Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Pharm Anal ; 14(3): 389-400, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38618248

ABSTRACT

Antibody-drug conjugates (ADCs) are a new type of targeting antibodies that conjugate with highly toxic anticancer drugs via chemical linkers to exert high specificity and efficient killing of tumor cells, thereby attracting considerable attention in precise oncology therapy. Cetuximab (Cet) is a typical antibody that offers the benefits of good targeting and safety for individuals with advanced and inoperable cutaneous squamous cell carcinoma (cSCC); however, its anti-tumor activity is limited to a single use. Cisplatin (CisPt) shows good curative effects; however, its adverse effects and non-tumor-targeting ability are major drawbacks. In this study, we designed and developed a new ADC based on a new cytotoxic platinum (IV) prodrug (C8Pt(IV)) and Cet. The so-called antibody-platinum (IV) prodrugs conjugates, named Cet-C8Pt(IV), showed excellent tumor targeting in cSCC. Specifically, it accurately delivered C8Pt(IV) into tumor cells to exert the combined anti-tumor effect of Cet and CisPt. Herein, metabolomic analysis showed that Cet-C8Pt(IV) promoted cellular apoptosis and increased DNA damage in cSCC cells by affecting the vitamin B6 metabolic pathway in tumor cells, thereby further enhancing the tumor-killing ability and providing a new strategy for clinical cancer treatment using antibody-platinum (IV) prodrugs conjugates.

2.
Adv Mater ; 36(19): e2312488, 2024 May.
Article in English | MEDLINE | ID: mdl-38301714

ABSTRACT

Drug resistance significantly hampers the clinical application of existing platinum-based anticancer drugs. New platinum medications that possess distinct mechanisms of action are highly desired for the treatment of Pt-resistant cancers. Herein, a nanoscale trans-platinum(II)-based supramolecular coordination self-assembly (Pt-TCPP-BA) is prepared via using trans-[PtCl2(pyridine)(NH3)] (transpyroplatin), tetracarboxylporphyrin (TCPP), and benzoic acid (BA) as building blocks to combat drug resistance in platinum-based chemotherapy. Mechanistic studies indicate that Pt-TCPP-BA shows a hydrogen-peroxide-responsive dissociation behavior along with the generation of bioactive trans-Pt(II) and TCPP-Pt species. Different from cisplatin, these degradation products interact with DNA via interstrand cross-links and small groove binding, and induce significant upregulation of cell-death-related proteins such as p53, cleaved caspase 3, p21, and phosphorylated H2A histone family member X in cisplatin-resistant cancer cells. As a result, Pt-TCPP-BA exhibits potent killing effects against Pt-resistant tumors both in vitro and in vivo. Overall, this work not only provides a new platinum drug for combating drug-resistant cancer but also offers a new paradigm for the development of platinum-based supramolecular anticancer drugs.


Subject(s)
Antineoplastic Agents , Platinum , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Humans , Cell Line, Tumor , Animals , Platinum/chemistry , Platinum/pharmacology , Drug Resistance, Neoplasm/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Mice , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Cisplatin/pharmacology , Cisplatin/chemistry , DNA/chemistry , DNA/metabolism , Apoptosis/drug effects
3.
Natl Sci Rev ; 11(1): nwae020, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38332843

ABSTRACT

Mounting evidence suggests that strategies combining DNA-damaging agents and stimulator of interferon genes (STING) agonists are promising cancer therapeutic regimens because they can amplify STING activation and remodel the immunosuppressive tumor microenvironment. However, a single molecular entity comprising both agents has not yet been developed. Herein, we designed two PtIV-MSA-2 conjugates (I and II) containing the DNA-damaging chemotherapeutic drug cisplatin and the innate immune-activating STING agonist MSA-2; these conjugates showed great potential as multispecific small-molecule drugs against pancreatic cancer. Mechanistic studies revealed that conjugate I upregulated the expression of transcripts associated with innate immunity and metabolism in cancer cells, significantly differing from cisplatin and MSA-2. An analysis of the tumor microenvironment demonstrated that conjugate I could enhance the infiltration of natural killer (NK) cells into tumors and promote the activation of T cells, NK cells and dendritic cells in tumor tissues. These findings indicated that conjugate I, which was created by incorporating a Pt chemotherapeutic drug and STING agonist into one molecule, is a promising and potent anticancer drug candidate, opening new avenues for small-molecule-based cancer metalloimmunotherapy.

