scholarly journals Synergistic Effect of Doxorubicin and siRNA-Mediated Silencing of Mcl-1 Using Cationic Niosomes against 3D MCF-7 Spheroids

Pharmaceutics ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 550
Author(s):  
Supusson Pengnam ◽  
Samarwadee Plianwong ◽  
Prasopchai Patrojanasophon ◽  
Widchaya Radchatawedchakoon ◽  
Boon-ek Yingyongnarongkul ◽  
...  

Chemotherapy is a vital option for cancer treatment; however, its therapeutic outcomes are limited by dose-dependent toxicity and the occurrence of chemoresistance. siRNAs have emerged as an attractive therapeutic option enabling specific interference with target genes. Combination therapy using chemotherapeutic agents along with gene therapy could be a potential strategy for cancer management, which not only improves therapeutic efficacy but also decreases untoward effects from dose reduction. In this study, a cationic niosome containing plier-like cationic lipid B was used to convey siRNA against anti-apoptotic mRNA into MCF-7 and MDA-MB-231 cells. Mcl-1 silencing markedly decreased the viability of MCF-7 cells and triggered apoptosis. Moreover, computer modeling suggested that the combination of doxorubicin (Dox) and Mcl-1 siRNA exhibited a synergistic relationship and enabled a dose reduction of each agent at 1.71 and 3.91 folds, respectively, to reach a 90% inhibitory effect when compared to single-agent treatments. Synergistic antitumor activity was further verified in a 3D spheroid culture which revealed, in contrast to single-agent treatment, the combination markedly decreased spheroid volume over time. Together, the combination therapy between Mcl-1 silencing and Dox exhibits a synergistic effect that may be exploited for novel breast cancer treatment.

Author(s):  
Khalid Umar Fakhri ◽  
Bader Saud Alotaibi ◽  
Sumit Kumar ◽  
Umesh Kumar ◽  
Subash C. Sonkar ◽  
...  

Several modern treatment procedures have been received to battle malignancy with the point of limiting lethality. Phytopharmaceuticals are auxiliary metabolites of plant origin which exclusively contain one or more substances as active ingredients or might be a blend of them. Analysts have excitedly attempted to diminish the lethality of current chemotherapeutic agents either by consolidating them with herbals or in utilizing herbals alone. Synergy is a procedure where a few substances participate to reach a consolidated impact that is more prominent than the entirety of their different impacts. It may be viewed as a characteristic straight technique that has developed ordinarily by nature to acquire more efficacies at a low cost. This chapter aims to present the fundamental mechanism of the activity of phytochemicals in combination therapy. This chapter additionally features the remarkable synergistic impacts of plant-drug cooperation with an emphasis on anticancer strategies.


Nutrients ◽  
2018 ◽  
Vol 10 (9) ◽  
pp. 1148 ◽  
Author(s):  
Jorge Hernandez-Valencia ◽  
Enrique Garcia-Villa ◽  
Aquetzalli Arenas-Hernandez ◽  
Jaime Garcia-Mena ◽  
Jose Diaz-Chavez ◽  
...  

Resistance to cisplatin (CDDP) is a major cause of cancer treatment failure, including human breast cancer. The tumor suppressor protein p53 is a key factor in the induction of cell cycle arrest, DNA repair, and apoptosis in response to cellular stimuli. This protein is phosphorylated in serine 15 and serine 20 during DNA damage repair or in serine 46 to induce apoptosis. Resveratrol (Resv) is a natural compound representing a promising chemosensitizer for cancer treatment that has been shown to sensitize tumor cells through upregulation and phosphorylation of p53 and inhibition of RAD51. We developed a CDDP-resistant MCF-7 cell line variant (MCF-7R) to investigate the effect of Resv in vitro in combination with CDDP over the role of p53 in overcoming CDDP resistance in MCF-7R cells. We have shown that Resv induces sensitivity to CDDP in MCF-7 and MCF-7R cells and that the downregulation of p53 protein expression and inhibition of p53 protein activity enhances resistance to CDDP in both cell lines. On the other hand, we found that Resv induces serine 20 (S20) phosphorylation in chemoresistant cells to activate p53 target genes such as PUMA and BAX, restoring apoptosis. It also changed the ratio between BCL-2 and BAX, where BCL-2 protein expression was decreased and at the same time BAX protein was increased. Interestingly, Resv attenuates CDDP-induced p53 phosphorylation in serine 15 (S15) and serine 46 (S46) probably through dephosphorylation and deactivation of ATM. It also activates different kinases, such as CK1, CHK2, and AMPK to induce phosphorylation of p53 in S20, suggesting a novel mechanism of p53 activation and chemosensitization to CDDP.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1374-1374 ◽  
Author(s):  
Marcus M. Schittenhelm ◽  
Kevin W. Yee ◽  
Kerstin M. Kampa ◽  
Michael C. Heinrich

