203 results on '"H Kaufmann"'
Search Results
2. Development and Validation of the Gene Expression Predictor of High-grade Serous Ovarian Carcinoma Molecular SubTYPE (PrOTYPE)
- Author
-
Sven Mahner, Robertson Mackenzie, Aline Talhouk, Linda E. Kelemen, Gottfried E. Konecny, Jennifer Alsop, Rosalind Glasspool, Chiu-Chen Tseng, Joy Hendley, Dennis J. Slamon, Jennifer A. Doherty, Andrew Berchuck, Anna H. Wu, Anna M. Piskorz, Chen Wang, Cristina Rodríguez-Antona, D.G.H. de Silva, Valerie Rhenius, Peter A. Fasching, Stacey J. Winham, Gary L. Keeney, Teodora Goranova, Joshy George, Jan Lubinski, Michelle J. Henderson, Rex C. Bentley, Jenny Lester, Sabine Behrens, Joellen M. Schildkraut, Michael E. Carney, Timothy Budden, David G. Huntsman, Oleg Oszurek, Michael S. Anglesio, Jacek Gronwald, Ruby Yun-Ju Huang, Martin Köbel, Javier Benitez, Martin Widschwendter, Melissa C. Larson, Raghwa Sharma, Clara Bodelon, Usha Menon, Janusz Menkiszak, Blake Gilks, María Josefa Mosteiro García, Jesús García-Donas, Wafaa Elatre, Scott H. Kaufmann, Paul Haluska, Pamela J. Thompson, Boris Winterhoff, Susan J. Ramus, Louise A. Brinton, Simon A. Gayther, Mary Anne Rossing, Georgia Chenevix-Trench, Hugh Luk, Jolanta Lissowska, Marc T. Goodman, Billy Chen, Beth Y. Karlan, Naveena Singh, Sian Fereday, Mark E. Sherman, Ana Osorio, Lynne R. Wilkens, Maria P. Intermaggio, Brenda Y. Hernandez, Britton Trabert, Esther Herpel, Mercedes Jimenez-Linan, Janine Senz, Geyi Liu, Celeste Leigh Pearce, Samuel C Y Leong, Iain A. McNeish, Isabelle Ray-Coquard, Susana Banerjee, Malcolm C. Pike, Liz-Anne Lewsley, Helen Steed, Honglin Song, Samantha Hinsley, David D.L. Bowtell, James D. Brenton, Holly R. Harris, Tuan Zea Tan, Cezary Cybulski, Alicia Beeghly-Fadiel, A. Toloczko, Nikilyn Nevins, Robert S. Brown, Darren Ennis, Stephanie Chen, Euan A. Stronach, José Palacios, Sandra Orsulic, Anna deFazio, Geoff Macintyre, Kara L. Cushing-Haugen, Mila Volchek, Aleksandra Gentry-Maharaj, Jenny Chang-Claude, Ellen L. Goode, Paul D.P. Pharoah, Hanwei Sudderuddin, Stefan Kommoss, Derek S. Chiu, Huei San Leong, Peter Sinn, Catherine J. Kennedy, Chloe Karpinskyj, Alison Brand, Amy Lum, Veronica Chow, Nicolas Wentzensen, Tayyebeh M. Nazeran, Nadia Traficante, Dustin Johnson, Yoke-Eng Chiew, Casey S. Greene, Jennifer M Koziak, Renée T. Fortner, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, Ovarian Cancer Action, Talhouk, Aline [0000-0001-7760-410X], George, Joshy [0000-0001-8510-8229], Wang, Chen [0000-0003-2638-3081], Tan, Tuan Zea [0000-0001-6624-1593], Behrens, Sabine [0000-0002-9714-104X], Bodelon, Clara [0000-0002-6578-2678], Brinton, Louise [0000-0003-3853-8562], Fortner, Renée T [0000-0002-1426-8505], García-Donas, Jesús [0000-0001-7731-3601], Gentry-Maharaj, Aleksandra [0000-0001-7270-9762], Glasspool, Rosalind [0000-0002-5000-1680], Greene, Casey S [0000-0001-8713-9213], Harris, Holly R [0000-0002-2572-6727], Kaufmann, Scott H [0000-0002-4900-7145], Kennedy, Catherine J [0000-0002-4465-5784], Köbel, Martin [0000-0002-6615-2037], Koziak, Jennifer M [0000-0001-5830-0397], Lissowska, Jolanta [0000-0003-2695-5799], McNeish, Iain A [0000-0002-9387-7586], Menkiszak, Janusz [0000-0001-8279-7196], Hinsley, Samantha [0000-0001-6903-4688], Pike, Malcolm C [0000-0003-4891-1199], Rodriguez-Antona, Cristina [0000-0001-8750-7338], Sinn, Peter [0000-0003-2836-6699], Trabert, Britton [0000-0002-1539-6090], Widschwendter, Martin [0000-0002-7778-8380], Winham, Stacey J [0000-0002-8492-9102], Brenton, James D [0000-0002-5738-6683], Brown, Robert [0000-0001-7960-5755], Chang-Claude, Jenny [0000-0001-8919-1971], deFazio, Anna [0000-0003-0057-4744], Fasching, Peter A [0000-0003-4885-8471], Kelemen, Linda E [0000-0003-4362-9784], Menon, Usha [0000-0003-3708-1732], Pharoah, Paul DP [0000-0001-8494-732X], Ramus, Susan J [0000-0003-0005-7798], Doherty, Jennifer A [0000-0002-1454-8187], Anglesio, Michael S [0000-0003-1639-5003], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Bevacizumab ,03 medical and health sciences ,Ovarian tumor ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Ovarian carcinoma ,Internal medicine ,medicine ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Stage (cooking) ,Aged ,Ovarian Neoplasms ,business.industry ,Cystadenoma, Serous ,Cancer ,Middle Aged ,Precision medicine ,Omics ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Serous fluid ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Neoplasm Grading ,Transcriptome ,business ,Algorithms ,medicine.drug - Abstract
Purpose: Gene expression–based molecular subtypes of high-grade serous tubo-ovarian cancer (HGSOC), demonstrated across multiple studies, may provide improved stratification for molecularly targeted trials. However, evaluation of clinical utility has been hindered by nonstandardized methods, which are not applicable in a clinical setting. We sought to generate a clinical grade minimal gene set assay for classification of individual tumor specimens into HGSOC subtypes and confirm previously reported subtype-associated features. Experimental Design: Adopting two independent approaches, we derived and internally validated algorithms for subtype prediction using published gene expression data from 1,650 tumors. We applied resulting models to NanoString data on 3,829 HGSOCs from the Ovarian Tumor Tissue Analysis consortium. We further developed, confirmed, and validated a reduced, minimal gene set predictor, with methods suitable for a single-patient setting. Results: Gene expression data were used to derive the predictor of high-grade serous ovarian carcinoma molecular subtype (PrOTYPE) assay. We established a de facto standard as a consensus of two parallel approaches. PrOTYPE subtypes are significantly associated with age, stage, residual disease, tumor-infiltrating lymphocytes, and outcome. The locked-down clinical grade PrOTYPE test includes a model with 55 genes that predicted gene expression subtype with >95% accuracy that was maintained in all analytic and biological validations. Conclusions: We validated the PrOTYPE assay following the Institute of Medicine guidelines for the development of omics-based tests. This fully defined and locked-down clinical grade assay will enable trial design with molecular subtype stratification and allow for objective assessment of the predictive value of HGSOC molecular subtypes in precision medicine applications. See related commentary by McMullen et al., p. 5271
- Published
- 2020
- Full Text
- View/download PDF
3. 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
4. Rare BRIP1 Missense Alleles Confer Risk for Ovarian and Breast Cancer
- Author
-
Marc R. Radke, Elizabeth M. Swisher, Cassandra L. Moyer, Jessica L. Gillespie, Paul J. Goodfellow, Rachel Doberstein, Jennifer Ivanovich, Scott H. Kaufmann, and Marcy E. Richardson
- Subjects
0301 basic medicine ,Cancer Research ,Mutation ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Population ,medicine.disease_cause ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Germline mutation ,Breast cancer ,Oncology ,030220 oncology & carcinogenesis ,Ovarian carcinoma ,medicine ,Cancer research ,Missense mutation ,business ,education ,Ovarian cancer ,Genetic testing - Abstract
Germline loss-of-function mutations in BRCA1 interacting protein C-terminal helicase 1 (BRIP1) are associated with ovarian carcinoma and may also contribute to breast cancer risk, particularly among patients who develop disease at an early age. Normal BRIP1 activity is required for DNA interstrand cross-link (ICL) repair and is thus central to the maintenance of genome stability. Although pathogenic mutations have been identified in BRIP1, genetic testing more often reveals missense variants, for which the impact on molecular function and subsequent roles in cancer risk are uncertain. Next-generation sequencing of germline DNA in 2,160 early-onset breast cancer and 1,199 patients with ovarian cancer revealed nearly 2% of patients carry a very rare missense variant (minor allele frequency < 0.0001) in BRIP1. This is 3-fold higher than the frequency of all rare BRIP1 missense alleles reported in more than 60,000 individuals of the general population (P < 0.0001, χ2 test). Using CRISPR-Cas9 gene editing technology and rescue assays, we functionally characterized 20 of these missense variants, focusing on the altered protein's ability to repair ICL damage. A total of 75% of the characterized variants rendered the protein hypomorph or null. In a clinical cohort of >117,000 patients with breast and ovarian cancer who underwent panel testing, the combined OR associated with BRIP1 hypomorph or null missense carriers compared with the general population was 2.30 (95% confidence interval, 1.60–3.30; P < 0.0001). These findings suggest that novel missense variants within the helicase domain of BRIP1 may confer risk for both breast and ovarian cancer and highlight the importance of functional testing for additional variants. Significance: Functional characterization of rare variants of uncertain significance in BRIP1 revealed that 75% demonstrate loss-of-function activity, suggesting rare missense alleles in BRIP1 confer risk for both breast and ovarian cancer.
