Todaro M et al. (JUN 2010)
Gastroenterology 138 6 2151--62
Colon cancer stem cells: promise of targeted therapy.
First developed for hematologic disorders,the concept of cancer stem cells (CSCs) was expanded to solid tumors,including colorectal cancer (CRC). The traditional model of colon carcinogenesis includes several steps that occur via mutational activation of oncogenes and inactivation of tumor suppressor genes. Intestinal epithelial cells exist for a shorter amount of time than that required to accumulate tumor-inducing genetic changes,so researchers have investigated the concept that CRC arises from the long-lived stem cells,rather than from the differentiated epithelial cells. Colon CSCs were originally identified through the expression of the CD133 glycoprotein using an antibody directed to its epitope AC133. It is not clear if CD133 is a marker of colon CSCs-other cell surface markers,such as epithelial-specific antigen,CD44,CD166,Musashi-1,CD29,CD24,leucine-rich repeat-containing G-protein-coupled receptor 5,and aldehyde dehydrogenase 1,have been proposed. In addition to initiating and sustaining tumor growth,CSCs are believed to mediate cancer relapse after chemotherapy. How can we identify and analyze colon CSCs and what agents are being designed to kill this chemotherapy-refractory population?
View Publication
Dylla SJ et al. (JAN 2008)
PloS one 3 6 e2428
Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy.
BACKGROUND: Patients generally die of cancer after the failure of current therapies to eliminate residual disease. A subpopulation of tumor cells,termed cancer stem cells (CSC),appears uniquely able to fuel the growth of phenotypically and histologically diverse tumors. It has been proposed,therefore,that failure to effectively treat cancer may in part be due to preferential resistance of these CSC to chemotherapeutic agents. The subpopulation of human colorectal tumor cells with an ESA(+)CD44(+) phenotype are uniquely responsible for tumorigenesis and have the capacity to generate heterogeneous tumors in a xenograft setting (i.e. CoCSC). We hypothesized that if non-tumorigenic cells are more susceptible to chemotherapeutic agents,then residual tumors might be expected to contain a higher frequency of CoCSC. METHODS AND FINDINGS: Xenogeneic tumors initiated with CoCSC were allowed to reach approximately 400 mm(3),at which point mice were randomized and chemotherapeutic regimens involving cyclophosphamide or Irinotecan were initiated. Data from individual tumor phenotypic analysis and serial transplants performed in limiting dilution show that residual tumors are enriched for cells with the CoCSC phenotype and have increased tumorigenic cell frequency. Moreover,the inherent ability of residual CoCSC to generate tumors appears preserved. Aldehyde dehydrogenase 1 gene expression and enzymatic activity are elevated in CoCSC and using an in vitro culture system that maintains CoCSC as demonstrated by serial transplants and lentiviral marking of single cell-derived clones,we further show that ALDH1 enzymatic activity is a major mediator of resistance to cyclophosphamide: a classical chemotherapeutic agent. CONCLUSIONS: CoCSC are enriched in colon tumors following chemotherapy and remain capable of rapidly regenerating tumors from which they originated. By focusing on the biology of CoCSC,major resistance mechanisms to specific chemotherapeutic agents can be attributed to specific genes,thereby suggesting avenues for improving cancer therapy.
View Publication
B. Ndreshkjana et al. ( 2019)
Cell death {\&} disease 10 6 379
Combination of 5-fluorouracil and thymoquinone targets stem cell gene signature in colorectal cancer cells.
