Michael M. Gottesman’s research while affiliated with National Cancer Institute and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (475)


Abcg2a antibody validation. (A) Immunoblot of total cell lysates and (B) immunohistochemistry of pellets of transfected HEK-293 cells expressing an empty vector, ABCG2, Abcg2a, Abcg2b, Abcg2c, or Abcg2d. Positive signal is only observed in Abcg2a-expressing cells. Scale bar = 100 μm
abcg2a and abcb4 are expressed in claudin-5 positive 5 dpf larval brain vasculature. A Paraffin-embedded 5 dpf larval zebrafish sections were probed with RNAscope probes (yellow) to detect abcg2a-d mRNA, an antibody against claudin-5 (green) and DAPI (blue). B Co-staining of the abcg2a probed section with an abcb4 RNAscope probe. Scale bar = 100 μm, inset scale bar = 10 μm
Correction: Abcg2a is the functional homolog of human ABCG2 expressed at the zebrafish blood–brain barrier
  • Article
  • Full-text available

January 2025

·

2 Reads

Fluids and Barriers of the CNS

·

William J. E. Frye

·

Robert W. Robey

·

[...]

·

Michael M. Gottesman
Download

EXTH-22. TARGETING MITOCHONDRIAL PROTEASE IN DIFFUSE GLIOMAS

November 2024

·

5 Reads

Neuro-Oncology

BACKGROUND Targeting mitochondrial function has emerged as a promising therapeutic strategy in cancer treatment. TR107 is characterized as a specific activator of caseinolytic protease proteolytic subunit (ClpP) in the mitochondria, potentially affecting oncogenic pathways. Here, we investigated the effects of TR107 on adult malignant gliomas, IDH-wildtype and IDH-mutant METHODS We utilized patient-derived glioma cells to determine the efficacy of TR107 with cell viability, proliferation, cell cycle, and apoptosis assays. Immunofluorescent staining and electron microscopy (EM) were employed to uncover organelle morphological changes upon treatment. Mitochondrial function and ATP production were assessed in live cells utilizing Seahorse analysis. Global proteomics and RNA sequencing were performed to identify treatment-induced dysregulated pathways in glioma cells RESULTS Both IDH-wildtype and IDH-mutant glioma cell lines were highly sensitive to TR107, showing at least 100 times lower IC50 compared to the related compound ONC201. Genetic knock out of ClpP revealed that the growth inhibitory effects of TR107 are ClpP-dependent. Interestingly, IDH-mutant cell lines showed a more profound disintegration of mitochondrial morphology and dysregulation of mitochondrial function evidenced by an enhanced suppression of oxidative phosphorylation, mitochondrial complexes expression, mtDNA copy number, complex I activity, and ATP production, compared to IDH-wildtype cells. Six days of treatment with TR107 led to a complete inhibition of cell growth and up to 97% of cell death in IDH-mutant cells, while 50% of IDH-wildtype cells remained viable. Western blot, EM, and RNAsequencing analyses uncovered autophagy as a potential pro-survival mechanism in IDH-wildtype cells. Importantly, multiple metabolic and DNA repair-related pathways were affected by the treatment, suggested by global proteomics and RNA sequencing. Finally, TR107 was not affected by ABC transporters, supporting its potential use in brain tumor patients CONCLUSION TR107 demonstrated potent cytotoxic effects in patient-derived glioma cells by disrupting mitochondrial structural and functional integrity. TR107 efficacy in vivo is under investigation.


Identification of DLK1, a Notch ligand, as an immunotherapeutic target and regulator of tumor cell plasticity and chemoresistance in adrenocortical carcinoma

October 2024

·

58 Reads

Immunotherapeutic targeting of cell surface proteins is an increasingly effective cancer therapy. However, given the limited number of current targets, the identification of new surface proteins, particularly those with biological importance, is critical. Here, we uncover delta-like non-canonical Notch ligand 1 (DLK1) as a cell surface protein with limited normal tissue expression and high expression in multiple refractory adult metastatic cancers including small cell lung cancer (SCLC) and adrenocortical carcinoma (ACC), a rare cancer with few effective therapies. In ACC, ADCT-701, a DLK1 targeting antibody-drug conjugate (ADC), shows potent in vitro activity among established cell lines and a new cohort of patient-derived organoids as well as robust in vivo anti-tumor responses in cell line-derived and patient-derived xenografts. However, ADCT-701 efficacy is overall limited in ACC due to high expression and activity of the drug efflux protein ABCB1 (MDR1, P-glycoprotein). In contrast, ADCT-701 is extremely potent and induces complete responses in DLK1+ ACC and SCLC in vivo models with low or no ABCB1 expression. Genetic deletion of DLK1 in ACC dramatically downregulates ABCB1 and increases ADC payload and chemotherapy sensitivity through NOTCH1-mediated adrenocortical de-differentiation. Single cell RNA-seq of ACC metastatic tumors reveals significantly decreased adrenocortical differentiation in DLK low or negative cells compared to DLK1 positive cells. This works identifies DLK1 as a novel immunotherapeutic target that regulates tumor cell plasticity and chemoresistance in ACC. Our data support targeting DLK1 with an ADC in ACC and neuroendocrine neoplasms in an active first-in-human phase I clinical trial ( NCT06041516 ).