- Published
- 2020
- Full Text
- View/download PDF
5. 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
6. 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
7. 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
8. Therapeutic options for mucinous ovarian carcinoma☆
- Author
-
Simone M Rowley, Scott H. Kaufmann, Yoke Eng Chiew, Yoland Antill, Michael Christie, C. Blake Gilks, Sally M Hunter, Andrew N. Stephens, Prue E. Allan, Michelle C. Torres, Cécile Le Page, Michael Churchman, Jan Pyman, Carolina Salazar, Kylie L. Gorringe, Martin Köbel, David D.L. Bowtell, Richard Lupat, Kathryn Alsop, Sian Fereday, Jason Li, Matthew Wakefield, Alison Maree Hadley, Nadia Traficante, Clare L. Scott, Magnus Zethoven, Sumitra Ananda, Kaushalya C. Amarasinghe, Charlie Gourley, Orla McNally, Georgina L Ryland, Jessica N. McAlpine, Hugo Saunders, Dane Cheasley, Joy Hendley, Catherine J. Kennedy, Timothy Semple, Niko Thio, Anna deFazio, and Ian G. Campbell
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Oncology ,medicine.medical_specialty ,Receptor, ErbB-3 ,Receptor, ErbB-2 ,medicine.disease_cause ,DNA Mismatch Repair ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Ovarian cancer ,Ovarian carcinoma ,Internal medicine ,medicine ,Missense mutation ,Sequencing ,Humans ,Molecular targeted therapy ,Copy-number variation ,Homologous Recombination ,Aged ,Neoplasm Staging ,Ovarian Neoplasms ,business.industry ,Obstetrics and Gynecology ,Precision oncology ,medicine.disease ,Adenocarcinoma, Mucinous ,Immunohistochemistry ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Genomic ,Adenocarcinoma ,DNA mismatch repair ,Female ,KRAS ,Therapy ,business - Abstract
Objective Mucinous ovarian carcinoma (MOC) is an uncommon ovarian cancer histotype that responds poorly to conventional chemotherapy regimens. Although long overall survival outcomes can occur with early detection and optimal surgical resection, recurrent and advanced disease are associated with extremely poor survival. There are no current guidelines specifically for the systemic management of recurrent MOC. We analyzed data from a large cohort of women with MOC to evaluate the potential for clinical utility from a range of systemic agents. Methods We analyzed gene copy number (n = 191) and DNA sequencing data (n = 184) from primary MOC to evaluate signatures of mismatch repair deficiency and homologous recombination deficiency, and other genetic events. Immunohistochemistry data were collated for ER, CK7, CK20, CDX2, HER2, PAX8 and p16 (n = 117–166). Results Molecular aberrations noted in MOC that suggest a match with current targeted therapies include amplification of ERBB2 (26.7%) and BRAF mutation (9%). Observed genetic events that suggest potential efficacy for agents currently in clinical trials include: KRAS/NRAS mutations (66%), TP53 missense mutation (49%), RNF43 mutation (11%), ARID1A mutation (10%), and PIK3CA/PTEN mutation (9%). Therapies exploiting homologous recombination deficiency (HRD) may not be effective in MOC, as only 1/191 had a high HRD score. Mismatch repair deficiency was similarly rare (1/184). Conclusions Although genetically diverse, MOC has several potential therapeutic targets. Importantly, the lack of response to platinum-based therapy observed clinically corresponds to the lack of a genomic signature associated with HRD, and MOC are thus also unlikely to respond to PARP inhibition.
- Published
- 2020
- Full Text
- View/download PDF
9. The molecular origin and taxonomy of mucinous ovarian carcinoma
- Author
-
Georgia Chenevix-Trench, Jan Pyman, David G. Hunstman, Hugo Saunders, Linda Mileshkin, Dane Cheasley, Alison Maree Hadley, Nadia Traficante, Clare L. Scott, Diane Provencher, Niko Thio, Clare G Fedele, Joy Hendley, Jason Li, Michael S. Anglesio, Prue E. Allan, Siobhan Hughes, Magnus Zethoven, Timothy Semple, Tom Jobling, Martin Köbel, Michael Christie, Goli Samimi, Kylie L. Gorringe, Anne Marie Mes-Masson, George Au-Yeung, Yoland Antill, Andrew N. Stephens, Cécile Le Page, Carolina Salazar, Kathryn Alsop, Anna deFazio, Kurosh Rahimi, Richard Lupat, Orla McNally, Georgina L Ryland, Jessica N. McAlpine, Ian G. Campbell, Renee Demeo, Rhiannon Dudley, Simone M Rowley, Michael Churchman, C. Blake Gilks, Gwo-Yaw Ho, Stephen B. Fox, Yoke Eng Chiew, Sally M Hunter, David D.L. Bowtell, Charlie Gourley, Catherine J. Kennedy, Zhongyue Xing, Scott H. Kaufmann, Nicole Fairweather, Kimberly R. Kalli, Alison Brand, Ragwha Sharma, Maret Böhm, Sian Fereday, Matthew Wakefield, Michelle C. Torres, Alice J. Sharpe, Neville F. Hacker, Sumitra Ananda, and Kaushalya C. Amarasinghe
- Subjects
0301 basic medicine ,Science ,General Physics and Astronomy ,02 engineering and technology ,Disease ,Carcinoma, Ovarian Epithelial ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cohort Studies ,03 medical and health sciences ,Ovarian cancer ,Ovarian carcinoma ,Cancer genomics ,medicine ,Carcinoma ,Humans ,lcsh:Science ,Survival analysis ,Ovarian Neoplasms ,Multidisciplinary ,business.industry ,Gene Expression Profiling ,Sequence Analysis, DNA ,General Chemistry ,021001 nanoscience & nanotechnology ,medicine.disease ,Adenocarcinoma, Mucinous ,Survival Analysis ,3. Good health ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,030104 developmental biology ,Mutation ,Etiology ,Cancer research ,Adenocarcinoma ,Female ,lcsh:Q ,0210 nano-technology ,business - Abstract
Mucinous ovarian carcinoma (MOC) is a unique subtype of ovarian cancer with an uncertain etiology, including whether it genuinely arises at the ovary or is metastatic disease from other organs. In addition, the molecular drivers of invasive progression, high-grade and metastatic disease are poorly defined. We perform genetic analysis of MOC across all histological grades, including benign and borderline mucinous ovarian tumors, and compare these to tumors from other potential extra-ovarian sites of origin. Here we show that MOC is distinct from tumors from other sites and supports a progressive model of evolution from borderline precursors to high-grade invasive MOC. Key drivers of progression identified are TP53 mutation and copy number aberrations, including a notable amplicon on 9p13. High copy number aberration burden is associated with worse prognosis in MOC. Our data conclusively demonstrate that MOC arise from benign and borderline precursors at the ovary and are not extra-ovarian metastases., Whether mucinous ovarian carcinoma (MOC) arises from cells at the ovary or from metastases from other primary sites is an unanswered question. Here, Cheasley et al perform a genetic analysis of the disease, showing that MOC arises at the ovary.
- Published
- 2019
- Full Text
- View/download PDF
10. Genes associated with bowel metastases in ovarian cancer
- Author
-
Ellen L. Goode, Francesco Multinu, Lynn C. Hartmann, Ling Jin, Tommaso Grassi, Andrea Mariani, Saravut J. Weroha, Julie Staub, Chen Wang, Debarshi Roy, Kimberly R. Kalli, Ann L. Oberg, Deok–Beom B. Jung, Michelle Torres, Daniel W. Visscher, Qing Zhang, Shaun M. Riska, Joseph E. Kumka, Scott H. Kaufmann, William A. Cliby, Viji Shridhar, Gunisha Sagar, and Vatsal P. Patel
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Mice, Nude ,Carcinoma, Ovarian Epithelial ,Article ,Metastasis ,Cohort Studies ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Internal medicine ,Intestinal Neoplasms ,medicine ,Animals ,Humans ,RNA, Neoplasm ,Ovarian Neoplasms ,Gene knockdown ,business.industry ,High-Throughput Nucleotide Sequencing ,Membrane Proteins ,Obstetrics and Gynecology ,Bowel resection ,medicine.disease ,Up-Regulation ,Bowel obstruction ,030104 developmental biology ,Real-time polymerase chain reaction ,Tumor progression ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Heterografts ,Immunohistochemistry ,Female ,Transcriptome ,Ovarian cancer ,business - Abstract
OBJECTIVE. This study is designed to identify genes and pathways that could promote metastasis to the bowel in high-grade serous ovarian cancer (OC) and evaluate their associations with clinical outcomes. METHODS. We performed RNA sequencing of OC primary tumors (PTs) and their corresponding bowel metastases (n = 21 discovery set; n = 18 replication set). Differentially expressed genes (DEGs) were those expressed at least 2-fold higher in bowel metastases (BMets) than PTs in at least 30% of patients (P < .05) with no increased expression in paired benign bowel tissue and were validated with quantitative reverse transcription PCR. Using an independent OC cohort (n = 333), associations between DEGs in PTs and surgical and clinical outcomes were performed. Immunohistochemistry and mouse xenograft studies were performed to confirm the role of LRRC15 in promoting metastasis. RESULTS. Among 27 DEGs in the discovery set, 21 were confirmed in the replication set: SFRP2, Col11A1, LRRC15, ADAM12, ADAMTS12, MFAP5, LUM, PLPP4, FAP, POSTN, GRP, MMP11, MMP13, C1QTNF3, EPYC, DIO2, KCNA1, NETO1, NTM, MYH13, and PVALB. Higher expression of more than half of the genes in the PT was associated with an increased requirement for bowel resection at primary surgery and an inability to achieve complete cytoreduction. Increased expression of LRRC15 in BMets was confirmed by immunohistochemistry and knockdown of LRRC15 significantly inhibited tumor progression in mice. CONCLUSIONS. We identified 21 genes that are overexpressed in bowel metastases among patients with OC. Our findings will help select potential molecular targets for the prevention and treatment of malignant bowel obstruction in OC.
- Published
- 2019
- Full Text
- View/download PDF
11. 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
12. 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
13. Porphyromonas somerae Invasion of Endometrial Cancer Cells
- Author
-
Taylor A. Crooks, Joseph D. Madison, Dana M. Walsh, William G. Herbert, Patricio R. Jeraldo, Nicholas Chia, William A. Cliby, Scott H. Kaufmann, and Marina R. S. Walther-Antonio
- Subjects
0301 basic medicine ,Microbiology (medical) ,In silico ,Biology ,medicine.disease_cause ,Microbiology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,intracellular invasion ,estradiol ,fumarate reductase ,medicine ,Porphyromonas somerae ,Uterine microbiome ,Original Research ,Endometrial cancer ,Cancer ,medicine.disease ,succinate dehydrogenase ,metabolomics ,Phenotype ,QR1-502 ,030104 developmental biology ,030220 oncology & carcinogenesis ,endometrial cancer ,Cancer research ,Intracellular - Abstract
Recent evidence suggests an association between endometrial cancer and the understudied bacterial species Porphyromonas somerae. This association was demonstrated in previous work that indicated a significantly enriched abundance of P. somerae in the uterine microbiome of endometrial cancer patients. Given the known associations of the Porphyromonas genus and oral cancer, we hypothesized that P. somerae may play a similar pathogenic role in endometrial cancer via intracellular activity. Before testing our hypothesis, we first characterized P. somerae biology, as current background data is limited. These novel characterizations include growth curves in liquid medium and susceptibility tests to antibiotics. We tested our hypothesis by examining growth changes in response to 17β-estradiol, a known risk factor for endometrial cancer, followed by metabolomic profiling in the presence and absence of 17β-estradiol. We found that P. somerae exhibits increased growth in the presence of 17β-estradiol of various concentrations. However, we did not find significant changes in metabolite levels in response to 17β-estradiol. To study direct host-microbe interactions, we used in vitro invasion assays under hypoxic conditions and found evidence for intracellular invasion of P. somerae in endometrial adenocarcinoma cells. We also examined these interactions in the presence of 17β-estradiol but did not observe changes in invasion frequency. Invasion was shown using three lines of evidence including visualization via differential staining and brightfield microscopy, increased frequency of bacterial recovery after co-culturing, and in silico methods to detail relevant genomic and transcriptomic components. These results underscore potential intracellular phenotypes of P. somerae within the uterine microbiome. Furthermore, these results raise new questions pertaining to the role of P. somerae in the progression of endometrial cancer.