Cancer stem cells (CSCs) residing in colorectal cancer tissues have tumorigenic capacity and contribute to chemotherapeutic resistance and disease relapse. It is well known that the survival of colorectal CSCs after 5-fluorouracil (5-FU)-based therapy leads to cancer recurrence. Thus CSCs represent a promising drug target. Here,we designed and synthesized novel hybrid molecules linking 5-FU with the plant-derived compound thymoquinone (TQ) and tested the potential of individual compounds and their combination to eliminate colorectal CSCs. Both,Combi and SARB hybrid showed augmented cytotoxicity against colorectal cancer cells,but were non-toxic to organoids prepared from healthy murine small intestine. NanoString analysis revealed a unique signature of deregulated gene expression in response to the combination of TQ and 5-FU (Combi) and SARB treatment. Importantly,two principle stem cell regulatory pathways WNT/{\ss}-Catenin and PI3K/AKT were found to be downregulated after Combi and hybrid treatment. Furthermore,both treatments strikingly eliminated CD133+ CSC population,accompanying the depleted self-renewal capacity by eradicating long-term propagated 3D tumor cell spheres at sub-toxic doses. In vivo xenografts on chicken eggs of SARB-treated HCT116 cells showed a prominent nuclear {\ss}-Catenin and E-cadherin staining. This was in line with the reduced transcriptional activity of {\ss}-Catenin and diminished cell adhesion under SARB exposure. In contrast to 5-FU,both,Combi and SARB treatment effectively reduced the angiogenic capacity of the remaining resistant tumor cells. Taken together,combination or hybridization of single compounds target simultaneously a broader spectrum of oncogenic pathways leading to an effective eradication of colorectal cancer cells.
View Publication
Rao R et al. (APR 2012)
Molecular cancer therapeutics 11 4 973--983
Combination of pan-histone deacetylase inhibitor and autophagy inhibitor exerts superior efficacy against triple-negative human breast cancer cells.
Histone deacetylase (HDAC) inhibitors (HDI) induce endoplasmic reticulum (ER) stress and apoptosis,while promoting autophagy,which promotes cancer cell survival when apoptosis is compromised. Here,we determined the in vitro and in vivo activity of the combination of the pan-HDI panobinostat and the autophagy inhibitor chloroquine against human estrogen/progesterone receptor and HER2 (triple)-negative breast cancer (TNBC) cells. Treatment of MB-231 and SUM159PT cells with panobinostat disrupted the hsp90/histone deacetylase 6/HSF1/p97 complex,resulting in the upregulation of hsp. This was accompanied by the induction of enhanced autophagic flux as evidenced by increased expression of LC3B-II and the degradation of the autophagic substrate p62. Treatment with panobinostat also induced the accumulation and colocalization of p62 with LC3B-II in cytosolic foci as evidenced by immunofluorescent confocal microscopy. Inhibition of panobinostat-induced autophagic flux by chloroquine markedly induced the accumulation of polyubiquitylated proteins and p62,caused synergistic cell death of MB-231 and SUM159PT cells,and inhibited mammosphere formation in MB-231 cells,compared with treatment with each agent alone. Finally,in mouse mammary fat pad xenografts of MB-231 cells,a tumor size-dependent induction of heat shock response,ER stress and autophagy were observed. Cotreatment with panobinostat and chloroquine resulted in reduced tumor burden and increased the survival of MB-231 breast cancer xenografts. Collectively,our findings show that cotreatment with an autophagy inhibitor and pan-HDI,for example,chloroquine and panobinostat results in accumulation of toxic polyubiquitylated proteins,exerts superior inhibitory effects on TNBC cell growth,and increases the survival of TNBC xenografts.