Self‐Assembled Verteporfin Nanoparticles for Photodynamic and Light‐Independent Therapy in Glioblastoma

September 2024

·

43 Reads

Verteporfin (VP) has been used for photodynamic therapy (PDT) for over 20 years, and new applications have brought it back into the spotlight. VP is hydrophobic and requires lipid carriers for clinical delivery as Visudyne. A nanosuspension of VP, termed NanoVP, that requires no carriers is developed, permitting delivery of VP alone in an aqueous solution. NanoVP is produced by solvent–antisolvent precipitation, with dimethyl sulfoxide as the preferable solvent of several screened. The initial formulation has a hydrodynamic diameter of 104 ± 6.0 nm, concentration of 133 ± 10 μm, polydispersity index (Pdi) of 0.12 ± 0.01, and zeta potential of −22.0 ± 0.93 mV. Seeking a concentration >500 μm, a zeta potential <−10 mV, a diameter <64 nm, and a Pdi < 0.2, eight synthesis parameters are probed, identifying three that modified nanoparticle diameter and three that modified nanoparticle dispersity. The diameter is tuned fourfold from 49.0 ± 4.4 to 195 ± 7.1 nm, and the solution concentration is increased by 6.3‐fold to 838 ± 45.0 μm. Finally, the bioavailability and anticancer capacity of NanoVP in glioblastoma are evaluated. In all, this provides a framework for the modification of amorphous nanoparticle properties and a new formulation for clinical use of VP.


Identification of NanoLuciferase Substrates Transported by Human ABCB1 and ABCG2 and Their Zebrafish Homologs at the Blood-Brain Barrier

September 2024

·

9 Reads

Molecular Pharmacology

ATP-binding cassette (ABC) transporters expressed at the blood-brain barrier (BBB) impede delivery of therapeutic agents to the brain, including agents to treat neurodegenerative diseases and primary and metastatic brain cancers. Two transporters, P-glycoprotein (P-gp, ABCB1) and ABCG2, are highly expressed at the BBB and are responsible for the efflux of numerous clinically useful chemotherapeutic agents, including irinotecan, paclitaxel, and doxorubicin. Based on a previous mouse model, we have generated transgenic zebrafish where expression of NanoLuciferase (NanoLuc) is controlled by the promoter of glial fibrillary acidic protein, leading to expression in zebrafish glia. To identify agents that disrupt the BBB including inhibitors of ABCB1 and ABCG2, we identified NanoLuc substrates that are also transported by P-gp, ABCG2, and their zebrafish homologs. These substrates will elevate the amount of bioluminescent light produced in the transgenic zebrafish with BBB disrpution. We transfected HEK-293 cells with both NanoLuc and human ABCB1 or ABCG2, or their zebrafish homologs Abcb4 and Abcg2a, which are functionally homologous to human P-gp and ABCG2, respectively, and expressed at the zebrafish BBB. We evaluated the brightness of ten NanoLuc substrates, then screened the eight brightest for their ability to be effluxed by the ABC transporters. We identified one ABCB1 substrate, two Abcb4 substrates, six ABCG2 substrates, and four Abcg2a substrates. These data will aid in the development of a transgenic zebrafish model of the BBB to identify novel BBB disruptors and should prove useful in the development of other animal models that use NanoLuc as a reporter. Significance Statement The ATP-Binding Cassette (ABC) transporters ABCB1 and ABCG2 at the blood-brain barrier (BBB) hinder pharmacological treatment of brain-related diseases. Consequently, there is a need for tools to identify BBB disruptors. We conducted a screen of ten NanoLuciferase substrates, identifying the brightest and those that were transported by human and zebrafish ABC transporters at the BBB. This work supports and complements our development of a transgenic zebrafish model, in which NanoLuciferase is expressed within glial cells, enabling detection of BBB disruption.