- Published
- 2021
- Full Text
- View/download PDF
14. 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
15. Targeting LRRC15 Inhibits Metastatic Dissemination of Ovarian Cancer
- Author
-
Sayantani Sarkar Bhattacharya, Scott H. Kaufmann, Anirban K. Mitra, Jamie N. Bakkum-Gamez, Deok-Beom Jung, Ling Jin, Julie Staub, Nagarajan Kannan, Bhaskar Roy, Prabhu Thirusangu, Yinan Xiao, James W. Purcell, S. John Weroha, Viji Shridhar, Xiaonan Hou, Upasana Ray, Debarshi Roy, and Subramanyam Dasari
- Subjects
Cancer Research ,Programmed cell death ,Integrins ,Immunoconjugates ,Carcinoma, Ovarian Epithelial ,Article ,Metastasis ,Focal adhesion ,Cell Line, Tumor ,Ascites ,medicine ,Cell Adhesion ,Humans ,Viability assay ,Ovarian Neoplasms ,business.industry ,Membrane Proteins ,medicine.disease ,Oncology ,Membrane protein ,Cell culture ,Cancer research ,Female ,medicine.symptom ,Ovarian cancer ,business - Abstract
Dissemination of ovarian cancer cells can lead to inoperable metastatic lesions in the bowel and omentum that cause patient death. Here we show that LRRC15, a type-I 15-leucine–rich repeat-containing membrane protein, highly overexpressed in ovarian cancer bowel metastases compared with matched primary tumors and acts as a potent promoter of omental metastasis. Complementary models of ovarian cancer demonstrated that LRRC15 expression leads to inhibition of anoikis-induced cell death and promotes adhesion and invasion through matrices that mimic omentum. Mechanistically, LRRC15 interacted with β1-integrin to stimulate activation of focal adhesion kinase (FAK) signaling. As a therapeutic proof of concept, targeting LRRC15 with the specific antibody–drug conjugate ABBV-085 in both early and late metastatic ovarian cancer cell line xenograft models prevented metastatic dissemination, and these results were corroborated in metastatic patient-derived ovarian cancer xenograft models. Furthermore, treatment of 3D-spheroid cultures of LRRC15-positive patient-derived ascites with ABBV-085 reduced cell viability. Overall, these data uncover a role for LRRC15 in promoting ovarian cancer metastasis and suggest a novel and promising therapy to target ovarian cancer metastases. Significance: This study identifies that LRRC15 activates β1-integrin/FAK signaling to promote ovarian cancer metastasis and shows that the LRRC15-targeted antibody–drug conjugate ABBV-085 suppresses ovarian cancer metastasis in preclinical models.
- Published
- 2021
16. 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
17. 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
18. Circulating CD14 + HLA‐DR lo/− monocytic cells as a biomarker for epithelial ovarian cancer progression
- Author
-
B.C. Orr, John Lewis Etter, Robert P. Edwards, Paul K. Wallace, Anna Kay Adamson, Matthew F. Buas, Kirsten B. Moysich, Ashley E. Stenzel, Jennifer M. Mongiovi, Sarah Taylor, Janine M. Joseph, Joseph D. Tario, Divjot Kaur, Ellen L. Goode, Esther Elishaev, Francesmary Modugno, Scott H. Kaufmann, Angela Laslavic, Stacey J. Winham, Amit A. Lugade, Scott I. Abrams, and Kunle Odunsi
- Subjects
0301 basic medicine ,030219 obstetrics & reproductive medicine ,business.industry ,medicine.medical_treatment ,Immunology ,CD33 ,Obstetrics and Gynecology ,Human leukocyte antigen ,Immunotherapy ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Reproductive Medicine ,Renal cell carcinoma ,Pancreatic cancer ,medicine ,Cancer research ,Immunology and Allergy ,Biomarker (medicine) ,Ovarian cancer ,business - Abstract
PROBLEM Previous studies identified circulating CD14+ HLA-DRlo/- monocytic cells as an immune suppressive subset in solid malignancies, such as prostate, renal cell carcinoma, and pancreatic cancer. Such monocytic cells have been implicated not only in tumour progression but also as a potential barrier for immunotherapy. This study examined the relationship between the frequency of circulating monocytic cells and epithelial ovarian cancer (EOC) progression pre- and post-frontline chemotherapy, defined by disease stage, which is a leading prognostic factor for this malignancy. METHOD OF STUDY Incident cases of 236 women with EOC were recruited and comprehensive flow cytometry was utilized to assess the frequency of peripheral blood CD33+ CD11b+ HLA-DR-/low CD14+ CD15- monocytic cells, henceforth termed CD14+ HLA-DRlo/- monocytic cells, prior to and after completion of frontline chemotherapy. Multivariable odds ratios (OR) were used to estimate the association between CD14+ HLA-DRlo/- monocytic cell percentages and disease stage. Wilcoxon signed-rank tests evaluated changes in these monocytic cell levels pre- and post-chemotherapy in a patient subset (n = 70). RESULTS Patients with elevated frequencies of circulating CD14+ HLA-DRlo/- monocytic cells at diagnosis were at 3.33-fold greater odds of having advanced stage (III/IV) EOC (CI: 1.04-10.64), with a significant trend in increasing CD14+ HLA-DRlo/- monocytic cell levels (P = .04). There was a 2.02% median decrease of these monocytic cells post-chemotherapy among a subset of patients with advanced stage disease (P
- Published
- 2020
- Full Text
- View/download PDF
19. Clinical and pathological associations of PTEN expression in ovarian cancer: a multicentre study from the Ovarian Tumour Tissue Analysis Consortium
- Author
-
Scott H. Kaufmann, Daniel Guimarães Tiezzi, Valerie Rhenius, Gary L. Keeney, Alexander Hein, Dominique-Laurent Couturier, Florin Andrei Taran, Robert Edwards, Aline Talhouk, Stacey J. Winham, Christiani Bisinoto de Sousa, Nadja Pejovic, Andreas D. Hartkopf, Ramona Erber, Brenda Y. Hernandez, Jenny Lester, Teri A. Longacre, Anna L. Paterson, Peter Sinn, Robin Crawford, Lene Lundvall, Hugh Luk, Roberta B. Ness, Adekunle Odunsi, Suha Deen, Javier Benitez, Alice S. Whittemore, Anna Fischer, Anthony N. Karnezis, Christine Chow, Ana Osorio, Ellen L. Goode, Stefan Kommoss, Angela Brooks-Wilson, Linda E. Kelemen, Tanja Pejovic, Simon A. Gayther, Peter A. Fasching, Beth Y. Karlan, Jurandyr Moreira de Andrade, Samuel Leung, David D.L. Bowtell, James D. Brenton, Aleksandra Vrvilo, Marc T. Goodman, Paul D.P. Pharoah, Sara Y. Brucker, Honglin Song, Jennifer M Koziak, Robert A. Vierkant, Naveena Singh, Valerie McGuire, Chloe Karpinskyj, Mercedes Jimenez-Linan, Aleksandra Gentry-Maharaj, Anna deFazio, Jenny Chang-Claude, Linda S. Cook, Francesmary Modugno, Filipe C. Martins, Estrid Høgdall, Prafull Ghatage, Marie Lyne Alcaraz, Susan J. Ramus, Annette Staebler, Jennifer Alsop, David G. Huntsman, Joseph H. Rothstein, Tayyebeh M. Nazeran, Cristina Rodríguez-Antona, Luis Robles-Díaz, Bryan M. McCauley, María Jesús Gómez García, Michael E. Carney, Michael S. Anglesio, Yurii B. Shvetsov, Claus Høgdall, Sian Fereday, Martin Köbel, Maria P. Intermaggio, Sandra Orsulic, Luis Paz-Ares, Mary Anne Brett, Weiva Sieh, Matthias W. Beckmann, Helen Steed, Arndt Hartmann, Michael Schneider, Lynne R. Wilkens, Esther Herpel, Jacek Gronwald, Francisco José Candido dos Reis, Nhu D. Le, Gregg Nelson, Kirsten B. Moysich, Nadia Traficante, Usha Menon, Candido Dos Reis, Francisco J [0000-0001-5758-5917], Brenton, James D [0000-0002-5738-6683], Apollo - University of Cambridge Repository, Candido dos Reis, Francisco J. [0000-0001-5758-5917], and Brenton, James D. [0000-0002-5738-6683]
- Subjects
Oncology ,Cancer Research ,692/4028/67/1517/1709 ,Carcinoma, Ovarian Epithelial ,Clear Cell ,Androgen ,ESTUDOS PROSPECTIVOS ,Cohort Studies ,Gene Knockout Techniques ,0302 clinical medicine ,Ovarian carcinoma ,Ovarian Epithelial ,Receptors ,Prospective Studies ,Progesterone ,Cancer ,Ovarian Neoplasms ,0303 health sciences ,Tumor ,Molecular medicine ,biology ,Hazard ratio ,article ,Age Factors ,Middle Aged ,Ovarian Cancer ,Serous fluid ,Receptors, Estrogen ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Public Health and Health Services ,Biomarker (medicine) ,Female ,Receptors, Progesterone ,medicine.medical_specialty ,Oncology and Carcinogenesis ,Down-Regulation ,Adenocarcinoma ,03 medical and health sciences ,Rare Diseases ,Ovarian cancer ,Internal medicine ,Progesterone receptor ,medicine ,Biomarkers, Tumor ,PTEN ,Humans ,Oncology & Carcinogenesis ,030304 developmental biology ,Neoplasm Staging ,business.industry ,Tumor Suppressor Proteins ,Carcinoma ,PTEN Phosphohydrolase ,medicine.disease ,Estrogen ,692/4017 ,Androgen receptor ,Tissue Array Analysis ,biology.protein ,business ,Biomarkers ,Adenocarcinoma, Clear Cell - Abstract
Background PTEN loss is a putative driver in histotypes of ovarian cancer (high-grade serous (HGSOC), endometrioid (ENOC), clear cell (CCOC), mucinous (MOC), low-grade serous (LGSOC)). We aimed to characterise PTEN expression as a biomarker in epithelial ovarian cancer in a large population-based study. Methods Tumours from 5400 patients from a multicentre observational, prospective cohort study of the Ovarian Tumour Tissue Analysis Consortium were used to evaluate associations between immunohistochemical PTEN patterns and overall survival time, age, stage, grade, residual tumour, CD8+ tumour-infiltrating lymphocytes (TIL) counts, expression of oestrogen receptor (ER), progesterone receptor (PR) and androgen receptor (AR) by means of Cox proportional hazard models and generalised Cochran–Mantel–Haenszel tests. Results Downregulation of cytoplasmic PTEN expression was most frequent in ENOC (most frequently in younger patients; p value = 0.0001) and CCOC and was associated with longer overall survival in HGSOC (hazard ratio: 0.78, 95% CI: 0.65–0.94, p value = 0.022). PTEN expression was associated with ER, PR and AR expression (p values: 0.0008, 0.062 and 0.0002, respectively) in HGSOC and with lower CD8 counts in CCOC (p value p value = 0.019) and associated with higher CD8 counts (p value = 0.0016). Conclusions PTEN loss is a frequent driver in ovarian carcinoma associating distinctly with expression of hormonal receptors and CD8+ TIL counts in HGSOC and CCOC histotypes.