Abstract FLT3 kinase inhibitors display promising pre-clinical efficacy in a variety of in vitro and animal models of FLT3-ITD+ AML. Tandutinib is a 4-piprazinylquinazoline compound that is a potent inhibitor of type III RTKs with a cellular IC50 of ~200 nM for FLT3. In a phase 2 study, Tandutinib demonstrated anti-leukemic activity in approximately half of the evaluable patients, although there were no partial or complete remissions. Therefore, optimal use of like Tandutinib will likely require combination therapy with standard cytotoxic agents such as cytarabine (Cy) and an anthracycline (e.g. daunorubicin, Dn). Notably, single agent Tandutinib has not been associated with myelosuppression, mucositis or cardiac toxicity--the dose limiting toxicities of AML chemotherapy. To determine the feasibility of combining Tandutinib and chemotherapy, we tested Tandutinib in combination with Cy and/or Dn. For our experiments, we utilized three cell lines containing FLT3 ITD mutations: a BaF3 cell line transduced with an ITD mutant FLT3 as well as MV 4–11 and MOLM 14 cell lines which have naturally occurring FLT3 ITD mutations. Data was analyzed using the statistical methods of Chou and Talalay to calculate combination indices (CI) for drug mixtures performed in a fixed dilution pattern. In all cell lines, calculated combination indices for inhibition of cellular proliferation and induction of apoptosis were much less than one, indicating a synergistic effect of combining Tandutinib with Cy or Dn. In addition, the combination of Tandutinib with Cy and Dn was also markedly synergistic. These results were confirmed using primary AML blasts (FLT3 ITD+). All of the above studies were performed using simultaneous treatment with Tandutinib and Cy and/or Dn. Previous in vitro studies utilizing a structurally unrelated FLT3 inhibitor, Lestaurtinib (CEP-701), demonstrated the potential importance of treatment sequencing for optimal killing of AML cells. Notably, treatment with Lestaurtinib with or following chemotherapy was found to be synergistic, whereas treatment with Lestaurtinib followed by chemotherapy was generally antagonistic. Therefore, we next determined whether sequencing of Tandutinib and chemotherapy had any effect on the efficacy of combination therapy. For these experiments, we tested two different sequencing regimens: monotherapy treatment with Tandutinib for 24 hours with the addition of Cy or Dn for an additional 48 hours; ormonotherapy with Cy or Dn for 24 hours with the addition of Tandutinib for an additional 48 hours. Both regimens produced a synergistic effect and had comparable efficacy. Therefore, unlike Lestaurtinib, the synergistic effect of Tandutinib and Cy/Dn appears to be independent of the sequence of drug administration. These data suggest that addition of Tandutinib to agents used in induction chemotherapy for AML could result in enhanced antileukemic effects. Our synergy experiments also suggest that combined use of Tandutinib and chemotherapy may allow dose reduction of chemotherapy (with resultant decreased side effects) without loss of antileukemic activity. Such a combined treatment approach may be particularly desirable for elderly AML patients who often have poor tolerance of standard AML chemotherapy regimens. Currently, the combination of Tandutinib + standard induction chemotherapy is being tested in a phase I/II study for treatment of newly diagnosed AML.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2651-2651
Author(s):  
Melissa G. Ooi ◽  
Patrick J. Hayden ◽  
Douglas W. McMillin ◽  
Joseph M. Negri ◽  
Jake Delmore ◽  
...  