View Publication
A. Shtilbans et al. (Dec 2024)
Frontiers in Neuroscience 18
Combination of tauroursodeoxycholic acid, co-enzyme Q10 and creatine demonstrates additive neuroprotective effects in in-vitro models of Parkinson’s disease
This study aimed to evaluate different combinations of three dietary supplements for potential additive or synergistic effects in an in vitro Parkinson’s Disease model. The complex and diverse processes leading to neurodegeneration in each patient with a neurodegenerative disorder cannot be effectively addressed by a single medication. Instead,various combinations of potentially neuroprotective agents targeting different disease mechanisms simultaneously may show improved additive or synergistic efficacy in slowing the disease progression and allowing the agents to be utilized at lower doses to minimize side effects. We evaluated four possible combinations of the three selected supplements: tauroursodeoxycholic acid (TUDCA),co-enzyme Q10 (CoQ10),and creatine,chosen for their effects on different targets that had previously shown neuroprotective effects in preclinical models. We evaluated the following combinations: (1) TUDCA+CoQ10,(2) TUDCA+Creatine,(3) CoQ10 + Creatine,and (4) TUDCA+CoQ10 + Creatine. We used induced pluripotent stem cell (iPSC) derived human dopaminergic neurons from a patient with Parkinson’s disease and healthy control,as well as microglial cells,to evaluate for an additive or synergistic effect of these combinations on neurodegeneration and neuroinflammation. We used neurofilament heavy chain,tubulin filament,and proinflammatory cytokines as metrics. We have identified a triple combination of these supplements that showed an additive protective effect across all these endpoints. Indeed,the agents in that combination could address the majority of the known pathways leading to neurodegeneration,such as accumulation of misfolded α -synuclein,mitochondrial dysfunction,reactive oxygen species,and neuroinflammation. We demonstrated that the combination of TUDCA,CoQ10,and creatine exerts an additive effect in in vitro models of a neurodegenerative disease,surpassing the efficacy of each compound individually. This combination shows strong potential as a candidate for further preclinical confirmatory studies and clinical trials as a neuroprotective treatment for patients with,or at risk for,Parkinson’s disease.
View Publication
S. Okabe et al. (Aug 2025)
Cancer Innovation 4 5
Combination of WEE1 Inhibitor and Vitamin K2 Enhances Therapeutic Efficacy in Chronic Myeloid Leukemia
Chronic myeloid leukemia (CML) is a clonal malignancy propelled by the BCR::ABL1 fusion gene originating from the Philadelphia chromosome. This gene activates ABL tyrosine kinase,which enhances the survival of leukemic cells. Although tyrosine kinase inhibitors (TKIs) have significantly advanced the treatment of CML,resistance to these inhibitors presents a substantial hurdle. Consequently,novel therapeutic strategies targeting resistance mechanisms independent of BCR::ABL1 are urgently needed. This study investigated the potential impact of combining WEE1 inhibitors,particularly MK‐1775,with vitamin K2 (VK2) in treating CML. To analyze differentially expressed and spliced transcripts in CML,we examined mRNA profiles from peripheral blood mononuclear cells of five patients with CML (during chronic and blast phases) and five healthy controls. The samples were analyzed using deep sequencing. Differential expression analyses were performed using RaNA‐Seq and Heatmapper,the latter of which was designed for complex data set visualizations. WEE1 controls the G2/M checkpoint to prevent early mitosis,and blocking it increases the cytotoxicity of agents that damage deoxyribonucleic acid,especially in cancers lacking p53. VK2,a micronutrient,exerts anticancer effects against various malignancies. Gene expression studies have indicated that PKMYT1 expression is elevated in CML but not WEE1 cells. MK‐1775 successfully halted the growth of both standard and TKI‐resistant CML cell lines by triggering apoptosis via caspase 3/7 activation. VK2 reduced the viability of CML cells and increased cytotoxicity. A combined regimen of MK‐1775 and VK2 markedly decreased colony growth,disrupted mitochondrial membrane potential,and increased death in CML cells,including those resistant to TKIs. The results suggest that a combination of MK‐1775 and VK2 represents a potentially effective treatment strategy for CML,especially in drug‐resistant cases.
View Publication
S. Adamia et al. (apr 2022)
Leukemia 36 4 1088--1101
Combination therapy targeting Erk1/2 and CDK4/6i in relapsed refractory multiple myeloma.