4.
Adv Mater ; 36(11): e2310456, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38092007

ABSTRACT

Pyroptosis, an emerging mechanism of programmed cell death, holds great potential to trigger a robust antitumor immune response. Platinum-based chemotherapeutic agents can induce pyroptosis via caspase-3 activation. However, these agents also enhance cyclooxygenase-2 (COX-2) expression in tumor tissues, leading to drug resistance and immune evasion in pancreatic cancer and significantly limiting the effectiveness of chemotherapy-induced pyroptosis. Here, an amphiphilic polymer (denoted as PHDT-Pt-In) containing both indomethacin (In, a COX-2 inhibitor) and platinum(IV) prodrug (Pt(IV)) is developed, which is responsive to glutathione (GSH). This polymer self-assemble into nanoparticles (denoted as Pt-In NP) that can disintegrate in cancer cells due to the GSH responsiveness, releasing In to inhibit the COX-2 expression, hence overcoming the chemoresistance and amplifying cisplatin-induced pyroptosis. In a pancreatic cancer mouse model, Pt-In NP significantly inhibit tumor growth and elicit both innate and adaptive immune responses. Moreover, when combined with anti-programmed death ligand (α-PD-L1) treatment, Pt-In NP demonstrate the ability to completely suppress metastatic tumors, transforming "cold tumors" into "hot tumors". Overall, the sustained release of Pt(IV) and In from Pt-In NP amplifies platinum-drug-induced pyroptosis to elicit long-term immune responses, hence presenting a generalizable strategy for pancreatic cancer.


Subject(s)
Nanoparticles , Pancreatic Neoplasms , Prodrugs , Animals , Mice , Prodrugs/pharmacology , Prodrugs/therapeutic use , Platinum , Cyclooxygenase 2 , Pyroptosis , Cisplatin/pharmacology , Nanoparticles/therapeutic use , Polymers , Pancreatic Neoplasms/drug therapy , Cell Line, Tumor
5.
Genes (Basel) ; 14(11)2023 Nov 13.
Article in English | MEDLINE | ID: mdl-38003016

ABSTRACT

The precise mechanism of resistance to anti-cancer drugs such as platinum drugs is not fully revealed. To reveal the mechanism of drug resistance, the molecular networks of anti-cancer drugs such as cisplatin, carboplatin, oxaliplatin, and arsenic trioxide were analyzed in several types of cancers. Since diffuse-type stomach adenocarcinoma, which has epithelial-mesenchymal transition (EMT)-like characteristics, is more malignant than intestinal-type stomach adenocarcinoma, the gene expression and molecular networks in diffuse- and intestinal-type stomach adenocarcinomas were analyzed. Analysis of carboplatin revealed the causal network in diffuse large B-cell lymphoma. The upstream regulators of the molecular networks of cisplatin-treated lung adenocarcinoma included the anti-cancer drug trichostatin A (TSA), a histone deacetylase inhibitor. The upstream regulator analysis of cisplatin revealed an increase in FAS, BTG2, SESN1, and CDKN1A, and the involvement of the tumor microenvironment pathway. The molecular networks were predicted to interact with several microRNAs, which may contribute to the identification of new drug targets for drug-resistant cancer. Analysis of oxaliplatin, a platinum drug, revealed that the SPINK1 pancreatic cancer pathway is inactivated in ischemic cardiomyopathy. The study showed the importance of the molecular networks of anti-cancer drugs and tumor microenvironment in the treatment of cancer resistant to anti-cancer drugs.