Abstract The “guardian of the genome” p53 is inactivated in most human cancers via missense mutations in its DNA binding core domain or via overexpression of the human homolog of Mdm2 (Hdm2), an E3 ubiquitin ligase that binds and ubiquitinates p53 thereby leading to its degradation via the ubiquitin/proteasome pathway. Direct inhibition of Mdm2 function could potentially stabilize p53 and activate the p53 apoptotic pathway, which could be a useful non-genotoxic approach for the treatment of cancer. Nutlin-3, a cis-imidazoline small molecule with affinity for the p53-binding pocket of Mdm2 and capable of disrupting the p53-Mdm2 interaction, can activate p53 and induce apoptosis in vitro in many malignancies, including multiple myeloma (MM) cells. We hypothesized that suppression of Mdm2-mediated p53 ubiquitination may synergize with accumulation of p53 triggered by bortezomib. To address this question, we evaluated the response of MM cells vs. select models of epithelial cancers to bortezomib and its combination with nutlin-3. We observed that breast (MDA-MB-231-luc); prostate (DU145); thyroid (SW579, FRO, WRO, TT) and colon (ARO/HT-29) cancer cell lines exhibited synergistic apoptotic response to the combination of sublethal concentrations of bortezomib plus nutlin-3. This synergistic killing effect was associated with synergistic increase in the expression of p53, p21, Mdm2, Bax, Noxa, PUMA and the cleavage of caspase-3, caspase-9 and PARP. In contrast, MM cell lines (MM-1S, MM-1R, NCI-H929, KMS-11) exhibited mostly an additive effect when treated with the combination of bortezomib plus nutlin-3. To further probe the biological relevance of these findings in the context of the bone microenvironment in MM, we compared the transcriptional profile of p53 target genes as well as the response to nutlin-3 in MM cells cultured alone vs. in the presence of bone marrow stromal cells (BMSCs). We observed that co-culture with BMSCs attenuates the response of MM cells to single-agent nutlin-3 and is associated, in p53 wild-type cells, with increased amplitude of the transcriptional signature of genes suppressed by activated p53, suggesting that BMSCs suppress p53 activity in MM cells. Overall, these results suggest a complex correlation between the Mdm2/p53 and proteasome pathways: many p53 mutant epithelial cancer models can become more bortezomib-responsive by Mdm2 inhibition, while MM cells, with higher baseline responsiveness to bortezomib and nutlin-3, typically do not exhibit significant increases in their response to the combination of these 2 drug classes compared to the single-agent treatments. These observations suggest that concurrent Mdm2 inhibition may extend the spectrum of bortezomib applications to tumor types with currently limited single-agent response to proteasome inhibition. In MM, p53 mutations have been historically considered to be present in only late stage disease (e.g. plasma cell leukemia/extramedullary MM), but their prevalence may increase in the future as more patients with advanced MM survive longer thanks to recently introduced drug classes (thalidomide, bortezomib, lenalidomide). In those MM patients who harbor p53 pathway lesions and have developed bortezomib-resistance/refractoriness, combinations of bortezomib with Mdm2 inhibitors may also represent in the future an intriguing potential therapeutic option that merits further preclinical and clinical evaluation.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. TPS8058-TPS8058 ◽  
Author(s):  
Sung-Soo Yoon ◽  
Elisabet Esteve Manasanch ◽  
Chang Ki Min ◽  
Jin Seok Kim ◽  
Robert S Hauptschein ◽  
...  