Oncogenic activated RAS mutations have been detected in 50% of de novo and 70% of relapsed multiple myeloma (MM) patients. Translocation t(11;14) involving IgH/CCDN1 and overexpression of cyclin-Ds are early events in MM pathogenesis,enhancing uncontrolled MM cell growth. We hypothesized that targeting both RAS/MAPK pathway molecules including Erk1/2 along with cyclin-Ds enhances MM cytotoxicity and minimizes side effects. Recent studies have demonstrated the high potency of Erk1/2 and CDK4/6 inhibitors in metastatic relapsed cancers,and here we tested anti-MM effects of the Erk1/2??+??CDK4/6 inhibitor combination. Our studies showed strong synergistic (IC???0.5) cytotoxicity of Erk1/2i??+??CDK4/6i in MM-cells. Erk1/2i??+??CDK4/6i treatment in a dose-dependent manner arrested MM-cells in the G0/G1 phase and activated mitochondrial apoptotic signaling. Our studies showed that Erk1/2i??+??CDK4/6i treatment-induced inhibition of key target molecules in Erk1/2 and CDK4/6 signaling,such as c-myc,p-RSK,p-S6,p-RB,and E2F1,suggesting on-target activity of these inhibitors. We identified Erk1/2i??+??CDK4/6i treatment associated five-gene signature which includes SNRPB and SLC25A5; these genes are involved in RNA processing and mitochondrial metabolism,respectively. Overall,our studies provide the preclinical framework for Erk1/2i??+??CDK4/6i combination clinical trials to target Ras+CDK pathways to improve patient outcome in MM.
View Publication
Radujkovic A et al. ( )
Anticancer research 26 3A 2169--77
Combination treatment of imatinib-sensitive and -resistant BCR-ABL-positive CML cells with imatinib and farnesyltransferase inhibitors.
BACKGROUND: Resistance to imatinib monotherapy frequently emerges in advanced stages of chronic myelogenous leukemia (CML),supporting the rationale for combination drug therapy. In the present study,the activities of the farnesyltransferase inhibitors (FTIs) L744,832 and LB42918,as single agents and in combination with imatinib,were investigated in different imatinib-sensitive and -resistant BCR-ABL-positive CML cells. MATERIALS AND METHODS: Growth inhibition of the cell lines and primary patient cells was assessed by MTT assays and colony-forming cell assays,respectively. Drug interactions were analyzed according to the median-effect method of Chou and Talalay. The determination of apoptotic cell death was performed by annexin V/propidium iodide staining. RESULTS: Combinations of both FTIs with imatinib displayed synergism or sensitization (potentiation) in all the cell lines tested. In primary chronic phase CML cells,additive and synergistic effects were discernible for the combination of imatinib plus L744,832 and imatinib plus LB42918,respectively. Annexin V/propidium iodide staining showed enhancement of imatinib-induced apoptosis with either drug combination,both in imatinib-sensitive and -resistant cells. CONCLUSION: The results indicated the potential of L744,832 and LB42918 as combination agents for CML patients on imatinib treatment.
View Publication
Albert BJ et al. (AUG 2017)
Scientific reports 7 1 7456
Combinations of isoform-targeted histone deacetylase inhibitors and bryostatin analogues display remarkable potency to activate latent HIV without global T-cell activation.
Current antiretroviral therapy (ART) for HIV/AIDS slows disease progression by reducing viral loads and increasing CD4 counts. Yet ART is not curative due to the persistence of CD4+ T-cell proviral reservoirs that chronically resupply active virus. Elimination of these reservoirs through the administration of synergistic combinations of latency reversing agents (LRAs),such as histone deacetylase (HDAC) inhibitors and protein kinase C (PKC) modulators,provides a promising strategy to reduce if not eradicate the viral reservoir. Here,we demonstrate that largazole and its analogues are isoform-targeted histone deacetylase inhibitors and potent LRAs. Significantly,these isoform-targeted HDAC inhibitors synergize with PKC modulators,namely bryostatin-1 analogues (bryologs). Implementation of this unprecedented LRA combination induces HIV-1 reactivation to unparalleled levels and avoids global T-cell activation within resting CD4+ T-cells.
View Publication
K. M. Glaser et al. ( 2022)
Frontiers in immunology 13 1039803
Combinatorial depletions of G-protein coupled receptor kinases in immune cells identify pleiotropic and cell type-specific functions.
G-protein coupled receptor kinases (GRKs) participate in the regulation of chemokine receptors by mediating receptor desensitization. They can be recruited to agonist-activated G-protein coupled receptors (GPCRs) and phosphorylate their intracellular parts,which eventually blocks signal propagation and often induces receptor internalization. However,there is growing evidence that GRKs can also control cellular functions beyond GPCR regulation. Immune cells commonly express two to four members of the GRK family (GRK2,GRK3,GRK5,GRK6) simultaneously,but we have very limited knowledge about their interplay in primary immune cells. In particular,we are missing comprehensive studies comparing the role of this GRK interplay for (a) multiple GPCRs within one leukocyte type,and (b) one specific GPCR between several immune cell subsets. To address this issue,we generated mouse models of single,combinatorial and complete GRK knockouts in four primary immune cell types (neutrophils,T cells,B cells and dendritic cells) and systematically addressed the functional consequences on GPCR-controlled cell migration and tissue localization. Our study shows that combinatorial depletions of GRKs have pleiotropic and cell-type specific effects in leukocytes,many of which could not be predicted. Neutrophils lacking all four GRK family members show increased chemotactic migration responses to a wide range of GPCR ligands,whereas combinatorial GRK depletions in other immune cell types lead to pro- and anti-migratory responses. Combined depletion of GRK2 and GRK6 in T cells and B cells shows distinct functional outcomes for (a) one GPCR type in different cell types,and (b) different GPCRs in one cell type. These GPCR-type and cell-type specific effects reflect in altered lymphocyte chemotaxis in vitro and localization in vivo. Lastly,we provide evidence that complete GRK deficiency impairs dendritic cell homeostasis,which unexpectedly results from defective dendritic cell differentiation and maturation in vitro and in vivo. Together,our findings demonstrate the complexity of GRK functions in immune cells,which go beyond GPCR desensitization in specific leukocyte types. Furthermore,they highlight the need for studying GRK functions in primary immune cells to address their specific roles in each leukocyte subset.
View Publication
Mei Y et al. (SEP 2010)
Nature materials 9 9 768--778
Combinatorial development of biomaterials for clonal growth of human pluripotent stem cells.
Both human embryonic stem cells and induced pluripotent stem cells can self-renew indefinitely in culture; however,present methods to clonally grow them are inefficient and poorly defined for genetic manipulation and therapeutic purposes. Here we develop the first chemically defined,xeno-free,feeder-free synthetic substrates to support robust self-renewal of fully dissociated human embryonic stem and induced pluripotent stem cells. Material properties including wettability,surface topography,surface chemistry and indentation elastic modulus of all polymeric substrates were quantified using high-throughput methods to develop structure-function relationships between material properties and biological performance. These analyses show that optimal human embryonic stem cell substrates are generated from monomers with high acrylate content,have a moderate wettability and employ integrin alpha(v)beta(3) and alpha(v)beta(5) engagement with adsorbed vitronectin to promote colony formation. The structure-function methodology employed herein provides a general framework for the combinatorial development of synthetic substrates for stem cell culture.
View Publication
(Oct 2024)
bioRxiv 379
Combinatorial effector targeting (COMET) for transcriptional modulation and locus-specific biochemistry
SUMMARYUnderstanding how human gene expression is coordinately regulated by functional units of proteins across the genome remains a major biological goal. Here,we present COMET,a high-throughput screening platform for combinatorial effector targeting for the identification of transcriptional modulators. We generate libraries of combinatorial dCas9-based fusion proteins,containing two to six effector domains,allowing us to systematically investigate more than 110,000 combinations of effector proteins at endogenous human loci for their influence on transcription. Importantly,we keep full proteins or domains intact,maintaining catalytic cores and surfaces for protein-protein interactions. We observe more than 5800 significant hits that modulate transcription,we demonstrate cell type specific transcriptional modulation,and we further investigate epistatic relationships between our effector combinations. We validate unexpected combinations as synergistic or buffering,emphasizing COMET as both a method for transcriptional effector discovery,and as a functional genomics tool for identifying novel domain interactions and directing locus-specific biochemistry.
View Publication