Subject(s)
Adenocarcinoma , Antineoplastic Agents , Immediate-Early Proteins , MicroRNAs , Humans , Cisplatin , Carboplatin/pharmacology , Platinum/pharmacology , Platinum/therapeutic use , Oxaliplatin/therapeutic use , MicroRNAs/genetics , MicroRNAs/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Tumor Microenvironment , Trypsin Inhibitor, Kazal Pancreatic , Tumor Suppressor Proteins
6.
Angew Chem Int Ed Engl ; 62(44): e202312170, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37710398

ABSTRACT

Regulating autophagy to control the homeostatic recycling process of cancer cells is a promising anticancer strategy. Golgi apparatus is a substrate of autophagy but the Golgi-autophagy (Golgiphagy) mediated antitumor pathway is rarely reported. Herein, we have developed a novel Golgi-targeted platinum (II) complex Pt3, which is ca. 20 times more cytotoxic to lung carcinoma than cisplatin and can completely eliminate tumors after intratumoral administration in vivo. Its nano-encapsulated system for tail vein administration also features a good anti-tumor effect. Mechanism studies indicate that Pt3 induces substantial Golgi stress, indicated by the fragmentation of Golgi structure, down-regulation of Golgi proteins (GM130, GRASP65/55), loss of Golgi-dependent transport and glycosylation. This triggers Golgiphagy but blocks the subsequent fusion of autophagosomes with lysosomes, that is a dual role in autophagy regulation, resulting in loss of proteostasis and apoptotic cell death. As far as we know, Pt3 is the first Golgi-targeted Pt complex that can trigger Golgi stress-mediated dual-regulation of autophagic flux and autophagy-apoptosis crosstalk for highly efficient cancer therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Platinum/pharmacology , Autophagy , Golgi Apparatus/metabolism , Cisplatin/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/metabolism , Neoplasms/metabolism
7.
Small ; 19(49): e2206688, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37606911

ABSTRACT

Non-small cell lung cancer (NSCLC) is the most common pathological type of lung cancer , accounting for approximately 85% of lung cancers. For more than 40 years, platinum (Pt)-based drugs are still one of the most widely used anticancer drugs even in the era of precision medicine and immunotherapy. However, the clinical limitations of Pt-based drugs, such as serious side effects and drug resistance, have not been well solved. This study constructs a new albumin-encapsulated Pt(IV) nanodrug (HSA@Pt(IV)) based on the Pt(IV) drug and nanodelivery system. The characterization of nanodrug and biological experiments demonstrate its excellent drug delivery and antitumor effects. The multi-omics analysis of the transcriptome and the ionome reveals that nanodrug can activate ferroptosis by affecting intracellular iron homeostasis in NSCLC. This study provides experimental evidence to suggest the potential of HSA@Pt(IV) as a nanodrug with clinical application.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Ferroptosis , Lung Neoplasms , Nanoparticles , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Albumins , Iron/pharmacology , Cell Line, Tumor
8.
J Control Release ; 361: 819-846, 2023 09.
Article in English | MEDLINE | ID: mdl-37597809

ABSTRACT

Owing to the unique DNA damaging cytotoxicity, platinum (Pt)-based chemotherapy has long been the first-line choice for clinical oncology. Unfortunately, Pt drugs are restricted by the severe dose-dependent toxicity and drug resistance. Correspondingly, Pt(IV) prodrugs are developed with the aim to improve the antitumor performance of Pt drugs. However, as "free" molecules, Pt(IV) prodrugs are still subject to unsatisfactory in vivo destiny and antitumor efficacy. Recently, Pt(IV) prodrug nanotherapeutics, inheriting both the merits of Pt(IV) prodrugs and nanotherapeutics, have emerged and demonstrated the promise to address the underexploited dilemma of Pt-based cancer therapy. Herein, we summarize the latest fronts of emerging Pt(IV) prodrug nanotherapeutics. First, the basic outlines of Pt(IV) prodrug nanotherapeutics are overviewed. Afterwards, how versatile Pt(IV) prodrug nanotherapeutics overcome the multiple biological barriers of antitumor drug delivery is introduced in detail. Moreover, advanced combination therapies based on multimodal Pt(IV) prodrug nanotherapeutics are discussed with special emphasis on the synergistic mechanisms. Finally, prospects and challenges of Pt(IV) prodrug nanotherapeutics for future clinical translation are spotlighted.