TPS8058 Background: CWP291, a novel peptidomimetic small molecule, has potent, selective inhibitory activity on a Wnt gene reporter, decreasing expression of β-catenin target genes, cyclin D1 and survivin. With broad anti-cancer efficacy in vitro, it significantly outperforms lenalidomide as a single agent combination in MM bone marrow engraftment models. Methods: This Phase 1a/1b study (NCT #02426723) was designed to define a well-tolerated dose of CWP291 as a single agent in subjects with R/R MM. CWP291 was administered IV over ≥30 minutes 2x weekly for 3 weeks out of a 4-week cycle, with standard 3+3 dose escalation design. But an important objective in terms of patient benefit and further clinical development was to explore activity of a combination regimen with lenalidomide. Thus, a novel study design allowed initiation of the Phase 1b as soon as CWP291 achieved a well-tolerated dose as a single agent. Combination therapy would start at one dose level lower. Enrollment of patients onto each arm was guided by Safety Review Committee assessments, including baseline laboratory values, performance status, extent of prior therapy, or prior adverse events related to lenalidomide. Results: Initiated September 2015, the starting dose was based on a prior Phase 1 study in AML, 198 mg/m2. There were 4 sites involved, and 11 patients enrolled over 12 months. Approval of the new design by regulatory authorities and IRBs was completed by November 2016. A well-tolerated single agent dose (297 mg/m2) was identified, allowing initiation of the Phase 1b at a dose of 198 mg/m2(one dose level lower) combined with lenalidomide. Four subjects were enrolled in ~2 months to the Phase 1b. Enrollment to both arms is continuing and the status of this study will be updated at presentation. Conclusions: The ability to consider combination therapy with a novel drug is clearly a motivation for patient participation in clinical trials; especially true in MM, as multiple new therapies are available. This trial design was approved and allowed based on assessment of individual patient safety and potential benefit. Rapid enrollment in the combination therapy arm may significantly foster development of novel agents with this study design. Clinical trial information: NCT #02426723.


Open Heart ◽  
2021 ◽  
Vol 8 (2) ◽  
pp. e001849
Author(s):  
Akshee Batra ◽  
Brijesh Patel ◽  
Daniel Addison ◽  
Lauren A Baldassarre ◽  
Nihar Desai ◽  
...  

ObjectiveAntimicrotubular agents are among the most commonly used classes of chemotherapeutic agents, but the risk of cardiovascular adverse events (CVAEs) remains unclear. Our objective was to study the CVAEs associated with antimicrotubular agents.MethodsThe Food and Drug Administration’s Adverse Event Reporting System was used to study CVAEs in adults from 1990 to 2020. Reported single-agent (only taxane or vinca alkaloid) CVAEs were compared with combination therapy (with at least one of the four major cardiotoxic drugs: anthracycline, HER2Neu inhibitors, tyrosine kinase inhibitors and checkpoint inhibitors) using adjusted polytomous logistic regression.ResultsOver 30 years, 134 398 adverse events were reported, of which 18 426 (13.4%) were CVAEs, with 74.1% due to taxanes and 25.9% due to vinca alkaloids. In 30 years, there has been a reduction in the proportion of reported CVAEs for taxanes from 15% to 11.8% (Cochran-Armitage P-trends <0.001) with no significant change in the proportion of reported CVAEs for vinca alkaloids (9.2%–11.7%; P-trends=0.06). The proportion of reported CVAEs was lower in both taxane and vinca alkaloid monotherapy versus combination therapy (reporting OR=0.50 and 0.55, respectively). Anthracyclines and HER2Neu inhibitor combinations with taxanes or vinca alkaloids primarily drove the higher burden of combination CVAEs. Hypertension requiring hospitalisation and heart failure was significantly lower in monotherapy versus combination antimicrotubular agent therapy.ConclusionsAntimicrotubular agents are associated with CVAEs, especially in combination chemotherapy regimens. Based on this study, we suggest routine cardiovascular assessment of patients with cancer before initiating antimicrotubular agents in combination therapy.


2020 ◽  
Vol 20 (8) ◽  
pp. 586-602
Author(s):  
Manzoor A. Mir ◽  
Hina Qayoom ◽  
Umar Mehraj ◽  
Safura Nisar ◽  
Basharat Bhat ◽  
...  