- Published
- 2020
- Full Text
- View/download PDF
20. 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
21. Pooled Clustering of High-Grade Serous Ovarian Cancer Gene Expression Leads to Novel Consensus Subtypes Associated with Survival and Surgical Outcomes
- Author
-
Ann L. Oberg, Scott H. Kaufmann, William A. Cliby, Chen Wang, Ellen L. Goode, Gary L. Keeney, Kimberly R. Kalli, Sebastian M. Armasu, Matthew J. Maurer, and Ethan P. Heinzen
- Subjects
Adult ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Concordance ,Disease ,Bioinformatics ,Article ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Internal medicine ,Gene expression ,medicine ,Humans ,Aged ,Aged, 80 and over ,Ovarian Neoplasms ,business.industry ,Mesenchymal stem cell ,Cancer ,Middle Aged ,Prognosis ,Debulking ,medicine.disease ,Neoadjuvant Therapy ,Subtyping ,Cystadenocarcinoma, Serous ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Neoplasm Grading ,business - Abstract
Purpose: Here we assess whether molecular subtyping identifies biological features of tumors that correlate with survival and surgical outcomes of high-grade serous ovarian cancer (HGSOC). Experimental Design: Consensus clustering of pooled mRNA expression data from over 2,000 HGSOC cases was used to define molecular subtypes of HGSOCs. This de novo classification scheme was then applied to 381 Mayo Clinic HGSOC patients with detailed survival and surgical outcome information. Results: Five molecular subtypes of HGSOC were identified. In the pooled dataset, three subtypes were largely concordant with prior studies describing proliferative, mesenchymal, and immunoreactive tumors (concordance > 70%), and the group of tumors previously described as differentiated type was segregated into two new types, one of which (anti-mesenchymal) had downregulation of genes that were typically upregulated in the mesenchymal subtype. Molecular subtypes were significantly associated with overall survival (P < 0.001) and with rate of optimal surgical debulking (≤1 cm, P = 1.9E−4) in the pooled dataset. Among stage III-C or IV Mayo Clinic patients, molecular subtypes were also significantly associated with overall survival (P = 0.001), as well as rate of complete surgical debulking (no residual disease; 16% in mesenchymal tumors compared with >28% in other subtypes; P = 0.02). Conclusions: HGSOC tumors may be categorized into five molecular subtypes that associate with overall survival and the extent of residual disease following debulking surgery. Because mesenchymal tumors may have features that were associated with less favorable surgical outcome, molecular subtyping may have future utility in guiding neoadjuvant treatment decisions for women with HGSOC. Clin Cancer Res; 23(15); 4077–85. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
22. RAS mutations drive proliferative chronic myelomonocytic leukemia via activation of a novel KMT2A-PLK1 axis
- Author
-
Matthew T. Howard, Klaus Geissler, Eric Padron, Isaac P. Horn, Ryan M. Carr, Nathalie Droin, Christopher Pin, Ajinkya Buradkar, Eric Solary, Mrinal M. Patnaik, Luciana L. Almada, Stephanie L. Safgren, Thomas E. Witzig, Kurt Berger, Jing Zhang, Andrey A. Yurchenko, David L. Marks, Alexis Vedder, Bonnie Alver, Xiaona You, Ismael Padioleau, Scott H. Kaufmann, Peter Valent, Sergey Nikolaev, Balasis Me, Ezequiel J. Tolosa, Martin E. Fernandez-Zapico, Giacomo Coltro, Terra L. Lasho, Temeida Graf, Abhishek A. Mangaonkar, Keith D. Robertson, Denis Vorobyev, and Moritz Binder
- Subjects
0303 health sciences ,biology ,Kinase ,Chronic myelomonocytic leukemia ,medicine.disease ,Phenotype ,3. Good health ,03 medical and health sciences ,Haematopoiesis ,Wee1 ,0302 clinical medicine ,KMT2A ,Monocytosis ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,Exome ,030304 developmental biology - Abstract
Chronic myelomonocytic leukemia (CMML) is an aggressive hematological malignancy with limited treatment options. Whole exome (WES) and targeted sequencing of several independent cohorts of CMML patients, comparing dysplastic (dCMML) to proliferative (pCMML) CMML, as well as paired chronic phase disease and acute leukemic transformation (LT), associate acquisition of oncogenic RAS pathway mutations, the most common being NRASG12D, with aggressive disease and with disease progression. Using patient derived progenitor colony assays and a NRASG12D-Vav-Cre mouse model, we further demonstrate the role of mutant RAS signaling in driving and maintaining pCMML phenotype. RNA-sequencing links RAS pathway mutations with an increased expression of genes encoding the mitotic checkpoint kinases PLK1 and WEE1. Further, we dmeoinstrated that non-mutated lysine methyltransferase KMT2A (MLL1) acts as mediator of NRAS-induced PLK1 and WEE1 expression. Finally, we demonstrate the translational value of our findings by showing that pharmacological PLK1 inhibition decreases monocytosis and hepatosplenomegaly while improving hematopoiesis in RAS mutant patient-derived xenografts. Hence, we define severe CMML as oncogenic RAS pathway-enriched malignancies, with a unique gene expression profile regulated by KMT2A, amenable to therapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
23. mTOR Targeting Agents for the Treatment of Lymphoma and Leukemia
- Author
-
Thomas E. Witzig, Andrea E. Wahner Hendrickson, and Scott H. Kaufmann
- Subjects
Leukemia ,business.industry ,medicine ,Cancer research ,medicine.disease ,business ,PI3K/AKT/mTOR pathway ,Lymphoma - Published
- 2019
- Full Text
- View/download PDF
24. 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
25. 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
26. Long-term survival of an ovarian cancer patient harboring a RAD51C missense mutation
- Author
-
Meghan R. Sullivan, Kara A. Bernstein, Weibin Wang, Scott H. Kaufmann, Yashpal Rawal, Elizabeth M. Swisher, Maria Jasin, Patrick Sung, Rohit Prakash, and Marc R. Radke
- Subjects
Insecta ,Mutation, Missense ,RAD51 ,ovarian neoplasm ,medicine.disease_cause ,Cell Line ,Gene Knockout Techniques ,Germline mutation ,XRCC3 ,medicine ,Animals ,Humans ,Missense mutation ,Germ-Line Mutation ,Ovarian Neoplasms ,Recombination, Genetic ,Mutation ,Binding Sites ,business.industry ,Cancer ,General Medicine ,medicine.disease ,DNA-Binding Proteins ,Drug Resistance, Neoplasm ,Cancer research ,RAD51C ,Female ,Rad51 Recombinase ,Transcriptome ,Ovarian cancer ,business ,Research Article - Abstract
Mutations in homologous recombination (HR) genes predispose to cancer but also sensitize to chemotherapeutics. Although therapy can initially be effective, cancers frequently cease responding, leading to recurrence and poor prognosis. Here we identify a germline mutation in RAD51C, a critical HR factor and known tumor suppressor, in an ovarian cancer patient with exceptionally long, progression-free survival. The RAD51C–T132P mutation is in a highly conserved residue within the nucleotide-binding site and interferes with single-strand DNA binding of the RAD51 paralog complex RAD51B–RAD51C–RAD51D–XRCC2 and association with another RAD51 paralog XRCC3. These biochemical defects lead to highly defective HR and drug sensitivity in tumor cells, ascribing RAD51C–T132P as a deleterious mutation that was likely causal for tumor formation. Conversely, its position within a critical site suggests that it is refractory to secondary mutations that would restore RAD51C gene function and lead to therapy resistance. A need for a greater understanding of the relationship between mutation position and reversion potential of HR genes is underscored, as it may help predict the effectiveness of therapies in patients with HR-deficient cancers.