Subject(s)
Neoplasms , Prodrugs , Humans , Prodrugs/therapeutic use , Neoplasms/drug therapy , Combined Modality Therapy , Drug Delivery Systems , Medical Oncology , Platinum/therapeutic use
9.
Clin. transl. oncol. (Print) ; 25(5): 1340-1352, mayo 2023. graf
Article in English | IBECS | ID: ibc-219518

ABSTRACT

Purpose The poor prognosis of ovarian cancer is largely due to platinum resistance. It has been demonstrated that nucleotide excision repair (NER) involving centrin-2(CETN2) is connected to platinum resistance in ovarian cancer. The molecular mechanism of CETN2 in ovarian cancer and the mechanism affecting the outcome of chemotherapy are unknown. Methods The protein–protein interaction (PPI) network was mapped after obtaining the interacting proteins of CETN2, and the interacting genes were subjected to enrichment analysis. To examine the relationship between CETN2 and platinum resistance, gene microarray data and clinical data related to platinum resistance in ovarian cancer were downloaded. The possible signaling pathway of CETN2 was investigated by Gene set enrichment analysis (GSEA). Immune infiltration analysis was performed. Immunohistochemistry (IHC) and quantitative real-time PCR (QRT-PCR) were used to examine the expression of CETN2 in clinical samples in relation to the effectiveness of chemotherapy. The capacity of CETN2 to predict chemotherapy results was proven by receiver operating characteristic (ROC) curves after the construction of two prediction models, the logistic regression model and the decision tree model. The impact of CETN2 on prognosis was examined using the Kaplan–Meier technique. Result CETN2 was associated with NER, oxidative phosphorylation (OXPHOS) and cell cycle pathways in ovarian cancer drug-resistant samples. In clinical samples, CETN2 showed its possible correlation with immune infiltration. The protein expression level of CETN2 was significantly higher in platinum-resistant patients than that in platinum-sensitive patients, and the expression level had some predictive value for chemotherapy outcome, and high CETN2 protein expression was associated with poorer progression-free survival (AU)


Subject(s)
Humans , Female , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Platinum/therapeutic use , Prognosis , DNA Repair , Drug Resistance, Neoplasm/genetics
10.
Curr Opin Chem Biol ; 74: 102303, 2023 06.
Article in English | MEDLINE | ID: mdl-37075513

ABSTRACT

The clinical application of Pt-based anticancer drugs has inspired the development of novel chemotherapeutic metallodrugs with improved efficacies. Pt(IV) prodrugs are one of the most promising successors of Pt(II) drugs and have displayed great anticancer performance. In particular, judicious modification of axial ligands endows Pt(IV) complexes with unique properties that enable them to overcome the limitations of conventional Pt(II) drugs. Herein, we summarize recent developments in Pt(IV) anticancer complexes, with a focus on their axial functionalization with other anticancer agents, immunotherapeutic agents, photosensitive ligands, peptides, and theranostic agents. We hope that this concise view of recently reported Pt(IV) coordination complexes will help researchers to design next-generation multi-functional anticancer agents based on a comprehensive Pt(IV) platform.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Prodrugs , Platinum/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Ligands , DNA Damage , Cell Line, Tumor
11.
Clin Transl Oncol ; 25(5): 1340-1352, 2023 May.
Article in English | MEDLINE | ID: mdl-36527574