Triple negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer accounting for 15-20% of cases and is defined by the lack of hormonal receptors viz., estrogen receptor (ER), progesterone receptor (PR) and expression of human epidermal growth receptor 2 (HER2). Treatment of TNBC is more challenging than other subtypes of breast cancer due to the lack of markers for the molecularly targeted therapies (ER, PR, and HER-2/ Neu), the conventional chemotherapeutic agents are still the mainstay of the therapeutic protocols of its patients. Despite, TNBC being more chemo-responsive than other subtypes, unfortunately, the initial good response to the chemotherapy eventually turns into a refractory drug-resistance. Using a monotherapy for the treatment of cancer, especially high-grade tumors like TNBC, is mostly worthless due to the inherent genetic instability of tumor cells to develop intrinsic and acquired resistance. Thus, a cocktail of two or more drugs with different mechanisms of action is more effective and could successfully control the disease. Furthermore, combination therapy reveals more, or at least the same, effectiveness with lower doses of every single agent and decreases the likelihood of chemoresistance. Herein, we shed light on the novel combinatorial approaches targeting PARP, EGFR, PI3K pathway, AR, and wnt signaling, HDAC, MEK pathway for efficient treatment of high-grade tumors like TNBC and decreasing the onset of resistance.


2011 ◽  
Vol 345 ◽  
pp. 304-309
Author(s):  
Guo Qing Chen ◽  
Zhi Wei Zhao

Multidrug resistance remains a major clinical obstacle to successful treatment of breast cancer and leads to poor prognosis for the patients. Recently studies have shown that microRNAs play an important role in breast cancer cell resistance to chemotherapeutic agents. In this study, microRNA expression profiles of MCF-7/AdrVp and MCF-7 were analyzed using microarray. Gene Ontology and pathways mapping tools were employed to analysis systemically the biological processes and signaling pathways affected by differential expression microRNAs. The results indicated that breast cancer cell resistant to chemotherapy was associated with a group of microRNAs. Gene Ontology and pathway mapping are valid and effective approach to analysis the function of microRNAs and the results could be a guideline for further investigation.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4894-4894
Author(s):  
Sabrin Albeituni ◽  
Katherine Verbist ◽  
Paige Tedrick ◽  
Heather Tillman ◽  
Kim E. Nichols

