49 results on '"Kaufmann, Scott H."'
Search Results
2. Abstract PR02: Proteogenomic approach to identify mechanisms of platinum refractoriness in high-grade serous ovarian cancers
- Author
-
Kennedy, Jacob J., primary, Chowdhury, Shrabanti, additional, Savage, Sara R., additional, Hou, Xiaonan, additional, Huntoon, Catherine J., additional, Ivey, Richard G., additional, Yu, Qing, additional, Lin, Chenwei, additional, Huang, Dongqing, additional, Zhao, Lei, additional, Voytovich, Uliana J., additional, Schoenherr, Regine M., additional, Shire, Zahra, additional, Skates, Steven J., additional, Whiteaker, Jeffrey R., additional, Hoofnagle, Andrew N., additional, Mok, Samuel C., additional, Zhang, Bing, additional, Karnitz, Larry M., additional, Weroha, S. John, additional, Gygi, Steven P., additional, Kaufmann, Scott H., additional, Wang, Pei, additional, Birrer, Michael J., additional, and Paulovich, Amanda G., additional
- Published
- 2020
- Full Text
- View/download PDF
3. Abstract 5885: Loss of RAD51C promoter hypermethylation confers PARP inhibitor resistance
- Author
-
Hurley, Rachel M., primary, Nesic, Ksenija, additional, McGehee, Cordelia, additional, Kondrashova, Olga, additional, Harrell, Maria I., additional, Schneider, Paula A., additional, Hou, Xiaonan, additional, Correia, Cristina, additional, Flatten, Karen S., additional, Zapparoli, Giada V., additional, Dobrovic, Alexander, additional, Lin, Kevin K., additional, Harding, Thomas C., additional, Hendrickson, Andrea E. Wahner, additional, Swisher, Elizabeth M., additional, Wakefield, Matthew, additional, Weroha, S. John, additional, Scott, Clare L., additional, and Kaufmann, Scott H., additional
- Published
- 2018
- Full Text
- View/download PDF
4. Abstract AP28:BRCA1andRAD51CPromoter Hypermethylation Confer Sensitivity to PARP Inhibitors in Patients with Platinum Sensitive Ovarian Carcinoma
- Author
-
Swisher, Elizabeth, primary, Harrell, Maria, additional, Lin, Kevin K., additional, Scott, Clare, additional, Goble, Sandra, additional, Oza, Amit, additional, Coleman, Robert L., additional, Konecny, Gottfried, additional, Tinker, Anna V., additional, O'Malley, David M., additional, Kristeleit, Rebecca, additional, Ma, Ling, additional, Brenton, James, additional, Bell-McGuinn, Katherine, additional, Oaknin, Ana, additional, Leary, Alexandra, additional, Mann, Elaina, additional, Giordano, Heidi, additional, Rapon, Mitch, additional, McNeish, Iain, additional, and Kaufmann, Scott H., additional
- Published
- 2017
- Full Text
- View/download PDF
5. Abstract IS04: PARP INHIBITOR COMBINATIONS FOR THE TREATMENT OF OVARIAN CANCER
- Author
-
Kaufmann, Scott H., primary, Hendrickson, Andrea E. Wahner, additional, Harrell, Maria I, additional, Menefee, Michael E., additional, Tanner, Edward J., additional, Mutch, David G, additional, Swisher, Elizabeth M., additional, and Karnitz, Larry M., additional
- Published
- 2017
- Full Text
- View/download PDF
6. Abstract 3479: CPX-351 (cytarabine:daunorubicin liposome for injection) anti-leukemia activity is potentiated by Chk1 inhibition
- Author
-
Vincelette, Nicole D., primary, Karnitz, Larry M., additional, Karp, Judith E., additional, Smith, Douglas B., additional, Hess, Allan D., additional, Mayer, Lawrence D., additional, and Kaufmann, Scott H., additional
- Published
- 2015
- Full Text
- View/download PDF
7. Abstract 38: Using molecularly characterized patient-derived models to delineate underlying drivers and vulnerabilities of epithelial ovarian cancer
- Author
-
Topp, Monique D., primary, Hartley, Lynne, additional, Cook, Michele, additional, Heong, Valerie, additional, Boehm, Emma, additional, McShane, Lauren, additional, Pyman, Jan, additional, McNally, Orla, additional, Ananda, Sumi, additional, Harell, Maria I., additional, Etemadmoghadam, Dariush, additional, Galletta, Laura, additional, Alsop, Kathryn, additional, Mitchell, Gillian, additional, Fox, Stephen B., additional, Kerr, Jeff B., additional, Hutt, Karla J., additional, Kaufmann, Scott H., additional, Swisher, Elizabeth M., additional, Bowtell, David D., additional, Wakefield, Matthew M., additional, and Scott, Clare L., additional
- Published
- 2014
- Full Text
- View/download PDF
8. ATR Inhibition Broadly Sensitizes Ovarian Cancer Cells to Chemotherapy Independent of BRCA Status.
- Author
-
Huntoon, Catherine J., Flatten, Karen S., Wahner Hendrickson, Andrea E., Huehls, Amelia M., Sutor, Shari L., Kaufmann, Scott H., and Karnitz, Larry M.
- Subjects
- *
CELLULAR signal transduction , *SERINE/THREONINE kinases , *OVARIAN cancer , *CANCER cells , *ANTINEOPLASTIC agents , *RNA interference , *PHOSPHORYLATION - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
9. Poly(ADP-Ribose) Polymerase Inhibition Synergizes with 5-Fluorodeoxyuridine but not 5-Fluorouracil in Ovarian Cancer Cells.
- Author
-
Huehls, Amelia M., Wagner, Jill M., Huntoon, Catherine J., Geng, Liyi, Erlichman, Charles, Patel, Anand G., Kaufmann, Scott H., and Karnitz, Larry M.
- Subjects
- *
OVARIAN cancer , *CANCER cells , *DNA damage , *CELL lines , *TUMORS , *RNA metabolism , *DNA metabolism - Abstract
5-Fluorouracil (5-FU) and 5-fluorodeoxyuridine (FdUrd, floxuridine) have activity in multiple tumors, and both agents undergo intracellular processing to active metabolites that disrupt RNA and DNA metabolism. These agents cause imbalances in deoxynucleotide triphosphate levels and the accumulation of uracil and 5-FU in the genome, events that activate the ATR- and ATM-dependent checkpoint signaling pathways and the base excision repair (BER) pathway. Here, we assessed which DNA damage response and repair processes influence 5- FU and FdUrd toxicity in ovarian cancer cells. These studies revealed that disabling the ATM, ATR, or BER pathways using small inhibitory RNAs did not affect 5-FU cytotoxicity. In stark contrast, ATR and a functional BER pathway protected FdUrd-treated cells. Consistent with a role for the BER pathway, the poly(ADP-ribose) polymerase (PARP) inhibitors ABT-888 (veliparib) and AZD2281 (olaparib) markedly synergized with FdUrd but not with 5-FU in ovarian cancer cell lines. Furthermore, ABT-888 synergized with FdUrd far more effectively than other agents commonly used to treat ovarian cancer. These findings underscore differences in the cytotoxic mechanisms of 5-FU and FdUrd and suggest that combining FdUrd and PARP inhibitors may be an innovative therapeutic strategy for ovarian tumors. Cancer Res; 71(14); 4944-54. ©2011 AACR. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
10. Concurrent RB1 Loss and BRCA Deficiency Predicts Enhanced Immunologic Response and Long-term Survival in Tubo-ovarian High-grade Serous Carcinoma.
- Author
-
Saner FAM, Takahashi K, Budden T, Pandey A, Ariyaratne D, Zwimpfer TA, Meagher NS, Fereday S, Twomey L, Pishas KI, Hoang T, Bolithon A, Traficante N, Alsop K, Christie EL, Kang EY, Nelson GS, Ghatage P, Lee CH, Riggan MJ, Alsop J, Beckmann MW, Boros J, Brand AH, Brooks-Wilson A, Carney ME, Coulson P, Courtney-Brooks M, Cushing-Haugen KL, Cybulski C, El-Bahrawy MA, Elishaev E, Erber R, Gayther SA, Gentry-Maharaj A, Gilks CB, Harnett PR, Harris HR, Hartmann A, Hein A, Hendley J, Hernandez BY, Jakubowska A, Jimenez-Linan M, Jones ME, Kaufmann SH, Kennedy CJ, Kluz T, Koziak JM, Kristjansdottir B, Le ND, Lener M, Lester J, Lubiński J, Mateoiu C, Orsulic S, Ruebner M, Schoemaker MJ, Shah M, Sharma R, Sherman ME, Shvetsov YB, Soong TR, Steed H, Sukumvanich P, Talhouk A, Taylor SE, Vierkant RA, Wang C, Widschwendter M, Wilkens LR, Winham SJ, Anglesio MS, Berchuck A, Brenton JD, Campbell I, Cook LS, Doherty JA, Fasching PA, Fortner RT, Goodman MT, Gronwald J, Huntsman DG, Karlan BY, Kelemen LE, Menon U, Modugno F, Pharoah PDP, Schildkraut JM, Sundfeldt K, Swerdlow AJ, Goode EL, DeFazio A, Köbel M, Ramus SJ, Bowtell DDL, and Garsed DW
- Subjects
- Humans, Female, Prognosis, Ubiquitin-Protein Ligases genetics, Neoplasm Grading, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Middle Aged, Germ-Line Mutation, Gene Expression Regulation, Neoplastic, Aged, Biomarkers, Tumor genetics, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Ovarian Neoplasms genetics, Ovarian Neoplasms mortality, Ovarian Neoplasms immunology, Ovarian Neoplasms pathology, BRCA2 Protein genetics, BRCA2 Protein deficiency, BRCA1 Protein genetics, BRCA1 Protein deficiency, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology, Cystadenocarcinoma, Serous mortality, Cystadenocarcinoma, Serous immunology, Retinoblastoma Binding Proteins genetics
- Abstract
Purpose: The purpose of this study was to evaluate RB1 expression and survival across ovarian carcinoma histotypes and how co-occurrence of BRCA1 or BRCA2 (BRCA) alterations and RB1 loss influences survival in tubo-ovarian high-grade serous carcinoma (HGSC)., Experimental Design: RB1 protein expression was classified by immunohistochemistry in ovarian carcinomas of 7,436 patients from the Ovarian Tumor Tissue Analysis consortium. We examined RB1 expression and germline BRCA status in a subset of 1,134 HGSC, and related genotype to overall survival (OS), tumor-infiltrating CD8+ lymphocytes, and transcriptomic subtypes. Using CRISPR-Cas9, we deleted RB1 in HGSC cells with and without BRCA1 alterations to model co-loss with treatment response. We performed whole-genome and transcriptome data analyses on 126 patients with primary HGSC to characterize tumors with concurrent BRCA deficiency and RB1 loss., Results: RB1 loss was associated with longer OS in HGSC but with poorer prognosis in endometrioid ovarian carcinoma. Patients with HGSC harboring both RB1 loss and pathogenic germline BRCA variants had superior OS compared with patients with either alteration alone, and their median OS was three times longer than those without pathogenic BRCA variants and retained RB1 expression (9.3 vs. 3.1 years). Enhanced sensitivity to cisplatin and paclitaxel was seen in BRCA1-altered cells with RB1 knockout. Combined RB1 loss and BRCA deficiency correlated with transcriptional markers of enhanced IFN response, cell-cycle deregulation, and reduced epithelial-mesenchymal transition. CD8+ lymphocytes were most prevalent in BRCA-deficient HGSC with co-loss of RB1., Conclusions: Co-occurrence of RB1 loss and BRCA deficiency was associated with exceptionally long survival in patients with HGSC, potentially due to better treatment response and immune stimulation., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
