolds and use it as a substitute to infarcted cardiac cells. With an ever-increasing risk of cardiac diseases there is an increasing need to have a stable and sustainable approach to cure such ailments. This review provides a comprehensive update on the cell sources and biomaterials essential for cardiac tissue engineering, ensuring their biocompatibility under a variety of conditions. Cells can be obtained from allogenic or autologous sources. The cells sources described in this paper are cardiomyocytes, a variety of stem cells, fibroblasts and parthenogenetic cells. Scaffolds to seed the cells should fulfill various criteria, the most important ones being non-immunogenicity, biodegradability and native tissue mimicking. There are several kinds of scaffolds including hydrogels, prefabricated matrices, decellularized scaffolds and cell sheets. Using this knowledge of cell sources and scaffold fabrication, production of various tissue engineered products is possible.


Subject(s)
Tissue Engineering , Biocompatible Materials , Humans , Hydrogels , Myocytes, Cardiac , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...