96 results on '"Kmieciak M"'
Search Results
2. Nonsense-mediated mRNA decay in mammalian cells: splicing, the pioneer round of translation, and mRNP remodeling
- Author
-
Lejeune, F., Chiu, S.-Y., Ranganathan, A., Kim, Y. K., Kmieciak, M., Gao, Q., and Maquat, L. E.
- Published
- 2004
3. Co-administration of the mTORC1/TORC2 inhibitor INK128 and the Bcl-2/Bcl-xL antagonist ABT-737 kills human myeloid leukemia cells through Mcl-1 down-regulation and AKT inactivation
- Author
-
Rahmani, M., primary, Aust, M. M., additional, Hawkins, E., additional, Parker, R. E., additional, Ross, M., additional, Kmieciak, M., additional, Reshko, L. B., additional, Rizzo, K. A., additional, Dumur, C. I., additional, Ferreira-Gonzalez, A., additional, and Grant, S., additional
- Published
- 2015
- Full Text
- View/download PDF
4. A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations
- Author
-
Zhou, L, primary, Zhang, Y, additional, Chen, S, additional, Kmieciak, M, additional, Leng, Y, additional, Lin, H, additional, Rizzo, K A, additional, Dumur, C I, additional, Ferreira-Gonzalez, A, additional, Dai, Y, additional, and Grant, S, additional
- Published
- 2014
- Full Text
- View/download PDF
5. OP0218 Regulation of auto-antibody production by auto-immune complexes on follicular dendritic cells
- Author
-
El Shikh, M.E., primary, Biggioggero, M., additional, El Sayed, R., additional, Kmieciak, M., additional, Manjili, M., additional, Szakal, A., additional, Meroni, P.L., additional, Pitzalis, C., additional, and Tew, J., additional
- Published
- 2013
- Full Text
- View/download PDF
6. Immunologic markers associated with favorable prognosis in breast cancer patients: Role of innate immune system.
- Author
-
Ascierto, M. L., primary, Kmieciak, M., additional, Idowu, M. O., additional, Bedognetti, D., additional, De Maria, A., additional, Bear, H. D., additional, Wang, E., additional, Marincola, F., additional, and Manjili, M., additional
- Published
- 2011
- Full Text
- View/download PDF
7. Cell receptor-ligand interaction, signalling, activation and apoptosis: 21. Pregnenolone Sulphate is Similar to Dexamethasone in Supressing the Unfettered Secretion of Hyaluronan: In Vitro Study on Cultured Synovial Fibroblasts from Patients with Longstanding Rheumatoid Arthritis
- Author
-
Ciurtin, C., primary, Majeed, Y., additional, Naylor, J., additional, Sukumar, P., additional, English, A., additional, Emery, P., additional, Beech, D., additional, Malik, N., additional, Lees, M., additional, Moradi, V., additional, Albrecht, W., additional, Laufer, S., additional, Schett, G., additional, Burnet, M., additional, Seed, M., additional, El Shikh, M. E., additional, El Sayed, R., additional, Kmieciak, M., additional, Manjili, M., additional, Szakal, A., additional, Pitzalis, C., additional, Tew, J., additional, Murphy, G., additional, Ryan, J., additional, Harney, S., additional, Shanahan, F., additional, Caplice, N., additional, and Molloy, M., additional
- Published
- 2011
- Full Text
- View/download PDF
8. Breakage of B cell tolerance by antigens on follicular dendritic cells
- Author
-
El Shikh, M. E., primary, El Sayed, R. M., additional, Kmieciak, M., additional, Manjili, M., additional, Szakal, A., additional, Pitzalis, C., additional, and Tew, J., additional
- Published
- 2011
- Full Text
- View/download PDF
9. Distinct Oligoclonal T Cells Are Associated With GVHD or GVHD-Free Responses in Patients With Hematologic Malignancies Following Stem Cell Transplantation
- Author
-
Toor, A.A., primary, Kmieciak, M., additional, Berrie, J.L., additional, Mallory, K.L., additional, Roberts, C.H., additional, Sabo, R., additional, Idowu, M., additional, Chung, H.M., additional, Veronica, B., additional, Clark, W.B., additional, McCarty, J.M., additional, Detwiler, M., additional, Kazim, L., additional, and Manjili, M.H., additional
- Published
- 2011
- Full Text
- View/download PDF
10. Database of emergency telephone calls - system tools for real-time registration and metadata searching.
- Author
-
Balcerek, J., Drgas, S., Kmieciak, M., Pawlowski, P., Konieczka, A., and Dabrowski, A.
- Published
- 2010
11. Investigation of kinetics of CH-radicals decay by cavity ring-down spectroscopy
- Author
-
Czyżewski, A., primary, Ernst, K., additional, Franssen, G., additional, Karasinski, G., additional, Kmieciak, M., additional, Lange, H., additional, Skubiszak, W., additional, and Stacewicz, T., additional
- Published
- 2002
- Full Text
- View/download PDF
12. Cloning and characterization of Arabidopsis thaliana AtNAP57--a homologue of yeast pseudouridine synthase Cbf5p.
- Author
-
Maceluch, J, primary, Kmieciak, M, additional, Szweykowska-Kulińska, Z, additional, and Jarmołowski, A, additional
- Published
- 2001
- Full Text
- View/download PDF
13. Comparison functioning and quality of life of patients with osteoarthritis and rheumatoid arthritis.
- Author
-
Bączyk, G., Samborski, P., Pieścikowska, J., Kmieciak, M., and Walkowiak, I.
- Subjects
QUALITY of life ,OSTEOARTHRITIS ,RHEUMATOID arthritis ,OUTPATIENT medical care ,GENDER ,AGE ,SCALING (Social sciences) ,PATIENTS - Abstract
Purpose: The aim of this study was to assess the quality of life of the osteoarthritis (OA) and rheumatoid arthritis (RA) patients of Outpatient Clinic Rehabilitation in Poznań. Material and methods: The study consisted of 97 OA patients, including 86 women and 11 men. Almost 123 patients with RA included of 102 women and 21 men. The mean age of the treated patients with OA was 11.50 and 11.10 years for RA patients. The Polish version of the Arthritis Impact Measurement Scales-2 (AIMS -2) was used to assess the quality of life. AIMS-2 scores range from 0-10, with 0 representing good quality of life, 10 representing poor quality of life. Results: It was showed that the mean score on the AIMS -2 for OA patients was: physical -- 3.53, affect -- 4.42, symptom -- 6.74, social interaction -- 3.33, role -- 4.20. Mean score on the AIMS-2 for RA patients was: physical -- 3.73, affect -- 4.48, symptom -- 7.09, social interaction -- 3.45, role -- 3.63. The quality of life depended on the sex of these patients. Women of OA and RA patients scored significantly higher in the physical state and symptom then men. Younger patients and suffering shorter than 5 years demonstrated higher evaluation of quality of life in the physical state. The assessment in most of the subscales of the AIMS-2 correlated significantly with Pain, Morning Stiffness and Grip Strength for OA and RA patients. Conclusion: This study showed that quality of life of OA and RA depends on gender, age and clinical variables. [ABSTRACT FROM AUTHOR]
- Published
- 2007
14. Ex vivo expansion of tumor-reactive T cells by means of bryostatin 1/ionomycin and the common gamma chain cytokines formulation
- Author
-
Kmieciak, M., Amir Ahmed Toor, Graham, L., Bear, H. D., and Manjili, M. H.
- Subjects
Mice ,Ionomycin ,T-Lymphocytes ,Immunology ,Animals ,Mammary Neoplasms, Experimental ,Female ,Mice, Transgenic ,Bryostatins ,Lymphocyte Activation ,Immunotherapy, Adoptive - Abstract
It was reported that breast cancer patients have pre-existing immune responses against their tumors(1,2). However, such immune responses fail to provide complete protection against the development or recurrence of breast cancer. To overcome this problem by increasing the frequency of tumor-reactive T cells, adoptive immunotherapy has been employed. A variety of protocols have been used for the expansion of tumor-specific T cells. These protocols, however, are restricted to the use of tumor antigens ex vivo for the activation of antigen-specific T cells. Very recently, common gamma chain cytokines such as IL-2, IL-7, IL-15, and IL-21 have been used alone or in combination for the enhancement of anti-tumor immune responses(3). However, it is not clear what formulation would work best for the expansion of tumor-reactive T cells. Here we present a protocol for the selective activation and expansion of tumor-reactive T cells from the FVBN202 transgenic mouse model of HER-2/neu positive breast carcinoma for use in adoptive T cell therapy of breast cancer. The protocol includes activation of T cells with bryostatin-1/ionomycin (B/I) and IL-2 in the absence of tumor antigens for 16 hours. B/I activation mimics intracellular signals that result in T cell activation by increasing protein kinase C activity and intracellular calcium, respectively(4). This protocol specifically activates tumor-specific T cells while killing irrelevant T cells. The B/I-activated T cells are cultured with IL-7 and IL-15 for 24 hours and then pulsed with IL-2. After 24 hours, T cells are washed, split, and cultured with IL-7+IL-15 for additional 4 days. Tumor-specificity and anti-tumor efficacy of the ex vivo expanded T cells is determined.
15. Database of emergency telephone calls - System tools for real-time registration and metadata searching
- Author
-
Julian Balcerek, Drgas, S., Kmieciak, M., Pawlowski, P., Konieczka, A., and Dabrowski, A.
16. Distinct signatures of the immune responses in low risk versus high risk neuroblastoma
- Author
-
Wang Ena, Ascierto Maria-Libera, Worschech Andrea, Kmieciak Maciej, Godder Kamar, Gowda Madhu, Marincola Francesco M, and Manjili Masoud H
- Subjects
Neuroblastoma ,innate immunity ,adaptive immunity ,prognostic biomarkers ,Medicine - Abstract
Abstract Background Over 90% of low risk (LR) neuroblastoma patients survive whereas less than 30% of high risk (HR) patients are long term survivors. Age (children younger than 18 months old) is associated with LR disease. Considering that adaptive immune system is well developed in older children, and that T cells were shown to be involved in tumor escape and progression of cancers, we sought to determine whether HR patients may tend to show a signature of adaptive immune responses compared to LR patients who tend to have diminished T-cell responses but an intact innate immune response. Methods We performed microarray analysis of RNA extracted from the tumor specimens of HR and LR patients. Flow cytometry was performed to determine the cellular constituents in the blood while multiplex cytokine array was used to detect the cytokine profile in patients' sera. A HR tumor cell line, SK-N-SH, was also used for detecting the response to IL-1β, a cytokines which is involved in the innate immune responses. Results Distinct patterns of gene expression were detected in HR and LR patients indicating an active T-cell response and a diminished adaptive immune response, respectively. A diminished adaptive immune response in LR patients was evident by higher levels of IL-10 in the sera. In addition, HR patients had lower levels of circulating myeloid derived suppressor cells (MDSC) compared with a control LR patient. LR patients showed slightly higher levels of cytokines of the innate immune responses. Treatment of the HR tumor line with IL-1β induced expression of cytokines of the innate immune responses. Conclusions This data suggests that adaptive immune responses may play an important role in the progression of HR disease whereas innate immune responses may be active in LR patients.
- Published
- 2011
- Full Text
- View/download PDF
17. Tumor escape and progression of HER-2/neu negative breast cancer under immune pressure
- Author
-
Wang Xiang-Yang, Wang Ena, Graham Laura, Ascierto Maria-Libera, Grimes Margaret M, Idowu Michael O, Payne Kyle K, Kmieciak Maciej, Bear Harry D, and Manjili Masoud H
- Subjects
Medicine - Abstract
Abstract Background Emerging data from pre-clinical and clinical studies suggest that HER-2/neu-specific T cell responses could induce HER-2/neu antigen loss in the tumor cells. These data suggest that patients with HER-2/neu negative breast cancer might have had HER-2/neu positive premalignant lesions in the past that progressed to HER-2/neu negative breast cancer under HER-2/neu-specific immune pressure. Methods We conducted a pilot study in patients with HER-2/neu positive and HER-2/neu negative breast cancers as well as a patient with ductal carcinoma in situ (DCIS). HER-2/neu expression was determined by FISH. HER-2/neu-specific T cell responses were determined by using IFN-γ ELISA. Expression of IFN-γ Rα in the tumors was determined by immunohistochemistry analysis of paraffin-embedded tissues. Results We determined that majority of (10 of 12) patients with HER-2/neu negative breast cancer had HER-2/neu-specific IFN-γ producing T cell responses which was stronger than those in patients with HER-2/neu positive tumors. Such immune responses were associated with nuclear translocation of IFN-γ Rα in their tumor cells. Patient with DCIS also showed HER-2/neu-specific T cell responses. Conclusion These data suggest that conducting retrospective studies in patients with HER-2/neu negative breast cancers and prospective studies in patients with HER-2/neu positive DCIS can determine whether HER-2/neu negative invasive carcinomas arise from HER-2/neu positive DCIS under the immune pressure.