11. Trisomy 8 Defines a Distinct Subtype of Myeloproliferative Neoplasms Driven by the MYC-Alarmin Axis.
- Author
-
Vincelette ND, Yu X, Kuykendall AT, Moon J, Su S, Cheng CH, Sammut R, Razabdouski TN, Nguyen HV, Eksioglu EA, Chan O, Al Ali N, Patel PC, Lee DH, Nakanishi S, Ferreira RB, Hyjek E, Mo Q, Cory S, Lawrence HR, Zhang L, Murphy DJ, Komrokji RS, Lee D, Kaufmann SH, Cleveland JL, and Yun S
- Subjects
- Humans, Animals, Mice, Primary Myelofibrosis genetics, Primary Myelofibrosis pathology, Primary Myelofibrosis metabolism, Signal Transduction genetics, Chromosomes, Human, Pair 8 genetics, Trisomy genetics, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Calgranulin B genetics, Calgranulin B metabolism, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology
- Abstract
Despite advances in understanding the genetic abnormalities in myeloproliferative neoplasms (MPN) and the development of JAK2 inhibitors, there is an urgent need to devise new treatment strategies, particularly for patients with triple-negative (TN) myelofibrosis (MF) who lack mutations in the JAK2 kinase pathway and have very poor clinical outcomes. Here we report that MYC copy number gain and increased MYC expression frequently occur in TN-MF and that MYC-directed activation of S100A9, an alarmin protein that plays pivotal roles in inflammation and innate immunity, is necessary and sufficient to drive development and progression of MF. Notably, the MYC-S100A9 circuit provokes a complex network of inflammatory signaling that involves numerous hematopoietic cell types in the bone marrow microenvironment. Accordingly, genetic ablation of S100A9 or treatment with small molecules targeting the MYC-S100A9 pathway effectively ameliorates MF phenotypes, highlighting the MYC-alarmin axis as a novel therapeutic vulnerability for this subgroup of MPNs. Significance: This study establishes that MYC expression is increased in TN-MPNs via trisomy 8, that a MYC-S100A9 circuit manifest in these cases is sufficient to provoke myelofibrosis and inflammation in diverse hematopoietic cell types in the BM niche, and that the MYC-S100A9 circuit is targetable in TN-MPNs., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
12. Targeting LRRC15 Inhibits Metastatic Dissemination of Ovarian Cancer.
- Author
-
Ray U, Jung DB, Jin L, Xiao Y, Dasari S, Sarkar Bhattacharya S, Thirusangu P, Staub JK, Roy D, Roy B, Weroha SJ, Hou X, Purcell JW, Bakkum-Gamez JN, Kaufmann SH, Kannan N, Mitra AK, and Shridhar V
- Subjects
- Carcinoma, Ovarian Epithelial, Cell Adhesion, Cell Line, Tumor, Female, Humans, Integrins, Membrane Proteins genetics, Membrane Proteins metabolism, Immunoconjugates pharmacology, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology
- 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., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
13. Repurposing Ceritinib Induces DNA Damage and Enhances PARP Inhibitor Responses in High-Grade Serous Ovarian Carcinoma.
- Author
-
Kanakkanthara A, Hou X, Ekstrom TL, Zanfagnin V, Huehls AM, Kelly RL, Ding H, Larson MC, Vasmatzis G, Oberg AL, Kaufmann SH, Mansfield AS, Weroha SJ, and Karnitz LM
- Subjects
- Animals, Carcinoma, Ovarian Epithelial pathology, Drug Resistance, Neoplasm drug effects, Drug Synergism, Female, Humans, Mice, Mice, SCID, Ovarian Neoplasms pathology, PC-3 Cells, Recombinational DNA Repair drug effects, Treatment Outcome, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents administration & dosage, Carcinoma, Ovarian Epithelial drug therapy, Carcinoma, Ovarian Epithelial metabolism, DNA Damage drug effects, Drug Repositioning methods, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Phthalazines administration & dosage, Piperazines administration & dosage, Poly(ADP-ribose) Polymerase Inhibitors administration & dosage, Protein Kinase Inhibitors administration & dosage, Pyrimidines administration & dosage, Sulfones administration & dosage
- Abstract
PARP inhibitors (PARPi) have activity in homologous recombination (HR) repair-deficient, high-grade serous ovarian cancers (HGSOC). However, even responsive tumors develop PARPi resistance, highlighting the need to delay or prevent the appearance of PARPi resistance. Here, we showed that the ALK kinase inhibitor ceritinib synergizes with PARPis by inhibiting complex I of the mitochondrial electron transport chain, which increases production of reactive oxygen species (ROS) and subsequent induction of oxidative DNA damage that is repaired in a PARP-dependent manner. In addition, combined treatment with ceritinib and PARPi synergized in HGSOC cell lines irrespective of HR status, and a combination of ceritinib with the PARPi olaparib induced tumor regression more effectively than olaparib alone in HGSOC patient-derived xenograft (PDX) models. Notably, the ceritinib and olaparib combination was most effective in PDX models with preexisting PARPi sensitivity and was well tolerated. These findings unveil suppression of mitochondrial respiration, accumulation of ROS, and subsequent induction of DNA damage as novel effects of ceritinib. They also suggest that the ceritinib and PARPi combination warrants further investigation as a means to enhance PARPi activity in HGSOC, particularly in tumors with preexisting HR defects. SIGNIFICANCE: The kinase inhibitor ceritinib synergizes with PARPi to induce tumor regression in ovarian cancer models, suggesting that ceritinib combined with PARPi may be an effective strategy for treating ovarian cancer., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
14. Acquired RAD51C Promoter Methylation Loss Causes PARP Inhibitor Resistance in High-Grade Serous Ovarian Carcinoma.
- Author
-
Nesic K, Kondrashova O, Hurley RM, McGehee CD, Vandenberg CJ, Ho GY, Lieschke E, Dall G, Bound N, Shield-Artin K, Radke M, Musafer A, Chai ZQ, Ghamsari MRE, Harrell MI, Kee D, Olesen I, McNally O, Traficante N, Australian Ovarian Cancer Study, DeFazio A, Bowtell DDL, Swisher EM, Weroha SJ, Nones K, Waddell N, Kaufmann SH, Dobrovic A, Wakefield MJ, and Scott CL
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Computational Biology, Cystadenocarcinoma, Serous drug therapy, Cystadenocarcinoma, Serous metabolism, Cystadenocarcinoma, Serous pathology, DNA-Binding Proteins metabolism, Disease Models, Animal, Female, Gene Expression Profiling, Gene Silencing, Homozygote, Humans, Mice, Neoplasm Grading, Neoplasm Staging, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Prognosis, Xenograft Model Antitumor Assays, Cystadenocarcinoma, Serous genetics, DNA Methylation, DNA-Binding Proteins genetics, Drug Resistance, Neoplasm genetics, Ovarian Neoplasms genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Promoter Regions, Genetic
- Abstract
In high-grade serous ovarian carcinoma (HGSC), deleterious mutations in DNA repair gene RAD51C are established drivers of defective homologous recombination and are emerging biomarkers of PARP inhibitor (PARPi) sensitivity. RAD51C promoter methylation (me RAD51C ) is detected at similar frequencies to mutations, yet its effects on PARPi responses remain unresolved.In this study, three HGSC patient-derived xenograft (PDX) models with methylation at most or all examined CpG sites in the RAD51C promoter show responses to PARPi. Both complete and heterogeneous methylation patterns were associated with RAD51C gene silencing and homologous recombination deficiency (HRD). PDX models lost me RAD51C following treatment with PARPi rucaparib or niraparib, where a single unmethylated copy of RAD51C was sufficient to drive PARPi resistance. Genomic copy number profiling of one of the PDX models using SNP arrays revealed that this resistance was acquired independently in two genetically distinct lineages.In a cohort of 12 patients with RAD51C -methylated HGSC, various patterns of me RAD51C were associated with genomic "scarring," indicative of HRD history, but exhibited no clear correlations with clinical outcome. Differences in methylation stability under treatment pressure were also observed between patients, where one HGSC was found to maintain me RAD51C after six lines of therapy (four platinum-based), whereas another HGSC sample was found to have heterozygous me RAD51C and elevated RAD51C gene expression (relative to homozygous me RAD51C controls) after only neoadjuvant chemotherapy.As me RAD51C loss in a single gene copy was sufficient to cause PARPi resistance in PDX, methylation zygosity should be carefully assessed in previously treated patients when considering PARPi therapy. SIGNIFICANCE: Homozygous RAD51C methylation is a positive predictive biomarker for sensitivity to PARP inhibitors, whereas a single unmethylated gene copy is sufficient to confer resistance., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
15. CDK2-Mediated Upregulation of TNFα as a Mechanism of Selective Cytotoxicity in Acute Leukemia.
- Author
-
Ding H, Vincelette ND, McGehee CD, Kohorst MA, Koh BD, Venkatachalam A, Meng XW, Schneider PA, Flatten KS, Peterson KL, Correia C, Lee SH, Patnaik M, Webster JA, Ghiaur G, Smith BD, Karp JE, Pratz KW, Li H, Karnitz LM, and Kaufmann SH
- Subjects
- Animals, Apoptosis, Cell Proliferation, Female, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Mice, Mice, Inbred NOD, Mice, SCID, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Tumor Cells, Cultured, Tumor Necrosis Factor-alpha genetics, Xenograft Model Antitumor Assays, Cyclin-Dependent Kinase 2 antagonists & inhibitors, Gene Expression Regulation, Neoplastic drug effects, Leukemia, Myeloid, Acute drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Pyrazines pharmacology, Pyrazoles pharmacology, Tumor Necrosis Factor-alpha metabolism
- Abstract
Although inhibitors of the kinases CHK1, ATR, and WEE1 are undergoing clinical testing, it remains unclear how these three classes of agents kill susceptible cells and whether they utilize the same cytotoxic mechanism. Here we observed that CHK1 inhibition induces apoptosis in a subset of acute leukemia cell lines in vitro , including TP53 -null acute myeloid leukemia (AML) and BCR/ABL-positive acute lymphoid leukemia (ALL), and inhibits leukemic colony formation in clinical AML samples ex vivo . In further studies, downregulation or inhibition of CHK1 triggered signaling in sensitive human acute leukemia cell lines that involved CDK2 activation followed by AP1-dependent TNF transactivation, TNFα production, and engagement of a TNFR1- and BID-dependent apoptotic pathway. AML lines that were intrinsically resistant to CHK1 inhibition exhibited high CHK1 expression and were sensitized by CHK1 downregulation. Signaling through this same CDK2-AP1- TNF cytotoxic pathway was also initiated by ATR or WEE1 inhibitors in vitro and during CHK1 inhibitor treatment of AML xenografts in vivo . Collectively, these observations not only identify new contributors to the antileukemic cell action of CHK1, ATR, and WEE1 inhibitors, but also delineate a previously undescribed pathway leading from aberrant CDK2 activation to death ligand-induced killing that can potentially be exploited for acute leukemia treatment. SIGNIFICANCE: This study demonstrates that replication checkpoint inhibitors can kill AML cells through a pathway involving AP1-mediated TNF gene activation and subsequent TP53-independent, TNFα-induced apoptosis, which can potentially be exploited clinically., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
16. TFEB links MYC signaling to epigenetic control of myeloid differentiation and acute myeloid leukemia.
- Author
-
Yun S, Vincelette ND, Yu X, Watson GW, Fernandez MR, Yang C, Hitosugi T, Cheng CH, Freischel AR, Zhang L, Li W, Hou H, Schaub FX, Vedder AR, Cen L, McGraw KL, Moon J, Murphy DJ, Ballabio A, Kaufmann SH, Berglund AE, and Cleveland JL
- Subjects
- Cell Differentiation genetics, Epigenesis, Genetic, Humans, Mutation, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute genetics, Proto-Oncogene Proteins c-myc genetics, Signal Transduction
- Abstract
MYC oncoproteins regulate transcription of genes directing cell proliferation, metabolism and tumorigenesis. A variety of alterations drive MYC expression in acute myeloid leukemia (AML) and enforced MYC expression in hematopoietic progenitors is sufficient to induce AML. Here we report that AML and myeloid progenitor cell growth and survival rely on MYC-directed suppression of Transcription Factor EB (TFEB), a master regulator of the autophagy-lysosome pathway. Notably, although originally identified as an oncogene, TFEB functions as a tumor suppressor in AML, where it provokes AML cell differentiation and death. These responses reflect TFEB control of myeloid epigenetic programs, by inducing expression of isocitrate dehydrogenase-1 (IDH1) and IDH2, resulting in global hydroxylation of 5-methycytosine. Finally, activating the TFEB-IDH1/IDH2-TET2 axis is revealed as a targetable vulnerability in AML. Thus, epigenetic control by a MYC-TFEB circuit dictates myeloid cell fate and is essential for maintenance of AML.
- Published
- 2021
- Full Text
- View/download PDF
17. Development and Validation of the Gene Expression Predictor of High-grade Serous Ovarian Carcinoma Molecular SubTYPE (PrOTYPE).
- Author
-
Talhouk A, George J, Wang C, Budden T, Tan TZ, Chiu DS, Kommoss S, Leong HS, Chen S, Intermaggio MP, Gilks B, Nazeran TM, Volchek M, Elatre W, Bentley RC, Senz J, Lum A, Chow V, Sudderuddin H, Mackenzie R, Leong SCY, Liu G, Johnson D, Chen B, Group A, Alsop J, Banerjee SN, Behrens S, Bodelon C, Brand AH, Brinton L, Carney ME, Chiew YE, Cushing-Haugen KL, Cybulski C, Ennis D, Fereday S, Fortner RT, García-Donas J, Gentry-Maharaj A, Glasspool R, Goranova T, Greene CS, Haluska P, Harris HR, Hendley J, Hernandez BY, Herpel E, Jimenez-Linan M, Karpinskyj C, Kaufmann SH, Keeney GL, Kennedy CJ, Köbel M, Koziak JM, Larson MC, Lester J, Lewsley LA, Lissowska J, Lubiński J, Luk H, Macintyre G, Mahner S, McNeish IA, Menkiszak J, Nevins N, Osorio A, Oszurek O, Palacios J, Hinsley S, Pearce CL, Pike MC, Piskorz AM, Ray-Coquard I, Rhenius V, Rodriguez-Antona C, Sharma R, Sherman ME, De Silva D, Singh N, Sinn P, Slamon D, Song H, Steed H, Stronach EA, Thompson PJ, Tołoczko A, Trabert B, Traficante N, Tseng CC, Widschwendter M, Wilkens LR, Winham SJ, Winterhoff B, Beeghly-Fadiel A, Benitez J, Berchuck A, Brenton JD, Brown R, Chang-Claude J, Chenevix-Trench G, deFazio A, Fasching PA, García MJ, Gayther SA, Goodman MT, Gronwald J, Henderson MJ, Karlan BY, Kelemen LE, Menon U, Orsulic S, Pharoah PDP, Wentzensen N, Wu AH, Schildkraut JM, Rossing MA, Konecny GE, Huntsman DG, Huang RY, Goode EL, Ramus SJ, Doherty JA, Bowtell DD, and Anglesio MS
- Subjects
- Aged, Algorithms, Cystadenoma, Serous classification, Cystadenoma, Serous pathology, Female, Gene Expression Regulation, Neoplastic genetics, Humans, Lymphocytes, Tumor-Infiltrating pathology, Middle Aged, Neoplasm Grading, Neoplasm, Residual classification, Neoplasm, Residual genetics, Neoplasm, Residual pathology, Ovarian Neoplasms classification, Ovarian Neoplasms pathology, Cystadenoma, Serous genetics, Neoplasm Proteins genetics, Ovarian Neoplasms genetics, Transcriptome genetics
- 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 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
18. The DNA Cytosine Deaminase APOBEC3B is a Molecular Determinant of Platinum Responsiveness in Clear Cell Ovarian Cancer.
- Author
-
Serebrenik AA, Argyris PP, Jarvis MC, Brown WL, Bazzaro M, Vogel RI, Erickson BK, Lee SH, Goergen KM, Maurer MJ, Heinzen EP, Oberg AL, Huang Y, Hou X, Weroha SJ, Kaufmann SH, and Harris RS
- Subjects
- Animals, Antineoplastic Agents pharmacology, Biomarkers, Tumor, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Cytidine Deaminase genetics, Disease Models, Animal, Drug Resistance, Neoplasm, Female, Gene Expression, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Mice, Minor Histocompatibility Antigens genetics, Ovarian Neoplasms drug therapy, Ovarian Neoplasms mortality, Ovarian Neoplasms pathology, Prognosis, Synthetic Lethal Mutations drug effects, Synthetic Lethal Mutations genetics, Xenograft Model Antitumor Assays, Cytidine Deaminase metabolism, Minor Histocompatibility Antigens metabolism, Ovarian Neoplasms metabolism, Platinum pharmacology
- 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., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
19. Anastrozole has an Association between Degree of Estrogen Suppression and Outcomes in Early Breast Cancer and is a Ligand for Estrogen Receptor α.
- Author
-
Ingle JN, Cairns J, Suman VJ, Shepherd LE, Fasching PA, Hoskin TL, Singh RJ, Desta Z, Kalari KR, Ellis MJ, Goss PE, Chen BE, Volz B, Barman P, Carlson EE, Haddad T, Goetz MP, Goodnature B, Cuellar ME, Walters MA, Correia C, Kaufmann SH, Weinshilboum RM, and Wang L
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Case-Control Studies, Clinical Trials, Phase III as Topic, Clinical Trials, Phase IV as Topic, Female, Follow-Up Studies, Humans, Middle Aged, Multicenter Studies as Topic, Prognosis, Prospective Studies, Randomized Controlled Trials as Topic, Anastrozole therapeutic use, Breast Neoplasms pathology, Estrogen Receptor alpha metabolism, Estrogens metabolism
- 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., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
20. Rare BRIP1 Missense Alleles Confer Risk for Ovarian and Breast Cancer.
- Author
-
Moyer CL, Ivanovich J, Gillespie JL, Doberstein R, Radke MR, Richardson ME, Kaufmann SH, Swisher EM, and Goodfellow PJ
- Subjects
- Adult, Age of Onset, Aged, Alleles, Breast Neoplasms blood, Breast Neoplasms diagnosis, Cohort Studies, DNA Mutational Analysis, Datasets as Topic, Female, Gene Frequency, Genetic Testing, Germ-Line Mutation, HeLa Cells, High-Throughput Nucleotide Sequencing, Humans, Loss of Function Mutation, Middle Aged, Mutation, Missense, Ovarian Neoplasms blood, Ovarian Neoplasms diagnosis, Breast Neoplasms genetics, Fanconi Anemia Complementation Group Proteins genetics, Genetic Predisposition to Disease, Ovarian Neoplasms genetics, RNA Helicases genetics
- 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, χ
2 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., (©2019 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
21. BRCA1 Deficiency Upregulates NNMT, Which Reprograms Metabolism and Sensitizes Ovarian Cancer Cells to Mitochondrial Metabolic Targeting Agents.
- Author
-
Kanakkanthara A, Kurmi K, Ekstrom TL, Hou X, Purfeerst ER, Heinzen EP, Correia C, Huntoon CJ, O'Brien D, Wahner Hendrickson AE, Dowdy SC, Li H, Oberg AL, Hitosugi T, Kaufmann SH, Weroha SJ, and Karnitz LM
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, BRCA1 Protein deficiency, Carcinoma, Ovarian Epithelial genetics, Carcinoma, Ovarian Epithelial pathology, Cell Line, Tumor, Cyclin-Dependent Kinases genetics, DNA Methylation, Energy Metabolism drug effects, Female, Gene Expression Regulation, Neoplastic, Humans, Hydrazones pharmacology, Hydrazones therapeutic use, Hydroxybenzoates pharmacology, Hydroxybenzoates therapeutic use, Mice, Mitochondria drug effects, Mitochondria metabolism, Mutation, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Ovary pathology, Oxidative Phosphorylation drug effects, Promoter Regions, Genetic genetics, Tigecycline pharmacology, Tigecycline therapeutic use, Triazoles pharmacology, Triazoles therapeutic use, Up-Regulation, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, BRCA1 Protein genetics, Carcinoma, Ovarian Epithelial drug therapy, Nicotinamide N-Methyltransferase metabolism, Ovarian Neoplasms drug therapy
- Abstract
BRCA1 plays a key role in homologous recombination (HR) DNA repair. Accordingly, changes that downregulate BRCA1, including BRCA1 mutations and reduced BRCA1 transcription, due to promoter hypermethylation or loss of the BRCA1 transcriptional regulator CDK12, disrupt HR in multiple cancers. In addition, BRCA1 has also been implicated in the regulation of metabolism. Here, we show that reducing BRCA1 expression, either by CDK12 or BRCA1 depletion, led to metabolic reprogramming of ovarian cancer cells, causing decreased mitochondrial respiration and reduced ATP levels. BRCA1 depletion drove this reprogramming by upregulating nicotinamide N-methyltransferase (NNMT). Notably, the metabolic alterations caused by BRCA1 depletion and NNMT upregulation sensitized ovarian cancer cells to agents that inhibit mitochondrial metabolism (VLX600 and tigecycline) and to agents that inhibit glucose import (WZB117). These observations suggest that inhibition of energy metabolism may be a potential strategy to selectively target BRCA1-deficient high-grade serous ovarian cancer, which is characterized by frequent BRCA1 loss and NNMT overexpression. SIGNIFICANCE: Loss of BRCA1 reprograms metabolism, creating a therapeutically targetable vulnerability in ovarian cancer., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
22. BRCA Reversion Mutations in Circulating Tumor DNA Predict Primary and Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma.
- Author
-
Lin KK, Harrell MI, Oza AM, Oaknin A, Ray-Coquard I, Tinker AV, Helman E, Radke MR, Say C, Vo LT, Mann E, Isaacson JD, Maloney L, O'Malley DM, Chambers SK, Kaufmann SH, Scott CL, Konecny GE, Coleman RL, Sun JX, Giordano H, Brenton JD, Harding TC, McNeish IA, and Swisher EM
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor genetics, Carcinoma, Ovarian Epithelial drug therapy, Carcinoma, Ovarian Epithelial genetics, Circulating Tumor DNA drug effects, Female, Follow-Up Studies, Humans, International Agencies, Middle Aged, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Prognosis, Survival Rate, BRCA1 Protein genetics, BRCA2 Protein genetics, Carcinoma, Ovarian Epithelial pathology, Circulating Tumor DNA genetics, Drug Resistance, Neoplasm genetics, Indoles therapeutic use, Mutation
- 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 ., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
23. A Phase I Clinical Trial of the Poly(ADP-ribose) Polymerase Inhibitor Veliparib and Weekly Topotecan in Patients with Solid Tumors.
- Author
-
Wahner Hendrickson AE, Menefee ME, Hartmann LC, Long HJ, Northfelt DW, Reid JM, Boakye-Agyeman F, Kayode O, Flatten KS, Harrell MI, Swisher EM, Poirier GG, Satele D, Allred J, Lensing JL, Chen A, Ji J, Zang Y, Erlichman C, Haluska P, and Kaufmann SH
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols, Area Under Curve, Benzimidazoles administration & dosage, Benzimidazoles adverse effects, Benzimidazoles pharmacokinetics, Disease-Free Survival, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Germ-Line Mutation, Humans, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear metabolism, Male, Middle Aged, Neoplasms genetics, Neoplasms metabolism, Neutropenia chemically induced, Poly(ADP-ribose) Polymerase Inhibitors administration & dosage, Poly(ADP-ribose) Polymerase Inhibitors adverse effects, Poly(ADP-ribose) Polymerase Inhibitors pharmacokinetics, Topotecan administration & dosage, Topotecan adverse effects, Topotecan pharmacokinetics, Neoplasms drug therapy
- 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/m
2 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. Clin Cancer Res; 24(4); 744-52. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
24. Secondary Somatic Mutations Restoring RAD51C and RAD51D Associated with Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma.
- Author
-
Kondrashova O, Nguyen M, Shield-Artin K, Tinker AV, Teng NNH, Harrell MI, Kuiper MJ, Ho GY, Barker H, Jasin M, Prakash R, Kass EM, Sullivan MR, Brunette GJ, Bernstein KA, Coleman RL, Floquet A, Friedlander M, Kichenadasse G, O'Malley DM, Oza A, Sun J, Robillard L, Maloney L, Bowtell D, Giordano H, Wakefield MJ, Kaufmann SH, Simmons AD, Harding TC, Raponi M, McNeish IA, Swisher EM, Lin KK, and Scott CL
- Subjects
- Animals, CHO Cells, Cell Line, Tumor, Cricetulus, Female, HEK293 Cells, Humans, Mutation, Ovarian Neoplasms genetics, DNA-Binding Proteins genetics, Drug Resistance, Neoplasm genetics, Indoles therapeutic use, Ovarian Neoplasms drug therapy, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use
- Abstract
High-grade epithelial ovarian carcinomas containing mutated BRCA1 or BRCA2 ( BRCA1/2 ) homologous recombination (HR) genes are sensitive to platinum-based chemotherapy and PARP inhibitors (PARPi), while restoration of HR function due to secondary mutations in BRCA1/2 has been recognized as an important resistance mechanism. We sequenced core HR pathway genes in 12 pairs of pretreatment and postprogression tumor biopsy samples collected from patients in ARIEL2 Part 1, a phase II study of the PARPi rucaparib as treatment for platinum-sensitive, relapsed ovarian carcinoma. In 6 of 12 pretreatment biopsies, a truncation mutation in BRCA1, RAD51C , or RAD51D was identified. In five of six paired postprogression biopsies, one or more secondary mutations restored the open reading frame. Four distinct secondary mutations and spatial heterogeneity were observed for RAD51C In vitro complementation assays and a patient-derived xenograft, as well as predictive molecular modeling, confirmed that resistance to rucaparib was associated with secondary mutations. Significance: Analyses of primary and secondary mutations in RAD51C and RAD51D provide evidence for these primary mutations in conferring PARPi sensitivity and secondary mutations as a mechanism of acquired PARPi resistance. PARPi resistance due to secondary mutations underpins the need for early delivery of PARPi therapy and for combination strategies. Cancer Discov; 7(9); 984-98. ©2017 AACR. See related commentary by Domchek, p. 937 See related article by Quigley et al., p. 999 See related article by Goodall et al., p. 1006 This article is highlighted in the In This Issue feature, p. 920 ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
25. Pooled Clustering of High-Grade Serous Ovarian Cancer Gene Expression Leads to Novel Consensus Subtypes Associated with Survival and Surgical Outcomes.
- Author
-
Wang C, Armasu SM, Kalli KR, Maurer MJ, Heinzen EP, Keeney GL, Cliby WA, Oberg AL, Kaufmann SH, and Goode EL
- Subjects
- Adult, Aged, Aged, 80 and over, Cystadenocarcinoma, Serous drug therapy, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous surgery, Disease-Free Survival, Female, Gene Expression Regulation, Neoplastic, Humans, Middle Aged, Neoadjuvant Therapy, Neoplasm Grading, Neoplasm, Residual drug therapy, Neoplasm, Residual genetics, Neoplasm, Residual surgery, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms surgery, Prognosis, Treatment Outcome, Cystadenocarcinoma, Serous pathology, Neoplasm Proteins genetics, Neoplasm, Residual pathology, Ovarian Neoplasms pathology
- 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 ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
26. A Phase I Study of Topotecan, Carboplatin and the PARP Inhibitor Veliparib in Acute Leukemias, Aggressive Myeloproliferative Neoplasms, and Chronic Myelomonocytic Leukemia.
- Author
-
Pratz KW, Rudek MA, Gojo I, Litzow MR, McDevitt MA, Ji J, Karnitz LM, Herman JG, Kinders RJ, Smith BD, Gore SD, Carraway HE, Showel MM, Gladstone DE, Levis MJ, Tsai HL, Rosner G, Chen A, Kaufmann SH, and Karp JE
- Subjects
- Adult, Aged, Benzimidazoles administration & dosage, Benzimidazoles adverse effects, Benzimidazoles pharmacokinetics, Carboplatin administration & dosage, Carboplatin adverse effects, Carboplatin pharmacokinetics, Disease-Free Survival, Dose-Response Relationship, Drug, Drug-Related Side Effects and Adverse Reactions pathology, Fanconi Anemia Complementation Group D2 Protein genetics, Female, Humans, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic pathology, Male, Middle Aged, Poly (ADP-Ribose) Polymerase-1 genetics, Topotecan administration & dosage, Topotecan adverse effects, Topotecan pharmacokinetics, Leukemia, Biphenotypic, Acute drug therapy, Leukemia, Myelomonocytic, Chronic drug therapy, Myeloproliferative Disorders drug therapy, Poly (ADP-Ribose) Polymerase-1 antagonists & inhibitors
- Abstract
Purpose: The PARP inhibitor veliparib delays DNA repair and potentiates cytotoxicity of multiple classes of chemotherapy drugs, including topoisomerase I inhibitors and platinating agents. This study evaluated veliparib incorporation into leukemia induction therapy using a previously described topotecan/carboplatin backbone. Experimental Design: Employing a 3+3 trial design, we administered escalating doses of veliparib combined with topotecan + carboplatin in relapsed or refractory acute leukemias, aggressive myeloproliferative neoplasms (MPN), and chronic myelomonocytic leukemia (CMML). Results: A total of 99 patients received veliparib 10-100 mg orally twice daily on days 1-8, 1-14, or 1-21 along with continuous infusion topotecan 1.0-1.2 mg/m
2 /d + carboplatin 120-150 mg/m2 /d on days 3-7. The MTD was veliparib 80 mg twice daily for up to 21 days with topotecan 1.2 mg/m2 /d + carboplatin 150 mg/m2 /d. Mucositis was dose limiting and correlated with high veliparib concentrations. The response rate was 33% overall (33/99: 14 CR, 11 CRi, 8 PR) but was 64% (14/22) for patients with antecedent or associated aggressive MPNs or CMML. Leukemias with baseline DNA repair defects, as evidenced by impaired DNA damage-induced FANCD2 monoubiquitination, had improved survival [HR = 0.56 (95% confidence interval, 0.27-0.92)]. A single 80-mg dose of veliparib, as well as veliparib in combination with topotecan + carboplatin, induced DNA damage as manifested by histone H2AX phosphorylation in CD34+ leukemia cells, with greater phosphorylation in cells from responders. Conclusions: The veliparib/topotecan/carboplatin combination warrants further investigation, particularly in patients with aggressive MPNs, CMML, and MPN- or CMML-related acute leukemias. Clin Cancer Res; 23(4); 899-907. ©2016 AACR ., (©2016 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
27. A Phase 1 Study of the PARP Inhibitor Veliparib in Combination with Temozolomide in Acute Myeloid Leukemia.
- Author
-
Gojo I, Beumer JH, Pratz KW, McDevitt MA, Baer MR, Blackford AL, Smith BD, Gore SD, Carraway HE, Showel MM, Levis MJ, Dezern AE, Gladstone DE, Ji JJ, Wang L, Kinders RJ, Pouquet M, Ali-Walbi I, Rudek MA, Poh W, Herman JG, Karnitz LM, Kaufmann SH, Chen A, and Karp JE
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacology, Benzimidazoles administration & dosage, Benzimidazoles adverse effects, Benzimidazoles pharmacology, DNA Methylation drug effects, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Dacarbazine administration & dosage, Dacarbazine adverse effects, Dacarbazine analogs & derivatives, Dacarbazine pharmacology, Drug Synergism, Esophagitis chemically induced, Female, Histones metabolism, Humans, Kaplan-Meier Estimate, Leukemia, Myelomonocytic, Chronic drug therapy, Male, Middle Aged, Mucositis chemically induced, Neoplasm Proteins analysis, Neoplasm Proteins antagonists & inhibitors, Phosphorylation drug effects, Poly (ADP-Ribose) Polymerase-1 analysis, Poly(ADP-ribose) Polymerase Inhibitors administration & dosage, Poly(ADP-ribose) Polymerase Inhibitors adverse effects, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Promoter Regions, Genetic drug effects, Promoter Regions, Genetic genetics, Protein Processing, Post-Translational drug effects, Remission Induction, Salvage Therapy, Temozolomide, Tumor Suppressor Proteins genetics, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
Purpose: In preclinical studies, the PARP inhibitor veliparib enhanced the antileukemic action of temozolomide through potentiation of DNA damage. Accordingly, we conducted a phase 1 study of temozolomide with escalating doses of veliparib in patients with relapsed, refractory acute myeloid leukemia (AML) or AML arising from aggressive myeloid malignancies., Experimental Design: Patients received veliparib [20-200 mg once a day on day 1 and twice daily on days 4-12 in cycle 1 (days 1-8 in cycle ≥2)] and temozolomide [150-200 mg/m
2 daily on days 3-9 in cycle 1 (days 1-5 in cycle ≥2)] every 28 to 56 days. Veliparib pharmacokinetics and pharmacodynamics [ability to inhibit poly(ADP-ribose) polymer (PAR) formation and induce H2AX phosphorylation] were assessed. Pretreatment levels of MGMT and PARP1 protein, methylation of the MGMT promoter, and integrity of the Fanconi anemia pathway were also examined., Results: Forty-eight patients were treated at seven dose levels. Dose-limiting toxicities were oral mucositis and esophagitis lasting >7 days. The MTD was veliparib 150 mg twice daily with temozolomide 200 mg/m2 daily. The complete response (CR) rate was 17% (8/48 patients). Veliparib exposure as well as inhibition of PAR polymer formation increased dose proportionately. A veliparib-induced increase in H2AX phosphorylation in CD34+ cells was observed in responders. Three of 4 patients with MGMT promoter methylation achieved CR., Conclusions: Veliparib plus temozolomide is well tolerated, with activity in advanced AML. Further evaluation of this regimen and of treatment-induced phosphorylation of H2AX and MGMT methylation as potential response predictors appears warranted. Clin Cancer Res; 23(3); 697-706. ©2016 AACR., Competing Interests: The authors of this manuscript report no relationship to disclose. The data in this manuscript were in part presented in the abstract/poster form at the American Society of Hematology Meeting, December 5–8, 2015, Orlando, FL, (©2016 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
28. APOBEC3G Expression Correlates with T-Cell Infiltration and Improved Clinical Outcomes in High-grade Serous Ovarian Carcinoma.
- Author
-
Leonard B, Starrett GJ, Maurer MJ, Oberg AL, Van Bockstal M, Van Dorpe J, De Wever O, Helleman J, Sieuwerts AM, Berns EM, Martens JW, Anderson BD, Brown WL, Kalli KR, Kaufmann SH, and Harris RS
- Subjects
- APOBEC-3G Deaminase metabolism, Biomarkers, Tumor, Cohort Studies, Cystadenocarcinoma, Serous mortality, Cystadenocarcinoma, Serous pathology, Female, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Lymphocyte Activation, Neoplasm Grading, Ovarian Neoplasms mortality, Ovarian Neoplasms pathology, Prognosis, Proportional Hazards Models, APOBEC-3G Deaminase genetics, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous immunology, Gene Expression, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Ovarian Neoplasms genetics, Ovarian Neoplasms immunology
- 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., Competing Interests: R.S.H. is a co-founder of ApoGen Biotechnologies Inc. The other authors have no conflicts of interest to disclose., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
29. Poly (ADP-Ribose) Polymerase Inhibitor Hypersensitivity in Aggressive Myeloproliferative Neoplasms.
- Author
-
Pratz KW, Koh BD, Patel AG, Flatten KS, Poh W, Herman JG, Dilley R, Harrell MI, Smith BD, Karp JE, Swisher EM, McDevitt MA, and Kaufmann SH
- Subjects
- Antineoplastic Agents therapeutic use, BRCA1 Protein genetics, Benzimidazoles adverse effects, Benzimidazoles pharmacology, DNA Damage, DNA Methylation, DNA Repair, Drug Tolerance genetics, Genomics methods, Humans, Janus Kinase 2 genetics, Mutation, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Antineoplastic Agents adverse effects, Drug Hypersensitivity etiology, Myeloproliferative Disorders complications, Poly(ADP-ribose) Polymerase Inhibitors adverse effects
- Abstract
Purpose: DNA repair defects have been previously reported in myeloproliferative neoplasms (MPN). Inhibitors of PARP have shown activity in solid tumors with defects in homologous recombination (HR). This study was performed to assess MPN sensitivity to PARP inhibitors ex vivo, Experimental Design: HR pathway integrity in circulating myeloid cells was evaluated by assessing the formation of RAD51 foci after treatment with ionizing radiation or PARP inhibitors. Sensitivity of MPN erythroid and myeloid progenitors to PARP inhibitors was evaluated using colony formation assays., Results: Six of 14 MPN primary samples had reduced formation of RAD51 foci after exposure to ionizing radiation, suggesting impaired HR. This phenotype was not associated with a specific MPN subtype, JAK2 mutation status, or karyotype. MPN samples showed increased sensitivity to the PARP inhibitors veliparib and olaparib compared with normal myeloid progenitors. This hypersensitivity, which was most pronounced in samples deficient in DNA damage-induced RAD51 foci, was observed predominantly in samples from patients with diagnoses of chronic myelogenous leukemia, chronic myelomonocytic leukemia, or unspecified myelodysplastic/MPN overlap syndromes., Conclusions: Like other neoplasms with HR defects, MPNs exhibit PARP inhibitor hypersensitivity compared with normal marrow. These results suggest that further preclinical and possibly clinical study of PARP inhibitors in MPNs is warranted. Clin Cancer Res; 22(15); 3894-902. ©2016 AACR., Competing Interests: No conflicts of interest exist for any authors of this manuscript., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