- Published
- 2021
- Full Text
- View/download PDF
27. Assessment of Drug Sensitivity in Hematopoietic Stem and Progenitor Cells from Acute Myelogenous Leukemia and Myelodysplastic Syndrome Ex Vivo
- Author
-
Scott H. Kaufmann, B. Douglas Smith, Allan D. Hess, Judith E. Karp, Laura Finn, James M. Foran, and Katherine L.B. Knorr
- Subjects
0301 basic medicine ,Myeloid ,Cell Survival ,Acute myelogenous leukemia ,MLN4924 ,Cell‐Based Drug Development, Screening, and Toxicology ,Cell Count ,Cyclopentanes ,03 medical and health sciences ,Myelogenous ,Translational Research Articles and Reviews ,hemic and lymphatic diseases ,Humans ,Medicine ,CD135 ,Progenitor cell ,Myeloid progenitor cells ,Cell Death ,business.industry ,Myelodysplastic syndromes ,Cytarabine ,Cell Biology ,General Medicine ,Hematopoietic Stem Cells ,medicine.disease ,3. Good health ,Leukemia, Myeloid, Acute ,Haematopoiesis ,Pyrimidines ,030104 developmental biology ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Cancer research ,Bone marrow ,Stem cell ,business ,Myelodysplastic syndrome ,Developmental Biology - Abstract
Current understanding suggests that malignant stem and progenitor cells must be reduced or eliminated for prolonged remissions in myeloid neoplasms such as acute myelogenous leukemia (AML) or myelodysplastic syndrome (MDS). Multicolor flow cytometry has been widely used to distinguish stem and myeloid progenitor cells from other populations in normal and malignant bone marrow. In this study, we present a method for assessing drug sensitivity in MDS and AML patient hematopoietic stem and myeloid progenitor cell populations ex vivo using the investigational Nedd8-activating enzyme inhibitor MLN4924 and standard-of-care agent cytarabine as examples. Utilizing a multicolor flow cytometry antibody panel for identification of hematopoietic stem cells, multipotent progenitors, common myeloid progenitors, granulocyte-monocyte progenitors, and megakaryocyte-erythroid progenitors present in mononuclear cell fractions isolated from bone marrow aspirates, we compare stem and progenitor cell counts after treatment for 24 hours with drug versus diluent. We demonstrate that MLN4924 exerts a cytotoxic effect on MDS and AML stem and progenitor cell populations, whereas cytarabine has more limited effects. Further application of this method for evaluating drug effects on these populations ex vivo and in vivo may inform rational design and selection of therapies in the clinical setting.
- Published
- 2016
- Full Text
- View/download PDF
28. 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
29. Abstract PR02: Proteogenomic approach to identify mechanisms of platinum refractoriness in high-grade serous ovarian cancers
- Author
-
Bing Zhang, Dongqing Huang, Sara R. Savage, Catherine J. Huntoon, Andrew N. Hoofnagle, Pei Wang, Steven P. Gygi, Shrabanti Chowdhury, Scott H. Kaufmann, Jeffrey R. Whiteaker, Xiaonan Hou, Regine M. Schoenherr, Samuel C. Mok, Amanda G. Paulovich, Uliana J. Voytovich, Michael J. Birrer, Steven J. Skates, Qing Yu, Jacob J. Kennedy, Lei Zhao, S. John Weroha, Zahra Shire, Chenwei Lin, Richard G. Ivey, and Larry M. Karnitz
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Proteomic Profiling ,Phosphoproteomics ,Cancer ,medicine.disease ,Proteomics ,Clinical trial ,Serous fluid ,Internal medicine ,Proteome ,medicine ,business ,Ovarian cancer - Abstract
Although high-grade serous ovarian cancers (HGSOC) are highly chemosensitive with an 85% initial response rate to platinum-based chemotherapy, 15% of patients are “exceptional nonresponders,” with platinum-refractory tumors that remain stable or progress during treatment. Unfortunately, we have no predictive biomarkers to identify refractory patients up front, and they receive futile chemotherapy through which most patients become too ill to be eligible for clinical trials. Hence, no progress has been made in treating these deadly tumors. The goal of this study is to identify mechanisms of platinum refractoriness to: i) predict refractory HGSOCs up front and ii) identify potential new drug targets in refractory disease to point to desperately needed new therapeutic approaches. Of note, 80-90% of patients who are initially platinum responsive will relapse and develop platinum-resistant disease, and it is possible that findings in platinum-refractory tumors might also provide insights into platinum-resistant tumors. Our NCI Clinical Proteomic Tumor Analysis Consortium (CPTAC)-funded approach combines genomic and proteomic (“proteogenomic”) analyses of both preclinical models (0, 8, and 24 hours post-platinum exposure) and treatment-naïve human tumors. For preclinical models, we studied a well-characterized collection of patient-derived xenograft (PDX) models (10 sensitive, 10 refractory), as well as intrapatient HGSOC cell line pairs derived from patients before and after the development of platinum resistance. For the PDX models, proteogenomic profiling included RNASeq, WES, global proteomics, and phosphoproteomics at all 3 time points (0, 8, 24 hours). For cell line models (3 sensitive, 3 resistant), proteogenomic profiling was performed at all 3 timepoints (0, 8, 24 hours), and experiments were performed in complete biologic triplicate. Analyses included RNASeq, WES, global proteomics, phosphoproteomics, ubiquitin proteome, acetylated proteome, and pTyr. A large collection of 275 human HGSOCs (an approximate equal balance of platinum sensitive and refractory tumors) is currently undergoing proteomic profiling, and genomic profiles (WGS, RNASeq) will be performed on a subset. In parallel, we have performed a comprehensive review of 31 years of published work on platinum responses of human cancers, identifying ~700 genes implicated in the response and scoring each gene with respect to strength of the published evidence. Using a Bayesian approach, we are integrating the curated candidates from the literature with our empirical proteogenomic datasets to identify a candidate signature for detecting platinum-refractory disease prior to chemotherapy. We are also performing gene-regulatory network analysis to identify potential drivers of chemo response. NextGen, targeted, multiplex, multiple reaction monitoring mass spectrometry-based assays are being developed to quantify proteins in the signature for validation studies, using independent patient cohorts. This abstract is also being presented as Poster A62. Citation Format: Jacob J. Kennedy, Shrabanti Chowdhury, Sara R. Savage, Xiaonan Hou, Catherine J. Huntoon, Richard G. Ivey, Qing Yu, Chenwei Lin, Dongqing Huang, Lei Zhao, Uliana J. Voytovich, Regine M. Schoenherr, Zahra Shire, Steven J. Skates, Jeffrey R. Whiteaker, Andrew N. Hoofnagle, Samuel C. Mok, Bing Zhang, Larry M. Karnitz, S. John Weroha, Steven P. Gygi, Scott H. Kaufmann, Pei Wang, Michael J. Birrer, Amanda G. Paulovich. Proteogenomic approach to identify mechanisms of platinum refractoriness in high-grade serous ovarian cancers [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 PR02.
- Published
- 2020
- Full Text
- View/download PDF
30. Abstract IA02: DNA repair gene promoter methylation patterns adapt and influence PARP inhibitor response
- Author
-
Maria I. Harrell, Hu Li, Giada V. Zapparoli, Clare L. Scott, Olga Kondrashova, Xiaonan Hou, Scott H. Kaufmann, John Weroha, Elizabeth M. Swisher, Cordelia D. McGehee, Ming E. Wong, Kevin A. Peterson, Kasenija Nesic, Vivian Negron, Matthew Wakefield, Ashan Musafer, Alexander Dobrovic, Rachel M. Hurley, Paula A. Schneider, and Melissa C. Southey
- Subjects
Cancer Research ,DNA repair ,Cancer ,Methylation ,Biology ,medicine.disease ,Oncology ,PARP inhibitor ,medicine ,Cancer research ,RAD51C ,Gene silencing ,Homologous recombination ,Gene - Abstract
PARP inhibitor (PARPi) resistance in high-grade serous ovarian carcinoma (HGSOC) can be acquired as a result of restored homologous recombination (HR) due to secondary or reversion mutations in HR genes, such as BRCA1, BRCA2, and RAD51C, or due to loss of BRCA1 promoter methylation (meBRCA1). We have demonstrated that homozygous meBRCA1 can be lost or reverted to heterozygous methylation following treatment with platinum-based chemotherapy, resulting in HR-competent PARPi-resistant tumors. RAD51C promoter methylation (meRAD51C) is detected in approximately 2% of HGSOC cases and, as with meBRCA1, is associated with gene silencing and HR deficiency. We are exploring PARPi response in meRAD51C preclinical models to determine clinical relevance. Two patient-derived xenograft (PDX) models of HGSOC with RAD51C gene silencing caused by meRAD51C have distinct meRAD51C profiles (measured by methylation-specific high-resolution melt analysis and targeted bisulfite next-generation sequencing), and different responses to PARPi treatment pressure. PDX PH039 loses methylation and regains RAD51C expression after only 2 cycles of PARPi retreatment (niraparib), resulting in PARPi-refractory tumors by cycle 3-4. Illumina EPIC methylation array analysis of PH039 revealed increasing global methylation losses following each round of PARPi treatment. Lack of meRAD51C stability and rapid development of PARPi resistance in PH039 may be due to the high degree of meRAD51C heterogeneity within the tumor favoring selection of pre-existing HR-competent clones under PARPi pressure. In contrast, PDX 183 has a relatively homogeneous and stable meRAD51C profile. We have multiple examples of using unique PDX models to demonstrate various important features of PARPi resistance, including loss of promoter methylation for BRCA1 vs. RAD51C. Thus, meRAD51C confers response to PARPi in HGSOC, but PARPi treatment pressure can cause loss of methylation and drug resistance in some tumors. The contrasting PARPi responses of these PDX provide a platform for the study of meRAD51C stability in vivo and may present therapeutic opportunities to improve meRAD51C durability and PARPi responses in patients. Citation Format: Kasenija Nesic, Rachel M Hurley, Cordelia McGehee, Olga Kondrashova, Maria I. Harrell, Giada V. Zapparoli, Ashan Musafer, Ming E. Wong, John Weroha, Xiaonan Hou, Hu Li, Vivian Negron, Kevin Peterson, Paula Schneider, Elizabeth M. Swisher, Melissa Southey, Alexander Dobrovic, Matthew Wakefield, Scott H. Kaufmann, Clare L. Scott. DNA repair gene promoter methylation patterns adapt and influence PARP inhibitor response [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 IA02.