ABSTRACT

PURPOSE: The poor prognosis of ovarian cancer is largely due to platinum resistance. It has been demonstrated that nucleotide excision repair (NER) involving centrin-2(CETN2) is connected to platinum resistance in ovarian cancer. The molecular mechanism of CETN2 in ovarian cancer and the mechanism affecting the outcome of chemotherapy are unknown. METHODS: The protein-protein interaction (PPI) network was mapped after obtaining the interacting proteins of CETN2, and the interacting genes were subjected to enrichment analysis. To examine the relationship between CETN2 and platinum resistance, gene microarray data and clinical data related to platinum resistance in ovarian cancer were downloaded. The possible signaling pathway of CETN2 was investigated by Gene set enrichment analysis (GSEA). Immune infiltration analysis was performed. Immunohistochemistry (IHC) and quantitative real-time PCR (QRT-PCR) were used to examine the expression of CETN2 in clinical samples in relation to the effectiveness of chemotherapy. The capacity of CETN2 to predict chemotherapy results was proven by receiver operating characteristic (ROC) curves after the construction of two prediction models, the logistic regression model and the decision tree model. The impact of CETN2 on prognosis was examined using the Kaplan-Meier technique. RESULTS: CETN2 was associated with NER, oxidative phosphorylation (OXPHOS) and cell cycle pathways in ovarian cancer drug-resistant samples. In clinical samples, CETN2 showed its possible correlation with immune infiltration. The protein expression level of CETN2 was significantly higher in platinum-resistant patients than that in platinum-sensitive patients, and the expression level had some predictive value for chemotherapy outcome, and high CETN2 protein expression was associated with poorer progression-free survival. CONCLUSIONS: CETN2 protein had a significant effect on ovarian cancer platinum sensitivity and prognosis, which may be related to the activation of NER, OXPHOS and cell cycle pathways upon CETN2 upregulation. Further research is necessary to determine the therapeutic application value of CETN2, which may be a new biomarker of chemoresponsiveness.


Subject(s)
Antineoplastic Agents , Carcinoma, Ovarian Epithelial , Drug Resistance, Neoplasm , Ovarian Neoplasms , Platinum Compounds , Female , Humans , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Prognosis , Platinum Compounds/pharmacology , Platinum Compounds/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
12.
Angew Chem Int Ed Engl ; 62(2): e202213337, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36259513

ABSTRACT

Triggering receptor expressed on myeloid cells-2 (TREM2) is a key pro-tumorigenic marker of tumor-infiltrating macrophages, showing potent immunosuppressive activity in tumor microenvironment. A platinum(IV) complex OPA derived from oxaliplatin (OP) and artesunate (ART) exhibited direct cytotoxicity against human colon cancer cells and immunomodulatory activity to inhibit TREM2 on macrophages in vitro and vivo. Furthermore, OPA deterred the tumor growth in mouse models bearing MC38 colorectal tumor by reducing the number of CD206+ and CX3 CR1+ immunosuppressive macrophages; it also promoted the expansion and infiltration of immunostimulatory dendritic, cytotoxic T, and natural killer cells. OPA is the first small-molecular TREM2 inhibitor capable of relieving immunosuppressive tumor microenvironment and enhancing chemical anticancer efficiency of a platinum drug, thus showing typical characteristics of a chemoimmunotherapeutic agent.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Animals , Mice , Humans , Platinum/pharmacology , Platinum/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Myeloid Cells/pathology , Macrophages , Immunosuppressive Agents/pharmacology , Colonic Neoplasms/drug therapy , Tumor Microenvironment , Membrane Glycoproteins , Receptors, Immunologic
13.
Cancer Res Commun ; 2(10): 1266-1281, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36467895

ABSTRACT

In recent years platinum (Pt) drugs have been found to be especially efficient to treat patients with cancers that lack a proper DNA damage response, e.g. due to dysfunctional BRCA1. Despite this knowledge, we are still missing helpful markers to predict Pt response in the clinic. We have previously shown that volume-regulated anion channels, containing the subunits LRRC8A and LRRC8D, promote the uptake of cisplatin and carboplatin in BRCA1-proficient cell lines. Here, we show that the loss of LRRC8A or LRRC8D significantly reduces the uptake of cis- and carboplatin in BRCA1;p53-deficient mouse mammary tumor cells. This results in reduced DNA damage and in vivo drug resistance. In contrast to Lrrc8a, the deletion of the Lrrc8d gene does not affect the viability and fertility of mice. Interestingly, Lrrc8d-/- mice tolerate a two-fold cisplatin maximum-tolerable dose. This allowed us to establish a mouse model for intensified Pt-based chemotherapy, and we found that an increased cisplatin dose eradicates BRCA1;p53-deficient tumors, whereas eradication is not possible in WT mice. Moreover, we show that decreased expression of LRRC8A/D in head and neck squamous cell carcinoma patients, who are treated with a Pt-based chemoradiotherapy, leads to decreased overall survival of the patients. In particular, high cumulative cisplatin dose treatments lost their efficacy in patients with a low LRRC8A/D expression in their cancers. Our data therefore suggest that LRRC8A and LRRC8D should be included in a prospective trial to predict the success of intensified cis- or car-boplatin-based chemotherapy.


