Argun Akcakanat’s research while affiliated with University of Texas MD Anderson Cancer Center 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 (91)


Efficacy of ATR Kinase Inhibitor Elimusertib Monotherapy or Combination in Tumors with DNA Damage Response Pathway and Other Genomic Alterations
  • Article

April 2025

·

13 Reads

Kaushik Varadarajan

·

·

Kurt W. Evans

·

[...]

·

Funda Meric-Bernstam

The ataxia telangiectasia and RAD3-related (ATR) kinase functions with ataxia telangiectasia mutated (ATM) kinase as a modulator of DNA damage response (DDR). We assessed the antitumor effects of the ATR inhibitor elimusertib (BAY 1895344) in patient-derived xenograft (PDX) models with DDR alterations. Antitumor activity was assessed by change in tumor volume (TV) from baseline. Responses were categorized as follows: partial response (PR): >30% decrease in TV; >20% increase in TV: progressive disease (PD); and non-PR/PD: stable disease (SD). Event-free survival was defined as time for tumor doubling (EFS-2). Of 21 PDX models tested, 11 had significant prolongation of EFS-2 with elimusertib monotherapy. Four models had a PR and 4 had SD. PR/SD was observed in 2 of 5 models with ATM loss on IHC and in models with a variety of alterations in DDR genes, including BRCA1/2 and ATM. Elimusertib prolonged EFS-2 in three of 5 models with known PARP inhibitor resistance. Pharmacodynamic studies conducted in 4 PDX models showed an increase in DNA damage markers. PI3K/mTOR pathway signaling increased in 2 of 4 models. The combination of the PI3K inhibitor copanlisib with elimusertib enhanced EFS-2 compared to monotherapy in 3 of 11 models tested. The combination of elimusertib with the poly (ADP-ribose) polymerase (PARP) inhibitor niraparib enhanced antitumor activity compared to single agents in PARP-resistant PDX models. Our study shows that ATR inhibition has antitumor activity, including in models with both intrinsic and acquired PARP inhibitor resistance. Further work is needed to better refine patient selection for ATR-based therapies.


Figure 1. TROP2 membrane expression in BCXs and matched tumor samples. A, TROP2 membrane expression (H-score) shown for 52 PDX models (as determined by IHC) relative to HR and HER2 status shown. HR + is defined as greater than 5% expression of estrogen receptor or progesterone receptor. B, Representative images of TROP2-positive (BCX.017) and -negative (BCX.011) IHC. C, TROP2 membrane expression (H-score) was assessed in 27 patient tumors and matched BCXs by IHC. D, TROP2 H-score in BCXs correlated with TROP2 expression in matched patients (r ¼ 0.6991; P < 0.0001).
Figure 3. Dato-DXd response correlates with TROP2 expression in BCXs. A, Percentage tumor growth change and growth inhibition (T/C ratio) for models shown for BCXs with 10 mg/kg Dato-DXd or 10 mg/kg IgG-DXd. Metrics shown were calculated at 21 days of treatment unless indicated otherwise. TROP2 and SLFN11 expression by IHC as well as genomic alterations in actionable genes are shown below the X-axis. TROP2 membranous expression was scored by H-score (0-300), and SLFN11 nuclear expression was scored by H-score. B, TROP2 membrane expression by IHC compared between BCXs that responded to Dato-DXd and BCXs that did not. C, TROP2 expression as determined by reverse phase protein array compared between BCXs that responded to Dato-DXd and BCXs that did not. D, TROP2 (TACSTD2) as determined by RNA-seq compared between BCXs that responded to Dato-DXd and BCXs that did not. E and F, Improvement in T/C and EFS with 10 mg/kg Dato-DXd vs. 10 mg/kg IgG-DXd correlates with TROP2 H-score.
Antitumor Activity and Biomarker Analysis for TROP2 Antibody–Drug Conjugate Datopotamab Deruxtecan in Patient-Derived Breast Cancer Xenograft Models
  • Article
  • Full-text available