- Published
- 2020
- Full Text
- View/download PDF
31. 53BP1 as a potential predictor of response in PARP inhibitor-treated homologous recombination-deficient ovarian cancer
- Author
-
Peter Ansell, Andrea E. Wahner Hendrickson, Rutger H. T. Koornstra, Scott H. Kaufmann, Matthew W. Dudley, Jourik A. Gietema, Krista M. Goergen, X. Wei Meng, Matthew J. Maurer, Karen S. Flatten, Carla D. Van Herpen, Martha W. den Hollander, Ann L. Oberg, Rachel M. Hurley, Agnes Jager, Maja J.A. de Jonge, Maria I. Harrell, Jill M. Wagner, Elizabeth M. Swisher, Stacie Peacock Shepherd, Daniel W. Visscher, Vivian Negron, Damage and Repair in Cancer Development and Cancer Treatment (DARE), Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Medical Oncology
- Subjects
0301 basic medicine ,DNA Repair ,Genes, BRCA2 ,Genes, BRCA1 ,Poly (ADP-Ribose) Polymerase-1 ,RAD51 ,MULTICENTER ,Cancer development and immune defence Radboud Institute for Health Sciences [Radboudumc 2] ,0302 clinical medicine ,PARP1 ,Medicine ,Homologous Recombination ,PARP inhibitors ,Ovarian Neoplasms ,Sulfonamides ,Obstetrics and Gynecology ,HR-deficiency ,OPEN-LABEL ,53BP1 ,POLYMERASE INHIBITORS ,Oncology ,NIRAPARIB ,030220 oncology & carcinogenesis ,Benzamides ,PARP inhibitor ,Female ,Tumor Suppressor p53-Binding Protein 1 ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,CARCINOMA ,DNA repair ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Ovarian cancer ,Cell Line, Tumor ,Carcinoma ,Humans ,Clonogenic assay ,REPAIR ,business.industry ,MUTATIONS ,DNA Repair Pathway ,medicine.disease ,PROFICIENT ,030104 developmental biology ,Drug Resistance, Neoplasm ,CELLS ,Cancer research ,DNA damage ,business ,RESISTANCE - Abstract
Contains fulltext : 202591.pdf (Publisher’s version ) (Closed access) OBJECTIVE: Poly(ADP-ribose) polymerase (PARP) inhibitors have shown substantial activity in homologous recombination- (HR-) deficient ovarian cancer and are undergoing testing in other HR-deficient tumors. For reasons that are incompletely understood, not all patients with HR-deficient cancers respond to these agents. Preclinical studies have demonstrated that changes in alternative DNA repair pathways affect PARP inhibitor (PARPi) sensitivity in ovarian cancer models. This has not previously been assessed in the clinical setting. METHODS: Clonogenic and plasmid-based HR repair assays were performed to compare BRCA1-mutant COV362 ovarian cancer cells with or without 53BP1 gene deletion. Archival biopsies from ovarian cancer patients in the phase I, open-label clinical trial of PARPi ABT-767 were stained for PARP1, RAD51, 53BP1 and multiple components of the nonhomologous end-joining (NHEJ) DNA repair pathway. Modified histochemistry- (H-) scores were determined for each repair protein in each sample. HRD score was determined from tumor DNA. RESULTS: 53BP1 deletion increased HR in BRCA1-mutant COV362 cells and decreased PARPi sensitivity in vitro. In 36 women with relapsed ovarian cancer, responses to the PARPi ABT-767 were observed exclusively in cancers with HR deficiency. In this subset, 7 of 18 patients (39%) had objective responses. The actual HRD score did not further correlate with change from baseline tumor volume (r=0.050; p=0.87). However, in the HR-deficient subset, decreased 53BP1 H-score was associated with decreased antitumor efficacy of ABT-767 (r=-0.69, p=0.004). CONCLUSION: Differences in complementary repair pathways, particularly 53BP1, correlate with PARPi response of HR-deficient ovarian cancers.
- Published
- 2019
32. 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
33. 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
34. A Phase I Study of the Farnesyltransferase Inhibitor Tipifarnib in Combination with the Epidermal Growth Factor Tyrosine Kinase Inhibitor Erlotinib in Patients with Advanced Solid Tumors
- Author
-
Joel M. Reid, Julian R. Molina, Scott H. Kaufmann, Alex A. Adjei, Jacob B. Allred, Jun Yin, Khalid Jazieh, Vun Sin Lim, and Matthew Bidwell Goetz
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Maximum Tolerated Dose ,medicine.drug_class ,Quinolones ,Tyrosine-kinase inhibitor ,Article ,03 medical and health sciences ,Erlotinib Hydrochloride ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Farnesyltranstransferase ,Humans ,Pharmacology (medical) ,Tissue Distribution ,Aged ,Pharmacology ,Aged, 80 and over ,Salvage Therapy ,business.industry ,Farnesyltransferase inhibitor ,Middle Aged ,medicine.disease ,Prognosis ,Rash ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Tolerability ,030220 oncology & carcinogenesis ,Tipifarnib ,Female ,Erlotinib ,medicine.symptom ,business ,Progressive disease ,medicine.drug - Abstract
INTRODUCTION: Based on preclinical cytotoxic synergy between tipifarnib and erlotinib, a phase I study of this combination was conducted in patients with advanced solid tumors to evaluate safety, tolerability, maximum tolerated dose (MTD) and preliminary evidence of efficacy. METHODS: Patient enrollment followed the traditional “3 + 3” dose escalation scheme, through 4 dose levels, ranging from tipifarnib 200 mg twice daily plus erlotinib 75 mg once daily to tipifarnib 300 mg twice daily plus erlotinib 150 mg once daily. After the MTD of the combination was identified, 12 additional patients were treated to better define the pharmacokinetics and pharmacodynamics of these agents. RESULTS: A total of 27 patients were enrolled in the study (dose escalation, 15; dose expansion, 12). Dose limiting toxicity was seen in one patient at dose level 4 (grade 3 diarrhea). The MTD was reached at erlotinib 150 mg once daily combined with tipifarnib 300 mg twice daily. The most common side effects of the combination of all grades were diarrhea (85.2%), fatigue (77.8%), rash (70.4%), and anorexia (59.3%). Overall, 2 patients (7.4%; with liver cancer and melanoma, respectively) had partial responses, 10 (37%) had stable disease, 11 had progressive disease (40.7%) and 4 stopped treatment prematurely for assessment. CONCLUSION: The combination of tipifarnib and erlotinib was well tolerated. Erlotinib 150 mg once daily for 28 days combined with tipifarnib 300 mg twice daily for 21 days was identified as the recommended phase 2 dose. Tipifarnib is currently being evaluated in HRAS mutant tumors, providing a potential opportunity to further test this combination.
- Published
- 2018
35. APOBEC3G Expression Correlates with T-Cell Infiltration and Improved Clinical Outcomes in High-grade Serous Ovarian Carcinoma
- Author
-
Anieta M. Sieuwerts, John W.M. Martens, Brandon Leonard, Els M.J.J. Berns, Brett D. Anderson, Matthew J. Maurer, Olivier De Wever, Scott H. Kaufmann, Ann L. Oberg, Mieke Van Bockstal, Reuben S. Harris, Kimberly R. Kalli, Jo Van Dorpe, Jozien Helleman, William L. Brown, Gabriel J. Starrett, Medical Oncology, and UCL - (SLuc) Service d'anatomie pathologique
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,viruses ,T cell ,Gene Expression ,APOBEC-3G Deaminase ,Kaplan-Meier Estimate ,Biology ,Lymphocyte Activation ,Article ,GZMB ,Cohort Studies ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Breast cancer ,Biomarkers, Tumor ,medicine ,Humans ,Proportional Hazards Models ,Ovarian Neoplasms ,Cancer ,biochemical phenomena, metabolism, and nutrition ,Prognosis ,medicine.disease ,Immunohistochemistry ,Cystadenocarcinoma, Serous ,CD8A ,Serous fluid ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Biomarker (medicine) ,Female ,Neoplasm Grading - Abstract
Purpose: APOBEC3 DNA cytosine deaminase family members normally defend against viruses and transposons. However, deregulated APOBEC3 activity causes mutations in cancer. Because of broad expression profiles and varying mixtures of normal and cancer cells in tumors, including immune cell infiltration, it is difficult to determine where different APOBEC3s are expressed. Here, we ask whether correlations exist between APOBEC3 expression and T-cell infiltration in high-grade serous ovarian cancer (HGSOC), and assess whether these correlations have prognostic value. Experimental Design: Transcripts for APOBEC3G, APOBEC3B, and the T-cell markers, CD3D, CD4, CD8A, GZMB, PRF1, and RNF128 were quantified by RT-qPCR for a cohort of 354 HGSOC patients. Expression values were correlated with each other and clinical parameters. Two additional cohorts were used to extend HGSOC clinical results. Immunoimaging was used to colocalize APOBEC3G and the T-cell marker CD3. TCGA data extended expression analyses to additional cancer types. Results: A surprising positive correlation was found for expression of APOBEC3G and several T cell genes in HGSOC. Immunohistochemistry and immunofluorescent imaging showed protein colocalization in tumor-infiltrating T lymphocytes. High APOBEC3G expression correlated with improved outcomes in multiple HGSOC cohorts. TCGA data analyses revealed that expression of APOBEC3D and APOBEC3H also correlates with CD3D across multiple cancer types. Conclusions: Our results identify APOBEC3G as a new candidate biomarker for tumor-infiltrating T lymphocytes and favorable prognoses for HGSOC. Our data also highlight the complexity of the tumor environment with respect to differential APOBEC family gene expression in both tumor and surrounding normal cell types. Clin Cancer Res; 22(18); 4746–55. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
36. 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
37. TP53mutations, tetraploidy and homologous recombination repair defects in early stage high-grade serous ovarian cancer
- Author
-
Jeremy Chien, R. Keira Cheetham, Yan W. Asmann, Ellen L. Goode, Jean Pierre A. Kocher, Lynn C. Hartmann, Scott H. Kaufmann, Russell J. Grocock, Julie M. Cunningham, Yaman Tarabishy, Steven N. Hart, John F. Peden, Ann L. Oberg, Ying Li, Kimberly R. Kalli, Rui Kuang, Marina Bibikova, David Bentley, Jaime I. Davila, Elizabeth J. Atkinson, Saurabh Baheti, Sabine Dietmann, Viji Shridhar, Chen Wang, Debra A. Bell, Takayo Ota, Elizabeth M. Swisher, Jian-Bing Fan, Hugues Sicotte, and Sean Humphray
- Subjects
p53 ,Mutation rate ,endocrine system diseases ,Somatic cell ,Loss of Heterozygosity ,DNA Primase ,Biology ,Loss of heterozygosity ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Mutation Rate ,Information and Computing Sciences ,Genetics ,Carcinoma ,medicine ,Humans ,2.1 Biological and endogenous factors ,neoplasms ,Cancer ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,Prevention ,Human Genome ,Recombinational DNA Repair ,Genomics ,Biological Sciences ,Genes, p53 ,medicine.disease ,BRCA2 Protein ,Molecular biology ,Ovarian Cancer ,3. Good health ,Tetraploidy ,Genes ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Ploidy ,Homologous recombination ,Ovarian cancer ,Environmental Sciences ,Biotechnology ,Developmental Biology - Abstract
To determine early somatic changes in high-grade serous ovarian cancer (HGSOC), we performed whole genome sequencing on a rare collection of 16 low stage HGSOCs. The majority showed extensive structural alterations (one had an ultramutated profile), exhibited high levels of p53 immunoreactivity, and harboured a TP53 mutation, deletion or inactivation. BRCA1 and BRCA2 mutations were observed in two tumors, with nine showing evidence of a homologous recombination (HR) defect. Combined Analysis with The Cancer Genome Atlas (TCGA) indicated that low and late stage HGSOCs have similar mutation and copy number profiles. We also found evidence that deleterious TP53 mutations are the earliest events, followed by deletions or loss of heterozygosity (LOH) of chromosomes carrying TP53, BRCA1 or BRCA2. Inactivation of HR appears to be an early event, as 62.5% of tumours showed a LOH pattern suggestive of HR defects. Three tumours with the highest ploidy had little genome-wide LOH, yet one of these had a homozygous somatic frame-shift BRCA2 mutation, suggesting that some carcinomas begin as tetraploid then descend into diploidy accompanied by genome-wide LOH. Lastly, we found evidence that structural variants (SV) cluster in HGSOC, but are absent in one ultramutated tumor, providing insights into the pathogenesis of low stage HGSOC.