Subject(s)
Cisplatin , Platinum , Mice , Animals , Cisplatin/pharmacology , Carboplatin/pharmacology , Platinum/metabolism , Tumor Suppressor Protein p53/genetics , Prospective Studies , Membrane Proteins/genetics , Anions/metabolism
14.
Cancers (Basel) ; 14(22)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36428691

ABSTRACT

Exposure to inorganic arsenic (As) is recognized as risk factor for basal cell carcinoma (BCC). We have followed-up 7000 adults for 6 years who were exposed to As and had manifest As skin toxicity. Of them, 1.7% developed BCC (males = 2.2%, females = 1.3%). In this study, we compared transcriptome-wide RNA sequencing data from the very first 26 BCC cases and healthy skin tissue from independent 16 individuals. Genes in " cell carcinoma pathway", "Hedgehog signaling pathway", and "Notch signaling pathway" were overexpressed in BCC, confirming the findings from earlier studies in BCC in other populations known to be exposed to As. However, we found that the overexpression of these known pathways was less pronounced in patients with high As exposure (urinary As creatinine ratio (UACR) > 192 µg/gm creatinine) than patients with low UACR. We also found that high UACR was associated with impaired DNA replication pathway, cellular response to different DNA damage repair mechanisms, and immune response. Transcriptomic data were not strongly suggestive of great potential for immune checkpoint inhibitors; however, it suggested lower chance of platinum drug resistance in BCC patients with high UACR compared high platinum drug resistance potential in patients with lower UACR.

15.
Front Med (Lausanne) ; 9: 942991, 2022.
Article in English | MEDLINE | ID: mdl-36016998

ABSTRACT

Necroptosis is a type of caspase-independent cell death, and it plays a critical role in regulating the development of cancer. To date, little is known about the role of necroptosis-related genes (NRGs) in clear cell renal cell carcinoma (ccRCC). In this study, we downloaded data regarding the expression of NRGs and overall survival (OS) from The Cancer Genome Atlas (TCGA) database and constructed a risk model to determine the prognostic features of necroptosis using COX regression analysis. Patients with ccRCC were divided into low-risk and high-risk groups based on their risk scores. Thereafter, Kaplan-Meier curves were used to evaluate OS, and receiver operating characteristic (ROC) curves were used to determine the accuracy of prediction. Stratified analyses were performed according to different clinical variables. Furthermore, we assessed the correlation between clinical variables and risk scores; the NRGs with differential expression were mainly enriched in positive regulation of intracellular transport and platinum resistance pathways. We constructed prognostic signatures for OS based on four NRGs and showed that the survival time was significantly longer in the low-risk groups than in the high-risk groups (p < 0.001). The area of the ROC curve for OS was 0.717, indicating excellent predictive accuracy of the established model. Therefore, a predictive model based on NRGs was constructed, which can predict the prognosis of patients and provides insights into the biological mechanisms underlying necroptosis in patients with ccRCC.