Abstract The Hemophagocytic lymphohistiocytoses (HLH) comprise a heterogeneous group of disorders characterized by abnormal and severely damaging immune responses. Patients with HLH experience episodes of inflammation associated with the hyper-activation of CD8+ T cells and macrophages, which overproduce pro-inflammatory cytokines including interferon (IFN)-γ, interleukins (IL)-12 and -18, -6 and Tumor Necrosis Factor-α. Current treatments include immunosuppressive medications (corticosteroids, anti-thymocyte globulin) with or without cytotoxic chemotherapeutic agents (etoposide). Despite the use of these drugs, >50% of HLH patients die from unbridled inflammation. To develop more effective treatments for HLH, our laboratory has focused on blocking the effects of cytokines, which drive inflammation and mediate morbidity and mortality in HLH. We recently demonstrated that targeting the Janus Kinases (JAK1/2) with the JAK inhibitor ruxolitinib (INCB018424) significantly ameliorates the clinical and laboratory manifestations of HLH in preclinical mouse models (R. Das et al., Blood, 2016). Despite its beneficial effects, ruxolitinib did not completely abrogate disease. We therefore sought to understand its mechanisms of action and further improve upon these results. In mice, IFN-γ is central to disease pathogenesis, as its neutralization reduces anemia and prolongs survival. To examine whether ruxolitinib might be acting primarily through the inhibition of this cytokine, we modeled HLH in perforin-deficient (Prf1-/-) mice that were infected with lymphocytic choriomeningitis virus (LCMV). Beginning on day 4 post infection (p.i.), mice were treated or not with ruxolitinib (90 mg/kg by oral gavage twice daily) or with an IFN-γ blocking antibody (0.5 mg intraperitoneally [i.p.] on days 4 and 7). On day 9 p.i., the end point for these studies, mice were examined for the manifestations of HLH. Compared to untreated LCMV-infected mice, which lost on average 16% of starting body weight, mice treated with ruxolitinib or IFN-γ blockade exhibited only 7% weight loss. Ruxolitinib and IFN-γ blockade also ameliorated LCMV-induced anemia and thrombocytopenia, with platelet counts with ruxolitinib treatment reaching levels comparable to those observed in uninfected animals. Of note, ruxolitinib treatment decreased splenomegaly by over 50% (p=0.0001), while IFN-γ blockade had no effect. When compared to mice that received either no treatment or IFN-γ blockade, mice treated with ruxolitinib also exhibited a reduction in inflammation, which was manifested as a significantly lower area of liver encompassed by inflammatory foci, as well as a marked reduction in the absolute number of total and gp33-tetramer reactive splenic and intrahepatic CD8+ T cells. Together, these results suggest that ruxolitinib exerts its effects through mechanisms that are in part independent of the inhibition of IFN-γ signaling. We next examined whether combining ruxolitinib with dexamethasone, a gold standard medication in HLH treatment, might further diminish inflammation. To this end, LCMV-infected Prf1-/- mice were treated or not with ruxolitinib (as above), dexamethasone (15mg/kg i.p. daily) or ruxolitinib and dexamethasone. All treatments resulted in a similar reduction in weight loss. Thrombocytopenia was abrogated with ruxolitinib treatment, either alone or in combination with dexamethasone. Mice treated with both drugs, however, did not develop splenomegaly and were less anemic than mice treated with either single agent. Strikingly, when compared to the livers of infected untreated mice, livers from mice treated with combination therapy exhibited a 99.7% decrease in the area encompassed by inflammatory foci. This was in contrast to mice treated with ruxolitinib or dexamethasone, which showed a 66.8% or 87% reduction, respectively. Consistent with these findings, combination therapy was also more potent in decreasing the numbers and to a lesser extent the frequencies of total and antigen-specific CD8+ T cells. These findings suggest that treatment with ruxolitinib and dexamethasone further limits inflammation and lessens the manifestations of HLH, which represents a serious clinical problem for which there has been limited progress in treatment. The results of these studies support the incorporation of ruxolitinib and dexamethasone into future clinical trials to improve the cure rate for HLH. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 25 (46) ◽  
pp. 4883-4892 ◽  
Author(s):  
Mitra Korani ◽  
Shahla Korani ◽  
Elham Zendehdel ◽  
Amin Reza Nikpoor ◽  
Mahmoud Reza Jaafari ◽  
...  

: Bortezomib (VELCADE®) is a boronate peptide and first-in-class proteasome inhibitor serving an important role in degenerating several intracellular proteins. It is a reversible inhibitor of the 26S proteasome, with antitumor activity and antiproliferative properties. This agent principally exerts its antineoplastic effects by inhibiting key players in the nuclear factor κB (NFκB) pathway involved in cell proliferation, apoptosis, and angiogenesis. This medication is used in the management of multiple myeloma. However, more recently, it has been used as a therapeutic option for mantle cell lymphoma. While promising, bortezomib has limited clinical applications due to its adverse effects (e.g., hematotoxicity and peripheral neuropathy) and low effectiveness in solid tumors resulting from its poor penetration into such masses and suboptimal pharmacokinetic parameters. Other limitations to bortezomib include its low chemical stability and bioavailability, which can be overcome by using nanoparticles for its delivery. Nanoparticle delivery systems can facilitate the targeted delivery of chemotherapeutic agents in high doses to the target site, while sparing healthy tissues. Therefore, this drug delivery system has provided a solution to circumvent the limitations faced with the delivery of traditional cancer chemotherapeutic agents. Our aim in this review was to describe polymer-based nanocarriers that can be used for the delivery of bortezomib in cancer chemotherapy.


Sign in / Sign up

Export Citation Format

Share Document