November 2024

·

22 Reads

·

2 Citations

Background: Datopotamab deruxtecan (Dato-DXd), is a humanized anti-TROP2 IgG1 monoclonal antibody linked to a potent topoisomerase I inhibitor payload (DXd). Dato-DXd has already shown antitumor activity in breast cancer; however, the determinants of response, including the importance of TROP2 expression, remain unclear. We tested the activity of Dato-DXd in a panel of breast cancer patient-derived xenografts (BCXs) varying in TROP2 expression. Methods: The antitumor activity of Dato-DXd and isotype-control-DXd (IgG-DXd) was assessed against 11 BCXs varying in TROP2 expression, 10 representing tumors post-neoadjuvant chemotherapy. Pharmacodynamic effects were assessed at 24 and 72 hours. The effects of TROP2 expression on Dato-DXd activity was assessed in vitro and in vivo using viral overexpression in BCX-derived cell lines. Results: Models differed in their sensitivity to both Dato-DXd and IgG-DXd. Dato-DXd (10 mg/kg) led to objective response in 4 (36%) models and statistically significant prolongation of event-free survival (EFS) in 8 (73%) models while IgG-DXd (10 mg/kg) led to response in 1 (9%) and prolonged EFS in 3 (27%) models. TROP2 RNA and protein was significantly higher in Dato-DXd-sensitive models. In isogenic cell lines derived from Dato-DXd-resistant BCXs, overexpression of TROP2 conferred Dato-DXd antitumor activity in vitro and in vivo. Dato-DXd increased γH2AX and phospho-KAP1 in the 2 Dato-DXd-sensitive BCXs but not in a Dato-DXd-resistant BCX. In Dato-DXd-sensitive models, antitumor activity was enhanced in combination with PARP inhibitor, olaparib. Conclusion: Dato-DXd is active in breast cancer models. Dato-DXd has TROP2 dependent and independent mediators of activity; however, high TROP2 expression enhances Dato-DXd antitumor activity.

Download

Heterogeneous Profile of ROR1 Protein Expression across Tumor Types

May 2024

·

52 Reads

·

3 Citations

Simple Summary The research explores the potential of the Wnt receptor ROR1 as a target for cancer treatment, an area gaining significant attention. Despite ongoing therapeutic investigations, the understanding of ROR1’s presence in different tissues remains limited. To address this, the study conducted immunohistochemistry analyses on a large variety of tumor types, including sarcomas and carcinomas, revealing a diverse ROR1 expression pattern. Notably, high ROR1 prevalence was observed in mesothelioma, liposarcoma, gastrointestinal stromal tumors, and uterine endometrioid carcinoma. Conversely, other cancers showed lower expression. These findings highlight the potential for ROR1-targeted therapies, particularly in mesothelioma, which exhibits frequent and high ROR1 expression, suggesting a novel therapeutic avenue. This research may inform the development of more effective cancer treatments targeting ROR1. Abstract The Wnt receptor ROR1 has generated increased interest as a cancer therapeutic target. Research on several therapeutic approaches involving this receptor is ongoing; however, ROR1 tissue expression remains understudied. We performed an immunohistochemistry analysis of ROR1 protein expression in a large cohort of multiple tumor and histologic types. We analyzed 12 anonymized multi-tumor tissue microarrays (TMAs), including mesothelioma, esophageal and upper gastrointestinal carcinomas, and uterine endometrioid carcinoma, among other tumor types. Additionally, we studied 5 different sarcoma types of TMAs and 6 patient-derived xenografts (PDX) TMAs developed from 19 different anatomic sites and tumor histologic types. A total of 1142 patient cases from different histologic types and 140 PDXs placed in TMAs were evaluated. Pathologists assessed the percentage of tumor cells in each case that were positive for ROR1 and the intensity of staining. For determining the prevalence of staining for each tumor type, a case was considered positive if >1% of its tumor cells showed ROR1 staining. Our immunohistochemistry assays revealed a heterogeneous ROR1 expression profile. A high prevalence of ROR1 expression was found in mesothelioma (84.6%), liposarcoma (36.1%), gastrointestinal stromal tumors (33.3%), and uterine endometrioid carcinoma (28.9%). Other histologic types such as breast, lung, renal cell, hepatocellular, urothelial carcinoma, and colon carcinomas; glioblastoma; cholangiocarcinoma; and leiomyosarcoma showed less ROR1 overall expression, ranging between 0.9 and 13%. No ROR1 expression was seen in mesenchymal chondrosarcoma, rhabdomyosarcoma, or gastric adenocarcinoma cases. Overall, ROR1 expression was relatively infrequent and low in most tumor types investigated; however, ROR1 expression was infrequent but high in selected tumor types, such as gastroesophageal GIST, suggesting that ROR1 prescreening may be preferable for those indications. Further, mesothelioma exhibited frequent and high levels of ROR1 expression, which represents a previously unrecognized therapeutic opportunity. These findings can contribute to the development of ROR1-targeted therapies.


Abstract 4536: MTAP loss alters the epigenetic landscape and demonstrates superior therapeutic sensitivity to concomitant PRMT5 and PARP inhibition in cholangiocarcinoma

March 2024

·

16 Reads

5’-methylthioadenosine phosphorylase (MTAP), is deleted in 15% of all cancers (estimated 100,000 patients per year in the United States), including 12-15% of cholangiocarcinoma (CCA) cases. MTAP deficiency accumulates 5’-methylthioadenosine (MTA) that subsequently binds protein arginine methyl transferase 5 (PRMT5) and inhibits its activity. This renders selective targeting of MTAP null tumors with agents (MRTX1719, AMG193, TNG462, TNG908) targeting MTA bound PRMT5 (PRMT5:MTA), while sparing surrounding normal MTAP wild-type (WT) tissue. Early signs of clinical activity was seen with MRTX1719, including objective responses in patients with MTAP loss tumors, including biliary tract cancer from the phase I/II study (NCT05245500). Despite these advances, prolonged targeted therapy poses the risk of development of acquired resistance demanding the need for better therapeutic strategies. To address this pressing need, we co-treated CCA cell lines with MRTX1719 and PARP inhibitor olaparib. We hypothesize that since PRMT5 is a critical regulator of splicing and DNA damage pathways, inhibition of PRMT5 disrupts these downstream processes creating an additional vulnerability to DNA damage inducing agents such as PARP inhibitors. Our preliminary data combining MRTX1719 with olaparib in three MTAP loss CCA cell lines (RBE, YSCCC, TFK1) show synergy (combination index; CI<1), and significant reduction in cell viability as well as colony formation ability when compared to monotherapy in vitro. To further gain mechanistic insights into the biology of MTAP loss, we stably expressed MTAP in MTAP null CCA cell line, RBE and demonstrated that MTAP loss drives tumorigenesis in vivo highlighting its tumor suppressor function in CCA. Mass spectrometry-based profiling of histone modifications from the isogenic RBE cell lines showed heightened methylation and reduced acetylation marks in the absence of MTAP. We also noted substantial differences in the distribution of splicing events, with increased intron retention, 3’ alternative splicing and decreased exon skipping as a result of MTAP loss. Similar differences in proportion of splicing events were also observed in MTAP loss CCA patient derived xenografts as compared to WT models. Collectively, these data implicate MTAP to be a crucial player in modulation of the chromatin and splicing events that may drive therapeutic response to PRMT5 inhibition. Citation Format: Pooja A. Shah, Ajay Kumar Saw, William Padron, Ming Zhao, Argun Akcakanat, Kurt Evans, Funda Meric-Bernstam, Milind Javle, Jordi Rodon, Kunal Rai. MTAP loss alters the epigenetic landscape and demonstrates superior therapeutic sensitivity to concomitant PRMT5 and PARP inhibition in cholangiocarcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 4536.


Establishment of co-clinical trial PDX models developed on the zanidatamab phase I study. A, Schematic of PDX development from pretreatment/screening biopsies on the zanidatamab phase I study. Tumors are implanted into NSG mice and early passage PDX tumors undergo WES, short-tandem repeat (STR) testing, CD45 IHC, and HER2 IHC prior to in vivo testing in nude mice. B, PFS durations are shorter in patients where PDXs were successfully established (blue) in comparison with patients where PDXs did not take (orange). C, Overall survival (OS) durations are shorter in patients where PDXs were successfully established (blue) in comparison with patients where PDXs did not take (orange). D, Diversity of HER2 expressing tumor types for which PDXs were developed. E, Spider plot showing percent (%) change in baseline as sum diameters of target lesions (y-axis) by weeks on trial (x-axis). CD45, leukocyte common antigen.
HER2 status and landscape of genomic alterations in patients and corresponding PDX models. PDXs were stratified by pretreatment and postprogression status, with prior therapies (chemotherapy, targeted therapy, and HER2 therapy) and primary tumor type reported. Waterfall plot depicting best response (percent change in baseline from sum of target lesions). PDX HER2 status reported by IHC (0, 1+, 2+, or 3+ expression) and ERBB2 amplification was assessed by whole-exome sequencing. Somatic alterations (mutations, indels, amplifications, and deletions) in cancer-related genes were reported by in descending frequency. Pre-zani, PDX developed before treatment; Post-zani, PDX developed after progression.
Zanidatamab activity in HER2-expressing xenografts correlates with patient clinical responses. Treatments were performed at 4, 8, and 16 mg/kg by intravenous tail-vein injection twice weekly. A, A cholangiocarcinoma pretreatment model (patient not enrolled in the trial). B, A gastric adenocarcinoma pretreatment model (patient had a PR on trial). C, A gastric adenocarcinoma pretreatment model (patient had a PR on trial). D, A colorectal carcinoma pretreatment model (patient had a PR on trial). E, A breast carcinoma pretreatment model (patient had a PD on trial). F, A cholangiocarcinoma pretreatment model (patient had a PD on trial). G and H, Two gallbladder adenocarcinoma postprogression models (developed from patients with who had PRs on trial before progressing). I, A colorectal carcinoma postprogression model (patient had a PR on trial before progressing). For each model, left plot represents growth curves shown as change in tumor volume, and the middle plot represents a waterfall plot of change in tumor volume at day 21 (except for PDX.003.285 where the waterfall plot shows day 24 and PDX.003.045 where waterfall plot shows day 14). On the right plot, the log-rank test was used to compare EFS-2 curves for each zanidatamab dose to the control arm.
Amplification of MET and MYC in the setting of acquired resistance to zanidatamab. A patient with HER2 3⁺/FISH⁺/ERBB2-amplified gallbladder cancer had NGS performed on a metastatic liver lesion and pre-zanidatamab PDX model (PDX.003.148) developed from an abdominal wall lesion which demonstrated ERBB2 amplification and HER2 3⁺ expression. The patient was enrolled in the zanidatamab phase I study and achieved a partial response best response −52%) and a post-zanidatamab PDX model (PDX.003.300) was developed at the time of progression. Both MET and MYC amplification emerged in the postprogression PDX model and were validated by IHC and FISH.
Overcoming acquired resistance to zanidatamab with MET inhibition. A, Three HER2⁺ cell lines were subjected to single-agent treatment with zanidatamab at serial dilutions for 72 hours, followed by SRB cell viability assay. IC50 was calculated using CalcuSyn. B, Three HER2⁺ cell lines were treated with zanidatamab and capmatinib or crizotinib at fixed ratios for 72 hours, followed by SRB cell viability assay. CI was calculated using CalcuSyn. C, HCC-1954 cells were seeded into 6-well plates at 1,000 cells/well. Cells were subjected to a single agent or combinatorial treatment at the indicated concentrations for 3 weeks, followed by crystal violet staining. Colony images were captured and colony areas were measured using ImageJ. D and E, Apoptosis assay. HCC-1954 cells were treated with single and combinatorial agents for 72 hours. Cells were probed with Annexin V FITC, followed by flow cytometry measurement. Percentage of Annexin V–positive apoptotic cells was calculated. T test was performed for P values. F, G and H,In vivo assessment of zanidatamab in combination with MET inhibitors, crizotinib (G) or capmatinib (H and I). Mice bearing PDX.003.300 were treated with 16 mg/kg zanidatamab with 6, 9, or 17 mg/kg capmatinib or 100 mg/kg crizotinib. High dose of crizotinib and capmatinib had strong antitumor effects on PDX.003.300. Notably, these two experiments were conducted together, using the same control groups but are being presented as two panels to help with visualization. The two lower doses of capmatinib (6 and 9 mg/kg) had fewer effects as single agent but demonstrated improved combination effects with zanidatamab.
Co-clinical Trial of Novel Bispecific Anti-HER2 Antibody Zanidatamab in Patient-Derived Xenografts

February 2024

·

41 Reads

·

16 Citations

Zanidatamab is a bispecific human epidermal growth factor receptor 2 (HER2)-targeted antibody that has demonstrated antitumor activity in a broad range of HER2-amplified/expressing solid tumors. We determined the antitumor activity of zanidatamab in patient-derived xenograft (PDX) models developed from pretreatment or postprogression biopsies on the first-in-human zanidatamab phase I study (NCT02892123). Of 36 tumors implanted, 19 PDX models were established (52.7% take rate) from 17 patients. Established PDXs represented a broad range of HER2-expressing cancers, and in vivo testing demonstrated an association between antitumor activity in PDXs and matched patients in 7 of 8 co-clinical models tested. We also identified amplification of MET as a potential mechanism of acquired resistance to zanidatamab and demonstrated that MET inhibitors have single-agent activity and can enhance zanidatamab activity in vitro and in vivo. These findings provide evidence that PDXs can be developed from pretreatment biopsies in clinical trials and may provide insight into mechanisms of resistance. Significance We demonstrate that PDXs can be developed from pretreatment and postprogression biopsies in clinical trials and may represent a powerful preclinical tool. We identified amplification of MET as a potential mechanism of acquired resistance to the HER2 inhibitor zanidatamab and MET inhibitors alone and in combination as a therapeutic strategy. This article is featured in Selected Articles from This Issue, p. 695


Fibroblast growth factor receptor (FGFR) alterations in breast cancer patient-derived xenografts (PDXs). (a) FGFR expression and genomic alterations from 22 PDXs generated from 21 patients. (b) Relative protein expression of breast cancer PDXs determined by reverse phase protein array (RPPA). (c) Relative FGFR1-4 mRNA expression in patient samples and matching PDX models. RNA expression is presented in log2-normalized reads per kilobase million (RPKM). A 2-sided paired Student t test was used to calculate P-values. (d) The correlation of expression between FGFR genes in patient samples and those in matching PDX models is shown. RNA expression is presented in log2-normalized RPKM. The Pearson correlation coefficient (r) was used to measure the statistical association between 2 variables.
Response of patient-derived xenografts (PDXs) to the fibroblast growth factor receptor (FGFR) inhibitor futibatinib. (a–i) Female mice bearing breast cancer PDXs (n = 5 per group) were treated orally with the vehicle control or futibatinib (15 mg/kg/day). The treatment was stopped at 28 days or when the tumor diameter reached 1.5 cm³, except for mice bearing the PDX.007 model, in which the treatment was stopped on day 110. Growth (left) and event-free survival duration (right, defined as days until tumor doubling) curves are shown for each model. Growth curve graphs show the mean change in TV (mm³) ± the standard errors of the means. (j) Tumor volume of mice in control and futibatinib groups were measured at the last day of vehicle treatment. Bars, error bars, and dots show mean tumor volume (mm³), SEM, and individual tumor volume of each mouse, respectively. C, control; F, futibatinib. (k) Relative tumor growth tumor volume treatment/control ratios (TV T/C ratio) were calculated on day 21. End point tumor volumes (TV) were calculated at the last day of vehicle treatment. Event free survival (EFS) was calculated at the last day of futibatinib treatment. *This model does not have RNAseq data.
Effects of FGFR2 alterations on cell sensitivity to fibroblast growth factor receptor (FGFR) inhibitors and FGFR signaling. Normal mammary epithelial MCF10A cells were transduced with FGFR2-BICC1 fusion, FGFR2 Y375C mutation, FGFR2 wild type (WT), and the vector control. (a) Cell viability was measured by a sulforhodamine B (SRB) assay. Bars show the mean OD570 ± the standard errors of the means. (b) Cells were cultured for 3 weeks. Cell colonies were stained, and the total colony area was quantitated. The comparisons of the total colony areas of the FGFR2 WT, FGFR2-BICC1, and FGFR2 Y375C cell lines to the vector control cell line. Bars show the mean total colony areas ± the standard errors of the means. Below, representative stained plates are shown. (c) Cells were and treated with a serial dilution of futibatinib for 4 days. Cell viability was assessed with an SRB assay, and the half-maximal inhibitory concentration (IC50) values were calculated. Bars show the mean IC50 values ± the standard errors of the means. (d) While being starved, cells were treated with fibroblast growth factor (FGF1) for 24 h and various doses of futibatinib for 4 days. Cell viability was assessed with an SRB assay, and IC50 values were calculated. Bars show mean IC50 values ± the standard errors of the means. (e) Cells were starved for 24 h and treated with futibatinib at 0.2 µM for 3 h 45 min, followed by FGF1 for 15 min. Immunoblotting was performed using antibodies against FGFR2, p-Akt (S473), Akt, p-ERK1/2 (T202/Y204), ERK1/2, and β-actin. (f) PDX.007CL cells were plated in spheroid plates and treated a panel of FGFR inhibitors for five days. A luminescence assay was used to determine cell viability and IC50 values were calculated. (g) MCF10A vector control and FGFR2 Y375C cells were treated with a panel of FGFR inhibitors for four days. Sulforhodamine B assay was used to determine cell viability and IC50 values were calculated.
FGFR mutations in patients with breast cancer. Breast cancer data in MD Anderson Cancer Center (MDACC), MSK-IMPACT, Metastatic Breast Cancer (MBC) Project, and The Cancer Genomics Project (TCGA) databases were downloaded and compiled. (a) The bars show the percentages of somatic FGFR mutations in patients with breast cancer in each of these databases. (b) The 4 databases were analyzed together, and all mutations on the individual genes were illustrated using the cBioPortal MutationMapper tool. (c) Percentages of samples with FGFR2 amplification (AMP) in each database. (d) Distribution of FGFR2 Y375C mutations in different tissues in the Catalogue of Somatic Mutations in Cancer (COSMIC) database.
Efficacy of futibatinib, an irreversible fibroblast growth factor receptor inhibitor, in FGFR-altered breast cancer