16.
Biochem Pharmacol ; 202: 115124, 2022 08.
Article in English | MEDLINE | ID: mdl-35688179

ABSTRACT

Among the new Pt complexes with anticancer properties, phenanthroline derivatives have aroused great interest due to their different mode of action compared to cisplatin. We previously examined cytotoxic effects of a new Pt(II)-complex containing 1,10-phenantroline (phen), [Pt(η1-C2H4OMe)(DMSO)(phen)]Cl, in a panel of eight human cancer cell lines, and showed that it exerted the greatest cytotoxic effect in the neuroblastoma SH-SY5Y cell line. In this study, the antiproliferative and antimetastatic potential of [Pt(η1-C2H4OMe)(DMSO)(phen)]+ (in short Pt-EtOMeSOphen) was investigated in neuroblastoma SH-SY5Y, SK-N-SH and SK-N-BE(2) cells. Pt-EtOMeSOphen provoked the early signs of apoptosis induction (cleavage of PARP and activation of caspases-9 and -7); it also increased the level of proapoptotic Bax protein whereas it decreased the level of the antiapoptotic Bcl-2 protein. The effects of Pt-EtOMeSOphen on migration and invasion processes were also evaluated. A decrease of cell migration/invasion by Pt-EtOMeSOphen was observed through 2D and 3D in vitro assays. Pt-EtOMeSOphen was found to exert its actions by decreasing MMP-9 and MMP-2 expressions and activities. Pt-EtOMeSOphen provoked the phosphorylation of both ERK1/2 and p38 MAPKs. All the effects of Pt-EtOMeSOphen on SH-SY5Y cell vitality, migration and metalloproteases activities described here were due to the activation of p38 MAPK since pharmacological p38 MAPK inhibition or small interfering RNAs to p38 MAPK mRNA blocked such effects. Results suggest that Pt-EtOMeSOphen inhibits neuroblastoma cancer cells survival, motility, and invasion. This could lead to the reduction of neuroblastoma metastatic potential.


Subject(s)
Antineoplastic Agents/pharmacology , Neuroblastoma , Platinum Compounds/pharmacology , Apoptosis , Cell Line, Tumor , Cell Movement , Dimethyl Sulfoxide/pharmacology , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Cancer Sci ; 112(11): 4655-4668, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34533854

ABSTRACT

Platinum-based regimens are the most widely used chemotherapy regimens, but cancer cells often develop resistance, which impedes therapy outcome for patients. Previous studies have shown that fibroblast growth factor 13 (FGF13) is associated with resistance to platinum drugs in HeLa cells. However, the mechanism and universality of this effect have not been clarified. Here, we found that FGF13 was associated with poor platinum-based chemotherapy outcomes in a variety of cancers, such as lung, endometrial, and cervical cancers, through bioinformatics analysis. We then found that FGF13 simultaneously regulates the expression and distribution of hCTR1 and ATP7A in cancer cells, causes reduced platinum influx, and promotes platinum sequestration and efflux upon cisplatin exposure. We subsequently observed that FGF13-mediated platinum resistance requires the microtubule-stabilizing effect of FGF13. Only overexpression of FGF13 with the -SMIYRQQQ- tubulin-binding domain could induce the platinum resistance effect. This phenomenon was also observed in SK-MES-1 cells, KLE cells, and 5637 cells. Our research reveals the mechanism of FGF13-induced platinum drug resistance and suggests that FGF13 can be a sensibilization target and prognostic biomarker for chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Copper Transporter 1/metabolism , Copper-Transporting ATPases/metabolism , Drug Resistance, Neoplasm , Fibroblast Growth Factors/physiology , A549 Cells , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cisplatin/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Microtubules/drug effects , Platinum Compounds/metabolism , Platinum Compounds/pharmacology , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
18.
ACS Appl Mater Interfaces ; 13(37): 44028-44040, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34499483