A Database Tool Integrating Genomic and Pharmacologic Data from Adrenocortical Carcinoma (ACC) Cell Lines, PDX, and Patient Samples

August 2024

·

66 Reads

·

1 Citation

Cancer Research Communications

Adrenocortical carcinoma (ACC) is a rare and highly heterogeneous disease with a notably poor prognosis due to significant challenges in diagnosis and treatment. Emphasizing on the importance of precision medicine, there is an increasing need for comprehensive genomic resources alongside well-developed experimental models to devise personalized therapeutic strategies. We present ACC_CellMinerCDB, a substantive genomic and drug sensitivity database (available at https://discover.nci.nih.gov/acc_cellminercdb) comprising ACC cell lines, patient-derived xenografts, surgical samples, and responses to more than 2,400 drugs examined by the NCI and National Center for Advancing Translational Sciences. This database exposes shared genomic pathways among ACC cell lines and surgical samples, thus authenticating the cell lines as research models. It also allows exploration of pertinent treatment markers such as MDR-1, SOAT1, MGMT, MMR, and SLFN11 and introduces the potential to repurpose agents like temozolomide for ACC therapy. ACC_CellMinerCDB provides the foundation for exploring larger preclinical ACC models. Significance ACC_CellMinerCDB, a comprehensive database of cell lines, patient-derived xenografts, surgical samples, and drug responses, reveals shared genomic pathways and treatment-relevant markers in ACC. This resource offers insights into potential therapeutic targets and the opportunity to repurpose existing drugs for ACC therapy.



Transcriptional Characterization of Resistance in Early Drug Response

July 2024

·

18 Reads

Therapeutic resistance is the leading cause of treatment failure and death from cancer. While resistance can be driven by genetic mutations, mounting evidence also points to an epigenetic basis of resistance. Much of this epigenetic, or non-genetic, resistance has been attributed to drug-resistant transcriptional cell states that are either induced by drug treatment or pre-exist in a fraction of cells selected by treatment. However, the extent to which long-term resistance is manifested in the early inherent cellular response to drugs is poorly understood, and which aspects, if any, of this early response to drug-induced transcriptional response are evolutionarily conserved properties of cells. To address these questions, we integrate datasets of long-term drug resistance and early drug response data across multiple cell lines with drug response and resistance data from bacteria and yeast. Our findings suggest that cancer cell states in both the drug naive populations as well as in populations shortly after treatment share transcriptional properties with fully established resistant cell populations, and CRISPR-cas9 knockout of transcription factors predicted to regulate the resistant transcriptional programs result in increased drug sensitivity. Furthermore, the resistance states manifested as early drug response are evolutionarily conserved. Finally, we show that early resistant states discriminate responders from non-responders across multiple human cancer trials.


A whole-genome CRISPR screen identifies the spindle accessory checkpoint as a locus of nab-paclitaxel resistance in a pancreatic cancer cell line

July 2024

·

17 Reads

·

1 Citation

Pancreatic adenocarcinoma is one of the most aggressive and lethal forms of cancer. Chemotherapy is the primary treatment for pancreatic cancer, but resistance to the drugs used remains a major challenge. A genome-wide CRISPR interference and knockout screen in the PANC-1 cell line with the drug nab-paclitaxel has identified a group of spindle assembly checkpoint (SAC) genes that enhance survival in nab-paclitaxel. Knockdown of these SAC genes (BUB1B, BUB3, and TTK) attenuates paclitaxel-induced cell death. Cells treated with the small molecule inhibitors BAY 1217389 or MPI 0479605, targeting the threonine tyrosine kinase (TTK), also enhance survival in paclitaxel. Overexpression of these SAC genes does not affect sensitivity to paclitaxel. These discoveries have helped to elucidate the mechanisms behind paclitaxel cytotoxicity. The outcomes of this investigation may pave the way for a deeper comprehension of the diverse responses of pancreatic cancer to therapies including paclitaxel. Additionally, they could facilitate the formulation of novel treatment approaches for pancreatic cancer.


High BTK expression in glioma tumors and vasculature. High BTK expression seen in grade IV gliomas with highest expression in glioblastoma tumors as noted by CGGA (left) and TCGA (right) plotted data (a). Representative low and high BTK expression staining in glioblastoma TMA. Low power magnification of BTK expression seen in tumor cells and high magnification is BTK expression of endothelium. Red arrows denote vascular staining (b). High BTK expression scoring predominately seen in both tissue and vascular staining, with table delineating sole versus dual BTK expression, ****p < 0.001 (c)
Ibrutinib disrupts brain endothelial integrity and inhibits ABC transporter function. Brain endothelial cell viability is not affected by ibrutinib treatment at varied doses (a). Dose-dependently, ibrutinib decreases brain endothelial cell–cell impedance, significantly 2 h after treatment seen with 5 and 10 µM with subsequent cell index plateau (*p < 0.05, **p < 0.005) (b). Bicellular junction protein ZO-1 and tricellular junction protein MarvelD2 was significantly decreased 2 h after 10 µM ibrutinib treatment (c). Junctional mRNA expression also decreased from 2 to 24 h after 10 µM treatment, without a washout period seen in tjp1 (ZO-1), MarvelD2 (tricellulin), Ocln (occludin), Cldn5 (claudin-5), Lsr (lipolysis stimulated lipoprotein receptor/angulin-1), and cldn3 (claudin-3) (d). High baseline BTK expression seen in both cytoplasm and nucleus of brain endothelial cells. Confirmatory immunostaining of ZO-1 expression demonstrated decreased tight junctional linear staining at 4 h, with rearrangement closer to baseline regarding adhesion expression by 24 h. (e). Silencing of BTK with siBTK results in decreased cell–cell impedance transiently and impaired tight junction gene expression (***p < 0.0005, ****p < 0.0001) (f, g). Ibrutinib dose-dependently inhibited Abcb1 function to increase rhodamine accumulation with higher FITC-H fluorescence measurement causing a shift of amplitude to the right, comparative to valspodar (ABCB1 inhibitor) treated cells (h). Monolayer endothelial cells treated with ibrutinib on transwells resulted in approximately 26% higher sodium fluorescein permeability compared with control treatment 24 h later (****p < 0.0001) (i)
Ibrutinib hinders glioma cell migration and Abcb1 efflux and viability in combination with doxil. Varied ibrutinib dosing does not influence S635 rat glioma cell viability after 24 h (a). Glioma cell migration was influenced greatest by 10 µM ibrutinib 36–48 h after treatment compared with control and 1 µM therapy as evidenced by decreased cell migration to serum containing fetal bovine-serum (*p < 0.05) (b). Rhodamine efflux as a measure of Abcb1 function demonstrates both 10 µM ibrutinib effectively decreased efflux akin to known inhibitor valspodar (c). Combination therapy of ibrutinib with doxil found dose-dependent cooperation to hinder cell viability after 48 h exposure to therapy (***p < 0.001****p < 0.0001) (d). Combined ibrutinib (10 µM) and doxil (10 and 100 µM) resulted in increased caspase3/7 apoptosis activity compared with single therapy (****p < 0.0001) (e)
Combination ibrutinib with doxil impairs glioma model growth and prolongs survival. Treatment schema denoting repeat drug therapies, image timing, doxil blood/tissue concentrations and survival studies (a). Repeat ibrutinib with doxil therapy does no influence doxil plasma concentrations yet increases brain tumor doxil concentrations by approximately 32% (*p < 0.05) (b). Brain MRIs reflect additive ibrutinib effect of ibrutinib compared to vehicle (left panel) doxil alone (left middle panel), ibrutinib alone (right middle panel) or ibrutinib + doxil (right panel) as seen by statistically significant volumetric decrease as denoted via graphed values (*p < 0.05) (c). H/E staining demonstrates the lessened tumor size with doxil treatment alone and with combination therapy (d). 3kD dextran extravasation studies revealed ibrutinib can increase the blood-tumor barrier to larger agents, specifically within the peritumoral tissue region (*p < 0.01) (e). Co-staining of CD31/Claudin-5 on tumor, peritumoral and distant sites areas did not demonstrate any statistical differences in co-localized expression. (f). Prolonged survival seen with combination therapy with a median survival of 27 days versus 16 days for control therapy (****p < 0.0001) (*p < 0.05) (g)
Ibrutinib aids doxil entry to hinder glioma progression. An intact blood–brain barrier limits doxil permeability through brain endothelial tight junction integrity and ABCB1 efflux activity. In the context of the blood-tumor barrier treated with ibrutinib, junctional expression is decreased, ABCB1 function is inhibited and doxil therapy entry is enhanced to delay glioma migration and growth for prolonged survival. Images created via Biorender
Ibrutinib disrupts blood-tumor barrier integrity and prolongs survival in rodent glioma model