November 2023

·

99 Reads

·

11 Citations

Several alterations in fibroblast growth factor receptor (FGFR) genes have been found in breast cancer; however, they have not been well characterized as therapeutic targets. Futibatinib (TAS-120; Taiho) is a novel, selective, pan-FGFR inhibitor that inhibits FGFR1-4 at nanomolar concentrations. We sought to determine futibatinib’s efficacy in breast cancer models. Nine breast cancer patient–derived xenografts (PDXs) with various FGFR1-4 alterations and expression levels were treated with futibatinib. Antitumor efficacy was evaluated by change in tumor volume and time to tumor doubling. Alterations indicating sensitization to futibatinib in vivo were further characterized in vitro. FGFR gene expression between patient tumors and matching PDXs was significantly correlated; however, overall PDXs had higher FGFR3-4 expression. Futibatinib inhibited tumor growth in 3 of 9 PDXs, with tumor stabilization in an FGFR2-amplified model and prolonged regression (> 110 days) in an FGFR2 Y375C mutant/amplified model. FGFR2 overexpression and, to a greater extent, FGFR2 Y375C expression in MCF10A cells enhanced cell growth and sensitivity to futibatinib. Per institutional and public databases, FGFR2 mutations and amplifications had a population frequency of 1.1%–2.6% and 1.5%–2.5%, respectively, in breast cancer patients. FGFR2 alterations in breast cancer may represent infrequent but highly promising targets for futibatinib.


Fig. 1 TROP2 and TACSTD2 expression in breast cancer. A Breast cancer FFPE sections were assessed for TROP2 expression by IHC. Representative images demonstrate no TROP2expression (top left, H-score 0), low TROP2 expression (top right, H-score 57), medium TROP2 expression (bottom left, H-score 140), and high TROP2 expression (bottom right, H-score 290). B TROP2 positivity in breast cancer when stratified by histologic subtype. Of 41 cases evaluated, only 2 of 5 (40%) of metaplastic breast cases were TROP2 positive. C TROP2 expression in TMA of 106 breast tumors. Of the 97 non-metaplastic cases, 2 (2.1%) were TROP2 negative, 42 had low TROP2 expression (43.3%), 42 had medium TROP2 expression, and 11 had high TROP2 expression. Of the 9 metaplastic cases, 5 (55.6%) had no TROP2 expression, 3 (33.3%) had low TROP2 expression, and 1 (11.1%) had high TROP2 expression. D Comparison of TACSTD2 expression in metaplastic vs. non-metaplastic breast tumors in TCGA. Expression of TACSTD2 was higher in non-metaplastic tumors (p < 0.001, t-test).
Epigenetically upregulating TROP2 and SLFN11 enhances therapeutic efficacy of TROP2 antibody drug conjugate sacitizumab govitecan

August 2023

·

57 Reads

·

9 Citations

npj Breast Cancer

TROP2 antibody drug conjugates (ADCs) are under active development. We seek to determine whether we can enhance activity of TROP2 ADCs by increasing TROP2 expression. In metaplastic breast cancers (MpBC), there is limited expression of TROP2, and downregulating transcription factor ZEB1 upregulates E-cad and TROP2, thus sensitizing cancers to TROP2 ADC sacituzumab govitecan (SG). Demethylating agent decitabine decreases DNA methyltransferase expression and TROP2 promoter methylation and subsequently increases TROP2 expression. Decitabine treatment as well as overexpression of TROP2 significantly enhance SG antitumor activity. Decitabine also increases SLFN11, a biomarker of topoisomerase 1 inhibitor (TOP1) sensitivity and is synergistic with SG which has a TOP1 payload, in TROP2-expressing SLFN11-low BC cells. In conclusion, TROP2 and SLFN11 expression can be epigenetically modulated and the combination of demethylating agent decitabine with TROP2 ADCs may represent a novel therapeutic approach for tumors with low TROP2 or SLFN11 expression.