ABSTRACT

Polymeric nanocapsules hold considerable applications in cancer drug delivery, but the synthesis of well-defined nanocapsules with a tunable drug release property remains a significant challenge in fabrication. Herein, we demonstrate a supramolecular complexation strategy to assemble small molecular platinum (Pt) compounds into well-defined nanocapsules with high drug loading, acidity-sensitivity, and tunable Pt releasing profile. The design utilizes poly(ethylene glycol)-dendritic polylysine-G4/amides to complex with Pt compounds, forming stable nanocapsules with diameters approximately ∼20 nm and membrane thickness around several nanometers. The stability, drug content, and release profiles are tunable by tailoring the dendritic structure. The designated polymer-Pt nanocapsules, PEG-G4/MSA-Pt, showed sustained blood retention, preferential tumor accumulation, enhanced cellular uptake, lysosomal drug release, and nuclear delivery capability. PEG-G4/MSA-Pt showed enhanced antitumor efficacy compared to free cisplatin and other nanocapsules, which stopped the progression of both A549 cell xenografts and patient-derived xenografts (PDXs) of hepatocellular carcinoma on a mice tumor model. Thus, we believe this strategy is promising for developing Pt-based nanomedicine for cancer drug delivery.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Carriers/chemistry , Nanocapsules/chemistry , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Dendrimers/chemistry , Dendrimers/pharmacokinetics , Drug Carriers/pharmacokinetics , Drug Liberation , Humans , Mice, Inbred BALB C , Platinum/chemistry , Platinum/pharmacokinetics , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polylysine/chemistry , Polylysine/pharmacokinetics , Xenograft Model Antitumor Assays
19.
Small ; 17(41): e2102494, 2021 10.
Article in English | MEDLINE | ID: mdl-34510754

ABSTRACT

Compared to traditional clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) system, CRISPR/dead Cas9 (dCas9) system can precisely regulate endogenous gene expression without damaging the host gene, representing a greater potential for cancer therapy. Cancer/testis antigen 45 (CT45) is proved to enhance platinum-based chemosensitivity for individualized ovarian cancer therapy. However, the development of a single nanocarrier codelivering CRISPR/dCas9 system and chemotherapeutics for synergistic cancer therapy still faces challenges. Herein, a reduction-sensitive fluorinated-Pt(IV) universal transfection nanoplatform (PtUTP-F) is developed for the CT45-targeted CRISPR/dCas9 activation to achieve synergistic and individualized treatment of ovarian cancer. Overcoming multiple physiological barriers, PtUTP-F condensed gene can efficiently transfect into different cells including 293T cells, A2780, SKOV3, A549, and A2780/cisplatin (DDP) cancer cells, which is superior to Lipofectamine 6000. With the responsive release of gene and Pt(II) in the intracellular reducing microenvironment, PtUTP-F/dCas9-CT45 can generate CRISPR/dCas9 activation of CT45 expression for protein phosphatase 4C (PP4C) activity inhibition to hinder the DNA repair pathway and thus enhances the sensitivity to Pt(II) drugs for individualized A2780 tumor therapy. The PtUTP-F not only represents a powerful nanoplatform for CRISPR/dCas9 system delivery but also initiates a novel strategy for synergistic and individualized treatment of CRISPR/dCas9-based gene therapy with chemotherapy.


Subject(s)
Ovarian Neoplasms , Cell Line, Tumor , Female , HEK293 Cells , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Transfection , Tumor Microenvironment
20.
Anticancer Res ; 41(5): 2653-2660, 2021 May.
Article in English | MEDLINE | ID: mdl-33952496

ABSTRACT

BACKGROUND/AIM: To predict the efficacy of platinum-containing chemotherapy, ERCC1 expression levels were investigated. Studies have shown changes in the performance of anti-ERCC1 antibodies; therefore, predicting chemotherapy efficacy by immunohistochemical assessment of ERCC1 is controversial. PATIENTS AND METHODS: Twenty-eight patients who received platinum-containing chemotherapy and underwent computed tomography evaluation 6-9 weeks after therapy initiation were retrospectively identified. The tumor samples were evaluated in 2012 and 2018 using the latest anti-ERCC1 antibodies available at those times. RESULTS: In 2012, the ERCC1 H-score was significantly higher in patients with disease progression than in patients without disease progression (p=0.019). Although the same trend was shown in 2018, there were some inconsistent results between the 2012 and 2018 samples. CONCLUSION: Patients with tumors showing low ERCC1 expression had a better disease control rate on platinum-containing chemotherapy. However, since the performance of the antibody changed over time, standardized technology to evaluate ERCC1 expression is needed.


Subject(s)
Antibodies, Anti-Idiotypic/genetics , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA-Binding Proteins/genetics , Endonucleases/genetics , Aged , Aged, 80 and over , Antibodies, Anti-Idiotypic/isolation & purification , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Platinum/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL