251 results on '"H Kaufmann"'
Search Results
2. Characterization of patients with long-term responses to rucaparib treatment in recurrent ovarian cancer
- Author
-
Carol Aghajanian, Lee-may Chen, Scott H. Kaufmann, Amit M. Oza, Geoffrey I. Shapiro, Rebecca Kristeleit, Sandra Goble, Iain A. McNeish, Diane Provencher, Ana Oaknin, Alexandra Leary, Lara Maloney, Elizabeth M. Swisher, Isabelle Ray-Coquard, Howard A. Burris, Stephen Welch, Kevin K. Lin, Tanya Kwan, Ronnie Shapira-Frommer, Anna V. Tinker, David M. O'Malley, Robert L. Coleman, Cancer Research UK, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Oncology ,Indoles ,ANTITUMOR-ACTIVITY ,MONOTHERAPY ,Loss of Heterozygosity ,DOUBLE-BLIND ,chemistry.chemical_compound ,Ovarian carcinoma ,Aged, 80 and over ,Ovarian Neoplasms ,Clinical Trials, Phase I as Topic ,BRCA1 Protein ,OLAPARIB ,Obstetrics & Gynecology ,Obstetrics and Gynecology ,Structural variant ,Genomics ,Middle Aged ,PARP inhibitor ,SURVIVAL ,RAD51C ,Female ,Safety ,Life Sciences & Biomedicine ,Adult ,medicine.medical_specialty ,CARCINOMA ,Duration of response ,Poly(ADP-ribose) Polymerase Inhibitors ,Clinical Trials, Phase II as Topic ,Internal medicine ,GERMLINE ,Post-hoc analysis ,medicine ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Rucaparib ,Aged ,BRCA2 Protein ,Science & Technology ,MUTATIONS ,business.industry ,BRCA1 ,chemistry ,PARP INHIBITOR RUCAPARIB ,Recurrent Ovarian Cancer ,1114 Paediatrics and Reproductive Medicine ,Neoplasm Recurrence, Local ,business ,Recurrent Ovarian Carcinoma ,Follow-Up Studies - Abstract
Objective. To describe molecular and clinical characteristics of patients with high-grade recurrent ovarian carcinoma (HGOC) who had long-term responses to the poly(ADP-ribose) polymerase (PARP) inhibitor rucaparib. Methods. This post hoc analysis pooled patients from Study 10 (NCT01482715; Parts 2A and 2B; n = 54) and ARIEL2 (NCT01891344; Parts 1 and 2; n = 491). Patients with investigator-assessed complete or partial response per RECIST were classified based on duration of response (DOR): long (≥1 year), intermediate (6 months to
- Published
- 2021
- Full Text
- View/download PDF
3. Acquired RAD51C Promoter Methylation Loss Causes PARP Inhibitor Resistance in High-Grade Serous Ovarian Carcinoma
- Author
-
Mohammad Reza Eftekhariyan Ghamsari, Nadia Traficante, Nirashaa Bound, Cordelia D. McGehee, Inger Olesen, Kristy Shield-Artin, Clare L. Scott, Elizabeth Lieschke, Rachel M. Hurley, Anna deFazio, Ashan Musafer, Nicola Waddell, Marc R. Radke, Matthew Wakefield, Alexander Dobrovic, Ksenija Nesic, Genevieve Dall, Maria I. Harrell, Olga Kondrashova, Damien Kee, S. John Weroha, Scott H. Kaufmann, Katia Nones, David D.L. Bowtell, Cassandra J. Vandenberg, Orla McNally, Elizabeth M. Swisher, Gwo-Yaw Ho, and Zi Qing Chai
- Subjects
Cancer Research ,DNA repair ,Methylation ,Biology ,chemistry.chemical_compound ,Germline mutation ,Oncology ,CpG site ,chemistry ,DNA methylation ,PARP inhibitor ,Cancer research ,RAD51C ,Rucaparib - Abstract
In high-grade serous ovarian carcinoma (HGSC), deleterious mutations in DNA repair gene RAD51C are established drivers of defective homologous recombination and are emerging biomarkers of PARP inhibitor (PARPi) sensitivity. RAD51C promoter methylation (meRAD51C) is detected at similar frequencies to mutations, yet its effects on PARPi responses remain unresolved. In this study, three HGSC patient-derived xenograft (PDX) models with methylation at most or all examined CpG sites in the RAD51C promoter show responses to PARPi. Both complete and heterogeneous methylation patterns were associated with RAD51C gene silencing and homologous recombination deficiency (HRD). PDX models lost meRAD51C following treatment with PARPi rucaparib or niraparib, where a single unmethylated copy of RAD51C was sufficient to drive PARPi resistance. Genomic copy number profiling of one of the PDX models using SNP arrays revealed that this resistance was acquired independently in two genetically distinct lineages. In a cohort of 12 patients with RAD51C-methylated HGSC, various patterns of meRAD51C were associated with genomic “scarring,” indicative of HRD history, but exhibited no clear correlations with clinical outcome. Differences in methylation stability under treatment pressure were also observed between patients, where one HGSC was found to maintain meRAD51C after six lines of therapy (four platinum-based), whereas another HGSC sample was found to have heterozygous meRAD51C and elevated RAD51C gene expression (relative to homozygous meRAD51C controls) after only neoadjuvant chemotherapy. As meRAD51C loss in a single gene copy was sufficient to cause PARPi resistance in PDX, methylation zygosity should be carefully assessed in previously treated patients when considering PARPi therapy. Significance: Homozygous RAD51C methylation is a positive predictive biomarker for sensitivity to PARP inhibitors, whereas a single unmethylated gene copy is sufficient to confer resistance.
- Published
- 2021
- Full Text
- View/download PDF
4. CDK2-Mediated Upregulation of TNFα as a Mechanism of Selective Cytotoxicity in Acute Leukemia
- Author
-
Cordelia D. McGehee, Brian D. Koh, Annapoorna Venkatachalam, Scott H. Kaufmann, Husheng Ding, Karen S. Flatten, Hu Li, B. Douglas Smith, Judith E. Karp, Cristina Correia, Mira A. Kohorst, Paula A. Schneider, Mrinal M. Patnaik, Jonathan Webster, Larry M. Karnitz, Nicole D. Vincelette, Gabriel Ghiaur, X. Wei Meng, Kevin L. Peterson, Keith W. Pratz, and Sun Hee Lee
- Subjects
0301 basic medicine ,Cancer Research ,Apoptosis ,Mice, SCID ,Article ,Mice ,03 medical and health sciences ,Transactivation ,0302 clinical medicine ,Downregulation and upregulation ,Mice, Inbred NOD ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,Animals ,Humans ,Cytotoxic T cell ,Cell Proliferation ,Acute leukemia ,Tumor Necrosis Factor-alpha ,Kinase ,Chemistry ,Cyclin-Dependent Kinase 2 ,breakpoint cluster region ,Myeloid leukemia ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,Pyrazines ,030220 oncology & carcinogenesis ,Cancer research ,Pyrazoles ,Female ,Tumor necrosis factor alpha ,biological phenomena, cell phenomena, and immunity - Abstract
Although inhibitors of the kinases CHK1, ATR, and WEE1 are undergoing clinical testing, it remains unclear how these three classes of agents kill susceptible cells and whether they utilize the same cytotoxic mechanism. Here we observed that CHK1 inhibition induces apoptosis in a subset of acute leukemia cell lines in vitro, including TP53-null acute myeloid leukemia (AML) and BCR/ABL–positive acute lymphoid leukemia (ALL), and inhibits leukemic colony formation in clinical AML samples ex vivo. In further studies, downregulation or inhibition of CHK1 triggered signaling in sensitive human acute leukemia cell lines that involved CDK2 activation followed by AP1-dependent TNF transactivation, TNFα production, and engagement of a TNFR1- and BID-dependent apoptotic pathway. AML lines that were intrinsically resistant to CHK1 inhibition exhibited high CHK1 expression and were sensitized by CHK1 downregulation. Signaling through this same CDK2–AP1–TNF cytotoxic pathway was also initiated by ATR or WEE1 inhibitors in vitro and during CHK1 inhibitor treatment of AML xenografts in vivo. Collectively, these observations not only identify new contributors to the antileukemic cell action of CHK1, ATR, and WEE1 inhibitors, but also delineate a previously undescribed pathway leading from aberrant CDK2 activation to death ligand–induced killing that can potentially be exploited for acute leukemia treatment. Significance: This study demonstrates that replication checkpoint inhibitors can kill AML cells through a pathway involving AP1-mediated TNF gene activation and subsequent TP53-independent, TNFα-induced apoptosis, which can potentially be exploited clinically.
- Published
- 2021
- Full Text
- View/download PDF
5. Anastrozole has an Association between Degree of Estrogen Suppression and Outcomes in Early Breast Cancer and is a Ligand for Estrogen Receptor α
- Author
-
Scott H. Kaufmann, James N. Ingle, Paul E. Goss, Michael A. Walters, Cristina Correia, Bingshu E. Chen, Matthew E. Cuellar, Tanya L. Hoskin, Matthew J. Ellis, Liewei Wang, Bernhard Volz, Ravinder J. Singh, Vera J. Suman, Richard M. Weinshilboum, Barbara Goodnature, Junmei Cairns, Krishna R. Kalari, Tufia C. Haddad, Matthew P. Goetz, Zeruesenay Desta, Erin E. Carlson, Poulami Barman, Peter A. Fasching, and Lois E. Shepherd
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Antineoplastic Agents, Hormonal ,medicine.drug_class ,Estrogen receptor ,Anastrozole ,Breast Neoplasms ,Clinical Trials, Phase IV as Topic ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Exemestane ,Internal medicine ,medicine ,Humans ,Multicenter Studies as Topic ,Prospective Studies ,Aromatase ,Aged ,Randomized Controlled Trials as Topic ,030304 developmental biology ,Aged, 80 and over ,0303 health sciences ,biology ,business.industry ,Estrogen receptor binding ,Letrozole ,Estrogen Receptor alpha ,Estrogens ,Middle Aged ,Prognosis ,medicine.disease ,Clinical Trials, Phase III as Topic ,chemistry ,Estrogen ,Case-Control Studies ,030220 oncology & carcinogenesis ,biology.protein ,Female ,business ,Follow-Up Studies ,medicine.drug - Abstract
Purpose:To determine if the degree of estrogen suppression with aromatase inhibitors (AI: anastrozole, exemestane, letrozole) is associated with efficacy in early-stage breast cancer, and to examine for differences in the mechanism of action between the three AIs.Experimental Design:Matched case–control studies [247 matched sets from MA.27 (anastrozole vs. exemestane) and PreFace (letrozole) trials] were undertaken to assess whether estrone (E1) or estradiol (E2) concentrations after 6 months of adjuvant therapy were associated with risk of an early breast cancer event (EBCE). Preclinical laboratory studies included luciferase activity, cell proliferation, radio-labeled ligand estrogen receptor binding, surface plasmon resonance ligand receptor binding, and nuclear magnetic resonance assays.Results:Women with E1 ≥1.3 pg/mL and E2 ≥0.5 pg/mL after 6 months of AI treatment had a 2.2-fold increase in risk (P = 0.0005) of an EBCE, and in the anastrozole subgroup, the increase in risk of an EBCE was 3.0-fold (P = 0.001). Preclinical laboratory studies examined mechanisms of action in addition to aromatase inhibition and showed that only anastrozole could directly bind to estrogen receptor α (ERα), activate estrogen response element-dependent transcription, and stimulate growth of an aromatase-deficient CYP19A1−/− T47D breast cancer cell line.Conclusions:This matched case–control clinical study revealed that levels of estrone and estradiol above identified thresholds after 6 months of adjuvant anastrozole treatment were associated with increased risk of an EBCE. Preclinical laboratory studies revealed that anastrozole, but not exemestane or letrozole, is a ligand for ERα. These findings represent potential steps towards individualized anastrozole therapy.
- Published
- 2020
- Full Text
- View/download PDF
6. FAM111A protects replication forks from protein obstacles via its trypsin-like domain
- Author
-
Scott H. Kaufmann, Thomas R. Caulfield, Sowmiya Palani, Yuka Machida, Yuichi J. Machida, Evette S. Radisky, and Yusuke Kojima
- Subjects
DNA Replication ,0301 basic medicine ,Virus genetics ,Poly ADP ribose polymerase ,medicine.medical_treatment ,Science ,Protein domain ,DNA, Single-Stranded ,General Physics and Astronomy ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,PARP1 ,Protein Domains ,Cell Line, Tumor ,medicine ,Chemotherapy ,Humans ,Trypsin ,lcsh:Science ,Polymerase ,Multidisciplinary ,Protease ,biology ,Chemistry ,DNA replication ,DNA ,Proteases ,Cell Cycle Checkpoints ,General Chemistry ,Enzymes ,Cell biology ,030104 developmental biology ,DNA Topoisomerases, Type I ,Proteasome ,030220 oncology & carcinogenesis ,Proteolysis ,Mutation ,biology.protein ,Receptors, Virus ,Camptothecin ,lcsh:Q ,Poly(ADP-ribose) Polymerases ,DNA Damage ,Protein Binding - Abstract
Persistent protein obstacles on genomic DNA, such as DNA-protein crosslinks (DPCs) and tight nucleoprotein complexes, can block replication forks. DPCs can be removed by the proteolytic activities of the metalloprotease SPRTN or the proteasome in a replication-coupled manner; however, additional proteolytic mechanisms may exist to cope with the diversity of protein obstacles. Here, we show that FAM111A, a PCNA-interacting protein, plays an important role in mitigating the effect of protein obstacles on replication forks. This function of FAM111A requires an intact trypsin-like protease domain, the PCNA interaction, and the DNA-binding domain that is necessary for protease activity in vivo. FAM111A, but not SPRTN, protects replication forks from stalling at poly(ADP-ribose) polymerase 1 (PARP1)-DNA complexes trapped by PARP inhibitors, thereby promoting cell survival after drug treatment. Altogether, our findings reveal a role of FAM111A in overcoming protein obstacles to replication forks, shedding light on cellular responses to anti-cancer therapies., DNA-protein crosslinks represent obstacles on genomic DNA that can hamper progression of replication forks. Here, the authors reveal that FAM111A, a PCNA-interacting protein, plays part in mitigating the effect of protein obstacles on replication forks.
- Published
- 2020
7. Therapeutics targeting BCL2 family proteins
- Author
-
Jia Jia, X. Wei Meng, Haiming Dai, Scott H. Kaufmann, and Kaiqin Ye
- Subjects
Subfamily ,Effector ,Venetoclax ,Endoplasmic reticulum ,Autophagy ,Computational biology ,Biology ,Small molecule ,chemistry.chemical_compound ,chemistry ,hemic and lymphatic diseases ,Mitochondrial fission ,biological phenomena, cell phenomena, and immunity ,Bacterial outer membrane - Abstract
BCL2 family proteins play an essential role in controlling mitochondrial outer membrane permeabilization which is the fatal and usually irreversible step of the mitochondrial apoptotic pathway. Moreover, BCL2 family proteins are also involved in the regulations of other cellular processes, such as autophagy, mitochondrial fission and fusion, and endoplasmic reticulum stress. Three proteins, BAK, BAX, and BOK, are the effector BCL2 family proteins that directly permeabilize the mitochondrial outer membrane while the antiapoptotic BCL2 family members and the BH3-only subfamily proteins regulate this process through protein–protein interactions. BCL2 family proteins are known to be important in the development of several diseases, especially cancer. Thus, much effort has gone into the development of drugs targeting BCL2 family proteins to treat cancer; developed agents include antisense oligonucleotides, modified peptides, and small molecules. Some of these efforts have been quite successful, with several candidates tested in clinical trials. Moreover, ABT-199 (venetoclax) was approved by the United States Food and Drug Administration to treat relapsed, chromosome 17p-deleted chronic lymphocytic leukemia in 2016, providing a huge inspiration for this field. In this chapter, we summarize the recent advances in BCL2 family studies, review the latest developments and clinical studies of drug candidates to target BCL2 family proteins, and discuss future directions of this class of antineoplastic agents.
- Published
- 2022
- Full Text
- View/download PDF
8. Correction: Constitutive BAK/MCL1 complexes predict paclitaxel and S63845 sensitivity of ovarian cancer
- Author
-
Cristina Correia, Dongyan Liu, Wangyu Wu, Chao Xu, Zhirong Liu, Tao Zhang, Zhiyou Fang, Yunjian Li, Chenggang Zhao, Zhiyang Zhao, Valentina Zafagnin, Xiaonan Hou, Haiming Dai, Saravut J. Weroha, Scott H. Kaufmann, and Hongzhi Wang
- Subjects
Cancer Research ,Chemotherapy ,QH573-671 ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,medicine.disease ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Paclitaxel ,chemistry ,Apoptosis ,Cancer research ,Medicine ,MCL1 ,Sensitivity (control systems) ,Cytology ,business ,Ovarian cancer - Published
- 2021
- Full Text
- View/download PDF
9. Constitutive BAK/MCL1 complexes predict paclitaxel and S63845 sensitivity of ovarian cancer
- Author
-
Scott H. Kaufmann, Valentina Zanfagnin, Tao Zhang, Haiming Dai, Zhiyang Zhao, Wangyu Wu, Zhirong Liu, Hongzhi Wang, Saravut J. Weroha, Chenggang Zhao, Chao Xu, Yunjian Li, Zhiyou Fang, Xiaonan Hou, Cristina Correia, and Dongyan Liu
- Subjects
Cancer Research ,Paclitaxel ,medicine.medical_treatment ,Immunology ,Mice, Nude ,Apoptosis ,Context (language use) ,Thiophenes ,Article ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Chemotherapy ,MCL1 ,RNA, Messenger ,Ovarian Neoplasms ,Mice, Inbred BALB C ,Bcl-2-Like Protein 11 ,QH573-671 ,Antagonist ,Correction ,Drug Synergism ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Pyrimidines ,bcl-2 Homologous Antagonist-Killer Protein ,chemistry ,Cell culture ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,Female ,biological phenomena, cell phenomena, and immunity ,Cytology ,Ovarian cancer ,Protein Binding - Abstract
We previously found that preformed complexes of BAK with antiapoptotic BCL2 proteins predict BH3 mimetic sensitivities in lymphohematopoietic cells. These complexes have not previously been examined in solid tumors or in the context of conventional anticancer drugs. Here we show the relative amount of BAK found in preformed complexes with MCL1 or BCLXL varies across ovarian cancer cell lines and patient-derived xenografts (PDXs). Cells bearing BAK/MCL1 complexes were more sensitive to paclitaxel and the MCL1 antagonist S63845. Likewise, PDX models with BAK/MCL1 complexes were more likely to respond to paclitaxel. Mechanistically, BIM induced by low paclitaxel concentrations interacted preferentially with MCL1 and displaced MCL1-bound BAK. Further studies indicated that cells with preformed BAK/MCL1 complexes were sensitive to the paclitaxel/S63845 combination, while cells without BAK/MCL1 complexes were not. Our study suggested that the assessment of BAK/MCL1 complexes might be useful for predicting response to paclitaxel alone or in combination with BH3 mimetics.