- Published
- 2011
- Full Text
- View/download PDF
18. Human T cells express CD25 and Foxp3 upon activation and exhibit effector/memory phenotypes without any regulatory/suppressor function
- Author
-
Godder Kamar, Graham Laura, Gowda Madhu, Kmieciak Maciej, Bear Harry D, Marincola Francesco M, and Manjili Masoud H
- Subjects
Medicine - Abstract
Abstract Background Foxp3 has been suggested to be a standard marker for murine Tregs whereas its role as marker for human Tregs is controversial. While some reports have shown that human Foxp3+ T cells had no regulatory function others have shown their role in the inhibition of T cell proliferation. Methods T cell activation was performed by means of brayostatin-1/ionomycin (B/I), mixed lymphocyte reaction (MLR), and CD3/CD28 activation. T cell proliferation was performed using BrdU and CFSE staining. Flow cytometry was performed to determine Foxp3 expression, cell proliferation, viabilities and phenotype analyses of T cells. Results Both CD4+ and CD8+ T cells expressed Foxp3 upon activation in vitro. Expression of Foxp3 remained more stable in CD4+CD25+ T cells compared to that in CD8+CD25+ T cells. The CD4+CD25+Foxp3+ T cells expressed CD44 and CD62L, showing their effector and memory phenotypes. Both FoxP3- responder T cells and CD4+FoxP3+ T cells underwent proliferation upon CD3/CD28 activation. Conclusion Expression of Foxp3 does not necessarily convey regulatory function in human CD4+CD25+ T cells. Increased FoxP3 on CD44+ effector and CD44+CD62L+ memory T cells upon stimulation suggest the activation-induced regulation of FoxP3 expression.
- Published
- 2009
- Full Text
- View/download PDF
19. Combined MEK1/2 and ATR inhibition promotes myeloma cell death through a STAT3-dependent mechanism in vitro and in vivo.
- Author
-
Li L, Hu X, Nkwocha J, Kmieciak M, Meads MB, Shain KH, Alugubelli RR, Silva AS, Mann H, Sudalagunta PR, Canevarolo RR, Zhou L, and Grant S
- Abstract
Mechanisms underlying potentiation of the anti-myeloma (MM) activity of ataxia telangiectasia Rad3 (ATR) antagonists by MAPK (Mitogen-activated protein kinases)-related extracellular kinase 1/2 (MEK1/2) inhibitors were investigated. Co-administration of the ATR inhibitor (ATRi) BAY1895344 (BAY) and MEK1/2 inhibitors, for example, cobimetinib, synergistically increased cell death in diverse MM cell lines. Mechanistically, BAY and cobimetinib blocked STAT3 Tyr705 and Ser727 phosphorylation, respectively, and dual dephosphorylation triggered marked STAT3 inactivation and downregulation of STAT3 (Signal transducer and activator of transcription 3) downstream targets (c-Myc and BCL-X
L ). Similar events occurred in highly bortezomib-resistant (PS-R) cells, in the presence of patient-derived conditioned medium, and with alternative ATR (e.g. M1774) and MEK1/2 (trametinib) inhibitors. Notably, constitutively active STAT3 c-MYC or BCL-XL ectopic expression significantly protected cells from BAY/cobimetinib. In contrast, transfection of cells with a dominant-negative form of STAT3 (Y705F) sensitized cells to cobimetinib, as did ATR shRNA knockdown. Conversely, MEK1/2 knockdown markedly increased ATRi sensitivity. The BAY/cobimetinib regimen was also active against primary CD138+ MM cells, but not normal CD34+ cells. Finally, the ATR inhibitor/cobimetinib regimen significantly improved survival in MM xenografts, including bortezomib-resistant models, with minimal toxicity. Collectively, these findings suggest that combined ATR/MEK1/2 inhibition triggers dual STAT3 Tyr705 and Ser727 dephosphorylation, pronounced downregulation of cytoprotective targets and MM cell death, warranting attention as a novel therapeutic strategy in MM., (© 2024 British Society for Haematology and John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