April 2024

·

79 Reads

·

1 Citation

Acta Neuropathologica Communications

In malignant glioma, cytotoxic drugs are often inhibited from accessing the tumor site due to the blood-tumor barrier (BTB). Ibrutinib, FDA-approved lymphoma agent, inhibits Bruton tyrosine kinase (BTK) and has previously been shown to independently impair aortic endothelial adhesion and increase rodent glioma model survival in combination with cytotoxic therapy. Yet additional research is required to understand ibrutinib’s effect on BTB function. In this study, we detail baseline BTK expression in glioma cells and its surrounding vasculature, then measure endothelial junctional expression/function changes with varied ibrutinib doses in vitro. Rat glioma cells and rodent glioma models were treated with ibrutinib alone (1–10 µM and 25 mg/kg) and in combination with doxil (10–100 µM and 3 mg/kg) to assess additive effects on viability, drug concentrations, tumor volume, endothelial junctional expression and survival. We found that ibrutinib, in a dose-dependent manner, decreased brain endothelial cell–cell adhesion over 24 h, without affecting endothelial cell viability (p < 0.005). Expression of tight junction gene and protein expression was decreased maximally 4 h after administration, along with inhibition of efflux transporter, ABCB1, activity. We demonstrated an additive effect of ibrutinib with doxil on rat glioma cells, as seen by a significant reduction in cell viability (p < 0.001) and increased CNS doxil concentration in the brain (56 ng/mL doxil alone vs. 74.6 ng/mL combination, p < 0.05). Finally, Ibrutinib, combined with doxil, prolonged median survival in rodent glioma models (27 vs. 16 days, p < 0.0001) with brain imaging showing a − 53% versus − 75% volume change with doxil alone versus combination therapy (p < 0.05). These findings indicate ibrutinib’s ability to increase brain endothelial permeability via junctional disruption and efflux inhibition, to increase BTB drug entry and prolong rodent glioma model survival. Our results motivate the need to identify other BTB modifiers, all with the intent of improving survival and reducing systemic toxicities.


Citations (63)


... Cancer cells, for example, can evolve to lose targeted antigens, thus evading the engineered CAR T cells, a phenomenon known as antigen-loss relapse (21). Efficacy can be increased by combining CAR T cell therapy with other vaccines, ICIs, oncolytic viruses, or small molecules such as ibrutinib or lenalidomide (21) that are brain penetrant (155,156). Furthermore, ibrutinib increases survival in rodent glioma models (156); lenalidomide may help prevent T cell exhaustion (21). Within the targeted tumor, diverse cell populations add to the complexity of immunotherapeutic approaches, but recent data indicates that immune triads-a close interaction between DCs, CD4 + T cells, and CD8 + T cells, working synergistically, can dramatically eliminate solid tumors by reprogramming the CD8 + T cell to become functional and tumor cytolytic for a range of cancers (41). ...

Reference:

The transformative potential of mRNA vaccines for glioblastoma and human cancer: technological advances and translation to clinical trials
Ibrutinib disrupts blood-tumor barrier integrity and prolongs survival in rodent glioma model

Acta Neuropathologica Communications

... Like humans, zebrafish have a functioning BBB comprised of endothelial cells that form tight junction complexes via claudin-5 and zona occludens-1 that physically limit drug entry into the brain (Jeong et al., 2008). Our group has recently characterized the different zebrafish homologs of human ABCB1 and ABCG2 and demonstrated that Abcb4 and Abcg2a (Thomas et al., 2024) have a substrate and inhibitor profile similar to those of their respective human equivalents and are expressed at the zebrafish BBB. With these functional similarities to the human BBB, and considering the potential for high throughput assays using zebrafish, they are a logical choice for a high-throughput model of the BBB . ...

Abcg2a is the functional homolog of human ABCG2 expressed at the zebrafish blood–brain barrier

Fluids and Barriers of the CNS

... Upon intravenous injection of the nanodrug, the photosensitizer is passively targeted to the tumor cells, enabling photodynamic therapy at the tumor location via laser irradiation [46]. The generated reactive oxygen species (ROS) are highly oxidative, capable of destroying cellular components like proteins, nucleic acids, and lipids, and producing apoptosis or necrosis [47,48]. ...