30. Tumorgrafts as in vivo surrogates for women with ovarian cancer.
- Author
-
Weroha SJ, Becker MA, Enderica-Gonzalez S, Harrington SC, Oberg AL, Maurer MJ, Perkins SE, AlHilli M, Butler KA, McKinstry S, Fink S, Jenkins RB, Hou X, Kalli KR, Goodman KM, Sarkaria JN, Karlan BY, Kumar A, Kaufmann SH, Hartmann LC, and Haluska P
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Biomarkers, Tumor, Chromosome Aberrations, Cluster Analysis, Comparative Genomic Hybridization, Disease Models, Animal, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Graft Survival, Humans, Mice, Middle Aged, Neoplasm Grading, Neoplasm Metastasis, Neoplasm Staging, Ovarian Neoplasms diagnosis, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Ultrasonography, Xenograft Model Antitumor Assays, Heterografts, Ovarian Neoplasms pathology
- Abstract
Purpose: Ovarian cancer has a high recurrence and mortality rate. A barrier to improved outcomes includes a lack of accurate models for preclinical testing of novel therapeutics., Experimental Design: Clinically relevant, patient-derived tumorgraft models were generated from sequential patients and the first 168 engrafted models are described. Fresh ovarian, primary peritoneal, and fallopian tube carcinomas were collected at the time of debulking surgery and injected intraperitoneally into severe combined immunodeficient mice., Results: Tumorgrafts demonstrated a 74% engraftment rate with microscopic fidelity of primary tumor characteristics. Low-passage tumorgrafts also showed comparable genomic aberrations with the corresponding primary tumor and exhibit gene set enrichment of multiple ovarian cancer molecular subtypes, similar to patient tumors. Importantly, each of these tumorgraft models is annotated with clinical data and for those that have been tested, response to platinum chemotherapy correlates with the source patient., Conclusions: Presented herein is the largest known living tumor bank of patient-derived, ovarian tumorgraft models that can be applied to the development of personalized cancer treatment., (©2014 AACR)
- Published
- 2014
- Full Text
- View/download PDF
31. APOBEC3B upregulation and genomic mutation patterns in serous ovarian carcinoma.
- Author
-
Leonard B, Hart SN, Burns MB, Carpenter MA, Temiz NA, Rathore A, Vogel RI, Nikas JB, Law EK, Brown WL, Li Y, Zhang Y, Maurer MJ, Oberg AL, Cunningham JM, Shridhar V, Bell DA, April C, Bentley D, Bibikova M, Cheetham RK, Fan JB, Grocock R, Humphray S, Kingsbury Z, Peden J, Chien J, Swisher EM, Hartmann LC, Kalli KR, Goode EL, Sicotte H, Kaufmann SH, and Harris RS
- Subjects
- Carcinoma, Ovarian Epithelial, Cell Line, Tumor, Cystadenocarcinoma, Serous enzymology, Cystadenocarcinoma, Serous pathology, Cytidine Deaminase biosynthesis, Cytidine Deaminase metabolism, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Genomics, Humans, Minor Histocompatibility Antigens, Neoplasms, Glandular and Epithelial enzymology, Neoplasms, Glandular and Epithelial pathology, Ovarian Neoplasms enzymology, Ovarian Neoplasms pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Up-Regulation, Cystadenocarcinoma, Serous genetics, Cytidine Deaminase genetics, Mutation, Neoplasms, Glandular and Epithelial genetics, Ovarian Neoplasms genetics
- 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., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
32. Contribution of Bcl-2 phosphorylation to Bak binding and drug resistance.
- Author
-
Dai H, Ding H, Meng XW, Lee SH, Schneider PA, and Kaufmann SH
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis Regulatory Proteins metabolism, Bcl-2-Like Protein 11, Cells, Cultured, Drug Evaluation, Preclinical, Humans, Jurkat Cells, K562 Cells, Membrane Proteins metabolism, Phosphorylation drug effects, Phosphorylation physiology, Protein Binding drug effects, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 genetics, RNA, Small Interfering pharmacology, CDC2 Protein Kinase metabolism, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, bcl-2 Homologous Antagonist-Killer Protein metabolism
- Abstract
Bcl-2 is phosphorylated on Ser(70) after treatment of cells with spindle poisons. On the basis of effects observed in cells overexpressing Bcl-2 S70E or S70A mutants, various studies have concluded that Ser(70) phosphorylation either enhances or diminishes Bcl-2 function. In the present study, the ability of phosphorylated Bcl-2, as well as the S70E and S70A mutants, to bind and neutralize proapoptotic Bcl-2 family members under cell-free conditions and in intact cells was examined in an attempt to resolve this controversy. Surface plasmon resonance indicated that phosphorylated Bcl-2, Bcl-2 S70E, and Bcl-2 S70A exhibit enhanced binding to Bim and Bak compared with unmodified Bcl-2. This enhanced binding reflected a readily detectable conformation change in the loop domain of Bcl-2. Furthermore, Bcl-2 S70E and S70A bound more Bak and Bim than wild-type Bcl-2 in pull-downs and afforded greater protection against several chemotherapeutic agents. Importantly, binding of endogenous Bcl-2 to Bim also increased during mitosis, when Bcl-2 is endogenously phosphorylated, and disruption of this mitotic Bcl-2/Bim binding with navitoclax or ABT-199, like Bcl-2 downregulation, enhanced the cytotoxicity of paclitaxel. Collectively, these results provide not only a mechanistic basis for the enhanced antiapoptotic activity of phosphorylated Bcl-2, but also an explanation for the ability of BH3 mimetics to enhance taxane sensitivity.
- Published
- 2013
- Full Text
- View/download PDF
33. Phase I and pharmacologic trial of cytosine arabinoside with the selective checkpoint 1 inhibitor Sch 900776 in refractory acute leukemias.
- Author
-
Karp JE, Thomas BM, Greer JM, Sorge C, Gore SD, Pratz KW, Smith BD, Flatten KS, Peterson K, Schneider P, Mackey K, Freshwater T, Levis MJ, McDevitt MA, Carraway HE, Gladstone DE, Showel MM, Loechner S, Parry DA, Horowitz JA, Isaacs R, and Kaufmann SH
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Bone Marrow drug effects, Bone Marrow metabolism, Checkpoint Kinase 1, Cytarabine administration & dosage, Drug Resistance, Neoplasm, Female, Histones metabolism, Humans, Male, Maximum Tolerated Dose, Middle Aged, Phosphorylation, Protein Kinases metabolism, Protein Processing, Post-Translational, Pyrazoles administration & dosage, Pyrimidines administration & dosage, Treatment Outcome, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
Purpose: Incorporation of cytarabine into DNA activates checkpoint kinase 1 (Chk1), which stabilizes stalled replication forks, induces S-phase slowing, and diminishes cytarabine cytotoxicity. The selective Chk1 inhibitor SCH 900776 abrogates cytarabine-induced S-phase arrest and enhances cytarabine cytotoxicity in acute leukemia cell lines and leukemic blasts in vitro. To extend these findings to the clinical setting, we have conducted a phase I study of cytarabine and SCH 900776., Experimental Design: Twenty-four adults with relapsed and refractory acute leukemias received timed sequential, continuous infusion cytarabine 2 g/m(2) over 72 hours (667 mg/m(2)/24 hours) beginning on day 1 and again on day 10. SCH 900776 was administered as a 15- to 30-minute infusion on days 2, 3, 11, and 12. The starting dose of SCH 900776 was 10 mg/m(2)/dose., Results: Dose-limiting toxicities consisting of corrected QT interval prolongation and grade 3 palmar-plantar erythrodysesthesia occurred at 140 mg flat dosing (dose level 5, equivalent to 80 mg/m(2)). Complete remissions occurred in 8 of 24 (33%) patients, with 7 of 8 at 40 mg/m(2) or higher. SCH 900776 did not accumulate at any dose level. Marrow blasts obtained pretreatment and during therapy showed increased phosphorylation of H2Ax after SCH 900776 beginning at 40 mg/m(2), consistent with unrepaired DNA damage., Conclusions: These data support a randomized phase II trial of cytarabine +/- SCH 900776 at a recommended flat dose of 100 mg (equivalent to 56 mg/m(2)) for adults with poor-risk leukemias. The trial (SP P05247) was registered at www.clinicaltrials.gov as NCT00907517., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
34. Effects of selective checkpoint kinase 1 inhibition on cytarabine cytotoxicity in acute myelogenous leukemia cells in vitro.
- Author
-
Schenk EL, Koh BD, Flatten KS, Peterson KL, Parry D, Hess AD, Smith BD, Karp JE, Karnitz LM, and Kaufmann SH
- Subjects
- Antineoplastic Combined Chemotherapy Protocols administration & dosage, Apoptosis drug effects, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Checkpoints drug effects, Cell Cycle Proteins antagonists & inhibitors, Cell Line, Tumor, Cell Proliferation drug effects, Checkpoint Kinase 1, Humans, Protein Kinase Inhibitors administration & dosage, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrazoles administration & dosage, Pyrimidines administration & dosage, Cell Cycle Proteins metabolism, Cytarabine administration & dosage, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Purpose: Previous studies have shown that the replication checkpoint, which involves the kinases ataxia telangiectasia mutated and Rad3 related (ATR) and Chk1, contributes to cytarabine resistance in cell lines. In the present study, we examined whether this checkpoint is activated in clinical acute myelogenous leukemia (AML) during cytarabine infusion in vivo and then assessed the impact of combining cytarabine with the recently described Chk1 inhibitor SCH 900776 in vitro., Experimental Design: AML marrow aspirates harvested before and during cytarabine infusion were examined by immunoblotting. Human AML lines treated with cytarabine in the absence or presence of SCH 900776 were assayed for checkpoint activation by immunoblotting, nucleotide incorporation into DNA, and flow cytometry. Long-term effects in AML lines, clinical AML isolates, and normal myeloid progenitors were assayed using clonogenic assays., Results: Immunoblotting revealed increased Chk1 phosphorylation, a marker of checkpoint activation, in more than half of Chk1-containing AMLs after 48 hours of cytarabine infusion. In human AML lines, SCH 900776 not only disrupted cytarabine-induced Chk1 activation and S-phase arrest but also markedly increased cytarabine-induced apoptosis. Clonogenic assays demonstrated that SCH 900776 enhanced the antiproliferative effects of cytarabine in AML cell lines and clinical AML samples at concentrations that had negligible impact on normal myeloid progenitors., Conclusions: These results not only provide evidence for cytarabine-induced S-phase checkpoint activation in AML in the clinical setting, but also show that a selective Chk1 inhibitor can overcome the S-phase checkpoint and enhance the cytotoxicity of cytarabine. Accordingly, further investigation of the cytarabine/SCH 900776 combination in AML appears warranted.
- Published
- 2012
- Full Text
- View/download PDF
35. Failure of iniparib to inhibit poly(ADP-Ribose) polymerase in vitro.
- Author
-
Patel AG, De Lorenzo SB, Flatten KS, Poirier GG, and Kaufmann SH
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Cell Line, Tumor, Drug Evaluation, Preclinical, Homologous Recombination, Humans, Mice, Phthalazines pharmacology, Piperazines pharmacology, Topoisomerase I Inhibitors pharmacology, Antineoplastic Agents pharmacology, Benzamides pharmacology, Benzimidazoles pharmacology, Poly(ADP-ribose) Polymerase Inhibitors
- Abstract
Purpose: Poly(ADP-ribose) polymerase (PARP) inhibitors are undergoing extensive clinical testing for their single-agent activity in homologous recombination (HR)-deficient tumors and ability to enhance the action of certain DNA-damaging agents. Compared with other PARP inhibitors in development, iniparib (4-iodo-3-nitrobenzamide) is notable for its simple structure and the reported ability of its intracellular metabolite 4-iodo-3-nitrosobenzamide to covalently inhibit PARP1 under cell-free conditions. The present preclinical studies were conducted to compare the actions iniparib with the more extensively characterized PARP inhibitors olaparib and veliparib., Experimental Design: The abilities of iniparib, olaparib, and veliparib to (i) selectively induce apoptosis or inhibit colony formation in HR-deficient cell lines, (ii) selectively sensitize HR-proficient cells to topoisomerase I poisons, and (iii) inhibit formation of poly(ADP-ribose) polymer (pADPr) in intact cells were compared., Results: Consistent with earlier reports, olaparib and veliparib selectively induced apoptosis and inhibited colony formation in cells lacking BRCA2 or ATM. Moreover, like earlier generation PARP inhibitors, olaparib and veliparib sensitized cells to the topoisomerase I poisons camptothecin and topotecan. Finally, olaparib and veliparib inhibited formation of pADPr in intact cells. In contrast, iniparib exhibited little or no ability to selectively kill HR-deficient cells, sensitize cells to topoisomerase I poisons, or inhibit pADPr formation in situ. In further experiments, iniparib also failed to sensitize cells to cisplatin, gemcitabine, or paclitaxel., Conclusions: While iniparib kills normal and neoplastic cells at high (>40 μmol/L) concentrations, its effects are unlikely to reflect PARP inhibition and should not be used to guide decisions about other PARP inhibitors.
- Published
- 2012
- Full Text
- View/download PDF
36. Heat shock protein 90 inhibition depletes LATS1 and LATS2, two regulators of the mammalian hippo tumor suppressor pathway.
- Author
-
Huntoon CJ, Nye MD, Geng L, Peterson KL, Flatten KS, Haluska P, Kaufmann SH, and Karnitz LM
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Blotting, Western, Cell Line, Tumor, Clinical Trials, Phase II as Topic, Connective Tissue Growth Factor metabolism, Female, Fluorescent Antibody Technique, HSP90 Heat-Shock Proteins genetics, HSP90 Heat-Shock Proteins metabolism, Humans, Immunoprecipitation, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Phosphoproteins metabolism, Phosphorylation drug effects, Plasmids, Protein Serine-Threonine Kinases genetics, Serine-Threonine Kinase 3, Transcription Factors, Tumor Suppressor Proteins genetics, YAP-Signaling Proteins, Benzoquinones pharmacology, HSP90 Heat-Shock Proteins antagonists & inhibitors, Lactams, Macrocyclic pharmacology, Protein Serine-Threonine Kinases metabolism, Signal Transduction drug effects, Tumor Suppressor Proteins metabolism
- Abstract
Heat shock protein 90 (HSP90), which regulates the functions of multiple oncogenic signaling pathways, has emerged as a novel anticancer therapeutic target, and multiple small-molecule HSP90 inhibitors are now in clinical trials. Although the effects of HSP90 inhibitors on oncogenic signaling pathways have been extensively studied, the effects of these agents on tumor suppressor signaling pathways are currently unknown. Here, we have examined how HSP90 inhibitors affect LATS1 and the related protein LATS2, two kinases that relay antiproliferative signals in the Hippo tumor suppressor pathway. Both LATS1 and LATS2 were depleted from cells treated with the HSP90 inhibitors 17-allylamino-17-demethoxygeldanamycin (17-AAG), radicicol, and PU-H71. Moreover, these kinases interacted with HSP90, and LATS1 isolated from 17-AAG-treated cells had reduced catalytic activity, thus showing that the kinase is a bona fide HSP90 client. Importantly, LATS1 signaling was disrupted by 17-AAG in tumor cell lines in vitro and clinical ovarian cancers in vivo as shown by reduced levels of LATS1 and decreased phosphorylation of the LATS substrate YAP, an oncoprotein transcriptional coactivator that regulates genes involved in cell and tissue growth, including the CTGF gene. Consistent with the reduced YAP phosphorylation, there were increased levels of CTGF, a secreted protein that is implicated in tumor proliferation, metastasis, and angiogenesis. Taken together, these results identify LATS1 and LATS2 as novel HSP90 clients and show that HSP90 inhibitors can disrupt the LATS tumor suppressor pathway in human cancer cells., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
37. Expression of insulin receptor isoform A and insulin-like growth factor-1 receptor in human acute myelogenous leukemia: effect of the dual-receptor inhibitor BMS-536924 in vitro.
- Author
-
Wahner Hendrickson AE, Haluska P, Schneider PA, Loegering DA, Peterson KL, Attar R, Smith BD, Erlichman C, Gottardis M, Karp JE, Carboni JM, and Kaufmann SH
- Subjects
- Cell Growth Processes drug effects, Cell Growth Processes physiology, HL-60 Cells, Humans, Insulin-Like Growth Factor I biosynthesis, K562 Cells, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology, Protein Isoforms, Protein Kinase Inhibitors pharmacology, Receptor, IGF Type 1 antagonists & inhibitors, Receptor, Insulin antagonists & inhibitors, Signal Transduction drug effects, Tumor Cells, Cultured, U937 Cells, Benzimidazoles pharmacology, Leukemia, Myeloid, Acute enzymology, Pyridones pharmacology, Receptor, IGF Type 1 biosynthesis, Receptor, Insulin biosynthesis
- Abstract
The insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R) are receptor tyrosine kinases that participate in mitogenic and antiapoptotic signaling in normal and neoplastic epithelia. In the present study, immunoblotting and reverse transcription-PCR demonstrated expression of IGF1R and IR isoform A in acute myelogenous leukemia (AML) cell lines as well as in >80% of clinical AML isolates. Treatment with insulin enhanced signaling through the Akt and MEK1/2 pathways as well as survival of serum-starved AML cell lines. Conversely, treatment with BMS-536924, a dual IGF1R/IR kinase inhibitor that is undergoing preclinical testing, inhibited constitutive receptor phosphorylation as well as downstream signaling through MEK1/2 and Akt. These changes inhibited proliferation and, in some AML cell lines, induced apoptosis at submicromolar concentrations. Likewise, BMS-536924 inhibited leukemic colony formation in CD34+ clinical AML samples in vitro. Collectively, these results not only indicate that expression of IGF1R and IR isoform A is common in AML but also show that interruption of signaling from these receptors inhibits proliferation in clinical AML isolates. Accordingly, further investigation of IGF1R/IR axis as a potential therapeutic target in AML appears warranted.
- Published
- 2009
- Full Text
- View/download PDF
38. Evaluation of lapatinib and topotecan combination therapy: tissue culture, murine xenograft, and phase I clinical trial data.
- Author
-
Molina JR, Kaufmann SH, Reid JM, Rubin SD, Gálvez-Peralta M, Friedman R, Flatten KS, Koch KM, Gilmer TM, Mullin RJ, Jewell RC, Felten SJ, Mandrekar S, Adjei AA, and Erlichman C
- Subjects
- Adult, Aged, Animals, Breast Neoplasms drug therapy, Cell Line, Tumor, Female, Humans, In Vitro Techniques, Lapatinib, Male, Maximum Tolerated Dose, Mice, Middle Aged, Quinazolines administration & dosage, Quinazolines adverse effects, Quinazolines pharmacokinetics, Topotecan administration & dosage, Topotecan adverse effects, Topotecan pharmacokinetics, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasms drug therapy
- Abstract
Purpose: Topotecan resistance can result from drug efflux by P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP) as well as survival signals initiated by epidermal growth factor receptor family members. The present studies were done to determine the effect of combining topotecan and the dual epidermal growth factor receptor/HER2 inhibitor lapatinib in tissue culture, a murine xenograft model, and a phase I clinical trial., Experimental Design: The effects of lapatinib on topotecan accumulation and cytotoxicity in vitro were examined in paired cell lines lacking or expressing Pgp or BCRP. Antiproliferative effects of the combination were assessed in mice bearing HER2+ BT474 breast cancer xenografts. Based on tolerability in this preclinical model, 37 patients with advanced-stage cancers received escalating doses of lapatinib and topotecan in a phase I trial., Results: Lapatinib increased topotecan accumulation in BCRP- or Pgp-expressing cells in vitro, and the combination showed enhanced efficacy in HER2+ BT474 xenografts. In the phase I study, nausea, vomiting, diarrhea, and fatigue were dose limiting. The maximum tolerated doses were 1,250 mg/d lapatinib by mouth for 21 or 28 days with 3.2 mg/m2 topotecan i.v. on days 1, 8, and 15 of 28-day cycles. Pharmacokinetic analyses showed that combined drug administration resulted in decreased topotecan clearance consistent with transporter-mediated interactions. Seventeen (46%) patients had disease stabilization., Conclusions: The lapatinib/topotecan combination is well tolerated and warrants further study.
- Published
- 2008
- Full Text
- View/download PDF
39. Resistance of cancers to immunologic cytotoxicity and adoptive immunotherapy via X-linked inhibitor of apoptosis protein expression and coexisting defects in mitochondrial death signaling.
- Author
-
Ravi R, Fuchs EJ, Jain A, Pham V, Yoshimura K, Prouser T, Jalla S, Zhou X, Garrett-Mayer E, Kaufmann SH, Schulick RD, Pardoll DM, and Bedi A
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma therapy, Animals, Apoptosis drug effects, Apoptosis immunology, Apoptosis Regulatory Proteins pharmacology, Caspases metabolism, Colonic Neoplasms genetics, Colonic Neoplasms metabolism, Colonic Neoplasms therapy, Cytotoxicity, Immunologic, Enzyme Activation, Female, Granzymes, HCT116 Cells, Humans, Interferon-gamma immunology, Interferon-gamma pharmacology, Membrane Glycoproteins pharmacology, Mice, Mice, Inbred BALB C, Mitochondria enzymology, Recombinant Proteins pharmacology, Serine Endopeptidases pharmacology, TNF-Related Apoptosis-Inducing Ligand, Transfection, Tumor Necrosis Factor-alpha pharmacology, X-Linked Inhibitor of Apoptosis Protein biosynthesis, X-Linked Inhibitor of Apoptosis Protein genetics, Xenograft Model Antitumor Assays, Adenocarcinoma immunology, Colonic Neoplasms immunology, Immunotherapy, Adoptive methods, Mitochondria immunology, X-Linked Inhibitor of Apoptosis Protein immunology
- Abstract
The ability of cancers to evade immune surveillance and resist immunotherapy raises a fundamental question of how tumor cells survive in the presence of a competent immune system. Studies to address this question have primarily focused on mechanisms by which tumor cells avoid recognition by or induce tolerance in the immune system. However, little is known about whether cancer cells also acquire an intrinsic ability to resist killing by immune effectors. We find that cancer cells enhance their ability to withstand an attack by cytotoxic immune effector cells via acquisition of specific genetic alterations that interfere with the shared mitochondrial death signaling pathway entrained by granzyme B, IFN-gamma, and Apo2 ligand/tumor necrosis factor-related apoptosis inducing ligand (Apo2L/TRAIL), three key mediators of immunologic cell-mediated cytotoxicity. We show that the coexistence of specific mitochondrial signaling defects (either deletion of Bax, overexpression of Bcl-x(L), or deletion of Smac) with expression of X-linked inhibitor of apoptosis protein decreases the sensitivity of cancer cells to IFN-gamma/Apo2L/TRAIL- or granzyme B-induced apoptosis, lymphocyte-mediated cytotoxicity in vitro, and adoptive cellular immunotherapy in vivo. Conversely, negating X-linked inhibitor of apoptosis protein expression or function in tumor cells with defective mitochondrial signaling enables direct activation of caspase-3/-7 by granzyme B or Apo2L/TRAIL, and restores their susceptibility to immunologic cytotoxicity. These findings identify an important mechanism by which cancers evade elimination by immune effector cells and suggest that cancer immunotherapy might be improved by concurrent strategies to alleviate or circumvent the intrinsic mitochondrial death signaling defects that help cancer cells resist immunologic cytotoxicity.
- Published
- 2006
- Full Text
- View/download PDF
40. In vitro and in vivo antitumor effects of the dual insulin-like growth factor-I/insulin receptor inhibitor, BMS-554417.
- Author
-
Haluska P, Carboni JM, Loegering DA, Lee FY, Wittman M, Saulnier MG, Frennesson DB, Kalli KR, Conover CA, Attar RM, Kaufmann SH, Gottardis M, and Erlichman C
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Caspases metabolism, Cell Growth Processes drug effects, Cell Line, Tumor, Cell Nucleus metabolism, Cyclin D1 metabolism, Dose-Response Relationship, Drug, Enzyme Activation, Female, G1 Phase drug effects, Humans, Insulin-Like Growth Factor I antagonists & inhibitors, Mice, Mice, Nude, Mitochondria drug effects, Mitochondria physiology, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Receptor, IGF Type 1 metabolism, Receptor, Insulin metabolism, S Phase drug effects, Piperazines pharmacology, Pyridones pharmacology, Receptor, IGF Type 1 antagonists & inhibitors, Receptor, Insulin antagonists & inhibitors
- Abstract
The insulin-like growth factor receptor (IGF-IR) and insulin receptor are either overactivated and/or overexpressed in a wide range of tumor types and contribute to tumorigenicity, proliferation, metastasis, and drug resistance. Here, we show that BMS-554417, a novel small molecule developed as an inhibitor of IGF-IR, inhibits IGF-IR and insulin receptor kinase activity and proliferation in vitro, and reduces tumor xenograft size in vivo. In a series of carcinoma cell lines, the IC50 for proliferation ranged from 120 nmol/L (Colo205) to >8.5 micromol/L (OV202). The addition of stimulatory ligands was unnecessary for the antiproliferative effect in MCF-7 and OV202 cells. BMS-554417 treatment inhibited IGF-IR and insulin receptor signaling through extracellular signal-related kinase as well as the phosphoinositide 3-kinase/Akt pathway, as evidenced by decreased Akt phosphorylation at Ser473. At doses that inhibited proliferation, the compound also caused a G0-G1 arrest and prevented nuclear accumulation of cyclin D1 in response to LR3 IGF-I. In Jurkat T-cell leukemia cells, this agent triggered apoptotic cell death via the mitochondrial pathway. BMS-554417 was orally bioavailable and significantly inhibited the growth of IGF1R-Sal tumor xenografts in vivo. BMS-554417 is a member of a novel class of IGF-IR/insulin receptor inhibitors that have potential clinical applications because of their antiproliferative and proapoptotic activity in vitro and in vivo.