Abstract 6180: Antitumor activity of the ATR inhibitor Elimusertib in patient-derived xenograft models with DNA damage response pathway alterations

April 2023

·

20 Reads

Introduction: The ataxia telangiectasia and RAD3-related (ATR) kinase is a key component of the DNA damage response (DDR) and functions in conjunction with ataxia telangiectasia mutated (ATM). ATM loss or functional deficiency may enhance reliance on ATR signaling. Preclinical and clinical studies have pointed to a potential synthetic lethality of ATR inhibition and ATM loss. In this study, we sought to further elaborate potential candidates for ATR inhibition by testing the anti-tumor effect of elimusertib (BAY 1895344) in patient-derived tumor xenograft (PDX) models with DDR alterations including ATM loss. Methods: Patient derived xenograft (PDX) models were implanted into athymic nu/nu mice. Once tumors reached an appropriate size of approximately 200-400mm3 treatments were started. Mice were randomized (N= 3-8) to different treatment arms for assessment of anti-tumor activity of elimusertib as monotherapy and in combination therapy with copanlisib. Monotherapy treatment was performed with 20 mg/kg and 40 mg/kg doses, applied twice daily for 3 days with 4 days treatment break (BID 3 days on/4 days off). Efficacy read-out was either Event-Free Survival (EFS-2), defined as time for doubling of tumor volume relative to baseline, or response based on RECIST criteria:(partial response [PR]= >30% decrease, stable disease [SD]= between 20% increase and 30% decrease, and progressive disease [PD]= >20% increase). For assessment of pharmacodynamic effects, tumors were harvested 10 days after treatment start and analyzed by immunohistochemistry (IHC) and RPPA (Reverse Phase Proteomics Array). Results: Elimusertib was tested in 21 PDX models with various DDR alterations. Of those 11 showed a statistically significant prolongation of event free survival compared to control. In 4 PDX models elimusertib reached a partial response (PR), and another 4 models showed stable disease (SD). Amongst the 5 models with ATM protein Ioss (IHC), one was PR and another one SD. In 3 of 5 PARP inhibitor resistant there was statistically significant prolongation of EFS upon elimusertib treatment vs control. We found an increase in DNA damage in tumors, as indicated by increased level of ƳH2AX (on IHC and RPPA). We also detected an increase in PI3K/mTOR pathway signaling as determined by p-MTOR and pS6 (RPPA) in 2 of 4 models (PR and PD model) with different sensitivity to ATRi and PARPi alone. We tested the combination of elimusertib with the PI3K inhibitor copanlisib achieved a statistically significant increase of EFS compared to monotherapy in 3 of 11 models tested. Conclusion ATR inhibition shows potent monotherapy activity in selected PDX models that harbor DDR defects, including models with intrinsic and acquired PARPi resistance. Further work is needed to identify markers predicting sensitivity or resistance that providing rationale for synergistic combination therapies. Citation Format: Kaushik Varadarajan, Christian X. Pico, Kurt Evans, Maria G. Raso, Yasmeen Rizvi, Xiaofeng Zheng, Timothy P. Diperi, Bailiang Wang, Stephen Scott, Ming Zhao, Argun Akcakanat, Antje M. Wengner, Timothy A. Yap, Funda Meric-Bernstam. Antitumor activity of the ATR inhibitor Elimusertib in patient-derived xenograft models with DNA damage response pathway alterations. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6180.



Citations (53)


... Rechallenge with platinum-based chemotherapy or taxane plus anti-angiogenic may be an effective option for selected populations. Patients with tumors resistant to first-line treatments remain a refractory population that requires to be enrolled in clinical trials [40,42]. ...

Reference:

Unexpected Long-Term Survival and Downstaging in Oligometastatic Non-Small Cell Lung Cancer Treated with Multimodal Therapy
Antitumor Activity and Biomarker Analysis for TROP2 Antibody–Drug Conjugate Datopotamab Deruxtecan in Patient-Derived Breast Cancer Xenograft Models

... Studies have demonstrated a high frequency of ROR1 expression in several tumor types, including mesothelioma, liposarcoma, GISTs, and uterine endometrioid carcinoma. This finding underscores the potential of targeting ROR1 for the treatment of these cancers [47]. Furthermore, the identification of Wnt-regulated long noncoding RNAs (lncRNAs), such as CCAT5 and CCAT2, has revealed their critical roles in regulating STAT3 activity and promoting tumor development. ...

Heterogeneous Profile of ROR1 Protein Expression across Tumor Types

... Alongside these agents, several other novel HER2-targeted ADCs, including XMT-1522 [55], SYD985 [135], BAT8001 [136], ZW49 [137], are undergoing clinical trials. The efficacy and safety of these therapies for patients with low HER2-expressing GC have attracted considerable interest (Table 2). ...

Co-clinical Trial of Novel Bispecific Anti-HER2 Antibody Zanidatamab in Patient-Derived Xenografts

... FGFR-1 is the most commonly altered (49%) followed by FGFR-3 (23%) and FGFR-2 (19%), while FGFR-4 is the least frequently altered (7%). Futibatinib is FDA approved as an irreversible FGFR inhibitor used to treat FGFR-altered breast cancer (Saridogan et al. 2023). ...