- Published
- 2021
- Full Text
- View/download PDF
10. Characterization of patients with BRCA mutated ovarian cancer who are eligible versus not eligible for PARP inhibitor maintenance therapy: exploratory analysis of the VELIA study
- Author
-
Peter Ansell, Karina Dahl Steffensen, Kathleen N. Moore, Aikou Okamoto, Christine K. Ratajczak, Danielle Sullivan, Robert L. Coleman, Minh H. Dinh, Brenden Chen, Scott H. Kaufmann, Bruce A. Bach, Elizabeth M. Swisher, and Michael A. Bookman
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,Performance status ,Veliparib ,business.industry ,medicine.medical_treatment ,Obstetrics and Gynecology ,medicine.disease ,Placebo ,Carboplatin ,Olaparib ,chemistry.chemical_compound ,chemistry ,Maintenance therapy ,Internal medicine ,otorhinolaryngologic diseases ,Medicine ,business ,Progressive disease - Abstract
Objectives: Previous phase 3 trials of front-line maintenance with the PARP inhibitors (PARPi) olaparib and niraparib only included patients (pts) with complete (CR) or partial (PR) response following front-line chemotherapy (CT); both agents were adopted as standard of care (SOC) in these pts. This prevented the systematic study of all pts who are eligible for PARPi maintenance therapy after front-line CT or to predict their responses. The phase 3 VELIA study (NCT02470585) added veliparib (VEL) to front-line CT at diagnosis, followed by VEL maintenance in the absence of progressive disease (PD), permitting treatment of a broader cohort of pts. This exploratory analysis evaluated baseline characteristics associated with first-line CT response, to understand if predictive variables exist that could identify those who would be eligible vs ineligible for SOC PARPi maintenance. Methods: Pts with untreated Stage III/IV ovarian cancer were randomized to carboplatin/paclitaxel (CP) + placebo (PBO) followed by PBO maintenance (control arm), CP + VEL followed by PBO maintenance (VEL combination only), or CP + VEL followed by VEL maintenance (VEL throughout). Pts without progression after combination treatment transitioned to assigned maintenance therapy. In this exploratory analysis, pts with germline or somatic BRCA mutations (BRCAm) were grouped as maintenance eligible (ME; with CR/PR or non-PD with surgical complete resection) or maintenance not eligible (MNE; with stable disease/progression/non-CR/non-PD). Progression-free survival (PFS) in pts starting maintenance was measured from initiation of maintenance treatment. Results: Of 1,140 randomized pts, 298 had germline or somatic BRCAm; of these, 278 could be classified according to maintenance eligibility. MNE pts (n=56) were distributed equally among the control, VEL combination only, and VEL throughout arms (21%, 19%, and 21% of evaluable pts, respectively). MNE subgroups were enriched for advanced disease (18-39% of MNE subgroups vs 13-19% of ME subgroups had Stage IV disease) and less favorable performance status (41-67% MNE vs 20-40% of ME pts had ECOG performance status ≥1). For ME pts (n=222), median PFS from initiation of maintenance was not reached in the VEL throughout arm vs 19.7 months in the control arm (HR 0.29, 95% CI 0.16-0.52, Figure). For MNE pts, median PFS was 16.4 months in the VEL throughout arm and 18.9 months in the control arm (HR 1.331, 95% CI 0.52-3.44); however, sample sizes (n=18 and n=16, respectively) limit a meaningful analysis of PFS in these pts. Download : Download high-res image (79KB) Download : Download full-size image Conclusions: The results of this exploratory analysis suggest that VEL has robust efficacy in pts with BRCA-mutated tumors eligible for SOC PARPi maintenance. Baseline characteristics of advanced disease and less favorable performance status were more common in MNE pts than ME pts. Molecular characterization is ongoing to identify mechanisms of sensitivity that predict eligibility for, and response to, PARPi therapy.
- Published
- 2021
- Full Text
- View/download PDF
11. ZC3H18 specifically binds and activates the BRCA1 promoter to facilitate homologous recombination in ovarian cancer
- Author
-
Ethan P. Heinzen, Larry M. Karnitz, Daniel R. O'Brien, Catherine J. Huntoon, Xiaonan Hou, Scott H. Kaufmann, Arun Kanakkanthara, Minzhi Zhang, Ann L. Oberg, and S. John Weroha
- Subjects
0301 basic medicine ,Transcription, Genetic ,endocrine system diseases ,DNA damage ,Science ,General Physics and Astronomy ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ovarian cancer ,Transcription (biology) ,Cell Line, Tumor ,Humans ,Tumour-suppressor proteins ,Homologous Recombination ,Promoter Regions, Genetic ,E2F ,skin and connective tissue diseases ,lcsh:Science ,E2F4 ,Ovarian Neoplasms ,Regulation of gene expression ,Multidisciplinary ,BRCA1 Protein ,RNA-Binding Proteins ,General Chemistry ,DNA Methylation ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,DNA methylation ,Female ,lcsh:Q ,Homologous recombination ,DNA ,DNA Damage - Abstract
Reduced BRCA1 expression causes homologous recombination (HR) repair defects in high-grade serous ovarian cancers (HGSOCs). Here, we demonstrate that BRCA1 is transcriptionally activated by a previously unknown function of ZC3H18. We show that ZC3H18 is a DNA-binding protein that interacts with an E2F site in the BRCA1 promoter where it facilitates recruitment of E2F4 to an adjacent E2F site to promote BRCA1 transcription. Consistent with ZC3H18 role in activating BRCA1 expression, ZC3H18 depletion induces BRCA1 promoter methylation, reduces BRCA1 expression, disrupts HR, and sensitizes cells to DNA crosslinkers and poly(ADP-ribose) polymerase inhibitors. Moreover, in patient-derived xenografts and primary HGSOC tumors, ZC3H18 and E2F4 mRNA levels are positively correlated with BRCA1 mRNA levels, further supporting ZC3H18 role in regulating BRCA1. Given that ZC3H18 lies within 16q24.2, a region with frequent copy number loss in HGSOC, these findings suggest that ZC3H18 copy number losses could contribute to HR defects in HGSOC., High-grade serous ovarian cancers (HGSOCs) have defects in homologous recombination despite a lack of BRCA1/2 mutations. Here, the authors show that ZC3H18 positively regulates BRCA1 transcription and its loss causes BRCA1 promoter methylation and increased HR deficiency in HGSOCs.
- Published
- 2019
- Full Text
- View/download PDF
12. Olaparib and α-specific PI3K inhibitor alpelisib for patients with epithelial ovarian cancer: a dose-escalation and dose-expansion phase 1b trial
- Author
-
Alan D. D'Andrea, Erica L. Mayer, Joyce F. Liu, Shannon N. Westin, Bose Kochupurakkal, Sarah Farooq, William T. Barry, Karen Cadoo, Lewis C. Cantley, Scott H. Kaufmann, Gerburg M. Wulf, Geoffrey I. Shapiro, Paul Kirschmeier, Panagiotis A. Konstantinopoulos, Gordon B. Mills, Ursula A. Matulonis, Eric P. Winer, Weixiu Luo, Robert L. Coleman, Julia Eismann, Sangeetha Palakurthi, Carol Aghajanian, Roisin E. O'Cearbhaill, Michael J. Birrer, Jennifer Curtis, Elizabeth M. Swisher, Christin Whalen, and Mary K. Buss
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Maximum Tolerated Dose ,Carcinoma, Ovarian Epithelial ,Poly(ADP-ribose) Polymerase Inhibitors ,Piperazines ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Carcinoma ,Humans ,Adverse effect ,Aged ,Phosphoinositide-3 Kinase Inhibitors ,Ovarian Neoplasms ,Dose-Response Relationship, Drug ,Genome, Human ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,Thiazoles ,Treatment Outcome ,030104 developmental biology ,chemistry ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Mutation ,PARP inhibitor ,Phthalazines ,Female ,Ovarian cancer ,business - Abstract
Summary Background Based on preclinical work, we found that combination of poly (ADP-ribose) polymerase (PARP) inhibitors with drugs that inhibit the homologous recombination repair (HRR) pathway (such as PI3K inhibitors) might sensitise HRR-proficient epithelial ovarian cancers to PARP inhibitors. We aimed to assess the safety and identify the recommended phase 2 dose of the PARP inhibitor olaparib in combination with the PI3K inhibitor alpelisib in patients with epithelial ovarian cancer and in patients with breast cancer. Methods In this multicentre, open-label, phase 1b trial following a 3 + 3 dose-escalation design, we recruited patients aged 18 years or older with the following key eligibility criteria: confirmed diagnosis of either recurrent ovarian, fallopian tube, or primary peritoneal cancer of high-grade serous histology; confirmed diagnosis of either recurrent ovarian, fallopian tube, or primary peritoneal cancer of any histology with known germline BRCA mutations; confirmed diagnosis of recurrent breast cancer of triple-negative histology; or confirmed diagnosis of recurrent breast cancer of any histology with known germline BRCA mutations. Additional patients with epithelial ovarian cancer were enrolled in a dose-expansion cohort. Four dose levels were planned: the starting dose level of alpelisib 250 mg once a day plus olaparib 100 mg twice a day (dose level 0); alpelisib 250 mg once a day plus olaparib 200 mg twice a day (dose level 1); alpelisib 300 mg once a day plus olaparib 200 mg twice a day (dose level 2); and alpelisib 200 mg once a day plus olaparib 200 mg twice a day (dose level 3). Both drugs were administered orally, in tablet formulation. The primary objective was to identify the maximum tolerated dose and the recommended phase 2 dose of the combination of alpelisib and olaparib for patients with epithelial ovarian cancer and patients with breast cancer. Analyses included all patients who received at least one dose of the study drugs. The trial is active, but closed to enrolment; follow-up for patients who completed treatment is ongoing. This trial is registered with ClinicalTrials.gov , number NCT01623349 . Findings Between Oct 3, 2014, and Dec 21, 2016, we enrolled 34 patients (28 in the dose-escalation cohort and six in the dose-expansion cohort); two in the dose-escalation cohort were ineligible at the day of scheduled study initiation. Maximum tolerated dose and recommended phase 2 dose were identified as alpelisib 200 mg once a day plus olaparib 200 mg twice a day (dose level 3). Considering all dose levels, the most common treatment-related grade 3–4 adverse events were hyperglycaemia (five [16%] of 32 patients), nausea (three [9%]), and increased alanine aminotransferase concentrations (three [9%]). No treatment-related deaths occurred. Dose-limiting toxic effects included hyperglycaemia and fever with decreased neutrophil count. Of the 28 patients with epithelial ovarian cancer, ten (36%) achieved a partial response and 14 (50%) had stable disease according to Response Evaluation Criteria in Solid Tumors 1.1. Interpretation Combining alpelisib and olaparib is feasible with no unexpected toxic effects. The observed activity provides preliminary clinical evidence of synergism between olaparib and alpelisib, particularly in epithelial ovarian cancer, and warrants further investigation. Funding Ovarian Cancer Dream Team (Stand Up To Cancer, Ovarian Cancer Research Alliance, National Ovarian Cancer Coalition), Breast Cancer Research Foundation, Novartis.
- Published
- 2019
- Full Text
- View/download PDF
13. Effect of CHK1 Inhibition on CPX-351 Cytotoxicity in vitro and ex vivo
- Author
-
Mira A. Kohorst, Karen S. Flatten, Scott H. Kaufmann, Jonathan Webster, Amelia M. Huehls, Nicole D. Vincelette, Rebecca L. Kelly, Larry M. Karnitz, Husheng Ding, Keith W. Pratz, and Allan D. Hess
- Subjects
0301 basic medicine ,animal structures ,Daunorubicin ,lcsh:Medicine ,Apoptosis ,In Vitro Techniques ,environment and public health ,Article ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Rabusertib ,medicine ,Tumor Cells, Cultured ,Humans ,CHEK1 ,lcsh:Science ,Protein Kinase Inhibitors ,Cell Proliferation ,Multidisciplinary ,Chemistry ,Cell growth ,lcsh:R ,Cytarabine ,Cell cycle ,3. Good health ,Prexasertib ,G2 Phase Cell Cycle Checkpoints ,Leukemia, Myeloid, Acute ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Checkpoint Kinase 1 ,Cancer research ,Leukocytes, Mononuclear ,lcsh:Q ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,Ex vivo ,medicine.drug - Abstract
CPX-351 is a liposomally encapsulated 5:1 molar ratio of cytarabine and daunorubicin that recently received regulatory approval for the treatment of therapy-related acute myeloid leukemia (AML) or AML with myelodysplasia-related changes based on improved overall survival compared to standard cytarabine/daunorubicin therapy. Checkpoint kinase 1 (CHK1), which is activated by DNA damage and replication stress, diminishes sensitivity to cytarabine and anthracyclines as single agents, suggesting that CHK1 inhibitors might increase the effectiveness of CPX-351. The present studies show that CPX-351 activates CHK1 as well as the S and G2/M cell cycle checkpoints. Conversely, CHK1 inhibition diminishes the cell cycle effects of CPX-351. Moreover, CHK1 knockdown or addition of a CHK1 inhibitor such as MK-8776, rabusertib or prexasertib enhances CPX-351-induced apoptosis in multiple TP53-null and TP53-wildtype AML cell lines. Likewise, CHK1 inhibition increases the antiproliferative effect of CPX-351 on primary AML specimens ex vivo, offering the possibility that CPX-351 may be well suited to combine with CHK1-targeted agents.
- Published
- 2019
- Full Text
- View/download PDF
14. Characterization of a RAD51C-silenced high-grade serous ovarian cancer model during development of PARP inhibitor resistance
- Author
-
Andrea E. Wahner Hendrickson, Ksenija Nesic, Hu Li, Xiaonan Hou, Rebecca L. Kelly, Thomas Harding, Clare L. Scott, Rachel M. Hurley, Paula A. Schneider, Melissa C. Southey, Matthew Wakefield, Karen S. Flatten, Jill M. Wagner, Larry M. Karnitz, Christian A. Ross, Cordelia D. McGehee, Alexander Dobrovic, Taylor M. Weiskittel, Paul Haluska, Scott H. Kaufmann, S. John Weroha, Annapoorna Venkatachalam, Cristina Correia, Marc A. Becker, Marc R. Radke, Kevin K. Lin, Kevin L. Peterson, X. Wei Meng, Nicholas M. Pathoulas, Elizabeth M. Swisher, Ee Ming Wong, Olga Kondrashova, and Iain A. McNeish
- Subjects
0301 basic medicine ,General Medicine ,Methylation ,Gene mutation ,Biology ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,In vivo ,030220 oncology & carcinogenesis ,PARP inhibitor ,medicine ,Cancer research ,RAD51C ,Rucaparib ,Ovarian cancer ,Homologous recombination - Abstract
Acquired PARP inhibitor (PARPi) resistance in BRCA1- or BRCA2-mutant ovarian cancer often results from secondary mutations that restore expression of functional protein. RAD51C is a less commonly studied ovarian cancer susceptibility gene whose promoter is sometimes methylated, leading to homologous recombination (HR) deficiency and PARPi sensitivity. For this study, the PARPi-sensitive patient-derived ovarian cancer xenograft PH039, which lacks HR gene mutations but harbors RAD51C promoter methylation, was selected for PARPi resistance by cyclical niraparib treatment in vivo. PH039 acquired PARPi resistance by the third treatment cycle and grew through subsequent treatment with either niraparib or rucaparib. Transcriptional profiling throughout the course of resistance development showed widespread pathway level changes along with a marked increase in RAD51C mRNA, which reflected loss of RAD51C promoter methylation. Analysis of ovarian cancer samples from the ARIEL2 Part 1 clinical trial of rucaparib monotherapy likewise indicated an association between loss of RAD51C methylation prior to on-study biopsy and limited response. Interestingly, the PARPi resistant PH039 model remained platinum sensitive. Collectively, these results not only indicate that PARPi treatment pressure can reverse RAD51C methylation and restore RAD51C expression, but also provide a model for studying the clinical observation that PARPi and platinum sensitivity are sometimes dissociated.
- Published
- 2021
- Full Text
- View/download PDF
15. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2)
- Author
-
Ling Ma, Tanya T. Kwan, Clare L. Scott, Isabelle Ray-Coquard, Ana Oaknin, Alexander Dobrovic, Robert L. Coleman, Iain A. McNeish, Andrea E. Wahner Hendrickson, Lee-may Chen, Alexandra Leary, Stephen Welch, Thomas Harding, Lara Maloney, Carol Aghajanian, Kevin K. Lin, Heidi Giordano, E. Dominy, Ashan Musafer, Gottfried E. Konecny, Scott H. Kaufmann, Diane Provencher, Julia A. Elvin, Oliver Dorigo, Sandra Goble, R Kristeleit, Douglas I. Lin, Anna V. Tinker, Amit M. Oza, Setsuko K. Chambers, David M. O'Malley, Elizabeth M. Swisher, Prafull Ghatage, Lan Thanh Vo, Institut Català de la Salut, [Swisher EM] University of Washington, Seattle, WA, USA. [Kwan TT] Clovis Oncology, Inc., Boulder, CO, USA. [Oza AM] Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. [Tinker AV] BC Cancer—Vancouver, Vancouver, BC, Canada. [Ray-Coquard I] GINECO, Centre Léon Bérard and University Claude Bernard, Lyon, France. [Oaknin A] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, Vall d'Hebron Barcelona Hospital Campus, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,Indoles ,endocrine system diseases ,General Physics and Astronomy ,neoplasias::neoplasias por localización::neoplasias de las glándulas endocrinas::neoplasias ováricas [ENFERMEDADES] ,Carcinoma, Ovarian Epithelial ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Poly (ADP-Ribose) Polymerase Inhibitor ,Tumour biomarkers ,chemistry.chemical_compound ,0302 clinical medicine ,Ovarian carcinoma ,Ovarian Epithelial ,80 and over ,Medicine ,Promoter Regions, Genetic ,Cancer ,Aged, 80 and over ,Ovarian Neoplasms ,neoplasias::procesos neoplásicos::recurrencia neoplásica local [ENFERMEDADES] ,Multidisciplinary ,BRCA1 Protein ,Ovaris - Càncer - Tractament ,Cell cycle ,Middle Aged ,BRCA2 Protein ,female genital diseases and pregnancy complications ,Ovarian Cancer ,DNA-Binding Proteins ,Local ,030220 oncology & carcinogenesis ,DNA methylation ,PARP inhibitor ,RAD51C ,Female ,Adult ,Science ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Antineoplastic Agents ,Poly(ADP-ribose) Polymerase Inhibitors ,General Biochemistry, Genetics and Molecular Biology ,Article ,Promoter Regions ,03 medical and health sciences ,Rare Diseases ,Genetic ,Clinical Research ,Ovarian cancer ,Neoplasms::Neoplastic Processes::Neoplasm Recurrence, Local [DISEASES] ,Genetics ,Humans ,Rucaparib ,Ovaris - Càncer - Recaiguda ,Aged ,Platinum ,business.industry ,Neoplasms::Neoplasms by Site::Endocrine Gland Neoplasms::Ovarian Neoplasms [DISEASES] ,Carcinoma ,General Chemistry ,DNA Methylation ,030104 developmental biology ,Good Health and Well Being ,Neoplasm Recurrence ,chemistry ,Cancer research ,Neoplasm Recurrence, Local ,business - Abstract
ARIEL2 (NCT01891344) is a single-arm, open-label phase 2 study of the PARP inhibitor (PARPi) rucaparib in relapsed high-grade ovarian carcinoma. In this post hoc exploratory biomarker analysis of pre- and post-platinum ARIEL2 samples, RAD51C and RAD51D mutations and high-level BRCA1 promoter methylation predict response to rucaparib, similar to BRCA1/BRCA2 mutations. BRCA1 methylation loss may be a major cross-resistance mechanism to platinum and PARPi. Genomic scars associated with homologous recombination deficiency are irreversible, persisting even as platinum resistance develops, and therefore are predictive of rucaparib response only in platinum-sensitive disease. The RAS, AKT, and cell cycle pathways may be additional modulators of PARPi sensitivity., The identification of biomarkers of response to PARP inhibitors can enable selection of appropriate ovarian cancer patients for treatment. In this study, the authors report clinical results and exploratory biomarker analyses from the ARIEL2 phase 2 clinical trial on the safety and efficacy of the PARP inhibitor rucaparib in patients with recurrent ovarian cancers
- Published
- 2021
16. Fatty acid synthase (FASN) regulates the mitochondrial priming of cancer cells
- Author
-
Kevin M. Regan, Elisabet Cuyàs, George Kemble, Chandra Mohan Kurapaty Venkatapoorna, Ruth Lupu, Barbara Schroeder, Joan Montero, Zeng Hu, Sara Verdura, Karen S. Flatten, Ingrid Espinoza, Javier A. Menendez, Aina Arbusà, Fernando Martín Silva, X. Wei Meng, Paula A. Schneider, Scott H. Kaufmann, and Travis Vander Steen
- Subjects
Cancer Research ,Programmed cell death ,Cancer cells ,Immunology ,Mice, Nude ,Transfection ,Cancer -- Treatment ,Article ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,Cancer -- Molecular aspects ,Puma ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Lipid signalling ,Càncer ,Cancer ,Navitoclax ,Càncer -- Aspectes moleculars ,biology ,QH573-671 ,Venetoclax ,Cell Biology ,Lipid membranes ,medicine.disease ,biology.organism_classification ,Membranes lipídiques ,Cancer metabolism ,Metabolisme ,Mitochondria ,Fatty acid synthase ,Metabolism ,chemistry ,Apoptosis ,Cancer cell ,biology.protein ,Cancer research ,Cèl·lules canceroses ,Female ,Càncer -- Tractament ,Fatty Acid Synthases ,Cytology - Abstract
Inhibitors of the lipogenic enzyme fatty acid synthase (FASN) have attracted much attention in the last decade as potential targeted cancer therapies. However, little is known about the molecular determinants of cancer cell sensitivity to FASN inhibitors (FASNis), which is a major roadblock to their therapeutic application. Here, we find that pharmacological starvation of endogenously produced FAs is a previously unrecognized metabolic stress that heightens mitochondrial apoptotic priming and favors cell death induction by BH3 mimetic inhibitors. Evaluation of the death decision circuits controlled by the BCL-2 family of proteins revealed that FASN inhibition is accompanied by the upregulation of the pro-death BH3-only proteins BIM, PUMA, and NOXA. Cell death triggered by FASN inhibition, which causally involves a palmitate/NADPH-related redox imbalance, is markedly diminished by concurrent loss of BIM or PUMA, suggesting that FASN activity controls cancer cell survival by fine-tuning the BH3 only proteins-dependent mitochondrial threshold for apoptosis. FASN inhibition results in a heightened mitochondrial apoptosis priming, shifting cells toward a primed-for-death state “addicted” to the anti-apoptotic protein BCL-2. Accordingly, co-administration of a FASNi synergistically augments the apoptosis-inducing activity of the dual BCL-XL/BCL-2 inhibitor ABT-263 (navitoclax) and the BCL-2 specific BH3-mimetic ABT-199 (venetoclax). FASN inhibition, however, fails to sensitize breast cancer cells to MCL-1- and BCL-XL-selective inhibitors such as S63845 and A1331852. A human breast cancer xenograft model evidenced that oral administration of the only clinically available FASNi drastically sensitizes FASN-addicted breast tumors to ineffective single-agents navitoclax and venetoclax in vivo. In summary, a novel FASN-driven facet of the mitochondrial priming mechanistically links the redox-buffering mechanism of FASN activity to the intrinsic apoptotic threshold in breast cancer cells. Combining next-generation FASNis with BCL-2-specific BH3 mimetics that directly activate the apoptotic machinery might generate more potent and longer-lasting antitumor responses in a clinical setting., The authors would like to thank Dr. Kenneth McCreath for editorial support. This work was supported by the NIH National Cancer Institute Grants R01 CA116623 (to Ruth Lupu) and R01 CA166741 (to Scott H. Kaufmann) and by the U.S. Department of Defense (DOD)-Breakthrough 3 Grants BC151072 and BC151072P1 (to Ruth Lupu). Work in the Menendez laboratory is supported by the Spanish Ministry of Science and Innovation (Grants SAF2016-80639-P and PID2019-10455GB-I00, Plan Nacional de l + D + I, founded by the European Regional Development Fund, Spain) and by an unrestricted research grant from the Fundació Oncolliga Girona (Lliga catalana d’ajuda al malalt de càncer, Girona). Joan Montero acknowledges support from the Ramon y Cajal Programme, Ministerio de Economía y Competitividad (RYC-2015-18357) and the Spanish National Plan “Retos Investigación” I + D + I (RTI2018-094533-A-I00) from the Ministerio de Ciencia, Innovación y Universidades. Elisabet Cuyàs holds a research contract “Miguel Servet” (CP20/00003) from the Instituto de Salud Carlos III, Spanish Ministry of Science and Innovation (Spain). All authors have read and agreed to the published version of the manuscript.
- Published
- 2021
17. Repurposing Ceritinib Induces DNA Damage and Enhances PARP Inhibitor Responses in High-Grade Serous Ovarian Carcinoma
- Author
-
Husheng Ding, Aaron S. Mansfield, Valentina Zanfagnin, S. John Weroha, Thomas L. Ekstrom, Melissa C. Larson, Scott H. Kaufmann, Larry M. Karnitz, Amelia M. Huehls, Arun Kanakkanthara, Xiaonan Hou, Rebecca L. Kelly, George Vasmatzis, and Ann L. Oberg
- Subjects
Cancer Research ,DNA damage ,Poly ADP ribose polymerase ,Antineoplastic Agents ,Mice, SCID ,Carcinoma, Ovarian Epithelial ,Poly(ADP-ribose) Polymerase Inhibitors ,Piperazines ,Article ,Olaparib ,chemistry.chemical_compound ,Mice ,medicine ,Animals ,Humans ,Sulfones ,Protein Kinase Inhibitors ,chemistry.chemical_classification ,Ovarian Neoplasms ,Reactive oxygen species ,Ceritinib ,Chemistry ,Kinase ,Drug Repositioning ,Recombinational DNA Repair ,Drug Synergism ,Xenograft Model Antitumor Assays ,Tumor Burden ,Pyrimidines ,Treatment Outcome ,Oncology ,Drug Resistance, Neoplasm ,PARP inhibitor ,PC-3 Cells ,Cancer research ,Phthalazines ,Female ,Homologous recombination ,medicine.drug ,DNA Damage - Abstract
PARP inhibitors (PARPi) have activity in homologous recombination (HR) repair-deficient, high-grade serous ovarian cancers (HGSOC). However, even responsive tumors develop PARPi resistance, highlighting the need to delay or prevent the appearance of PARPi resistance. Here, we showed that the ALK kinase inhibitor ceritinib synergizes with PARPis by inhibiting complex I of the mitochondrial electron transport chain, which increases production of reactive oxygen species (ROS) and subsequent induction of oxidative DNA damage that is repaired in a PARP-dependent manner. In addition, combined treatment with ceritinib and PARPi synergized in HGSOC cell lines irrespective of HR status, and a combination of ceritinib with the PARPi olaparib induced tumor regression more effectively than olaparib alone in HGSOC patient-derived xenograft (PDX) models. Notably, the ceritinib and olaparib combination was most effective in PDX models with preexisting PARPi sensitivity and was well tolerated. These findings unveil suppression of mitochondrial respiration, accumulation of ROS, and subsequent induction of DNA damage as novel effects of ceritinib. They also suggest that the ceritinib and PARPi combination warrants further investigation as a means to enhance PARPi activity in HGSOC, particularly in tumors with preexisting HR defects. Significance: The kinase inhibitor ceritinib synergizes with PARPi to induce tumor regression in ovarian cancer models, suggesting that ceritinib combined with PARPi may be an effective strategy for treating ovarian cancer.
- Published
- 2021
18. Preexisting TP53 -Variant Clonal Hematopoiesis and Risk of Secondary Myeloid Neoplasms in Patients with High-grade Ovarian Cancer Treated with Rucaparib
- Author
-
Elizabeth M. Swisher, Tanya T. Kwan, Lan-Thanh Vo, Sandra Goble, Nicoletta Colombo, Lara Maloney, Anna V. Tinker, Eric Allan Severson, Kevin K. Lin, Scott H. Kaufmann, Iain A. McNeish, Robert L. Coleman, Domenica Lorusso, Johanne I Weberpals, Isabelle Ray-Coquard, Amit M. Oza, Ana Oaknin, Andrew Dean, Carol Aghajanian, Jonathan A. Ledermann, Thomas Harding, Kwan, T, Oza, A, Tinker, A, Ray-Coquard, I, Oaknin, A, Aghajanian, C, Lorusso, D, Colombo, N, Dean, A, Weberpals, J, Severson, E, Vo, L, Goble, S, Maloney, L, Harding, T, Kaufmann, S, Ledermann, J, Coleman, R, Mcneish, I, Lin, K, and Swisher, E
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Indoles ,Myeloid ,medicine.medical_treatment ,Poly (ADP-Ribose) Polymerase-1 ,Genome-wide association study ,Poly(ADP-ribose) Polymerase Inhibitors ,chemistry.chemical_compound ,Neoplasms ,Internal medicine ,ARIEL3 ,medicine ,Humans ,Rucaparib ,Original Investigation ,Retrospective Studies ,Ovarian Neoplasms ,Chemotherapy ,Myeloproliferative Disorders ,business.industry ,Middle Aged ,medicine.disease ,Serous fluid ,medicine.anatomical_structure ,chemistry ,Fallopian tube cancer ,Benzamides ,Female ,Clonal Hematopoiesis ,Tumor Suppressor Protein p53 ,Ovarian cancer ,business ,Fallopian tube - Abstract
IMPORTANCE: A total of 1% to 3% of patients treated with a poly(adenosine diphosphate–ribose) polymerase inhibitor for high-grade ovarian cancer (HGOC) develop therapy-related myeloid neoplasms (t-MNs), which are rare but often fatal conditions. Although the cause of these t-MNs is unknown, clonal hematopoiesis of indeterminate potential (CHIP) variants can increase the risk of primary myeloid malignant neoplasms and are more frequent among patients with solid tumors. OBJECTIVES: To examine whether preexisting CHIP variants are associated with the development of t-MNs after rucaparib treatment and how these CHIP variants are affected by treatment. DESIGN, SETTING, AND PARTICIPANTS: This retrospective genetic association study used peripheral blood cell (PBC) samples collected before rucaparib treatment from patients in the multicenter, single-arm ARIEL2 (Study of Rucaparib in Patients With Platinum-Sensitive, Relapsed, High-Grade Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancer) (n = 491; between October 30, 2013, and August 9, 2016) and the multicenter, placebo-controlled, double-blind ARIEL3 (Study of Rucaparib as Switch Maintenance Following Platinum-Based Chemotherapy in Patients With Platinum-Sensitive, High-Grade Serous or Endometrioid Epithelial Ovarian, Primary Peritoneal or Fallopian Tube Cancer) (n = 561; between April 7, 2014, and July 19, 2016), which tested rucaparib as HGOC therapy in the treatment and maintenance settings, respectively. The follow-up data cutoff date was September 1, 2019. Of 1052 patients in ARIEL2 and ARIEL3, PBC samples from 20 patients who developed t-MNs (cases) and 44 randomly selected patients who did not (controls) were analyzed for the presence of CHIP variants using targeted next-generation sequencing. Additional longitudinal analysis was performed on available ARIEL2 samples collected during treatment and at the end of treatment. MAIN OUTCOMES AND MEASURES: Enrichment analysis of preexisting variants in 10 predefined CHIP-associated genes in cases relative to controls; association with clinical correlates. RESULTS: Among 1052 patients (mean [SE] age, 61.7 [0.3] years) enrolled and dosed in ARIEL2 and ARIEL3, 22 (2.1%) developed t-MNs. The t-MNs were associated with longer overall exposure to prior platinum therapies (13.2 vs 9.0 months in ARIEL2, P = .04; 12.4 vs 9.6 months in ARIEL3, P = .003). The presence of homologous recombination repair gene variants in the tumor, either germline or somatic, was associated with increased prevalence of t-MNs (15 [4.1%] of 369 patients with HGOC associated with an HRR gene variant vs 7 [1.0%] of 683 patients with wild-type HGOC, P = .002). The prevalence of preexisting CHIP variants in TP53 but not other CHIP-associated genes at a variant allele frequency of 1% or greater was significantly higher in PBCs from cases vs controls (9 [45.0%] of 20 cases vs 6 [13.6%] of 44 controls, P = .009). TP53 CHIP was associated with longer prior exposure to platinum (mean 14.0 months of 15 TP53 CHIP cases vs 11.1 months of 49 non-TP53 CHIP cases; P = .02). Longitudinal analysis showed that preexisting TP53 CHIP variants expanded in patients who developed t-MNs. CONCLUSIONS AND RELEVANCE: The findings of this genetic association study suggest that preexisting TP53 CHIP variants may be associated with t-MNs after rucaparib treatment.
- Published
- 2021
19. A knowledge-based, validated classifier for the identification of aliphatic and aromatic plastics by WorldView-3 satellite data
- Author
-
Shanyu Zhou, Theres Kuester, Mathias Bochow, Niklas Bohn, H. Kaufmann, and Maximilian Brell
- Subjects
Polypropylene ,Brightness ,Materials science ,Pixel ,Soil Science ,Hyperspectral imaging ,Geology ,chemistry.chemical_compound ,chemistry ,visual_art ,visual_art.visual_art_medium ,Polystyrene ,Computers in Earth Sciences ,Polycarbonate ,Image resolution ,Classifier (UML) ,Remote sensing - Abstract
Although the C H chains of petroleum derivatives display unique absorption features in the short-wave infrared (SWIR), it is a challenge to identify plastics on terrestrial surfaces. The diverse reflectance spectra caused by chemically varying polymer types and their different kinds of brightness and transparencies, which are, moreover, influenced further by the respective surface backgrounds. This paper investigates the capability of WorldView-3 (WV-3) satellite data, characterized by a high spatial resolution and equipped with eight distinct and relatively narrow SWIR bands suitable for global monitoring of different types of plastic materials. To meet the objective, hyperspectral measurements and simulations were conducted in the laboratory and by aircraft campaigns, based on the JPL-ECOSTRESS, USGS, and inhouse hyperspectral libraries, all of which are convolved to the spectral response functions of the WV-3 system. Experiments further supported the analyses wherein different plastic materials were placed on different backgrounds, and scaled percentages of plastics per pixel were modeled to determine the minimum detectable fractions. To determine the detectability of plastics with various chemical and physical properties and different fractions against diverse backgrounds, a knowledge-based classifier was developed, the routines of which are based on diagnostic spectral features in the SWIR range. The classifier shows outstanding results on various background scenarios for lab experimental imagery as well as for airborne data and it is further able to mask non-plastic materials. Three clusters of plastic materials can clearly be identified, based on spectra and imagery: The first cluster identifies aliphatic compounds, comprising polyethylene (PE), polyvinylchloride (PVC), ethylene vinyl acetate copolymer (EVAC), polypropylene (PP), polyoxymethylene (POM), polymethyl methacrylate (PMMA), and polyamide (PA). The second and third clusters are diagnostic for aromatic hydrocarbons, including polyethylene terephthalate (PET), polystyrene (PS), polycarbonate (PC), and styrene-acrylonitrile (SAN), respectively separated from polybutylene adipate terephthalate (PBAT), acrylonitrile butadiene styrene (ABS), and polyurethane (PU). The robustness of the classifier is examined on the basis of simulated spectra derived from our HySimCaR model, which has been developed in-house. The model simulates radiation transfer by using virtual 3D scenarios and ray tracing, hence, enables the analysis of the influence of various factors, such as material brightness, transparency, and fractional coverage as well as different background materials. We validated our results by laboratory and simulated datasets and by tests using airborne data recorded at four distinct sites with different surface characteristics. The results of the classifier were further compared to results produced by another signature-based method, the spectral angle mapper (SAM) and a commonly used technique, the maximum likelihood estimation (MLE). Finally, we applied and successfully tested the classifier on WV-3 imagery of sites known for a high abundance of plastics in Almeria (Spain), Cairo (Egypt), and Accra, (Ghana, West Africa). Both airborne and WV-3 data were atmospherically corrected and transferred to “at-surface reflectances”. The results prove the combination of WV-3 data and the newly designed classifier to be an efficient and reliable approach to globally monitor and identify three clusters of plastic materials at various fractions on different backgrounds.
- Published
- 2021
20. Characterization of a RAD51C-Silenced High Grade Serous Ovarian Cancer Model During PARP Inhibitor Resistance Development
- Author
-
Rebecca L. Kelly, Alexander Dobrovic, Matthew Wakefield, Kevin L. Peterson, Christian A. Ross, Taylor M. Weiskittel, Xiaonan Hou, Annapoorna Venkatachalam, Clare L. Scott, Thomas Harding, S. John Weroha, Karen S. Flatten, Larry M. Karnitz, Jill M. Wagner, Kevin K. Lin, Andrea E. Wahner Hendrickson, Cristina Correia, Hu Li, Scott H. Kaufmann, X. Wei Meng, Paul Haluska, Rachel M. Hurley, Olga Kondrashova, Paula A. Schneider, Ksenija Nesic, Marc R. Radke, Nicholas M. Pathoulas, Iain A. McNeish, Cordelia D. McGehee, and Elizabeth M. Swisher
- Subjects
Messenger RNA ,Methylation ,Biology ,Gene mutation ,medicine.disease ,chemistry.chemical_compound ,chemistry ,In vivo ,PARP inhibitor ,medicine ,Cancer research ,RAD51C ,Ovarian cancer ,Rucaparib - Abstract
Acquired PARP inhibitor (PARPi) resistance in BRCA1- or BRCA2-mutant ovarian cancer often results from secondary mutations that restore expression of functional protein. RAD51C is a less commonly studied ovarian cancer susceptibility gene whose promoter is sometimes methylated in the tumor, leading to homologous recombination deficiency and PARPi sensitivity. For this study, the PARPi-sensitive patient-derived xenograft PH039, which lacks demonstrable repair gene mutations but harbors RAD51C promoter methylation, was selected for PARPi resistance by repeated 21-day niraparib treatments in vivo. PH039 acquired PARPi resistance by the third cycle of treatment and demonstrated unimpeded growth during subsequent exposure to either niraparib or rucaparib. Transcriptional profiling throughout the time course of resistance development showed widespread pathway level changes along with a marked increase in RAD51C mRNA, which reflected loss of RAD51C promoter methylation. Analysis of RAD51C methylation in patient tumor samples from the ARIEL2 Part 1 clinical trial of rucaparib monotherapy likewise indicated that loss of RAD51C methylation prior to on-study biopsy was associated with limited response. Interestingly, the PARPi resistant PH039 model remained platinum sensitive. Collectively, these results not only indicate that PARPi treatment pressure can reverse RAD51C methylation and restore RAD51C expression, but also provide an important model for studying the clinical observation that PARPi and platinum sensitivity are sometimes dissociated.
- Published
- 2020
- Full Text
- View/download PDF
21. Acquired RAD51C promoter methylation loss causes PARP inhibitor resistance in high grade serous ovarian carcinoma
- Author
-
Nadia Traficante, Damien Kee, Clare L. Scott, David D.L. Bowtell, Kristy Shield-Artin, Nirashaa Bound, Gwo-Yaw Ho, Ksenija Nesic, Alexander Dobrovic, Cordelia D. McGehee, Mohammad Reza Eftekhariyan Ghamsari, Ashan Musafer, Nicola Waddell, Maria I. Harrell, Rachel M. Hurley, Orla McNally, S. John Weroha, Zi Qing Chai, Cassandra J. Vandenberg, Inger Olesen, Scott H. Kaufmann, Katia Nones, Anna deFazio, Genevieve Dall, Olga Kondrashova, Elizabeth Lieschke, Marc R. Radke, Elizabeth M. Swisher, and Matthew Wakefield
- Subjects
Chemotherapy ,medicine.medical_treatment ,Methylation ,Biology ,chemistry.chemical_compound ,Serous fluid ,chemistry ,Ovarian carcinoma ,PARP inhibitor ,Cancer research ,medicine ,Gene silencing ,RAD51C ,Rucaparib - Abstract
While loss of BRCA1 promoter methylation has been shown to cause PARP inhibitor (PARPi) resistance in high-grade serous ovarian carcinoma (HGSC), the impacts of RAD51C methylation (meRAD51C) remain unresolved. In this study, three PARPi-responsive HGSC patient-derived xenografts (PDX) with RAD51C gene silencing and homologous recombination deficiency were found to have either homogeneous or heterogeneous patterns of meRAD51C. PDX could lose meRAD51C following PARPi treatment (rucaparib/niraparib), where a single unmethylated RAD51C copy was sufficient to drive PARPi-resistance. Genomic profiling revealed this resistance was acquired independently in two distinct PDX lineages. Furthermore, we describe a patient sample where 1/3 RAD51C gene copies lost methylation following neoadjuvant chemotherapy. We show meRAD51C is a positive predictive biomarker for PARPi response and should be screened for routinely in patients. However, methylation loss in a single gene copy is sufficient to cause PARPi resistance and should be carefully assessed in previously treated patients considering PARPi therapy.
- Published
- 2020
- Full Text
- View/download PDF
22. Reactivating latent HIV with PKC agonists induces resistance to apoptosis and is associated with phosphorylation and activation of BCL2
- Author
-
Anisha Misra, Aswath P. Chandrasekar, Cristina Correia, Sekar Natesampillai, Andrew D. Badley, Nathan W. Cummins, Kevin L. Peterson, Scott H. Kaufmann, Ying Li, Alecia Alto, Andrea J. French, and Ashton Krogman
- Subjects
RNA viruses ,CD4-Positive T-Lymphocytes ,Apoptosis ,HIV Infections ,Pathology and Laboratory Medicine ,Biochemistry ,chemistry.chemical_compound ,White Blood Cells ,Latent Virus ,Immunodeficiency Viruses ,Animal Cells ,Medicine and Health Sciences ,Post-Translational Modification ,Phosphorylation ,Biology (General) ,Protein Kinase C ,0303 health sciences ,Cell Death ,T Cells ,030302 biochemistry & molecular biology ,Cell biology ,Virus Latency ,Proto-Oncogene Proteins c-bcl-2 ,Cell Processes ,Medical Microbiology ,Viral Pathogens ,Viruses ,Infectious diseases ,bcl-Associated Death Protein ,Cellular Types ,Pathogens ,Research Article ,Medical conditions ,Programmed cell death ,QH301-705.5 ,Immune Cells ,Immunology ,Viral diseases ,Microbiology ,03 medical and health sciences ,In vivo ,Virology ,Retroviruses ,Genetics ,Humans ,T Helper Cells ,Prostratin ,Molecular Biology ,Microbial Pathogens ,Protein kinase C ,030304 developmental biology ,Blood Cells ,Lentivirus ,Organisms ,Biology and Life Sciences ,HIV ,Proteins ,Cell Biology ,RC581-607 ,Viral Replication ,chemistry ,HIV-1 ,Parasitology ,Virus Activation ,Immunologic diseases. Allergy ,Ex vivo - Abstract
Eradication of HIV-1 by the “kick and kill” strategy requires reactivation of latent virus to cause death of infected cells by either HIV-induced or immune-mediated apoptosis. To date this strategy has been unsuccessful, possibly due to insufficient cell death in reactivated cells to effectively reduce HIV-1 reservoir size. As a possible cause for this cell death resistance, we examined whether leading latency reversal agents (LRAs) affected apoptosis sensitivity of CD4 T cells. Multiple LRAs of different classes inhibited apoptosis in CD4 T cells. Protein kinase C (PKC) agonists bryostatin-1 and prostratin induced phosphorylation and enhanced neutralizing capability of the anti-apoptotic protein BCL2 in a PKC-dependent manner, leading to resistance to apoptosis induced by both intrinsic and extrinsic death stimuli. Furthermore, HIV-1 producing CD4 T cells expressed more BCL2 than uninfected cells, both in vivo and after ex vivo reactivation. Therefore, activation of BCL2 likely contributes to HIV-1 persistence after latency reversal with PKC agonists. The effects of LRAs on apoptosis sensitivity should be considered in designing HIV cure strategies predicated upon the “kick and kill” paradigm., Author summary The major barrier to an HIV cure is the latent viral reservoir. We questioned why some drugs that reactivate latent HIV fail to reduce the viral reservoir size. We show that some HIV latency reversal agents, particularly PKC agonists such as bryostatin-1, activate the anti-apoptotic BCL2 protein in CD4 T cells. This unintended biologic effect inhibits apoptosis and, thereby, may promote HIV persistence despite viral reactivation. It is therefore important to screen potential latency reversal agents for off-target effects that might promote survival of HIV infected cells.
- Published
- 2020
23. Characterization of an alternative BAK-binding site for BH3 peptides
- Author
-
Junfeng Wang, Haiming Dai, Yuan Ping Pang, Hongbin Sun, Kaiqin Ye, Meng Chen, Bo Wu, Scott H. Kaufmann, Hongzhi Wang, Wei X. Meng, and Jia Gao
- Subjects
0301 basic medicine ,Magnetic Resonance Spectroscopy ,Science ,Protein domain ,General Physics and Astronomy ,Apoptosis ,Plasma protein binding ,Molecular dynamics ,Molecular Dynamics Simulation ,Jurkat cells ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Jurkat Cells ,0302 clinical medicine ,Protein Domains ,hemic and lymphatic diseases ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,lcsh:Science ,neoplasms ,Peptide sequence ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Multidisciplinary ,Binding Sites ,Sequence Homology, Amino Acid ,Chemistry ,Mutagenesis ,Signal transducing adaptor protein ,General Chemistry ,Cell biology ,030104 developmental biology ,bcl-2 Homologous Antagonist-Killer Protein ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,Mitochondrial Membranes ,Mutation ,lcsh:Q ,biological phenomena, cell phenomena, and immunity ,Bacterial outer membrane ,Peptides ,Apoptosis Regulatory Proteins ,Solution-state NMR ,Protein Binding - Abstract
Many cellular stresses are transduced into apoptotic signals through modification or up-regulation of the BH3-only subfamily of BCL2 proteins. Through direct or indirect mechanisms, these proteins activate BAK and BAX to permeabilize the mitochondrial outer membrane. While the BH3-only proteins BIM, PUMA, and tBID have been confirmed to directly activate BAK through its canonical BH3 binding groove, whether the BH3-only proteins BMF, HRK or BIK can directly activate BAK is less clear. Here we show that BMF and HRK bind and directly activate BAK. Through NMR studies, site-directed mutagenesis, and advanced molecular dynamics simulations, we also find that BAK activation by BMF and possibly HRK involves a previously unrecognized binding groove formed by BAK α4, α6, and α7 helices. Alterations in this groove decrease the ability of BMF and HRK to bind BAK, permeabilize membranes and induce apoptosis, suggesting a potential role for this BH3-binding site in BAK activation., Mitochondrial apoptosis is controlled by BCL2 family proteins, and the BH3-only proteins often act as sensors that transmit apoptotic signals. Here the authors show how the BH3-only proteins BMF and HRK can directly activate the BCL2 protein BAK and interact with BAK through an alternative binding groove.
- Published
- 2020
24. A randomized trial of three novel regimens for recurrent acute myeloid leukemia demonstrates the continuing challenge of treating this difficult disease
- Author
-
Xin V. Wang, Jessica K. Altman, Martin S. Tallman, Keith W. Pratz, Hillard M. Lazarus, Jacob M. Rowe, Scott H. Kaufmann, Mark R. Litzow, Han Win Tun, Edward R. Broun, Martin Carroll, Witold B. Rybka, Rhett P. Ketterling, Selina M. Luger, Judith E. Karp, Elisabeth Paietta, and Yanming Zhang
- Subjects
Male ,medicine.medical_specialty ,Gastrointestinal Diseases ,medicine.medical_treatment ,Article ,Disease-Free Survival ,Carboplatin ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Piperidines ,Randomized controlled trial ,Recurrence ,law ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Etoposide ,Aged ,Flavonoids ,Salvage Therapy ,Sirolimus ,Chemotherapy ,Mitoxantrone ,business.industry ,Remission Induction ,Cytarabine ,Hematology ,Middle Aged ,Alvocidib ,Hematologic Diseases ,Clinical trial ,Leukemia, Myeloid, Acute ,Regimen ,chemistry ,030220 oncology & carcinogenesis ,Female ,Topotecan ,Tumor Lysis Syndrome ,business ,Follow-Up Studies ,030215 immunology ,medicine.drug - Abstract
To improve the outcome of relapsed/refractory acute myeloid leukemia (AML), a randomized phase II trial of three novel regimens was conducted. Ninety patients were enrolled and were in first relapse or were refractory to induction/re-induction chemotherapy. They were randomized to the following regimens: carboplatin-topotecan (CT), each by continuous infusion for five days; alvocidib (formerly flavopiridol), cytarabine, and mitoxantrone (FLAM) in a timed sequential regimen; or sirolimus combined with mitoxantrone, etoposide, and cytarabine (S-MEC). The primary objective was attainment of a complete remission (CR). A Simon two-stage design was used for each of the three arms. The median age of the patients in the FLAM arm was older at 62 years compared to 55 years for the CT arm and the S-MEC arm. The overall response was 14% in the CT arm (5/35, 90% CI 7%−35%), 28% in the FLAM arm (10/36, 90% CI, 16%−43%), and 16% in the S-MEC arm (3/19, 90% CI, 4%−36%). There were nine treatment-related deaths, seven of which occurred in the FLAM arm with four of these in elderly patients. We conclude that the FLAM regimen had an encouraging response rate and should be considered for further clinical development but should be used with caution in elderly patients.
- Published
- 2018
- Full Text
- View/download PDF
25. Fibroblast growth factor receptor inhibition induces loss of matrix MCL1 and necrosis in cholangiocarcinoma
- Author
-
Petra Hirsova, Gregory J. Gores, Scott H. Kaufmann, Ayano Kabashima, Mark J. Truty, Sumera Rizvi, Matthew C. Hernandez, and Steven F. Bronk
- Subjects
Male ,0301 basic medicine ,Programmed cell death ,Indazoles ,Indoles ,Necrosis ,FGFR Inhibition ,Cell ,Mice, SCID ,Article ,Cholangiocarcinoma ,Mice ,03 medical and health sciences ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Extracellular ,Animals ,Humans ,Receptor ,Sulfonamides ,Cell Death ,Hepatology ,Chemistry ,Receptors, Fibroblast Growth Factor ,Xenograft Model Antitumor Assays ,Mitochondria ,030104 developmental biology ,medicine.anatomical_structure ,Bile Duct Neoplasms ,Cell culture ,Cancer cell ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,medicine.symptom ,Oxidation-Reduction - Abstract
Background & Aims Myeloid cell leukemia 1 (MCL1), a prosurvival member of the BCL2 protein family, has a pivotal role in human cholangiocarcinoma (CCA) cell survival. We previously reported that fibroblast growth factor receptor (FGFR) signalling mediates MCL1-dependent survival of CCA cells in vitro and in vivo. However, the mode and mechanisms of cell death in this model were not delineated. Methods Human CCA cell lines were treated with the pan-FGFR inhibitor LY2874455 and the mode of cell death examined by several complementary assays. Mitochondrial oxidative metabolism was examined using a XF24 extracellular flux analyser. The efficiency of FGFR inhibition in patient-derived xenografts (PDX) was also assessed. Results CCA cells expressed two species of MCL1, a full-length form localised to the outer mitochondrial membrane, and an N terminus-truncated species compartmentalised within the mitochondrial matrix. The pan-FGFR inhibitor LY2874455 induced non-apoptotic cell death in the CCA cell lines associated with cellular depletion of both MCL1 species. The cell death was accompanied by failure of mitochondrial oxidative metabolism and was most consistent with necrosis. Enforced expression of N terminus-truncated MCL1 targeted to the mitochondrial matrix, but not full-length MCL1 targeted to the outer mitochondrial membrane, rescued cell death and mitochondrial function. LY2874455 treatment of PDX-bearing mice was associated with tumour cell loss of MCL1 and cell necrosis. Conclusions FGFR inhibition induces loss of matrix MCL1, resulting in cell necrosis. These observations support a heretofore unidentified, alternative MCL1 survival function, namely prevention of cell necrosis, and have implications for treatment of human CCA. Lay summary Herein, we report that therapeutic inhibition of a cell receptor expressed by bile duct cancer cells resulted in the loss of a critical survival protein termed MCL1. Cellular depletion of MCL1 resulted in the death of the cancer cells by a process characterised by cell rupture. Cell death by this process can stimulate the immune system and has implications for combination therapy using receptor inhibition with immunotherapy.
- Published
- 2018
- Full Text
- View/download PDF
26. A Phase I Study of Pevonedistat, Azacitidine and Venetoclax for Patients with Relapsed/Refractory Acute Myelogenous Leukemia (AML)
- Author
-
Karen Carlson, Mark R. Litzow, Arielle Baim, Althea Thomas, Aniko Szabo, Laura C. Michaelis, Alexandra M. Harrington, Sameem Abedin, Sonia Maldonado-Schmidt, Walter L. Longo, Lyndsey Runaas, Alexander Hinman, Ehab Atallah, Scott H. Kaufmann, and Guru Subramanian Guru Murthy
- Subjects
Oncology ,medicine.medical_specialty ,Acute myelogenous leukemia (AML) ,business.industry ,Venetoclax ,Immunology ,Azacitidine ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Phase i study ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Relapsed refractory ,Medicine ,business ,medicine.drug - Abstract
Background: Outcomes of patients with relapsed/refractory AML (RR-AML) have remained poor. Therapy with venetoclax based combinations in this setting leads to CR/CRi rates of 21-49% (DiNardo CD et al. Am J Hematol 2018, Aldoss I et al. Haematologica 2018, Stahl M et al. Blood Adv 2020). Preclinical studies with BCL-2 inhibitors indicate potential mechanisms of drug resistance including overexpression of the anti-apoptotic protein MCL-1 (Konopleva et al. Cancer Cell. 2006). Pevonedistat is a first in class inhibitor of Nedd8 activating enzyme that induces the pro-apoptotic protein NOXA leading to neutralization of MCL-1 and apoptosis. Preclinical studies evaluating the combination of pevonedistat and venetoclax against AML cell lines have demonstrated a synergistic effect (Knorr KL et al. Cell Death Differ. 2015). Hence, we designed a phase I study to assess the safety and tolerability of adding pevonedistat to the combination of azacitidine and venetoclax in patients with RR-AML. Study design and methods: We conducted a phase I study with the combination of pevonedistat, venetoclax and azacitidine in patients with RR-AML. Patients aged 18 years or above with morphologically documented RR-AML, ECOG performance status 0-2 and adequate organ function were eligible. Major exclusion criteria were isolated extramedullary relapse, hematopoietic cell transplantation (HCT) within 100 days of enrollment, and active acute GVHD. Previous therapy with hypomethylating agents (HMA) or venetoclax was not an exclusion criterion. The dose escalation phase was conducted using 3+3 design. Treatment included azacitidine (75 mg/m 2 daily x 7 days), venetoclax (400 mg daily x 28 days), and pevonedistat in escalating doses (10-20 mg/m 2 IV days 1,3,5 of each cycle) with a cycle length of 28 days. Pevonedistat was given at 10 mg/m 2 dose in cohort 1, 15 mg/m 2 dose in cohort 2 and 20 mg/m 2 dose in cohort 3. The primary endpoint is to determine the recommended phase 2 dose (RP2D) and toxicity profile of pevonedistat, azacitidine and venetoclax. Other endpoints included determination of response rates, duration of response, survival, pharmacokinetics, correlation of response rates with AML genomic profile, correlation of pretreatment levels of BCL2, BCLXL, MCL1, BAX or BAK with response, determination of changes in NOXA (PMAIP1) mRNA and protein expression pre-and post-pevonedistat treatment, evaluation of BH3 mimetic profiling on bone marrow samples by flow cytometry and assessing the sensitivity of leukemia and leukemic stem/progenitor cells to pevonedistat ex vivo. Results: Thirteen patients participated in the dose escalation phase, 12 of whom were evaluable. Median age was 69 years (61-91), 30.8% had secondary/therapy related AML, 69.2% with adverse risk disease, 53.8% previously received venetoclax/HMA and 23.1% had relapse after prior allogeneic hematopoietic cell transplantation (HCT) (Table 1). Seven patients were enrolled into cohort 1 (pevonedistat 10 mg/m 2 dose) of which one was not evaluable, three patients enrolled into cohort 2 (pevonedistat 15 mg/m 2 dose), and three patients enrolled into cohort 3 (pevonedistat 20 mg/m 2 dose). Grade 3 or higher AEs included febrile neutropenia (23%), infection (15%), anemia (38%), neutropenia (54%), thrombocytopenia (38%). There was 1 dose limiting toxicity (DLT) in cohort 1 (atrial fibrillation) that triggered cohort expansion. However, subsequent patients did not experience DLT despite planned dose escalation. Of the 12 evaluable total patients, CR/CRi was observed in 5 (41.6%) patients. Notably, patients with RR-AML who were venetoclax/HMA naïve had a CR/CRi 83.3% (5/6 patients). The response rates for each cohort are summarized in Table 2. Three of the five patients with CR/CRi (60%) achieved MRD negativity by flow cytometry. Four patients who achieved CR/CRi proceeded to allogeneic HCT after therapy. Median OS of the cohort was 5.4 months (1.8-14) and median OS was not reached in patients with CR/CRi. Conclusions: The addition of pevonedistat to venetoclax and azacitidine backbone is safe and well tolerated in patients with RR-AML. Dose escalation yielded encouraging efficacy in venetoclax/HMA naïve RR-AML patients. The study is currently in the dose expansion phase. Details on correlative studies examining mechanisms of therapeutic efficacy and resistance will be reported in the main meeting. Figure 1 Figure 1. Disclosures Guru Murthy: Cardinal health: Honoraria; TG Therapeutics: Other: Advisory board meeting; DAVA Oncology: Honoraria; CancerExpertNow: Honoraria; Qessential: Honoraria; Techspert: Consultancy; Curio Sciences: Honoraria; Guidepoint: Consultancy. Abedin: AltruBio: Research Funding; Helsinn: Research Funding; Amgen: Honoraria; Pfizer: Research Funding; Agios: Honoraria; Actinium: Research Funding; Astellas Pharma Inc.: Research Funding. Litzow: Jazz: Other: Advisory Board; Pluristem: Research Funding; Actinium: Research Funding; Amgen: Research Funding; Astellas: Research Funding; AbbVie: Research Funding; Omeros: Other: Advisory Board; Biosight: Other: Data monitoring committee. Atallah: Novartis: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Research Funding; BMS: Honoraria, Speakers Bureau; Takeda: Consultancy, Research Funding; Abbvie: Consultancy, Speakers Bureau; Amgen: Consultancy.
- Published
- 2021
- Full Text
- View/download PDF
27. Histone deacetylase inhibitors reduce differentiating osteoblast-mediated protection of acute myeloid leukemia cells from cytarabine
- Author
-
Rosalie M. Sterner, Aref Al-Kali, Mrinal M. Patnaik, Naseema Gangat, Scott H. Kaufmann, Andre J. van Wijnen, Mark R. Litzow, Kimberly N. Kremer, Jennifer J. Westendorf, and Karen E. Hedin
- Subjects
0301 basic medicine ,panobinostat ,medicine.medical_treatment ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,AML ,Panobinostat ,hemic and lymphatic diseases ,medicine ,Vorinostat ,neoplasms ,Chemotherapy ,business.industry ,HDACi ,Myeloid leukemia ,Osteoblast ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,vorinostat ,030220 oncology & carcinogenesis ,Immunology ,Cytarabine ,osteoblast ,Histone deacetylase ,Bone marrow ,business ,medicine.drug ,Research Paper - Abstract
The bone marrow microenvironment protects acute myeloid leukemia (AML) cells during chemotherapy and is a major factor in relapse. Here, we examined which type(s) of bone marrow cells are responsible for the relapse of AML following treatment with cytarabine (Ara-C), and we identified a means to inhibit this protection. To determine the protective cell type(s), AML cells were treated with Ara-C, and AML cell survival in the presence or absence of osteoblast lineage cells was assessed. Cultured AML cells and patient bone marrow isolates were each significantly protected from Ara-C-induced apoptosis by co-culture with differentiating osteoblasts. Moreover, pretreating differentiating osteoblasts with the histone deacetylase inhibitors (HDACi) vorinostat and panobinostat abrogated the ability of the differentiating osteoblasts to protect AML cells. Together, our results indicate that differentiating osteoblasts have the potential to promote residual AML in the bone marrow following standard chemotherapy and act via a mechanism requiring HDACi-sensitive gene expression. Using HDACi to target the leukemic microenvironment in combination with Ara-C could potentially improve treatment of AML. Moreover, other strategies for manipulating bone marrow osteoblasts may also help eradicate AML cells and reduce relapse.
- Published
- 2017
28. Resistance to venetoclax and hypomethylating agents in acute myeloid leukemia
- Author
-
Antoine N. Saliba, Scott H. Kaufmann, and August J. John
- Subjects
Oncology ,azacitidine ,medicine.medical_specialty ,hypomethylating agents ,business.industry ,Venetoclax ,Azacitidine ,Myeloid leukemia ,Decitabine ,acute myeloid leukemia ,Article ,Treatment failure ,resistance ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Medicine ,business ,decitabine ,medicine.drug - Abstract
Despite the success of the combination of venetoclax with the hypomethylating agents (HMA) decitabine or azacitidine in inducing remission in older, previously untreated patients with acute myeloid leukemia (AML), resistance - primary or secondary - still constitutes a significant roadblock in the quest to prolong the duration of response. Here we review the proposed and proven mechanisms of resistance to venetoclax monotherapy, HMA monotherapy, and the doublet of venetoclax and HMA for the treatment of AML. We approach the mechanisms of resistance to HMAs and venetoclax in the light of the agents’ mechanisms of action. We briefly describe potential therapeutic strategies to circumvent resistance to this promising combination, including alternative scheduling or the addition of other agents to the HMA and venetoclax backbone. Understanding the mechanisms of action and evolving resistance in AML remains a priority in order to maximize the benefit from novel drugs and combinations, identify new therapeutic targets, define potential prognostic markers, and avoid treatment failure.
- Published
- 2020
- Full Text
- View/download PDF
29. DNA Repair, Apoptotic Pathways, CDK Inhibitors
- Author
-
Scott H. Kaufmann and Mira A. Kohorst
- Subjects
biology ,DNA repair ,Apoptosis ,Chemistry ,Cyclin-dependent kinase ,Cancer research ,biology.protein - Published
- 2019
- Full Text
- View/download PDF
30. The DNA Cytosine Deaminase APOBEC3B is a Molecular Determinant of Platinum Responsiveness in Clear Cell Ovarian Cancer
- Author
-
Matthew C. Jarvis, Scott H. Kaufmann, Ethan P. Heinzen, William L. Brown, Rachel Isaksson Vogel, S. John Weroha, Sun Hee Lee, Krista M. Goergen, Artur A. Serebrenik, Prokopios P. Argyris, Ann L. Oberg, Britt K. Erickson, Xiaonan Hou, Reuben S. Harris, Martina Bazzaro, Yajue Huang, and Matthew J. Maurer
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,Gene Expression ,Antineoplastic Agents ,Kaplan-Meier Estimate ,Biology ,Article ,Minor Histocompatibility Antigens ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Cytidine Deaminase ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Clear-cell ovarian carcinoma ,Platinum ,Cisplatin ,Ovarian Neoplasms ,Cytosine deaminase ,Cancer ,medicine.disease ,Prognosis ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Carboplatin ,Disease Models, Animal ,030104 developmental biology ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Biomarker (medicine) ,Female ,Ovarian cancer ,Synthetic Lethal Mutations ,Clear cell ,medicine.drug - Abstract
Purpose: Clear cell ovarian carcinoma (CCOC) is an aggressive disease that often demonstrates resistance to standard chemotherapies. Approximately 25% of patients with CCOC show a strong APOBEC mutation signature. Here, we determine which APOBEC3 enzymes are expressed in CCOC, establish clinical correlates, and identify a new biomarker for detection and intervention. Experimental Designs: APOBEC3 expression was analyzed by IHC and qRT-PCR in a pilot set of CCOC specimens (n = 9 tumors). The IHC analysis of APOBEC3B was extended to a larger cohort to identify clinical correlates (n = 48). Dose-response experiments with platinum-based drugs in CCOC cell lines and carboplatin treatment of patient-derived xenografts (PDXs) were done to address mechanistic linkages. Results: One DNA deaminase, APOBEC3B, is overexpressed in a formidable subset of CCOC tumors and is low or absent in normal ovarian and fallopian tube epithelial tissues. High APOBEC3B expression associates with improved progression-free survival (P = 0.026) and moderately with overall survival (P = 0.057). Cell-based studies link APOBEC3B activity and subsequent uracil processing to sensitivity to cisplatin and carboplatin. PDX studies extend this mechanistic relationship to CCOC tissues. Conclusions: These studies demonstrate that APOBEC3B is overexpressed in a subset of CCOC and, contrary to initial expectations, associated with improved (not worse) clinical outcomes. A likely molecular explanation is that APOBEC3B-induced DNA damage sensitizes cells to additional genotoxic stress by cisplatin. Thus, APOBEC3B is a molecular determinant and a candidate predictive biomarker of the therapeutic response to platinum-based chemotherapy. These findings may have broader translational relevance, as APOBEC3B is overexpressed in many different cancer types.
- Published
- 2019
31. BRCA1 Deficiency Upregulates NNMT, Which Reprograms Metabolism and Sensitizes Ovarian Cancer Cells to Mitochondrial Metabolic Targeting Agents
- Author
-
Taro Hitosugi, Catherine J. Huntoon, Daniel R. O'Brien, Cristina Correia, Xiaonan Hou, Scott H. Kaufmann, Hu Li, Arun Kanakkanthara, Thomas L. Ekstrom, Ethan P. Heinzen, Ann L. Oberg, S. John Weroha, Emma R. Purfeerst, Andrea E. Wahner Hendrickson, Kiran Kurmi, Sean C. Dowdy, and Larry M. Karnitz
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system diseases ,DNA repair ,Carcinoma, Ovarian Epithelial ,Tigecycline ,Oxidative Phosphorylation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,Glucose import ,Antineoplastic Combined Chemotherapy Protocols ,Transcriptional regulation ,Hydroxybenzoates ,Nicotinamide N-Methyltransferase ,Animals ,Humans ,skin and connective tissue diseases ,Promoter Regions, Genetic ,Regulation of gene expression ,Ovarian Neoplasms ,Chemistry ,BRCA1 Protein ,Ovary ,Hydrazones ,DNA Methylation ,Triazoles ,Xenograft Model Antitumor Assays ,Cyclin-Dependent Kinases ,Mitochondria ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Mutation ,Cancer research ,Female ,Homologous recombination ,Energy Metabolism ,Reprogramming - Abstract
BRCA1 plays a key role in homologous recombination (HR) DNA repair. Accordingly, changes that downregulate BRCA1, including BRCA1 mutations and reduced BRCA1 transcription, due to promoter hypermethylation or loss of the BRCA1 transcriptional regulator CDK12, disrupt HR in multiple cancers. In addition, BRCA1 has also been implicated in the regulation of metabolism. Here, we show that reducing BRCA1 expression, either by CDK12 or BRCA1 depletion, led to metabolic reprogramming of ovarian cancer cells, causing decreased mitochondrial respiration and reduced ATP levels. BRCA1 depletion drove this reprogramming by upregulating nicotinamide N-methyltransferase (NNMT). Notably, the metabolic alterations caused by BRCA1 depletion and NNMT upregulation sensitized ovarian cancer cells to agents that inhibit mitochondrial metabolism (VLX600 and tigecycline) and to agents that inhibit glucose import (WZB117). These observations suggest that inhibition of energy metabolism may be a potential strategy to selectively target BRCA1-deficient high-grade serous ovarian cancer, which is characterized by frequent BRCA1 loss and NNMT overexpression. Significance: Loss of BRCA1 reprograms metabolism, creating a therapeutically targetable vulnerability in ovarian cancer.
- Published
- 2019
32. Abstract 1426: Multiomic analysis identifies CPT1A and fatty acid oxidation as a potential therapeutic target in platinum-refractory high grade serous ovarian cancer
- Author
-
Scott H. Kaufmann, Hong Wang, Bing Zhang, Amanda G. Paulovich, Pei Wang, Richard G. Ivey, Sara R. Savage, Michael J. Birrer, Catherine J. Huntoon, Andrew N. Hoofnagle, Anna Calinawan, Steven P. Gygi, Shrabanti Chowdhury, Larry M. Karnitz, Jacob J. Kennedy, Uliana J. Voytovich, S. John Weroha, Zahra Shire, Chenwei Lin, Jeffrey R. Whiteaker, Zachary T. Herbert, Dongqing Hugang, Qing Yu, and Xiaonan Hou
- Subjects
Cancer Research ,endocrine system diseases ,business.industry ,chemistry.chemical_element ,Cancer ,medicine.disease ,female genital diseases and pregnancy complications ,Carboplatin ,chemistry.chemical_compound ,Oncology ,chemistry ,Cell culture ,In vivo ,Platinum resistance ,Serous ovarian cancer ,Cancer research ,Medicine ,business ,Platinum ,Beta oxidation - Abstract
Platinum-based DNA cross-linking agents are widely used anti-cancer drugs. Tumor resistance to platinum compounds is a major determinant of patient survival, including in high grade serous ovarian cancer (HGSOC). Remarkably, despite >30 years of literature on platinum responses in human cancer, none of these findings is used clinically as a predictive biomarker to stratify patients for platinum resistance, nor exploited therapeutically to treat platinum-resistant disease. Thus, understanding mechanisms of platinum resistance is an urgent goal, both to identify predictive biomarkers of platinum response (to spare patients with platinum-resistant tumors futile platinum therapy) and to develop efficacious therapies for platinum-resistant disease. To better understand mechanisms underlying platinum resistance in HGSOC, we performed comprehensive, dynamic (+/-carboplatin), multiomic profiling of DNA, RNA, protein and post-translational modifications (phosphorylation, ubiquitination, acetylation) to identify the cellular networks that respond to platinum treatment and associate with platinum resistance in three HGSOC intra-patient cell line pairs (PEA1S/PEA2R, PEO1S/PEO4R, PEO14S/PEO23R). The cell line pairs were derived from ascites or pleural effusions (Langdon et al., 1988) from three patients both before (PEA1S, PEO1S, PEO14S) and after (PEA2R, PEO4R, PEO23R, respectively) their tumors became clinically platinum resistant (i.e., in vivo development of resistance). The molecular profiles revealed extensive responses to carboplatin and differential responses between platinum-sensitive and platinum-resistant cells. Higher oxidative phosphorylation and fatty acid oxidation (FAO) pathway expression were observed in the platinum-resistant cells, which was further validated in patient-derived xenograft (PDX) models. We show that pharmacologic inhibition or CRISPR knockout of CPT1A, which represents a rate limiting step of FAO, sensitize HGSOC cells to platinum. Thus, FAO, and CPT1A in particular, represent a candidate therapeutic target to overcome platinum resistance in HGSOC. Citation Format: Hong Wang, Dongqing Hugang, Shrabanti Chowdhury, Sara Savage, Richard Ivey, Jacob Kennedy, Jeffrey Whiteaker, Chenwei Lin, Xiaonan Hou, Catherine Huntoon, Uliana Voytovich, Zahra Shire, Qing Yu, Steven Gygi, Andrew Hoofnagle, Zachary Herbert, Anna Calinawan, Larry Karnitz, S. John Weroha, Scott Kaufmann, Bing Zhang, Pei Wang, Michael Birrer, Amanda Paulovich. Multiomic analysis identifies CPT1A and fatty acid oxidation as a potential therapeutic target in platinum-refractory high grade serous ovarian cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1426.
- Published
- 2021
- Full Text
- View/download PDF
33. In vivo anti-tumor activity of the PARP inhibitor niraparib in homologous recombination deficient and proficient ovarian carcinoma
- Author
-
Sarah McKinstry, Rachel M. Hurley, Scott H. Kaufmann, Ann L. Oberg, Keith M. Wilcoxen, X. Wei Meng, Sean C. Harrington, Maria I. Harrell, Mariam M. AlHilli, S. John Weroha, Karen S. Flatten, Matt J. Maurer, Kieran M. Hawthorne, Paul Haluska, Elizabeth M. Swisher, Marc A. Becker, Xiaonan Hou, and Kimberly R. Kalli
- Subjects
0301 basic medicine ,Indazoles ,endocrine system diseases ,DNA repair ,Genes, BRCA2 ,RAD51 ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,PARP1 ,Piperidines ,medicine ,Humans ,Homologous Recombination ,Promoter Regions, Genetic ,Ovarian Neoplasms ,business.industry ,Obstetrics and Gynecology ,medicine.disease ,Carboplatin ,DNA-Binding Proteins ,030104 developmental biology ,Oncology ,chemistry ,Paclitaxel ,030220 oncology & carcinogenesis ,PARP inhibitor ,Cancer research ,RAD51C ,Female ,Ovarian cancer ,business - Abstract
Objective Poly(ADP-ribose) polymerase (PARP) inhibitors have yielded encouraging responses in high-grade serous ovarian carcinomas (HGSOCs), but the optimal treatment setting remains unknown. We assessed the effect of niraparib on HGSOC patient-derived xenograft (PDX) models as well as the relationship between certain markers of homologous recombination (HR) status, including BRCA1 / 2 mutations and formation of RAD51 foci after DNA damage, and response of these PDXs to niraparib in vivo . Methods Massively parallel sequencing was performed on HGSOCs to identify mutations contributing to HR deficiency. HR pathway integrity was assessed using fluorescence microscopy-based RAD51 focus formation assays. Effects of niraparib (MK-4827) on treatment-naive PDX tumor growth as monotherapy, in combination with carboplatin/paclitaxel, and as maintenance therapy were assessed by transabdominal ultrasound. Niraparib responses were correlated with changes in levels of poly(ADP-ribose), PARP1, and repair proteins by western blotting. Results Five PDX models were evaluated in vivo . Tumor regressions were induced by single-agent niraparib in one of two PDX models with deleterious BRCA2 mutations and in a PDX with RAD51C promoter methylation. Diminished formation of RAD51 foci failed to predict response, but Artemis loss was associated with resistance. Niraparib generally failed to enhance responses to carboplatin/paclitaxel chemotherapy, but maintenance niraparib therapy delayed progression in a BRCA2 -deficient PDX. Conclusions Mutations in HR genes are neither necessary nor sufficient to predict response to niraparib. Assessment of repair status through multiple complementary assays is needed to guide PARP inhibitor therapy, design future clinical trials and identify ovarian cancer patients most likely to benefit from PARP inhibition.
- Published
- 2016
- Full Text
- View/download PDF
34. Immunodetection of human topoisomerase I-DNA covalent complexes
- Author
-
Paula A. Schneider, Kevin L. Peterson, Anand G. Patel, Thomas G. Beito, Karen S. Flatten, Scott H. Kaufmann, Angela L. Perkins, and Daniel A. Harki
- Subjects
0301 basic medicine ,Apoptosis ,Histones ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,DNA Breaks, Double-Stranded ,Cytotoxicity ,Nucleic Acid Enzymes ,Antibodies, Monoclonal ,3. Good health ,Gene Expression Regulation, Neoplastic ,DNA Topoisomerases, Type I ,Biochemistry ,Covalent bond ,030220 oncology & carcinogenesis ,Protein Binding ,medicine.drug ,Molecular Sequence Data ,Biology ,Topoisomerase-I Inhibitor ,Structure-Activity Relationship ,03 medical and health sciences ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Structure–activity relationship ,Amino Acid Sequence ,Benzodioxoles ,Cisplatin ,Topoisomerase ,DNA ,HCT116 Cells ,Isoquinolines ,Antineoplastic Agents, Phytogenic ,030104 developmental biology ,chemistry ,biology.protein ,Rad51 Recombinase ,Topoisomerase I Inhibitors ,K562 Cells ,Topotecan ,Sequence Alignment ,Camptothecin - Abstract
A number of established and investigational anticancer drugs slow the religation step of DNA topoisomerase I (topo I). These agents induce cytotoxicity by stabilizing topo I-DNA covalent complexes, which in turn interact with advancing replication forks or transcription complexes to generate lethal lesions. Despite the importance of topo I-DNA covalent complexes, it has been difficult to detect these lesions within intact cells and tumors. Here, we report development of a monoclonal antibody that specifically recognizes covalent topo I-DNA complexes, but not free topo I or DNA, by immunoblotting, immunofluorescence or flow cytometry. Utilizing this antibody, we demonstrate readily detectable topo I-DNA covalent complexes after treatment with camptothecins, indenoisoquinolines and cisplatin but not nucleoside analogues. Topotecan-induced topo I-DNA complexes peak at 15-30 min after drug addition and then decrease, whereas indotecan-induced complexes persist for at least 4 h. Interestingly, simultaneous staining for covalent topo I-DNA complexes, phospho-H2AX and Rad51 suggests that topotecan-induced DNA double-strand breaks occur at sites distinct from stabilized topo I-DNA covalent complexes. These studies not only provide new insight into the action of topo I-directed agents, but also illustrate a strategy that can be applied to study additional topoisomerases and their inhibitors in vitro and in vivo.
- Published
- 2016
- Full Text
- View/download PDF
35. Exploring the relationship between homologous recombination score and progression-free survival in BRCA wildtype ovarian carcinoma: Analysis of veliparib plus carboplatin/paclitaxel in the velia study
- Author
-
Kirsten Timms, Gini F. Fleming, Robert L. Coleman, Michael A. Bookman, Danielle Sullivan, Scott H. Kaufmann, Michael J. Birrer, Bruce A. Bach, Douglas A. Levine, Minh H. Dinh, Vasudha Sehgal, Kathleen N. Moore, Peter Ansell, Carol Aghajanian, Elizabeth M. Swisher, and Brenden Chen
- Subjects
Veliparib ,business.industry ,Wild type ,Obstetrics and Gynecology ,Carboplatin/paclitaxel ,chemistry.chemical_compound ,Oncology ,chemistry ,Ovarian carcinoma ,Cancer research ,Medicine ,Progression-free survival ,Homologous recombination ,business - Published
- 2020
- Full Text
- View/download PDF
36. Abstract CT114: Pharmacokinetic analysis of navitoclax in combination with sorafenib in patients with relapsed or refractory solid organ tumors
- Author
-
Emily J. Koubek, Mitesh J. Borad, Yixing Jiang, Jun Yin, Carrie Strand, Sarah A. Buhrow, Renee M. McGovern, Scott H. Kaufmann, Naoko Takebe, Brian A. Costello, Renee A. Schoon, Alex A. Adjei, and Joel M. Reid
- Subjects
Sorafenib ,Volume of distribution ,Cancer Research ,Navitoclax ,business.industry ,Phases of clinical research ,Pharmacology ,chemistry.chemical_compound ,Oncology ,Pharmacokinetics ,chemistry ,Pharmacodynamics ,Concomitant ,medicine ,Dosing ,business ,medicine.drug - Abstract
Prior in vitro work in the human hepatoma cell lines Huh7 and HEP3B indicated that combination treatment of the BH3 mimetic, navitoclax, and the multi-kinase inhibitor, sorafenib, was more effective at inducing apoptosis than either compound alone. This current study was part of a phase I clinical trial to determine the maximum tolerated dose of concurrent navitoclax and sorafenib treatment in patients with relapsed or refractory solid organ tumors and sought to characterize the pharmacokinetics (PK) and pharmacodynamics (PD) of both agents in combination. Cleavage of cytokeratin 18 by caspase 3 was used as a biomarker for therapy-induced activation of apoptosis to determine if combination treatment results in an increase in apoptosis as previously observed in in vitro work. PK were assessed in 26 patients (ages 32 - 80) enrolled in a phase I clinical trial (NCT02143401). 150 mg of oral navitoclax was administered once daily for a seven day run-in (beginning Day -7) prior to 150 mg (Dose level 1) or 200 mg (Dose level 2) navitoclax concomitant treatment with 400 mg of twice daily oral sorafenib (Day 1). Samples for pharmacokinetic analysis were obtained on Day -7 and Day 1 prior to dosing and at 1, 2, 4, 8-12, and 24 hours post dosing. PK parameters were estimated by noncompartmental analysis using Phoenix® WinNonlin® Version 6.4. To examine the PD of both agents in combination, cytokeratin 18 cleavage was quantified by ELISA after the 7-day navitoclax run-in and on Day 2 or 4 and Day 8 of navitoclax and sorafenib combination treatment. PK estimates of maximum concentration, exposure, volume of distribution, and clearance for navitoclax on Day -7 and Day 1 were not significantly different, indicating a lack of navitoclax accumulation. Finally, no correlation was found between navitoclax exposure and apoptosis as indicated by cytokeratin-18 cleavage. This work was supported by the NCI Cancer Center Support Grant P30 CA15083 and NCI Experimental Therapeutics Phase I Grant UM1 CA186686. Citation Format: Emily J. Koubek, Brian A. Costello, Jun Yin, Renee M. McGovern, Sarah A. Buhrow, Renee A. Schoon, Carrie A. Strand, Yixing Jiang, Mitesh J. Borad, Naoko Takebe, Scott H. Kaufmann, Alex A. Adjei, Joel M. Reid. Pharmacokinetic analysis of navitoclax in combination with sorafenib in patients with relapsed or refractory solid organ tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT114.
- Published
- 2020
- Full Text
- View/download PDF
37. Abstract A33: Loss of ZC3H18 disrupts homologous recombination repair and sensitizes ovarian cancer cells to PARP inhibitors and DNA cross-linking agents
- Author
-
Ann L. Oberg, Scott H. Kaufmann, Arun Kanakkanthara, Minzhi Zhang, Catherine J. Huntoon, Larry M. Karnitz, Ethan P. Heinzen, Xiaonan Hou, and John Weroha
- Subjects
Cancer Research ,Oncology ,Crosslinking of DNA ,Chemistry ,Poly ADP ribose polymerase ,Cancer research ,Ovarian cancer cells ,Homologous recombination - Abstract
About 50% of high-grade serous ovarian cancers (HGSOC) have defects in homologous recombination (HR) DNA repair. Intriguingly, HR-defective tumors are highly susceptible to poly(ADP-ribose) polymerase (PARP) inhibitors and platinum-based chemotherapy. Mutations in BRCA1/2 or epigenetic silencing of BRCA1 via promoter hypermethylation contributes to ~30% of the HR defects, but other genetic alterations that cause HR deficiency in HGSOC remain unclear. To identify novel genes that may regulate HR, we studied Zinc Finger CCCH-Type Containing 18 (ZC3H18), encoded by ZC3H18. Notably, ZC3H18 lies within 16q24.2, a region with frequent copy number loss in HGSOC. Here we demonstrate that BRCA1 is transcriptionally activated by a previously unknown function of ZC3H18. We show that ZC3H18 interacts with an E2F site in the BRCA1 promoter where it facilitates recruitment of E2F4 to an adjacent E2F site to promote BRCA1 transcription. Consistent with the role of ZC3H18 in promoting BRCA1 expression, ZC3H18 depletion induces BRCA1 promoter methylation, reduces BRCA1 expression, disrupts HR, and sensitizes ovarian cancer cells to PARP inhibitors and platinum agents. Moreover, in patient-derived xenografts and primary HGSOC tumors, ZC3H18 mRNA levels were positively correlated with BRCA1 mRNA levels, further supporting the role of ZC3H18 in regulating BRCA1. Together, these findings suggest that loss and/or reduced ZC3H18 expression may help identify HGSOC patients most likely to benefit from PARP inhibitor and platinum-based therapies. Summary of the Unpublished Data/Significance/Innovation: These studies are innovative on several fronts. First, although BRCA1 methylation is a frequent event that mediates tumorigenesis and therapeutic responses in HGSOC, how BRCA1 methylation is regulated remains unknown. These studies will be the first to provide a mechanistic basis for how BRCA1 is methylated in HGSOC. Second, our studies are the first to show a role for ZC3H18 in any cancer. Third, we show that ZC3H18 regulates HR and that its depletion sensitizes ovarian cancer cells to PARP inhibitors and platinum therapy. Thus, these studies will provide important basis for developing ZC3H18 loss/decreased abundance as a novel biomarker for ovarian cancers that may respond to PARP inhibitors and platinum agents. Citation Format: Arun Kanakkanthara, Catherine J. Huntoon, Xiaonan Hou, Minzhi Zhang, Ethan P Heinzen, Ann L. Oberg, John S. Weroha, Scott H. Kaufmann, Larry M. Karnitz. Loss of ZC3H18 disrupts homologous recombination repair and sensitizes ovarian cancer cells to PARP inhibitors and DNA cross-linking agents [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research; 2019 Sep 13-16, 2019; Atlanta, GA. Philadelphia (PA): AACR; Clin Cancer Res 2020;26(13_Suppl):Abstract nr A33.
- Published
- 2020
- Full Text
- View/download PDF
38. BRCA reversion mutations in circulating tumor DNA predict primary and acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma
- Author
-
Gottfried E. Konecny, Robert L. Coleman, Anna V. Tinker, Heidi Giordano, Carmen Say, Scott H. Kaufmann, Jeffrey D. Isaacson, Amit M. Oza, Ana Oaknin, Marc R. Radke, Elizabeth M. Swisher, Maria I. Harrell, Setsuko K. Chambers, Iain A. McNeish, Elena Helman, Isabelle Ray-Coquard, Lara Maloney, David M. O'Malley, Lan Thanh Vo, Kevin K. Lin, Clare L. Scott, Thomas Harding, James D. Brenton, Elaina Mann, James Sun, and Cancer Research UK
- Subjects
0301 basic medicine ,MAINTENANCE THERAPY ,endocrine system diseases ,Reversion ,medicine.disease_cause ,SECONDARY MUTATIONS ,BREAST ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Germline mutation ,medicine ,Carcinoma ,1112 Oncology and Carcinogenesis ,skin and connective tissue diseases ,Rucaparib ,Mutation ,Science & Technology ,business.industry ,OLAPARIB ,GERMLINE MUTATIONS ,CHEMOTHERAPY ,BRCA2 Protein ,medicine.disease ,female genital diseases and pregnancy complications ,030104 developmental biology ,chemistry ,Oncology ,030220 oncology & carcinogenesis ,CELL-FREE DNA ,PARP inhibitor ,Cancer research ,business ,Life Sciences & Biomedicine - Abstract
A key resistance mechanism to platinum-based chemotherapies and PARP inhibitors in BRCA-mutant cancers is the acquisition of BRCA reversion mutations that restore protein function. To estimate the prevalence of BRCA reversion mutations in high-grade ovarian carcinoma (HGOC), we performed targeted next-generation sequencing of circulating cell-free DNA (cfDNA) extracted from pretreatment and postprogression plasma in patients with deleterious germline or somatic BRCA mutations treated with the PARP inhibitor rucaparib. BRCA reversion mutations were identified in pretreatment cfDNA from 18% (2/11) of platinum-refractory and 13% (5/38) of platinum-resistant cancers, compared with 2% (1/48) of platinum-sensitive cancers (P = 0.049). Patients without BRCA reversion mutations detected in pretreatment cfDNA had significantly longer rucaparib progression-free survival than those with reversion mutations (median, 9.0 vs. 1.8 months; HR, 0.12; P < 0.0001). To study acquired resistance, we sequenced 78 postprogression cfDNA, identifying eight additional patients with BRCA reversion mutations not found in pretreatment cfDNA. Significance: BRCA reversion mutations are detected in cfDNA from platinum-resistant or platinum-refractory HGOC and are associated with decreased clinical benefit from rucaparib treatment. Sequencing of cfDNA can detect multiple BRCA reversion mutations, highlighting the ability to capture multiclonal heterogeneity. This article is highlighted in the In This Issue feature, p. 151
- Published
- 2018
39. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial
- Author
-
Robert L. Coleman, Heidi Giordano, Anne Floquet, Clare L. Scott, Jeff Isaacson, Andrew R. Allen, Ana Oaknin, Elizabeth M. Swisher, Scott H. Kaufmann, Anna V. Tinker, David M. O'Malley, Mitch Raponi, Ling Ma, Lara Maloney, Thomas Harding, Janiel M. Cragun, Lindsey Rolfe, Rebecca Kristeleit, Isabelle Ray-Coquard, James D. Brenton, Maria I. Harrell, Kevin K. Lin, Iain A. McNeish, Roman Yelensky, Sandra Goble, Amit M. Oza, Alexandra Leary, Gottfried E. Konecny, Katherine M. Bell-McGuinn, James Sun, Elaina Mann, Brenton, James [0000-0002-5738-6683], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Oncology ,Indoles ,endocrine system diseases ,HOMOLOGOUS RECOMBINATION DEFICIENCY ,Drug Resistance ,POLY(ADP-RIBOSE) POLYMERASE ,Carcinoma, Ovarian Epithelial ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,Ovarian Epithelial ,Neoplasms, Glandular and Epithelial ,Prospective Studies ,RANDOMIZED PHASE-2 ,Peritoneal Neoplasms ,Cancer ,Ovarian Neoplasms ,BRCA1 Protein ,OLAPARIB ,Glandular and Epithelial ,Middle Aged ,Prognosis ,CANCER ,TUMORS ,PROSTATE-CANCER ,Ovarian Cancer ,Survival Rate ,Local ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,PARP inhibitor ,DNA-REPAIR ,Female ,Development of treatments and therapeutic interventions ,Poly(ADP-ribose) Polymerases ,Life Sciences & Biomedicine ,medicine.medical_specialty ,HETEROZYGOSITY ,Oncology and Carcinogenesis ,BRCA MUTATION ,Antineoplastic Agents ,Poly(ADP-ribose) Polymerase Inhibitors ,BREAST ,Olaparib ,03 medical and health sciences ,Rare Diseases ,Internal medicine ,Carcinoma ,medicine ,Genetics ,Fallopian Tube Neoplasms ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Rucaparib ,REPAIR DEFECTS ,Survival rate ,Germ-Line Mutation ,Aged ,Neoplasm Staging ,Platinum ,Gynecology ,BRCA2 Protein ,Salvage Therapy ,Science & Technology ,MUTANT-CELLS ,business.industry ,BRCA mutation ,Evaluation of treatments and therapeutic interventions ,International Agencies ,medicine.disease ,NEGATIVE BREAST-CANCER ,030104 developmental biology ,Neoplasm Recurrence ,Good Health and Well Being ,GENOMIC LOSS ,chemistry ,Drug Resistance, Neoplasm ,Neoplasm ,Neoplasm Recurrence, Local ,INHIBITORS ,business ,Ovarian cancer ,Follow-Up Studies - Abstract
© 2017 Elsevier Ltd Background Poly(ADP-ribose) polymerase (PARP) inhibitors have activity in ovarian carcinomas with homologous recombination deficiency. Along with BRCA1 and BRCA2 (BRCA) mutations genomic loss of heterozygosity (LOH) might also represent homologous recombination deficiency. In ARIEL2, we assessed the ability of tumour genomic LOH, quantified with a next-generation sequencing assay, to predict response to rucaparib, an oral PARP inhibitor. Methods ARIEL2 is an international, multicentre, two-part, phase 2, open-label study done at 49 hospitals and cancer centres in Australia, Canada, France, Spain, the UK, and the USA. In ARIEL2 Part 1, patients with recurrent, platinum-sensitive, high-grade ovarian carcinoma were classified into one of three predefined homologous recombination deficiency subgroups on the basis of tumour mutational analysis: BRCA mutant (deleterious germline or somatic), BRCA wild-type and LOH high (LOH high group), or BRCA wild-type and LOH low (LOH low group). We prespecified a cutoff of 14% or more genomic LOH for LOH high. Patients began treatment with oral rucaparib at 600 mg twice per day for continuous 28 day cycles until disease progression or any other reason for discontinuation. The primary endpoint was progression-free survival. All patients treated with at least one dose of rucaparib were included in the safety analyses and all treated patients who were classified were included in the primary endpoint analysis. This trial is registered with ClinicalTrials.gov, number NCT01891344. Enrolment into ARIEL2 Part 1 is complete, although an extension (Part 2) is ongoing. Findings 256 patients were screened and 206 were enrolled between Oct 30, 2013, and Dec 19, 2014. At the data cutoff date (Jan 18, 2016), 204 patients had received rucaparib, with 28 patients remaining in the study. 192 patients could be classified into one of the three predefined homologous recombination deficiency subgroups: BRCA mutant (n=40), LOH high (n=82), or LOH low (n=70). Tumours from 12 patients were established as BRCA wild-type, but could not be classified for LOH, because of insufficient neoplastic nuclei in the sample. The median duration of treatment for the 204 patients was 5·7 months (IQR 2·8–10·1). 24 patients in the BRCA mutant subgroup, 56 patients in the LOH high subgroup, and 59 patients in the LOH low subgroup had disease progression or died. Median progression-free survival after rucaparib treatment was 12·8 months (95% CI 9·0–14·7) in the BRCA mutant subgroup, 5·7 months (5·3–7·6) in the LOH high subgroup, and 5·2 months (3·6–5·5) in the LOH low subgroup. Progression-free survival was significantly longer in the BRCA mutant (hazard ratio 0·27, 95% CI 0·16–0·44, p
- Published
- 2018
- Full Text
- View/download PDF
40. MLN4924 induces Noxa upregulation in acute myelogenous leukemia and synergizes with Bcl-2 inhibitors
- Author
-
Allan D. Hess, Xue Wei Meng, Judith E. Karp, Scott H. Kaufmann, B D Smith, Paula A. Schneider, Katherine L. B. Knorr, and Haiming Dai
- Subjects
Antineoplastic Agents ,Apoptosis ,HL-60 Cells ,Cyclopentanes ,Proto-Oncogene Proteins c-myc ,Myelogenous ,chemistry.chemical_compound ,Downregulation and upregulation ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,RNA, Small Interfering ,Molecular Biology ,Sulfonamides ,Original Paper ,Gene knockdown ,Navitoclax ,biology ,Drug Synergism ,Cell Biology ,Bridged Bicyclo Compounds, Heterocyclic ,medicine.disease ,Up-Regulation ,Leukemia, Myeloid, Acute ,Leukemia ,Pyrimidines ,Pevonedistat ,Proto-Oncogene Proteins c-bcl-2 ,chemistry ,biology.protein ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,RNA Interference ,Cullin - Abstract
MLN4924 (pevonedistat), an inhibitor of the Nedd8 activating enzyme (NAE), has exhibited promising clinical activity in acute myelogenous leukemia (AML). Here we demonstrate that MLN4924 induces apoptosis in AML cell lines and clinical samples via a mechanism distinct from those observed in other malignancies. Inactivation of E3 cullin ring ligases (CRLs) through NAE inhibition causes accumulation of the CRL substrate c-Myc, which transactivates the PMAIP1 gene encoding Noxa, leading to increased Noxa protein, Bax and Bak activation, and subsequent apoptotic changes. Importantly, c-Myc knockdown diminishes Noxa induction; and Noxa siRNA diminishes MLN4924-induced killing. Because Noxa also neutralizes Mcl-1, an anti-apoptotic Bcl-2 paralog often upregulated in resistant AML, further experiments have examined the effect of combining MLN4924 with BH3 mimetics that target other anti-apoptotic proteins. In combination with ABT-199 or ABT-263 (navitoclax), MLN4924 exerts a synergistic cytotoxic effect. Collectively, these results provide new insight into MLN4924-induced engagement of the apoptotic machinery that could help guide further exploration of MLN4924 for AML.
- Published
- 2015
- Full Text
- View/download PDF
41. Ketamine and ketamine metabolites as novel estrogen receptor ligands: Induction of cytochrome P450 and AMPA glutamate receptor gene expression
- Author
-
Rima Kaddurah-Daouk, Scott H. Kaufmann, Ming Fen Ho, Richard M. Weinshilboum, Liewei Wang, James N. Ingle, and Cristina Correia
- Subjects
0301 basic medicine ,CYP2B6 ,medicine.drug_class ,Estrogen receptor ,AMPA receptor ,Pharmacology ,Neurotransmission ,Biochemistry ,Article ,Cell Line ,Cytochrome P-450 CYP2A6 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Ketamine ,RNA, Messenger ,Receptors, AMPA ,Cloning, Molecular ,Chemistry ,Glutamate receptor ,Cytochrome P-450 CYP2B6 ,030104 developmental biology ,Gene Expression Regulation ,Receptors, Estrogen ,Estrogen ,Astrocytes ,Estrogen receptor alpha ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Major depressive disorder (MDD) is the most common psychiatric illness worldwide, and it displays a striking sex-dependent difference in incidence, with two thirds of MDD patients being women. Ketamine treatment can produce rapid antidepressant effects in MDD patients, effects that are mediated—at least partially—through glutamatergic neurotransmission. Two active metabolites of ketamine, ( 2R,6R )-hydroxynorketamine (HNK) and ( 2S,6S)- HNK, also appear to play a key role in ketamine’s rapid antidepressant effects through the activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors. In the present study, we demonstrated that estrogen plus ketamine or estrogen plus active ketamine metabolites displayed additive effects on the induction of the expression of AMPA receptor subunits. In parallel, the expression of estrogen receptor alpha (ERα) was also significantly upregulated. Even more striking, radioligand binding assays demonstrated that [ 3 H]-ketamine can directly bind to ERα ( K D : 344.5 ± 13 nM). Furthermore, ketamine and its (2 R ,6 R )-HNK and (2 S ,6 S )-HNK metabolites displayed similar affinity for ERα (IC 50 : 2.31 ± 0.1, 3.40 ± 0.2, and 3.53 ± 0.2 µM, respectively) as determined by [ 3 H]-ketamine displacement assays. Finally, induction of AMPA receptors by either estrogens or ketamine and its metabolites was lost when ERα was knocked down or silenced pharmacologically. These results suggest a positive feedback loop by which estrogens can augment the effects of ketamine and its (2 R ,6 R )-HNK and (2 S ,6 S )-HNK metabolites on the ERα-induced transcription of CYP2A6 and CYP2B6, estrogen inducible enzymes that catalyze ketamine’s biotransformation to form the two active metabolites. These observations provide novel insight into ketamine’s molecular mechanism(s) of action and have potential implications for the treatment of MDD.
- Published
- 2018
42. Gadolinium-enhanced cardiac MR exams of human subjects are associated with significant increases in the DNA repair marker 53BP1, but not the damage marker γH2AX
- Author
-
David F. Kallmes, Sylvain V. Costes, Tamara M. Hudson, Dana Schroeder, Jennifer S. McDonald, Philip M. Young, Jacob B. Ekins, Anthony S. Tin, Robert J. McDonald, Aiming Lu, Ramanathan Kadirvel, Scott H. Kaufmann, and Kevin M. Kallmes
- Subjects
0301 basic medicine ,Male ,Pathology ,DNA Repair ,Physiology ,medicine.medical_treatment ,lcsh:Medicine ,Gadolinium ,030204 cardiovascular system & hematology ,Biochemistry ,Diagnostic Radiology ,Histones ,White Blood Cells ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Lymphocytes ,Prospective Studies ,Prospective cohort study ,lcsh:Science ,Multidisciplinary ,medicine.diagnostic_test ,Radiology and Imaging ,Heart ,Middle Aged ,Magnetic Resonance Imaging ,Body Fluids ,Nucleic acids ,Chemistry ,Blood ,Physical Sciences ,Female ,Cellular Types ,Anatomy ,Cardiomyopathies ,Tumor Suppressor p53-Binding Protein 1 ,Research Article ,Chemical Elements ,Adult ,medicine.medical_specialty ,DNA damage ,Imaging Techniques ,Immune Cells ,Immunology ,Cardiology ,Malignancy ,Research and Analysis Methods ,Drug Absorption ,Peripheral blood mononuclear cell ,03 medical and health sciences ,Diagnostic Medicine ,medicine ,Genetics ,Humans ,Clinical significance ,Pharmacokinetics ,Aged ,Retrospective Studies ,Pharmacology ,Chemotherapy ,Blood Cells ,Biology and life sciences ,business.industry ,lcsh:R ,Correction ,Magnetic resonance imaging ,DNA ,Cell Biology ,medicine.disease ,Radiation therapy ,030104 developmental biology ,lcsh:Q ,business ,Biomarkers ,DNA Damage - Abstract
Magnetic resonance imaging is considered low risk, yet recent studies have raised a concern of potential damage to DNA in peripheral blood leukocytes. This prospective Institutional Review Board-approved study examined potential double-strand DNA damage by analyzing changes in the DNA damage and repair markers γH2AX and 53BP1 in patients who underwent a 1.5 T gadolinium-enhanced cardiac magnetic resonance (MR) exam. Sixty patients were enrolled (median age 55 years, 39 males). Patients with history of malignancy or who were receiving chemotherapy, radiation therapy, or steroids were excluded. MR sequence data were recorded and blood samples obtained immediately before and after MR exposure. An automated immunofluorescence assay quantified γH2AX or 53BP1 foci number in isolated peripheral blood mononuclear cells. Changes in foci number were analyzed using the Wilcoxon signed-rank test. Clinical and MR procedural characteristics were compared between patients who had a >10% increase in γH2AX or 53BP1 foci numbers and patients who did not. The number of γH2AX foci did not significantly change following cardiac MR (median foci per cell pre-MR = 0.11, post-MR = 0.11, p = .90), but the number of 53BP1 foci significantly increased following MR (median foci per cell pre-MR = 0.46, post-MR = 0.54, p = .0140). Clinical and MR characteristics did not differ significantly between patients who had at least a 10% increase in foci per cell and those who did not. We conclude that MR exposure leads to a small (median 25%) increase in 53BP1 foci, however the clinical relevance of this increase is unknown and may be attributable to normal variation instead of MR exposure.
- Published
- 2018
43. Randomized Phase II Trial of Cytosine Arabinoside with and without the CHK1 inhibitor MK-8776 in Relapsed and Refractory Acute Myeloid Leukemia
- Author
-
Larry M. Karnitz, Mrinal M. Patnaik, Scott H. Kaufmann, Ivana Gojo, Jonathan Webster, Lawrence E. Morris, Amanda L. Blackford, Judith E. Karp, L. Austin Doyle, B. Douglas Smith, Lihua Wang, Raoul Tibes, Gary L. Rosner, Robert J. Kinders, Mark R. Litzow, and Catherine J. Huntoon
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Antineoplastic Agents ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Refractory ,medicine ,Humans ,CHEK1 ,Cytotoxicity ,Aged ,Genetics ,business.industry ,Cytarabine ,Myeloid leukemia ,Hematology ,Middle Aged ,medicine.disease ,CHK1 Inhibitor MK-8776 ,carbohydrates (lipids) ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Pyrimidines ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Checkpoint Kinase 1 ,Cancer research ,Pyrazoles ,Female ,biological phenomena, cell phenomena, and immunity ,Neoplasm Recurrence, Local ,business ,Cytosine ,medicine.drug - Abstract
Cytosine arabinoside (AraC) remains the backbone of most treatment regimens for acute myeloid leukemia (AML). Incorporation of AraC into DNA activates checkpoint kinase 1 (Chk1), leading to cell-cycle arrest and diminished AraC cytotoxicity, which can be reversed by the selective Chk1 inhibitor MK-8776. Building on a Phase I trial, we conducted a phase II trial comparing timed sequential AraC with or without MK-8776.Patients with relapsed or primary refractory AML were randomized 1:1 to receive either AraC with MK-8776 (Arm A); or AraC alone (Arm B).32 patients were treated: 14 assigned to Arm A and 18 to Arm B. There were 5 (36%) complete responses (CR/CRi) and 1 (7%) partial response (PR) in Arm A, and 8 (44%) CR/CRis and 1 (6%) PR in Arm B. Median survival did not differ significantly between the two groups (5.9months in Arm A vs. 4.5 months in Arm B). MK-8776 led to a robust increase in DNA damage in circulating leukemic blasts as measured by increased γ-H2AX (16.9%±6.1% prior and 36.4%±6.8% at one hour after MK-8776 infusion, p=0.016).Response rates and survival were similar between the two groups in spite of evidence that MK-8776 augmented DNA damage in circulating leukemic blasts. Better than expected results in the control arm using timed sequential AraC and truncated patient enrollment may have limited the ability to detect clinical benefit from the combination.