- Published
- 2006
- Full Text
- View/download PDF
41. Phase I and pharmacologic study of infusional topotecan and Carboplatin in relapsed and refractory acute leukemia.
- Author
-
Kaufmann SH, Karp JE, Letendre L, Kottke TJ, Safgren S, Greer J, Gojo I, Atherton P, Svingen PA, Loegering DA, Litzow MR, Sloan JA, Reid JM, Ames MM, Adjei AA, and Erlichman C
- Subjects
- Acute Disease, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Carboplatin administration & dosage, Carboplatin adverse effects, Carboplatin pharmacokinetics, Cell Cycle Proteins metabolism, Combined Modality Therapy, DNA Topoisomerases, Type I metabolism, Dose-Response Relationship, Drug, Drug Resistance, Neoplasm, Female, HL-60 Cells, Hematopoietic Stem Cell Transplantation, Humans, Immunoblotting, Infusions, Intravenous, Leukemia metabolism, Male, Middle Aged, Neoplasm Recurrence, Local, Proliferating Cell Nuclear Antigen metabolism, Topotecan administration & dosage, Topotecan adverse effects, Topotecan pharmacokinetics, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia drug therapy
- Abstract
Purpose: To assess the maximum tolerated dose, toxicities, pharmacokinetics, and antileukemic activity of topotecan and carboplatin in adults with recurrent or refractory acute leukemias., Experimental Design: Patients received topotecan and carboplatin by 5-day continuous infusion at nine dose levels. Patients achieving a complete remission received up to two additional courses for consolidation. Plasma topotecan and ultrafilterable platinum were assayed on days 1 to 5. In addition, pretreatment levels of various polypeptides in leukemic cells were examined by immunoblotting to assess possible correlations with response., Results: Fifty-one patients received a total of 69 courses of therapy. Dose-limiting toxicity consisted of grade 4/5 typhlitis and grade 3/4 mucositis after one course of therapy or grade 4 neutropenia and thrombocytopenia lasting >50 days when a second course was administered on day 21. Among 45 evaluable patients, there were 7 complete remissions, 2 partial remissions, 1 incomplete complete remission, and 1 reversion to chronic-phase chronic myelogenous leukemia. Topotecan steady-state plasma concentrations increased with dose. No accumulation of topotecan or ultrafilterable platinum occurred between days 1 and 5 of therapy. Leukemic cell levels of topoisomerase I, checkpoint kinase 1, checkpoint kinase 2, and Mcl-1 correlated with proliferating cell nuclear antigen but not with response. In contrast, low Bcl-2 expression correlated with response (P = 0.014, Mann-Whitney U test)., Conclusions: The maximum tolerated dose was 1.6 mg/m(2)/d topotecan plus 150 mg/m(2)/d carboplatin. The complete remission rate in a heavily pretreated population was 16% (33% at the highest three dose levels). Responses seem to correlate with low pretreatment blast cell Bcl-2 expression.
- Published
- 2005
- Full Text
- View/download PDF
42. Phase 1 trial of flavopiridol combined with cisplatin or carboplatin in patients with advanced malignancies with the assessment of pharmacokinetic and pharmacodynamic end points.
- Author
-
Bible KC, Lensing JL, Nelson SA, Lee YK, Reid JM, Ames MM, Isham CR, Piens J, Rubin SL, Rubin J, Kaufmann SH, Atherton PJ, Sloan JA, Daiss MK, Adjei AA, and Erlichman C
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Area Under Curve, Carboplatin administration & dosage, Carboplatin adverse effects, Carboplatin pharmacokinetics, Cell Line, Tumor, Cisplatin administration & dosage, Cisplatin adverse effects, Cisplatin pharmacokinetics, Cohort Studies, Diarrhea chemically induced, Dose-Response Relationship, Drug, Female, Flavonoids administration & dosage, Flavonoids adverse effects, Flavonoids pharmacokinetics, Humans, Immunoblotting, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear metabolism, Male, Middle Aged, Nausea chemically induced, Neoplasms blood, Neoplasms pathology, Piperidines administration & dosage, Piperidines adverse effects, Piperidines pharmacokinetics, Proto-Oncogene Proteins c-bcl-2 blood, STAT3 Transcription Factor blood, Treatment Outcome, Tumor Suppressor Protein p53 blood, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasms drug therapy
- Abstract
Purpose: Flavopiridol, a cyclin-dependent kinase inhibitor, transcription inhibitor, and DNA-interacting agent, was combined with cisplatin or carboplatin to establish toxicities, evaluate pharmacokinetics, and examine its effects on patient cancers and levels of selected polypeptides in patient peripheral blood mononuclear cells (PBMC)., Experimental Design: Therapy was given every 3 weeks. Stage I: cisplatin was fixed at 30 mg/m2 with escalating flavopiridol. Stage II: flavopiridol was fixed at the stage I maximum tolerated dose (MTD) with escalation of cisplatin. Stage III: flavopiridol was fixed at the stage I MTD with escalation of carboplatin., Results: Thirty-nine patients were treated with 136 cycles of chemotherapy. Neutropenia was seen in only 11% of patients. Grade 3 flavopiridol/CDDP toxicities were nausea (30%), vomiting (19%), diarrhea (15%), dehydration (15%), and neutropenia (10%). Flavopiridol combined with carboplatin resulted in unexpectedly high toxicities and one treatment-related death. Stable disease (>3 months) was seen in 34% of treated patients, but there were no objective responses. The stage II MTD was 60 mg/m2 cisplatin and 100 mg/m2/24 hours flavopiridol. As given, CDDP did not alter flavopiridol pharmacokinetics. Flavopiridol induced increased p53 and pSTAT3 levels in patient PBMCs but had no effects on cyclin D1, phosphoRNA polymerase II, or Mcl-1., Conclusions: Flavopiridol and cisplatin can be safely combined in the treatment of cancer patients. Unexpected toxicity in flavopiridol/carboplatin-treated patients attenuates enthusiasm for this alternative combination. Analysis of polypeptide levels in patient PBMCs suggests that flavopiridol may be affecting some, but not all, of its known in vitro molecular targets in vivo.
- Published
- 2005
- Full Text
- View/download PDF
43. A phase I and pharmacologic trial of two schedules of the proteasome inhibitor, PS-341 (bortezomib, velcade), in patients with advanced cancer.
- Author
-
Dy GK, Thomas JP, Wilding G, Bruzek L, Mandrekar S, Erlichman C, Alberti D, Binger K, Pitot HC, Alberts SR, Hanson LJ, Marnocha R, Tutsch K, Kaufmann SH, and Adjei AA
- Subjects
- Adult, Aged, Aged, 80 and over, Anemia chemically induced, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacology, Blotting, Western, Boronic Acids adverse effects, Boronic Acids pharmacology, Bortezomib, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cyclin E metabolism, Cyclin-Dependent Kinase Inhibitor p27, Diarrhea chemically induced, Dose-Response Relationship, Drug, Drug Administration Schedule, Fatigue chemically induced, Female, Humans, I-kappa B Proteins metabolism, Male, Middle Aged, NF-kappa B metabolism, Nausea chemically induced, Neoplasms metabolism, Neoplasms pathology, Proteasome Endopeptidase Complex metabolism, Proteasome Inhibitors, Proto-Oncogene Proteins c-bcl-2 metabolism, Pyrazines adverse effects, Pyrazines pharmacology, Thrombocytopenia chemically induced, Treatment Outcome, Tumor Suppressor Proteins metabolism, Vomiting chemically induced, bcl-2-Associated X Protein, Antineoplastic Agents therapeutic use, Boronic Acids therapeutic use, Neoplasms drug therapy, Pyrazines therapeutic use
- Abstract
Purpose: To define the toxicities, pharmacodynamics, and clinical activity of the proteasome inhibitor, PS-341 (bortezomib), in patients with advanced malignancies., Patients and Methods: Twenty-eight patients (14 male and 14 female) received PS-341 twice weekly for 4 of 6 weeks (schedule I). Because toxicity necessitated dose omissions on this schedule, 16 additional patients (12 male and 4 female) received PS-341 twice weekly for 2 of every 3 weeks (schedule II). A total of 73 courses of treatment was given (median, 2; range, 1-4). Inhibition of 20S proteasome activity in peripheral blood mononuclear cells (PBMC) and accumulation of proteasome-targeted polypeptides in tumor tissue were evaluated as pharmacodynamic markers of PS-341 activity., Results: The most common toxicity was thrombocytopenia, which was dose limiting at 1.7 mg/m2 (schedule I) and 1.6 mg/m2 (schedule II), respectively. Sensory neuropathy was dose-limiting in a patient in schedule I. Grade > or =3 toxicities for schedule I were constipation, fatigue, myalgia, and sensory neuropathy. Grade > or =3 toxicities for schedule II were dehydration resulting from diarrhea, nausea and vomiting, fatigue, hypoglycemia, and hypotension. The maximum tolerated dose was 1.5 mg/m2 for both schedules. Reversible dose-dependent decreases in 20S proteasome activity in PBMCs were observed, with 36% inhibition at 0.5 mg/m2, 52% at 0.9 mg/m2, and 75% at 1.25 mg/m2. Accumulation of proteasome-targeted polypeptides was detected in tumor samples after treatment with PS-341. A patient with multiple myeloma had a partial response., Conclusion: PS-341 given 1.5 mg/m2 twice weekly for 2 of every 3 weeks is well tolerated and should be further studied.
- Published
- 2005
- Full Text
- View/download PDF
44. Gene expression profiles predict early relapse in ovarian cancer after platinum-paclitaxel chemotherapy.
- Author
-
Hartmann LC, Lu KH, Linette GP, Cliby WA, Kalli KR, Gershenson D, Bast RC, Stec J, Iartchouk N, Smith DI, Ross JS, Hoersch S, Shridhar V, Lillie J, Kaufmann SH, Clark EA, and Damokosh AI
- Subjects
- Adult, Aged, Aged, 80 and over, Carcinoma, Endometrioid drug therapy, Carcinoma, Endometrioid genetics, Carcinoma, Endometrioid pathology, Cystadenocarcinoma, Serous drug therapy, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology, Female, Gene Expression Profiling, Humans, Middle Aged, Neoplasm Recurrence, Local metabolism, Neoplasm Staging, Oligonucleotide Array Sequence Analysis, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology, Paclitaxel administration & dosage, Platinum administration & dosage, Survival Rate, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor analysis, Gene Expression Regulation, Neoplastic, Neoplasm Recurrence, Local pathology, Ovarian Neoplasms genetics
- Abstract
Purpose: Women with advanced epithelial ovarian cancer are routinely treated with platinum-paclitaxel chemotherapy following cytoreductive surgery, yet only approximately 20% achieve long-term disease-free survival. We hypothesized that differences in gene expression before treatment could distinguish patients with short versus long time to recurrence after administration of platinum-paclitaxel combination chemotherapy., Experimental Design: To test this hypothesis, gene expression profiling of 79 primary surgically resected tumors from women with advanced-stage, high-grade epithelial ovarian cancer was done using cDNA microarrays containing 30,721 genes. Supervised learning algorithms were applied in an effort to develop a binary classifier that could discriminate women at risk for early (< or =21 months) versus late (>21 months) relapse after initial chemotherapy., Results: A 14-gene predictive model was developed using a set of training samples (n = 51) and subsequently tested using an independent set of test samples (n = 28). This model correctly predicted the outcome of 24 of the 28 test samples (86% accuracy) with 95% positive predictive value for early relapse., Conclusions: Predictive markers for early recurrence can be identified for platinum-paclitaxel combination chemotherapy in primary ovarian carcinoma. The proposed 14-gene model requires further validation.
- Published
- 2005
- Full Text
- View/download PDF
45. A phase I trial of the novel farnesyl protein transferase inhibitor, BMS-214662, in combination with paclitaxel and carboplatin in patients with advanced cancer.