Efficacy of futibatinib, an irreversible fibroblast growth factor receptor inhibitor, in FGFR-altered breast cancer

... Both preclinically and clinically, drug combinations are being explored to pharmacologically modulate or enhance target expression. For example, in a preclinical model, decitabine enhanced TROP2 expression in a metaplastic breast cancer model to potentiate the effect of sacituzumab govitecan 62 . While sacituzumab govitecan is currently FDA approved independent of TROP2 expression, methods to enhance target engagement may prove efficacious. ...

Epigenetically upregulating TROP2 and SLFN11 enhances therapeutic efficacy of TROP2 antibody drug conjugate sacitizumab govitecan

npj Breast Cancer

... Indeed, the field is moving in this direction; to bridge the gap between information gained from preclinical PDX studies and clinical trials, recent work published by the NCI PDXNet consortium presented recommendations that deal with the standardization of assessment of PDX growth and antitumour activity 143 . Having universally adopted standards for evaluating responses, and ways to make PDX models and data available publicly around the world, could lessen the time taken to identify novel therapies that translate to meaningful therapeutic responses for patients 144 . Additional publicly available data and validated model repositories include CancerModels.org, ...

PDXNet portal: patient-derived Xenograft model, data, workflow and tool discovery

NAR Cancer

... To address this, murine double minute 2 (MDM2) inhibitors have been developed to prevent p53 degradation and promote cancer cell death. Although these inhibitors have shown promise in early-phase trials across various cancers, the results so far in advanced liposarcoma have been disappointing, and specific data for chondrosarcoma are still lacking [20][21][22]. On the other hand, CDKN2A mutations are more amenable to therapeutic inhibition [23]. ...

Combined MEK/MDM2 inhibition demonstrates antitumor efficacy in TP53 wild-type thyroid and colorectal cancers with MAPK alterations

... Therefore, hyperkalemia may cause abnormal aggregation or degradation of neurofilament proteins by affecting cytoskeletal stability, which in turn causes increased release of sNfL, as has been demonstrated in a variety of neurological disorders. In addition, hyperkalemia may affect cell signaling pathways, such as activating microglia by activating N-methyl-D-aspartate (NMDA) receptors and increasing calcium inward flow, and may also activate microglia by increasing the release of inflammatory factors, such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6), which can directly stimulate microglia, prompting them to produce more inflammatory mediators, further exacerbating neuroinflammation and causing nerve damage (Bagheri-Yarmand et al., 2021;Liu et al., 2024;Merighi et al., 2022). Finally, hyperkalemia may trigger oxidative stress through several aspects, including affecting the process of intra-and extracellular ion exchange, leading to changes in cellular membrane potential and affecting normal cellular function and the normal operation of antioxidant defense mechanisms; the ionic changes triggered by hyperkalemia may also lead to mitochondrial dysfunction, affecting the process of oxidative phosphorylation, which may increase the production of reactive oxygen species (ROS); hyperkalemia can also affect the activity of antioxidant enzymes, such as superoxide dismutase (SOD) and catalase (CAT), which are the main defense mechanisms for scavenging ROS in the body. ...

RAC1 Alterations Induce Acquired Dabrafenib Resistance in Association with Anaplastic Transformation in a Papillary Thyroid Cancer Patient

... Beyond glycolysis, mitochondrial metabolism plays a dual role in resistance. While OXPHOS inhibition through metformin or IACS-10759 (a mitochondrial complex I inhibitor) depletes NAD + and suppresses DNA repair, making cells more dependent on PARPmediated repair and thus more sensitive to PARPi [107][108][109]. OXPHOS inhibition can also exacerbate oxidative stress, further diminishing cancer cell viability [110]. ...

Oxidative Phosphorylation Is a Metabolic Vulnerability in Chemotherapy-Resistant Triple-Negative Breast Cancer

... One CS PDX (CF8X) model is present in the patient-derived model repository of the National Cancer Institute (NCI-PDMR) (https://pdmr.cancer.gov) and one (WU-0064) in the PDXNet Consortium, both were used for extensive genomic and molecular studies [81]. Two conventional CS PDX models, CTG-1255 and CTG-2383 are commercially available at Champions Oncology, Inc. (Hackensack, NJ, USA) [82], and eight conventional CS PDXs are available at the Jackson Laboratory for drug screening studies [83]. ...

Comprehensive characterization of 536 patient-derived xenograft models prioritizes candidatesfor targeted treatment