Carrier‐Free, Amorphous Verteporfin Nanodrug for Enhanced Photodynamic Cancer Therapy and Brain Drug Delivery

... METTL7A and METTL7B were renamed TMT1A and TMT1B, respectively, to reflect their activity. We discovered that TMT1A was overexpressed in a series of cell lines selected for resistance to romidepsin, a histone deacetylase (HDAC) inhibitor (HDACi) [9]. Romidepsin is a prodrug that harbors a disulfide bond that must be reduced to yield the active form of the molecule. ...

The methyltransferases METTL7A and METTL7B confer resistance to thiol-based histone deacetylase inhibitors
  • Citing Article
  • December 2023

Molecular Cancer Therapeutics

... In animals, drug resistance mechanisms often involve dynamic interactions between immune responses and cellular adaptations. Animals under the influence of external stresses can influence drug resistance by modulating the expression of membrane proteins that alter the composition and function of cell membranes (Thomas et al. 2023;Duvivier et al. 2024). For example, in mouse models, UBP1 can produce drug resistance to anti-malarial drugs such as mefloquine, benzfluorenol, and piperaquine by altering the localisation and direction of substrate transport of MDR1 (Xu, Lin, et al. 2024). ...

Progress in characterizing ABC multidrug transporters in zebrafish
  • Citing Article
  • December 2023

Drug Resistance Updates

... The MddH protein shared 31/50% and 34/53% amino acid sequence identity/similarity to the human thiol S-methyltransferases TMT1A and TMT1B, respectively, and their AlphaFold 26 predicted structures were similar, particularly in their central and C-terminal regions, containing the conserved central GxGxG binding motif 21 for SAM binding ( Supplementary Fig. 2a). The predicted MddH structure was comparatively more compact than for TMT1A and TMT1B, lacked an extended N-terminal 'hooked' helical region and a conserved aspartate residue at position 98 previously implicated in SAM binding 21 , which was a glutamate at position 63 in MddH ( Supplementary Fig. 2b). These observations support the hypothesis that MddH was a thiol S-methyltransferase similar to TMT1A and TMT1B. ...

The thiol methyltransferase activity of TMT1A (METTL7A) is conserved across species

... Among the important recent milestones is indotecan receiving the FDA orphan drug designation in malignant glioma [198]. A proof-of-concept study [199] showed PTEN-deficient glioma cells particularly sensitive to the drug; it also showed synergistic cytotoxicity with niraparib, another PARP inhibitor. Additionally, SLFN11 was shown as one of the dominant drug determinants for the three investigational indenoisoquinolines, showing synergism with a PARP inhibitor olaparib [200]. ...

Combined inhibition of TOP1 and PARP: a synergistic therapeutic strategy for glioblastoma with PTEN deficiency
  • Citing Article
  • August 2023

Neuro-Oncology Advances

... A prominent concern in oncological treatment is the phenomenon of drug resistance, which can be delineated into intrinsic and acquired categories. Intrinsic resistance pertains to the inherent capacity of cancer cells to endure the effects of pharmacological agents, often attributable to preexisting genetic mutations or the activation of survival pathways [5,6]. For example, numerous tumors overexpress ATP-binding cassette (ABC) transporters, which actively eliminate chemotherapeutic agents, consequently diminishing their intracellular concentrations and therapeutic efficacy [7]. ...

New mechanisms of multidrug resistance: an introduction to the Cancer Drug Resistance special collection

Cancer Drug Resistance

... While ER has been shown to be a substrate of P-gp in certain P-gp-expressing tumor cells [22], in this study, NCI/ADR-RES cells showed no resistance to ER or RSL3 ( Figure 1A,C). While ER may also be a substrate for BCRP [23], our studies indicated that BCRP-expressing MCF-7/MXR cells were more sensitive to both ER and RSL3 compared to the parent MCF-7 cells ( Figure 1B,D), suggesting that ER was not a substrate for BCRP in MCF-7/MXR cells. ...

The multidrug resistance transporter P-glycoprotein confers resistance to ferroptosis inducers

Cancer Drug Resistance

... However, these studies were not specifically performed in mIDH malignancies, and researchers are optimistic that more recently designed NAMPT-directed therapies and combinations may yield improved results [134]. Lastly, high throughput drug screening revealed that mIDH1 glioma cells may be susceptible to the CDK9 inhibitor zotiraciclib, possibly due to increased vulnerability to ETC dysfunction [113,135]. If deemed safe and effective in ongoing clinical trials in glioma, then its use in mIDH1/2 CCA warrants consideration. ...

Exploiting the therapeutic vulnerability of IDH-mutant gliomas with zotiraciclib
  • Citing Preprint
  • July 2023