- Published
- 2017
44. Getting a GRP on histone deacetylase inhibitor selectivity
- Author
-
Husheng Ding, Scott H. Kaufmann, and Cristina Correia
- Subjects
BCL2 mutations ,Histone deacetylase 5 ,HDAC11 ,Chemistry ,Histone deacetylase 2 ,medicine.drug_class ,Histone deacetylase inhibitor ,RASGRP1 ,Editorial ,Oncology ,HDAC inhibitor ,Cancer research ,medicine ,RAF1 ,Selectivity ,BIM upregulation - Published
- 2017
45. A Phase I Clinical Trial of the Poly(ADP-ribose) Polymerase Inhibitor Veliparib and Weekly Topotecan in Patients with Solid Tumors
- Author
-
Guy G. Poirer, Michael E. Menefee, Paul Haluska, Maria I. Harrell, Joel M. Reid, Donald W. Northfelt, Jiuping Ji, Janet Lensing, Charles Erlichman, Andrea E. Wahner Hendrickson, Karen S. Flatten, Scott H. Kaufmann, Lynn C. Hartmann, Yiping Zang, Felix Boakye-Agyeman, Daniel Satele, Harry J. Long, Elizabeth M. Swisher, Olumide Kayode, Alice P. Chen, and Jake Allred
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Neutropenia ,Veliparib ,Phases of clinical research ,Gene mutation ,Poly(ADP-ribose) Polymerase Inhibitors ,Disease-Free Survival ,Drug Administration Schedule ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Germ-Line Mutation ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Area Under Curve ,Leukocytes, Mononuclear ,Topotecan ,Benzimidazoles ,Female ,business ,medicine.drug - Abstract
Purpose: To determine the dose limiting toxicities (DLT), maximum tolerated dose (MTD), and recommended phase II dose (RP2D) of veliparib in combination with weekly topotecan in patients with solid tumors. Correlative studies were included to assess the impact of topotecan and veliparib on poly(ADP-ribose) levels in peripheral blood mononuclear cells, serum pharmacokinetics of both agents, and potential association of germline repair gene mutations with outcome. Experimental Design: Eligible patients had metastatic nonhematologic malignancies with measurable disease. Using a 3 + 3 design, patients were treated with veliparib orally twice daily on days 1–3, 8–10, and 15–17 and topotecan intravenously on days 2, 9, and 16 every 28 days. Tumor responses were assessed by RECIST. Results: Of 58 patients enrolled, 51 were evaluable for the primary endpoint. The MTD and RP2D was veliparib 300 mg twice daily on days 1–3, 8–10, and 15–17 along with topotecan 3 mg/m2 on days 2, 9, and 16 of a 28-day cycle. DLTs were grade 4 neutropenia lasting >5 days. The median number of cycles was 2 (1–26). The objective response rate was 10%, with 1 complete and 4 partial responses. Twenty-two patients (42%) had stable disease ranging from 4 to 26 cycles. Patients with germline BRCA1, BRCA2, or RAD51D mutations remained on study longer than those without homologous recombination repair (HRR) gene mutations (median 4 vs. 2 cycles). Conclusions: Weekly topotecan in combination with veliparib has a manageable safety profile and appears to warrant further investigation.
- Published
- 2017
46. Poly(ADP-ribose) Polymerase Inhibitors Sensitize Cancer Cells to Death Receptor-mediated Apoptosis by Enhancing Death Receptor Expression
- Author
-
Karen S. Flatten, Scott H. Kaufmann, Jin San Zhang, Brian D. Koh, B. Douglas Smith, Allan D. Hess, Judith E. Karp, Daniel D. Billadeau, X. Wei Meng, and Paula A. Schneider
- Subjects
Veliparib ,Sp1 Transcription Factor ,Receptor expression ,Apoptosis ,Poly(ADP-ribose) Polymerase Inhibitors ,Biology ,Response Elements ,Biochemistry ,Poly (ADP-Ribose) Polymerase Inhibitor ,Olaparib ,TNF-Related Apoptosis-Inducing Ligand ,chemistry.chemical_compound ,PARP1 ,Neoplasms ,Humans ,fas Receptor ,Enzyme Inhibitors ,Receptor ,Molecular Biology ,Cell Biology ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,chemistry ,PARP inhibitor ,Cancer research ,Poly(ADP-ribose) Polymerases ,K562 Cells ,Signal Transduction - Abstract
Recombinant human tumor necrosis factor-α-related apoptosis inducing ligand (TRAIL), agonistic monoclonal antibodies to TRAIL receptors, and small molecule TRAIL receptor agonists are in various stages of preclinical and early phase clinical testing as potential anticancer drugs. Accordingly, there is substantial interest in understanding factors that affect sensitivity to these agents. In the present study we observed that the poly(ADP-ribose) polymerase (PARP) inhibitors olaparib and veliparib sensitize the myeloid leukemia cell lines ML-1 and K562, the ovarian cancer line PEO1, non-small cell lung cancer line A549, and a majority of clinical AML isolates, but not normal marrow, to TRAIL. Further analysis demonstrated that PARP inhibitor treatment results in activation of the FAS and TNFRSF10B (death receptor 5 (DR5)) promoters, increased Fas and DR5 mRNA, and elevated cell surface expression of these receptors in sensitized cells. Chromatin immunoprecipitation demonstrated enhanced binding of the transcription factor Sp1 to the TNFRSF10B promoter in the presence of PARP inhibitor. Knockdown of PARP1 or PARP2 (but not PARP3 and PARP4) not only increased expression of Fas and DR5 at the mRNA and protein level, but also recapitulated the sensitizing effects of the PARP inhibition. Conversely, Sp1 knockdown diminished the PARP inhibitor effects. In view of the fact that TRAIL is part of the armamentarium of natural killer cells, these observations identify a new facet of PARP inhibitor action while simultaneously providing the mechanistic underpinnings of a novel therapeutic combination that warrants further investigation.
- Published
- 2014
- Full Text
- View/download PDF
47. Loss of HSulf-1 expression enhances tumorigenicity by inhibiting Bim expression in ovarian cancer
- Author
-
Viji Shridhar, Ashwani Khurana, Debarshi Roy, Xiaoping He, and Scott H. Kaufmann
- Subjects
Cancer Research ,medicine.medical_specialty ,Gene knockdown ,Kinase ,Biology ,medicine.disease ,medicine.disease_cause ,chemistry.chemical_compound ,Endocrinology ,Oncology ,chemistry ,Downregulation and upregulation ,RNA interference ,Internal medicine ,medicine ,Cancer research ,LY294002 ,Ectopic expression ,Ovarian cancer ,Carcinogenesis - Abstract
The expression of human Sulfatase1 (HSulf-1) is downregulated in the majority of primary ovarian cancer tumors, but the functional consequence of this downregulation remains unclear. Using two different shRNAs (Sh1 and Sh2), HSulf-1 expression was stably downregulated in ovarian cancer OV202 cells. We found that HSulf-1-deficient OV202 Sh1 and Sh2 cells formed colonies in soft agar. In contrast, nontargeting control (NTC) shRNA-transduced OV202 cells did not form any colonies. Moreover, subcutaneous injection of OV202 HSulf-1-deficient cells resulted in tumor formation in nude mice, whereas OV202 NTC cells did not. Also, ectopic expression of HSulf-1 in ovarian cancer SKOV3 cells significantly suppressed tumor growth in nude mice. Here, we show that HSulf-1-deficient OV202 cells have markedly decreased expression of proapoptotic Bim protein, which can be rescued by restoring HSulf-1 expression in OV202 Sh1 cells. Enhanced expression of HSulf-1 in HSulf-1-deficient SKOV3 cells resulted in increased Bim expression. Decreased Bim levels after loss of HSulf-1 were due to increased p-ERK, because inhibition of ERK activity with PD98059 resulted in increased Bim expression. However, treatment with a PI3 kinase/AKT inhibitor, LY294002, failed to show any change in Bim protein level. Importantly, rescuing Bim expression in HSulf-1 knockdown cells significantly retarded tumor growth in nude mice. Collectively, these results suggest that loss of HSulf-1 expression promotes tumorigenicity in ovarian cancer through regulating Bim expression.
- Published
- 2014
- Full Text
- View/download PDF
48. A Multisite Phase Ib Study of Pevonedistat, Azacitidine and Venetoclax (PAVE) for the Treatment of Subjects with Acute Myelogenous Leukemia (AML)
- Author
-
Sameem Abedin, Scott H. Kaufmann, Ehab Atallah, Aniko Szabo, Ashish Anshu, Karen-Sue B. Carlson, Lyndsey Runaas, Arielle Baim, Alexander Hinman, Laura C. Michaelis, and Guru Subramanian Guru Murthy
- Subjects
Oncology ,Acute promyelocytic leukemia ,medicine.medical_specialty ,Acute myelogenous leukemia (AML) ,Venetoclax ,business.industry ,medicine.medical_treatment ,Immunology ,Azacitidine ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Transplantation ,chemistry.chemical_compound ,Leukemia ,chemistry ,Internal medicine ,medicine ,Clinical endpoint ,business ,medicine.drug - Abstract
Background: Outcomes of patients with AML have remained poor despite the availability of cytotoxic chemotherapy, hypomethylating agents (HMAs) and targeted therapies. HMAs, such as azacitidine, in combination with Bcl-2 inhibitors like venetoclax have demonstrated response rates of 67% in newly diagnosed AML and 21% in relapsed/refractory (RR) AML (DiNardo et al. Blood 2019 and Am J Hematol 2018). While the combination of azacitidine and venetoclax is efficacious in AML, preclinical studies indicate potential mechanisms of drug resistance including overexpression of MCL-1, an anti-apoptotic protein (Konopleva et al. Cancer Cell. 2006). Pevonedistat is a first in class inhibitor of Nedd8 activating enzyme that has demonstrated activity against AML (Swords RT et al. Blood. 2010). Pevonedistat induces NOXA, a pro-apoptotic protein leading to neutralization of MCL-1 inducing apoptosis (Wang et al. Biochem Biophys Res Commun. 2017). Preclinical studies evaluating the combination of pevonedistat and venetoclax against AML cell lines have demonstrated synergistic effect (Knoor KL et al. Cell Death Differ. 2015). Hence, we hypothesize that the addition of pevonedistat to the combination of azacitidine and venetoclax would enhance the therapeutic efficacy by overcoming resistance to apoptosis. Study design and methods: This is an investigator-initiated phase Ib study evaluating the safety of pevonedistat, azacitidine and venetoclax. Patients aged 18 years or above with morphologically documented AML (de novo, secondary or therapy-related), ECOG performance status 0-2 and adequate organ function are eligible for the study. Major exclusion criteria are patients with isolated extramedullary relapse, hematopoietic cell transplantation (HCT) within 100 days of enrollment, active acute GVHD, veno-occlusive disease, acute promyelocytic leukemia, liver cirrhosis and severe liver impairment. While the dose escalation phase is available only for patients with RR-AML, the dose expansion phase can also include newly diagnosed AML patients who are ineligible for intensive induction. The study is planned to be conducted at Medical College of Wisconsin, Mayo Clinic and University of Pennsylvania. The primary endpoint is to determine the recommended phase 2 dose (RP2D) and toxicity profile of pevonedistat, azacitidine and venetoclax. The secondary endpoints include determination of response rates, duration of response, survival and pharmacokinetics. Exploratory endpoints include correlation of response rates with AML genomic profile, correlation of pretreatment levels of BCL2, BCLXL, MCL1, BAX or BAK with response, determination of changes in NOXA (PMAIP1) mRNA and protein expression pre-and post-pevonedistat treatment, evaluation of BH3 mimetic profiling on bone marrow samples by flow cytometry and assessing the sensitivity of leukemia and leukemic stem/progenitor cells to pevonedistat ex vivo. The study will follow 3+3 design with dose escalation (Arms A and B), de-escalation in case of dose limiting toxicity (DLT) (arms Z and Y) and dose expansion phase (figure 1). Patients will be entered sequentially to each dose level, starting with dose level 0. The DLT observation period for dose-escalation will be 1 cycle. The maximal tolerated dose (MTD) will be defined as the highest dose level at which none of the first 3 treated subjects, or no more than 1 of the first 6 treated subjects experiences a DLT. A minimum of 9 and a maximum of 24 patients will be needed for the dose escalation phase and 6 patients for the dose expansion phase. Response rate, duration of response and exploratory endpoints will be analyzed using descriptive statistics. Kaplan-Meier method will be used to determine survival. Disclosures Guru Murthy: Cardinal Health Inc.: Honoraria. Michaelis:Incyte: Consultancy, Research Funding; Pfizer: Equity Ownership, Research Funding; Novartis: Consultancy; Macrogeneics: Research Funding; Millenium: Research Funding; BMS: Research Funding; Celgene: Consultancy, Research Funding; JAZZ: Other: Data Safety Monitoring Board, uncompensated, Research Funding; TG Therapeutics: Consultancy, Research Funding; Janssen: Research Funding; ASTEX: Research Funding; Bioline: Research Funding. Abedin:Jazz Pharmaceuticals: Honoraria; Agios: Honoraria; Helsinn Healthcare: Research Funding; Pfizer Inc: Research Funding; Actinium Pharmaceuticals: Research Funding. Runaas:Agios: Honoraria; Blueprint Medicine: Honoraria. Atallah:Jazz: Consultancy; Novartis: Consultancy; Takeda: Consultancy, Research Funding; Pfizer: Consultancy; Jazz: Consultancy; Helsinn: Consultancy; Helsinn: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
49. Topoisomerases and cancer chemotherapy: recent advances and unanswered questions
- Author
-
Mary-Ann Bjornsti and Scott H. Kaufmann
- Subjects
DNA Replication ,Ribonucleotide ,Cell division ,Review ,Eukaryotic chromosome structure ,DNA-activated protease ,General Biochemistry, Genetics and Molecular Biology ,Chromosome segregation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,030304 developmental biology ,0303 health sciences ,DNA-protein crosslink ,topoisomerase poison ,General Immunology and Microbiology ,biology ,Chemistry ,Topoisomerase ,DNA replication ,DNA ,Articles ,General Medicine ,DNA supercoiling ,3. Good health ,Biochemistry ,030220 oncology & carcinogenesis ,biology.protein ,DNA supercoil ,DNA Topoisomerases ,DNA Damage ,chromatin organization - Abstract
DNA topoisomerases are enzymes that catalyze changes in the torsional and flexural strain of DNA molecules. Earlier studies implicated these enzymes in a variety of processes in both prokaryotes and eukaryotes, including DNA replication, transcription, recombination, and chromosome segregation. Studies performed over the past 3 years have provided new insight into the roles of various topoisomerases in maintaining eukaryotic chromosome structure and facilitating the decatenation of daughter chromosomes at cell division. In addition, recent studies have demonstrated that the incorporation of ribonucleotides into DNA results in trapping of topoisomerase I (TOP1)–DNA covalent complexes during aborted ribonucleotide removal. Importantly, such trapped TOP1–DNA covalent complexes, formed either during ribonucleotide removal or as a consequence of drug action, activate several repair processes, including processes involving the recently described nuclear proteases SPARTAN and GCNA-1. A variety of new TOP1 inhibitors and formulations, including antibody–drug conjugates and PEGylated complexes, exert their anticancer effects by also trapping these TOP1–DNA covalent complexes. Here we review recent developments and identify further questions raised by these new findings.
- Published
- 2019
- Full Text
- View/download PDF
50. ATR Inhibition Broadly Sensitizes Ovarian Cancer Cells to Chemotherapy Independent of BRCA Status
- Author
-
Larry M. Karnitz, Scott H. Kaufmann, Shari L. Sutor, Catherine J. Huntoon, Karen S. Flatten, Amelia M. Huehls, and Andrea E. Wahner Hendrickson
- Subjects
Cancer Research ,Veliparib ,Cell Survival ,Immunoblotting ,Antineoplastic Agents ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Poly(ADP-ribose) Polymerase Inhibitors ,Protein Serine-Threonine Kinases ,Biology ,Deoxycytidine ,Poly (ADP-Ribose) Polymerase Inhibitor ,Article ,chemistry.chemical_compound ,Cell Line, Tumor ,medicine ,Humans ,cdc25 Phosphatases ,Sulfones ,CHEK1 ,Phosphorylation ,BRCA2 Protein ,Ovarian Neoplasms ,Cisplatin ,Dose-Response Relationship, Drug ,BRCA1 Protein ,Kinase ,medicine.disease ,Gemcitabine ,Pyrimidines ,Oncology ,chemistry ,Pyrazines ,Checkpoint Kinase 1 ,Cancer research ,Pyrazoles ,Benzimidazoles ,Female ,RNA Interference ,Topotecan ,Poly(ADP-ribose) Polymerases ,biological phenomena, cell phenomena, and immunity ,Ovarian cancer ,Protein Kinases ,Signal Transduction ,medicine.drug - Abstract
Replication stress and DNA damage activate the ATR-Chk1 checkpoint signaling pathway that licenses repair and cell survival processes. In this study, we examined the respective roles of the ATR and Chk1 kinases in ovarian cancer cells using genetic and pharmacologic inhibitors in combination with cisplatin, topotecan, gemcitabine, and the PARP inhibitor veliparib (ABT-888), four agents with clinical activity in ovarian cancer. RNA interference (RNAi)–mediated depletion or inhibition of ATR sensitized ovarian cancer cells to all four agents. In contrast, while cisplatin, topotecan, and gemcitabine each activated Chk1, RNAi-mediated depletion or inhibition of this kinase in cells sensitized them only to gemcitabine. Unexpectedly, we found that neither the ATR kinase inhibitor VE-821 nor the Chk1 inhibitor MK-8776 blocked ATR-mediated Chk1 phosphorylation or autophosphorylation, two commonly used readouts for inhibition of the ATR-Chk1 pathway. Instead, their ability to sensitize cells correlated with enhanced CDC25A levels. In addition, we also found that VE-821 could further sensitize BRCA1-depleted cells to cisplatin, topotecan, and veliparib beyond the potent sensitization already caused by their deficiency in homologous recombination. Taken together, our results established that ATR and Chk1 inhibitors differentially sensitize ovarian cancer cells to commonly used chemotherapy agents and that Chk1 phosphorylation status may not offer a reliable marker for inhibition of the ATR-Chk1 pathway. A key implication of our work is the clinical rationale it provides to evaluate ATR inhibitors in combination with PARP inhibitors in BRCA1/2-deficient cells. Cancer Res; 73(12); 3683–91. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.