- Author
-
Dy GK, Bruzek LM, Croghan GA, Mandrekar S, Erlichman C, Peethambaram P, Pitot HC, Hanson LJ, Reid JM, Furth A, Cheng S, Martell RE, Kaufmann SH, and Adjei AA
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Benzodiazepines pharmacology, Carboplatin administration & dosage, Drug Administration Schedule, Female, Humans, Imidazoles pharmacology, Male, Maximum Tolerated Dose, Middle Aged, Paclitaxel administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Benzodiazepines adverse effects, Benzodiazepines pharmacokinetics, Imidazoles adverse effects, Imidazoles pharmacokinetics, Neoplasms drug therapy
- Abstract
Purpose: This phase I study was conducted to determine the toxicities, pharmacokinetics, and pharmacodynamics of BMS-214662, a farnesyl transferase inhibitor, in combination with paclitaxel and carboplatin, in patients with advanced solid tumors., Experimental Design: Patients with solid tumors received one of six escalating dose levels of BMS-214662 infused over 1 hour given following paclitaxel and carboplatin on the first day of a 21-day cycle. Toxicities were graded by the National Cancer Institute common toxicity criteria and recorded as maximum grade per patient for each treatment cycle. Inhibition of farnesyl transferase activity in peripheral blood mononuclear cells (PBMCs) was evaluated. Accumulation of unfarnesylated HDJ-2 in PBMCs of patients was evaluated as a marker of farnesyl transferase inhibition by BMS-214662., Results: Thirty patients received 141 cycles of treatment through six dose levels. Dose-limiting toxicities were neutropenia, thrombocytopenia, nausea, and vomiting. There was no pharmacokinetic interaction between BMS-214662 and paclitaxel. The maximum tolerated dose was established as BMS-214662 (160 mg/m(2)), paclitaxel (225 mg/m(2)) and carboplatin (area under the curve = 6 on day 1), every 21 days. Inhibition of HDJ-2 farnesylation in PBMCs of patients was shown. One measurable partial response was observed in a patient with taxane-resistant esophageal cancer. There was partial regression of evaluable disease in two other patients (endometrial and ovarian cancer). Stable disease (> 4 cycles) occurred in eight other patients., Conclusions: The combination of BMS-214662 with paclitaxel and carboplatin was well tolerated, with broad activity in solid tumors. There was no correlation between dose level and accumulation of unfarnesylated HDJ-2 in PBMCs nor tumor response.
- Published
- 2005
- Full Text
- View/download PDF
46. Components of the cell death machine and drug sensitivity of the National Cancer Institute Cell Line Panel.
- Author
-
Svingen PA, Loegering D, Rodriquez J, Meng XW, Mesner PW Jr, Holbeck S, Monks A, Krajewski S, Scudiero DA, Sausville EA, Reed JC, Lazebnik YA, and Kaufmann SH
- Subjects
- Apoptotic Protease-Activating Factor 1, Drug Screening Assays, Antitumor, Humans, Immunoblotting, Proteins analysis, Apoptosis drug effects, Caspases biosynthesis, Caspases genetics, Neoplasms genetics, Neoplasms pathology, Proteins genetics, Tumor Cells, Cultured
- Abstract
Purpose: According to some studies, susceptibility of cells to anticancer drug-induced apoptosis is markedly inhibited by targeted deletion of genes encoding apoptotic protease activating factor 1 (Apaf-1) or certain caspases. Information about levels of these polypeptides in common cancer cell types and any possible correlation with drug sensitivity in the absence of gene deletion is currently fragmentary., Experimental Design: Immunoblotting was used to estimate levels of Apaf-1 as well as procaspase-2, -3, -6, -7, -8, and -9 in the 60-cell-line panel used for drug screening by the National Cancer Institute. Sensitivity of the same lines to >80,000 compounds was determined with 48-hour sulforhodamine B binding assays. Additional 6-day assays were performed for selected agents., Results: Levels of Apaf-1 and procaspases varied widely. Apaf-1 and procaspase-9, which are implicated in caspase activation after treatment of cells with various anticancer drugs, were detectable in all of the cell lines, with levels of Apaf-1 ranging from approximately 1 x 10(5) to 2 x 10(6) molecules per cell and procaspase-9 from approximately 5 x 10(3) to approximately 1.6 x 10(5) molecules per cell. Procaspase-8 levels ranged from 1.7 x 10(5) to 8 x 10(6) molecules per cell. Procaspase-3, a major effector caspase, varied from undetectable to approximately 1.6 x 10(6) molecules per cell. Correlations between levels of these polypeptides and sensitivity to any of a variety of experimental or conventional antineoplastic agents in either 2-day or 6-day cytotoxicity assays were weak at best., Conclusions: With the exception of caspase-3, all of the components of the core cell-death machinery are expressed in all of the cell lines examined. Despite variations in expression, levels of any one component are not a major determinant of drug sensitivity in these cells in vitro.
- Published
- 2004
- Full Text
- View/download PDF
47. Mcl-1 mediates tumor necrosis factor-related apoptosis-inducing ligand resistance in human cholangiocarcinoma cells.
- Author
-
Taniai M, Grambihler A, Higuchi H, Werneburg N, Bronk SF, Farrugia DJ, Kaufmann SH, and Gores GJ
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis physiology, Apoptosis Regulatory Proteins, CASP8 and FADD-Like Apoptosis Regulating Protein, Carrier Proteins biosynthesis, Cell Line, Tumor, Cholangiocarcinoma genetics, Cholangiocarcinoma metabolism, Cholangiocarcinoma pathology, Down-Regulation, Drug Resistance, Neoplasm, Drug Synergism, Flavonoids pharmacology, Genes, bcl-2 genetics, Humans, Membrane Glycoproteins pharmacology, Mitochondria drug effects, Mitochondria physiology, Myeloid Cell Leukemia Sequence 1 Protein, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Piperidines pharmacology, Proto-Oncogene Proteins c-bcl-2 genetics, RNA, Small Interfering genetics, Receptors, Tumor Necrosis Factor biosynthesis, TNF-Related Apoptosis-Inducing Ligand, Tumor Necrosis Factor-alpha pharmacology, bcl-X Protein, Antineoplastic Agents antagonists & inhibitors, Apoptosis drug effects, Cholangiocarcinoma drug therapy, Intracellular Signaling Peptides and Proteins, Membrane Glycoproteins antagonists & inhibitors, Neoplasm Proteins physiology, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Cholangiocarcinomas are usually fatal neoplasms originating from bile duct epithelia. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising agent for cancer therapy, including cholangiocarcinoma. However, many cholangiocarcinoma cells are resistant to TRAIL-mediated apoptosis. Thus, our aim was to examine the intracellular mechanisms responsible for TRAIL resistance in human cholangiocarcinoma cell lines. Three TRAIL-resistant human cholangiocarcinoma cell lines were identified. All of the cell lines expressed TRAIL receptor 1/death receptor 4 (TRAIL-R1/DR4) and TRAIL-R2/DR5. Expression of TRAIL decoy receptors and the antiapoptotic cellular FLICE-inhibitory protein (cFLIP) was inconsistent across the cell lines. Of the antiapoptotic Bcl-2 family of proteins profiled (Bcl-2, Bcl-x(L), and Mcl-1), Mcl-1 was uniquely overexpressed by the cell lines. When small-interfering-RNA (siRNA) technology was used to knock down expression of Bcl-2, Bcl-x(L), and Mcl-1, only the Mcl-1-siRNA sensitized the cells to TRAIL-mediated apoptosis. In a cell line stably transfected with Mcl-1-small-hairpin-RNA (Mcl-1-shRNA), Mcl-1 depletion sensitized cells to TRAIL-mediated apoptosis despite Bcl-2 expression. TRAIL-mediated apoptosis in the stably transfected cells was associated with mitochondrial depolarization, Bax activation, cytochrome c release from mitochondria, and caspase activation. Finally, flavopiridol, an anticancer drug that rapidly down-regulates Mcl-1, also sensitized cells to TRAIL cytotoxicity. In conclusion, these studies not only demonstrate that Mcl-1 mediates TRAIL resistance in cholangiocarcinoma cells by blocking the mitochondrial pathway of cell death but also identify two strategies for circumventing this resistance.
- Published
- 2004
- Full Text
- View/download PDF
48. A Phase I trial of the farnesyl protein transferase inhibitor R115777 in combination with gemcitabine and cisplatin in patients with advanced cancer.
- Author
-
Adjei AA, Croghan GA, Erlichman C, Marks RS, Reid JM, Sloan JA, Pitot HC, Alberts SR, Goldberg RM, Hanson LJ, Bruzek LM, Atherton P, Thibault A, Palmer PA, and Kaufmann SH
- Subjects
- Adult, Aged, Alkyl and Aryl Transferases antagonists & inhibitors, Dose-Response Relationship, Drug, Electrolytes, Enzyme Inhibitors pharmacology, Farnesyltranstransferase, Female, Humans, Immunohistochemistry, Male, Maximum Tolerated Dose, Middle Aged, Mouth Mucosa metabolism, Quinolones adverse effects, Time Factors, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cisplatin administration & dosage, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Enzyme Inhibitors therapeutic use, Neoplasms drug therapy, Quinolones administration & dosage
- Abstract
Purpose: This Phase I study was undertaken to define the toxicity, pharmacodynamics, and maximum tolerated dose of the combination of R115777, a farnesyl transferase inhibitor, with gemcitabine and cisplatin in patients with advanced solid tumors., Patients and Methods: Thirty patients with solid tumors received a median of 2.5 cycles (range 1-30+) through five dose levels. R115777 was administered p.o. twice daily for 14 days. Gemcitabine was infused 15 min after the ingestion of R115777 on days 1 and 8. Cisplatin was administered starting 30 min after completion of the gemcitabine infusion on day 1. Cycles were repeated every 21 days. Toxicities were graded by the National Cancer Institute Common Toxicity Criteria and recorded as maximum grade per patient for each treatment cycle. At the maximum tolerated dose, accumulation of prelamin A in buccal mucosa cells of patients was evaluated as a marker of farnesyl transferase inhibition by R115777., Results: Neutropenia and thrombocytopenia were the most common toxicities. Dose-limiting toxicity in cycle 1 was myelosuppression with thrombocytopenia alone (4 patients), neutropenia alone (1 patient), or a combination of both (3 patients). Common nonhematologic toxicities were anorexia, rash, nausea, vomiting, and fatigue, none of which was dose limiting in the first cycle. At the maximum tolerated dose, defined as R115777 300 mg twice daily p.o., 1000 mg/m(2) gemcitabine, and 75 mg/m(2) cisplatin, inhibition of prelamin A farnesylation in buccal mucosa cells of patients was demonstrated, confirming that R115777 inhibits protein farnesylation in vivo. Nine objective responses (one complete response and eight partial responses) were documented in 27 evaluable patients., Conclusion: The combination of R115777 with gemcitabine and cisplatin was well tolerated and showed evidence of antitumor activity. The maximum tolerated dose of R115777 successfully inhibits farnesyltransferase in patients in vivo. This combination warrants further evaluation in a number of tumor types.
- Published
- 2003
49. Bile acids inhibit Mcl-1 protein turnover via an epidermal growth factor receptor/Raf-1-dependent mechanism.
- Author
-
Yoon JH, Werneburg NW, Higuchi H, Canbay AE, Kaufmann SH, Akgul C, Edwards SW, and Gores GJ
- Subjects
- Apoptosis drug effects, Apoptosis physiology, Cholangiocarcinoma metabolism, ErbB Receptors antagonists & inhibitors, Humans, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System physiology, Myeloid Cell Leukemia Sequence 1 Protein, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Proto-Oncogene Proteins c-raf antagonists & inhibitors, RNA, Messenger biosynthesis, RNA, Messenger genetics, Transcription, Genetic drug effects, Tumor Cells, Cultured, Deoxycholic Acid pharmacology, ErbB Receptors physiology, Neoplasm Proteins metabolism, Proto-Oncogene Proteins c-bcl-2, Proto-Oncogene Proteins c-raf physiology
- Abstract
Bile acids have been implicated in biliary tract carcinogenesis, in part, by activating the epidermal growth factor receptor (EGFR). Overexpression of Mcl-1, a potent antiapoptotic protein of the Bcl-2 family, has also been reported in cholangiocarcinomas. Because receptor tyrosine kinases like EGFR may modulate antiapoptotic protein expression, we examined the hypothesis that bile acids modulate Mcl-1 expression levels via EGFR. Deoxycholate increased cellular Mcl-1 protein in a concentration-dependent manner. The deoxycholate-mediated increase of cellular Mcl-1 protein was blocked equally by EGFR tyrosine kinase inhibitors or an EGFR-neutralizing antibody. Although inhibition of mitogen-activated protein kinases did not attenuate the deoxycholate-associated increase in Mcl-1 protein, the Raf-1 inhibitor, BAY 37-9751, effectively blocked the cellular increase of this protein. Neither Mcl-1 transcriptional activity nor its mRNA stability was altered by deoxycholate treatment. However, Mcl-1 protein stability was increased by bile acid treatment, an effect duplicated by proteasome inhibition. Deoxycholate prolongation of Mcl-1 turnover was blocked by either EGFR inhibitors or the Raf-1 inhibitor. Whereas the deoxycholate-induced increase in Mcl-1 reduced Fas-mediated apoptosis, the Raf-1 inhibitor potentiated Fas apoptosis. Our results demonstrate that bile acids block Mcl-1 protein degradation via activation of an EGFR/Raf-1 cascade resulting in its cellular accumulation. Raf-1 inhibitors block this increase of Mcl-1 and render the cells more susceptible to apoptosis, a potential therapeutic strategy for cholangiocarcinomas.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.