- Published
- 2015
- Full Text
- View/download PDF
38. 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
39. BCL2 mutations are associated with increased risk of transformation and shortened survival in follicular lymphoma
- Author
-
James R. Cerhan, Xiaosheng Wu, Diane F. Jelinek, Anne J. Novak, Cristina Correia, Grzegorz S. Nowakowski, Steven D. Gore, Husheng Ding, Judith E. Karp, Matthew J. Maurer, Ahmet Dogan, X. Wei Meng, Thomas M. Habermann, Paula A. Schneider, Amy K. Church, Scott H. Kaufmann, William R. Macon, Andrew L. Feldman, Neil E. Kay, Thomas E. Witzig, Haiming Dai, and Susan L. Slager
- Subjects
Adult ,Male ,Immunology ,Follicular lymphoma ,Immunoglobulins ,Chromosomal translocation ,Locus (genetics) ,Aggressive lymphoma ,Biochemistry ,Disease-Free Survival ,Gene Expression Regulation, Enzymologic ,Cohort Studies ,Risk Factors ,immune system diseases ,Cytidine Deaminase ,hemic and lymphatic diseases ,Prevalence ,medicine ,Humans ,Neoplasm ,neoplasms ,Lymphoma, Follicular ,Survival rate ,Aged ,Aged, 80 and over ,Chromosomes, Human, Pair 14 ,Lymphoid Neoplasia ,biology ,business.industry ,Age Factors ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Lymphoma ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Cell Transformation, Neoplastic ,Proto-Oncogene Proteins c-bcl-2 ,Mutation ,biology.protein ,Female ,biological phenomena, cell phenomena, and immunity ,Antibody ,Chromosomes, Human, Pair 18 ,business - Abstract
Follicular lymphoma (FL), an indolent neoplasm caused by a t(14;18) chromosomal translocation that juxtaposes the BCL2 gene and immunoglobulin locus, has a variable clinical course and frequently undergoes transformation to an aggressive lymphoma. Although BCL2 mutations have been previously described, their relationship to FL progression remains unclear. In this study, we evaluated the frequency and nature of BCL2 mutations in 2 independent cohorts of grade 1 and 2 FLs, along with the correlation between BCL2 mutations, transformation risk, and survival. The prevalence of BCL2 coding sequence mutations was 12% in FL at diagnosis and 53% at transformation (P.0001). The presence of these BCL2 mutations at diagnosis correlated with an increased risk of transformation (hazard ratio 3.6; 95% CI, 2.0-6.2; P.0001) and increased risk of death due to lymphoma (median survival of 9.5 years with BCL2 mutations vs 20.4 years without; P = .012). In a multivariate analysis, BCL2 mutations and high FL international prognostic index were independent risk factors for transformation and death due to lymphoma. Some mutant Bcl-2 proteins exhibited enhanced antiapoptotic capacity in vitro. Accordingly, BCL2 mutations can affect antiapoptotic Bcl-2 function, are associated with increased activation-induced cytidine deaminase expression, and correlate with increased risk of transformation and death due to lymphoma.
- Published
- 2015
- Full Text
- View/download PDF
40. 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
41. 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
42. Osteoblasts Protect AML Cells From SDF-1-Induced Apoptosis
- Author
-
Amel Dudakovic, Meghan E. McGee-Lawrence, Scott H. Kaufmann, Allan D. Hess, Kimberly N. Kremer, B. Douglas Smith, Jennifer J. Westendorf, Rachael L. Philips, Judith E. Karp, Andre J. van Wijnen, and Karen E. Hedin
- Subjects
U937 cell ,Cellular differentiation ,Mesenchymal stem cell ,Osteoblast ,Cell Biology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Leukemia ,medicine.anatomical_structure ,hemic and lymphatic diseases ,medicine ,Cancer research ,MC3T3 ,Bone marrow ,Stem cell ,Molecular Biology - Abstract
The bone marrow provides a protective environment for acute myeloid leukemia (AML) cells that often allows leukemic stem cells to survive standard chemotherapeutic regimens. Targeting these leukemic stem cells within the bone marrow is critical for preventing relapse. We recently demonstrated that SDF-1, a chemokine abundant in the bone marrow, induces apoptosis in AML cell lines and in patient samples expressing high levels of its receptor, CXCR4. Here we show that a subset of osteoblast lineage cells within the bone marrow can protect AML cells from undergoing apoptosis in response to the SDF-1 naturally present in that location. In co-culture systems, osteoblasts at various stages of differentiation protected AML cell lines and patient isolates from SDF-1-induced apoptosis. The differentiation of the osteoblast cell lines, MC3T3 and W-20-17, mediated this protection via a cell contact-independent mechanism. In contrast, bone marrow-derived mesenchymal cells, the precursors of osteoblasts, induced apoptosis in AML cells via a CXCR4-dependent mechanism and failed to protect AML cells from exogenously added SDF-1. These results indicate that osteoblasts in the process of differentiation potently inhibit the SDF-1-driven apoptotic pathway of CXCR4-expressing AML cells residing in the bone marrow. Drugs targeting this protective mechanism could potentially provide a new approach to treating AML by enhancing the SDF-1-induced apoptosis of AML cells residing within the bone marrow microenvironment.
- Published
- 2014
- Full Text
- View/download PDF
43. 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
44. 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
45. Clinical Categorization of Chronic Myelomonocytic Leukemia into Proliferative and Dysplastic Subtypes Correlates with Distinct Genomic, Transcriptomic and Epigenomic Signatures
- Author
-
Matthew T. Howard, Isaac P. Horn, Naseema Gangat, Terra L. Lasho, Ryan M. Carr, Bonnie Alver, Klaus Geissler, Christopher Pin, Animesh Pardanani, Mark R. Litzow, Kurt Berger, Guadalupe Belen Antelo, Moritz Binder, Martin E. Fernandez-Zapico, Thomas E. Witzig, Mrinal M. Patnaik, Stephanie L. Safgren, David L. Marks, Ezequiel J. Tolosa, Luciana L. Almada, Michelle A. Elliott, Aref Al-Kali, Scott H. Kaufmann, Ayalew Tefferi, Abhishek A. Mangaonkar, Giacomo Coltro, and Keith D. Robertson
- Subjects
biology ,Immunology ,Chronic myelomonocytic leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Genome ,Gene expression profiling ,Transcriptome ,Histone ,medicine.anatomical_structure ,Categorization ,biology.protein ,Cancer research ,medicine ,Bone marrow ,Epigenomics - Abstract
Introduction: Chronic myelomonocytic leukemia (CMML), an aggressive myeloid malignancy, can be categorized into two subtypes, proliferative CMML (pCMML) and dysplastic (dCMML), based on a white blood cell (WBC) count of ≥ 13 x 109/L for the former (Arber et al. Blood 2016). While this WBC cut off is somewhat arbitrary, patients with pCMML have unique phenotypic features and a shorter survival. We carried out this study to assess the genomic, transcriptomic and epigenetic landscapes of these two CMML subtypes. Methods: Peripheral blood (PB) and bone marrow (BM) mononuclear cells (MNC) were obtained from WHO-defined CMML patients. Next generation sequencing (NGS) using a 36-gene panel was performed on 350 patients with Illumina HiSeq4000 platform with median read depth of 400X. RNA sequencing (RNA-seq) was performed on 25 patients by bulk whole transcriptome sequencing using Illumina TruSeq. DNA immunoprecipitation sequencing (DIP-seq) was performed on 18 patients using 5-methylcystocine (5mC), 5-hydroxymethylcytosine (5hmC) and bridging monoclonal antibodies with subsequent paired-end sequencing using HiSeq4000. Chromatin immunoprecipitation sequencing (ChIP-seq) was performed on 30 patients with Illumina HiSeq2500 to a depth of 25 million for histone 3 lysine 4 monomethylation (H3K4me1) and histone 3 lysine 4 trimethylation (H3K4me3) and 50 million reads for histone 3 lysine 27 trimethylation (H3K27me3) and Input per sample. Results: Five hundred and seventy-three patients with WHO defined CMML were included; median age 71 years (range 18-95 years), 67% males. 282 patients had pCMML (49%), while 291 (51%) had dCMML. As pre-defined, patients with pCMML were more likely to have higher absolute monocyte counts (p Conclusions: Despite the somewhat arbitrary WBC distinction between pCMML and dCMML, clear phenotypic, genetic, transcriptomic, epigenetic and survival differences exist between the two subtypes, providing strong biological rationale for this distinction. pCMML patients have a higher frequency of oncogenic RAS pathway mutations, a unique transcriptomic profile enriched in mitotic check point kinases and a unique chromatin configuration with global and sequence specific enrichment in H3K4me1, with no significant global differences in 5mC, 5hmC, or H3K4me3 and H3K27me3 occupancy. Figure 1 Disclosures Geissler: AOP: Honoraria; Pfizer: Honoraria; AstraZeneca: Honoraria; Novartis: Honoraria; Celgene: Honoraria; Roche: Honoraria; Abbvie: Honoraria; Ratiopharm: Honoraria; Amgen: Honoraria. Al-Kali:Astex Pharmaceuticals, Inc.: Research Funding. Patnaik:Stem Line Pharmaceuticals.: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2019
- Full Text
- View/download PDF
46. APOBEC3B Upregulation and Genomic Mutation Patterns in Serous Ovarian Carcinoma
- Author
-
Hugues Sicotte, Craig April, Julie M. Cunningham, Emily K. Law, Ann L. Oberg, Brandon Leonard, David Bentley, Ellen L. Goode, Marina Bibikova, Michael A. Carpenter, Anurag Rathore, Lynn C. Hartmann, Jason B. Nikas, Sean Humphray, Russell J. Grocock, Michael B. Burns, Rachel Isaksson Vogel, Yuji Zhang, Matthew J. Maurer, Debra A. Bell, Scott H. Kaufmann, Steven N. Hart, John F. Peden, Jeremy Chien, Ying Li, Zoya Kingsbury, Elizabeth M. Swisher, R. Keira Cheetham, Jian-Bing Fan, William L. Brown, Viji Shridhar, Nuri A. Temiz, Kimberly R. Kalli, and Reuben S. Harris
- Subjects
Genome instability ,Cancer Research ,endocrine system diseases ,Messenger ,Carcinoma, Ovarian Epithelial ,medicine.disease_cause ,Neoplasms ,Ovarian Epithelial ,Ovarian carcinoma ,2.1 Biological and endogenous factors ,Neoplasms, Glandular and Epithelial ,Aetiology ,Transversion ,Cancer ,Ovarian Neoplasms ,Mutation ,Tumor ,Glandular and Epithelial ,Genomics ,female genital diseases and pregnancy complications ,Ovarian Cancer ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Serous fluid ,Oncology ,Gene Knockdown Techniques ,Female ,Oncology and Carcinogenesis ,Cystadenocarcinoma ,Biology ,Article ,Cell Line ,Minor Histocompatibility Antigens ,Rare Diseases ,Cell Line, Tumor ,Cytidine Deaminase ,Genetics ,medicine ,Carcinoma ,Humans ,Oncology & Carcinogenesis ,RNA, Messenger ,Neoplastic ,Gene Expression Profiling ,Human Genome ,Serous ,medicine.disease ,Molecular biology ,Cystadenocarcinoma, Serous ,Gene Expression Regulation ,Kataegis ,Cancer research ,RNA ,Ovarian cancer - Abstract
Ovarian cancer is a clinically and molecularly heterogeneous disease. The driving forces behind this variability are unknown. Here, we report wide variation in the expression of the DNA cytosine deaminase APOBEC3B, with elevated expression in the majority of ovarian cancer cell lines (three SDs above the mean of normal ovarian surface epithelial cells) and high-grade primary ovarian cancers. APOBEC3B is active in the nucleus of several ovarian cancer cell lines and elicits a biochemical preference for deamination of cytosines in 5′-TC dinucleotides. Importantly, examination of whole-genome sequence from 16 ovarian cancers reveals that APOBEC3B expression correlates with total mutation load as well as elevated levels of transversion mutations. In particular, high APOBEC3B expression correlates with C-to-A and C-to-G transversion mutations within 5′-TC dinucleotide motifs in early-stage high-grade serous ovarian cancer genomes, suggesting that APOBEC3B-catalyzed genomic uracil lesions are further processed by downstream DNA "repair" enzymes including error-prone translesion polymerases. These data identify a potential role for APOBEC3B in serous ovarian cancer genomic instability. Cancer Res; 73(24); 7222–31. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
47. Phase 1 study of sorafenib in combination with bortezomib in patients with advanced malignancies
- Author
-
James R. Jett, Scott H. Kaufmann, Keith C. Bible, Shaji Kumar, Alex A. Adjei, Angelina Tan, Fernando Quevedo, Timothy J. Moynihan, Ronald L. Richardson, Svetomir N. Markovic, Randolph S. Marks, Charles Erlichman, Rui Qin, Gary A. Croghan, and Julian R. Molina
- Subjects
Adult ,Male ,Niacinamide ,Oncology ,Sorafenib ,medicine.medical_specialty ,Maximum Tolerated Dose ,Chronic lymphocytic leukemia ,Antineoplastic Agents ,Pharmacology ,Article ,Bortezomib ,Young Adult ,Neoplasms ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Pharmacology (medical) ,Protein Kinase Inhibitors ,neoplasms ,Multiple myeloma ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Phenylurea Compounds ,Melanoma ,Middle Aged ,medicine.disease ,Boronic Acids ,Treatment Outcome ,Pyrazines ,Proteasome inhibitor ,Vomiting ,Female ,medicine.symptom ,business ,Off Treatment ,medicine.drug - Abstract
Background. Sorafenib (a VEGFR and multi-targeted kinase inhibitor) and Bortezomib (a proteasome inhibitor) have clinical antineoplastic activities as single agents, and combine synergistically in preclinical models. Methods. This Phase I study was undertaken to define the toxicity and the maximum tolerated doses (MTD) of the combination in patients with advanced solid tumors. Patients with cytologic or histologic proof of unresectable solid tumors were treated with escalating doses of sorafenib (twice daily) and bortezomib (days 1, 4, 8 and 11 intravenously) with 21-day cycles. Results. Fourteen patients (7 males, median age 65, range 24–74), with renal (3), lung (3), pancreas (2), and breast, adrenal gland, melanoma, spindle cell tumor, chronic lymphocytic leukemia and multiple myeloma (1 each) were enrolled. All patients are off treatment, 10 due to disease progression. DLT was seen in two patients (one grade 3 abdominal pain and grade 4 lipase elevation; one with grade 3 vomiting) at sorafenib 200 mg twice daily and bortezomib 1.3 mg/m2, establishing the MTD. No grade 4 hematologic or grade 5 toxicities were seen. One patient with renal cell cancer had a partial response and 5 patients attained stable disease. Conclusions. The combination of sorafenib and bortezomib was tolerated well. The recommended phase 2 doses are sorafenib 200 mg twice daily continuously with bortezomib 1 mg/m2 on days 1, 4, 8, 11 (21 day cycles). The combination shows preliminary signs of efficacy, supporting phase 2 studies.
- Published
- 2013
- Full Text
- View/download PDF
48. 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
49. Maintenance of the HIV Reservoir Is Antagonized by Selective BCL2 Inhibition
- Author
-
Andrew D. Badley, Fatma Aboulnasr, Sekar Natesampillai, Scott H. Kaufmann, Nathan W. Cummins, and Amy M. Sainski-Nguyen
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Programmed cell death ,medicine.medical_treatment ,Immunology ,Apoptosis ,Biology ,Virus Replication ,Microbiology ,Jurkat cells ,03 medical and health sciences ,chemistry.chemical_compound ,Jurkat Cells ,Virology ,Virus latency ,medicine ,Humans ,Cells, Cultured ,Sulfonamides ,Protease ,Cell Death ,Venetoclax ,Bcl-2 family ,virus diseases ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Virus Latency ,030104 developmental biology ,chemistry ,Viral replication ,Proto-Oncogene Proteins c-bcl-2 ,Insect Science ,HIV-1 ,Pathogenesis and Immunity - Abstract
Decay of the HIV reservoir is slowed over time in part by expansion of the pool of HIV-infected cells. This expansion reflects homeostatic proliferation of infected cells by interleukin-7 (IL-7) or antigenic stimulation, as well as new rounds of infection of susceptible target cells. As novel therapies are being developed to accelerate the decay of the latent HIV reservoir, it will be important to identify interventions that prevent expansion and/or repopulation of the latent HIV reservoir. Our previous studies showed that HIV protease cleaves the host protein procaspase 8 to generate Casp8p41, which can bind and activate Bak to induce apoptosis of infected cells. In circumstances where expression of the anti-apoptotic protein BCL2 is high, Casp8p41 instead binds BCL2, and cell death does not occur. This effect can be overcome by treating cells with the clinically approved BCL2 antagonist venetoclax, which prevents Casp8p41 from binding BCL2, thereby allowing Casp8p41 to bind Bak and kill the infected cell. Here we assess whether the events that maintain the HIV reservoir are also antagonized by venetoclax. Using the J-Lat 10.6 model of persistent infection, we demonstrate that proliferation and HIV expression are countered by the use of venetoclax, which causes preferential killing of the HIV-expressing cells. Similarly, during new rounds of infection of primary CD4 T cells, venetoclax causes selective killing of HIV-infected cells, resulting in decreased numbers of HIV DNA-containing cells. IMPORTANCE Cure of HIV infection requires an intervention that reduces the HIV reservoir size. A variety of approaches are being tested for their ability to impact HIV reservoir size. Even if successful, however, these approaches will need to be combined with additional complementary approaches that prevent replenishment or repopulation of the HIV reservoir. Our previous studies have shown that the FDA-approved BCL2 antagonist venetoclax has a beneficial effect on the HIV reservoir size following HIV reactivation. Here we demonstrate that venetoclax also has a beneficial effect on HIV reservoir size in a model of homeostatic proliferation of HIV as well as in acute spreading infection of HIV in primary CD4 T cells. These results suggest that venetoclax, either alone or in combination with other approaches to reducing HIV reservoir size, is a compound worthy of further study for its effects on HIV reservoir size.
- Published
- 2017
50. Spartan deficiency causes accumulation of Topoisomerase 1 cleavage complexes and tumorigenesis
- Author
-
Hyun Ja Nam, Yusuke Kojima, Reeja S. Maskey, Thomas C. Smyrk, Annyoceli Santiago, Yuka Machida, Scott H. Kaufmann, Cynthia J. Sieben, Myoung Shin Kim, Yuichi J. Machida, Karen S. Flatten, Jan M. van Deursen, Darren J. Baker, and Kevin L. Peterson
- Subjects
Male ,0301 basic medicine ,Genome instability ,Carcinoma, Hepatocellular ,Carcinogenesis ,Chromosomal Proteins, Non-Histone ,DNA damage ,Gene Expression ,Genome Integrity, Repair and Replication ,Biology ,medicine.disease_cause ,DNA Adducts ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Progeria ,Germline mutation ,Genetics ,medicine ,Animals ,Humans ,Mice, Knockout ,Topoisomerase ,Liver Neoplasms ,Syndrome ,Aneuploidy ,medicine.disease ,Phenotype ,Molecular biology ,DNA-Binding Proteins ,Disease Models, Animal ,030104 developmental biology ,DNA Topoisomerases, Type I ,Liver ,chemistry ,Proteolysis ,Cancer research ,biology.protein ,Female ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,DNA - Abstract
Contains fulltext : 174445.pdf (Publisher’s version ) (Open Access) Germline mutations in SPRTN cause Ruijs-Aalfs syndrome (RJALS), a disorder characterized by genome instability, progeria and early onset hepatocellular carcinoma. Spartan, the protein encoded by SPRTN, is a nuclear metalloprotease that is involved in the repair of DNA-protein crosslinks (DPCs). Although Sprtn hypomorphic mice recapitulate key progeroid phenotypes of RJALS, whether this model expressing low amounts of Spartan is prone to DPC repair defects and spontaneous tumors is unknown. Here, we showed that the livers of Sprtn hypomorphic mice accumulate DPCs containing Topoisomerase 1 covalently linked to DNA. Furthermore, these mice exhibited DNA damage, aneuploidy and spontaneous tumorigenesis in the liver. Collectively, these findings provide evidence that partial loss of Spartan impairs DPC repair and tumor suppression.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.