20. A phase 1 study of regorafenib and sildenafil in adults with advanced solid tumors.
- Author
-
Poklepovic AS, Gordon SW, Kothadia S, McGuire WP, Thacker LR, Deng X, Tombes MB, Shrader E, Hudson D, Bandyopadhyay D, Ryan AA, Kmieciak M, Smith S, and Dent P
- Subjects
- Adult, Female, Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Maximum Tolerated Dose, Phenylurea Compounds adverse effects, Pyridines therapeutic use, Sildenafil Citrate adverse effects, Genital Neoplasms, Female chemically induced, Genital Neoplasms, Female drug therapy, Neoplasms drug therapy, Neoplasms pathology
- Abstract
The purpose of this study is to establish the recommended phase 2 dose for regorafenib in combination with sildenafil for patients with advanced solid tumors. Secondary outcomes included identification of antitumor effects of regorafenib and sildenafil, toxicity of the combination, determination of PDE5 expression in tumor samples, and the impact of sildenafil on the pharmacokinetics of regorafenib. This study was a phase 1, open-label single-arm dose-escalation trial using a 3 + 3 design. Additional patients were enrolled at the maximum tolerated dose (MTD) until a total of 12 patients were treated at the MTD. A total of 29 patients were treated in this study. The median duration of treatment was 8 weeks. The recommended phase 2 doses determined in this study are regorafenib 160 mg daily with sildenafil 100 mg daily. The most common toxicities included palmar-plantar erythrodysesthesia syndrome (20 patients, 69%) and hypophosphatemia (18 patients, 62%). Two patients (7%) experienced grade 4 lipase increase. Objective responses were not observed; however, 14 patients (48%) had a period of stable disease during the study. Stable disease for up to 12 months was observed in patients with ovarian cancer as well as up to 20 months for a patient with cervical cancer. The combination of regorafenib and sildenafil at the recommended phase 2 dose is safe and generally well tolerated. Disease control in patients with gynecologic malignancies was especially encouraging. Further evaluation of the combination of regorafenib and sildenafil in gynecologic malignancies is warranted. Clinical Trial Registration Number: NCT02466802., (Copyright © 2024 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
21. Phase 2 Study of Sorafenib, Valproic Acid, and Sildenafil in the Treatment of Recurrent High-Grade Glioma.
- Author
-
Poklepovic AS, Shah P, Tombes MB, Shrader E, Bandyopadhyay D, Deng X, Roberts CH, Ryan AA, Hudson D, Sankala H, Kmieciak M, Dent P, and Malkin MG
- Abstract
Here we report the results of a single-center phase 2 clinical trial combining sorafenib tosylate, valproic acid, and sildenafil for the treatment of patients with recurrent high-grade glioma (NCT01817751). Clinical toxicities were grade 1 and grade 2, with one grade 3 toxicity for maculopapular rash (6.4%). For all evaluable patients, the median progression-free survival was 3.65 months and overall survival (OS) 10.0 months. There was promising evidence showing clinical activity and benefit. In the 33 evaluable patients, low protein levels of the chaperone GRP78 (HSPA5) was significantly associated with a better OS (p < 0.0026). A correlation between the expression of PDGFRα and OS approached significance (p < 0.0728). Five patients presently have a mean OS of 73.6 months and remain alive. This is the first therapeutic intervention glioblastoma trial to significantly associate GRP78 expression to OS. Our data suggest that the combination of sorafenib tosylate, valproic acid, and sildenafil requires additional clinical development in the recurrent glioma population., Competing Interests: The authors have no conflict of interest to disclose.
- Published
- 2024
- Full Text
- View/download PDF
22. Non-canonical role for the ataxia-telangiectasia-Rad3 pathway in STAT3 activation in human multiple myeloma cells.
- Author
-
Li L, Hu X, Nkwocha J, Sharma K, Kmieciak M, Mann H, Zhou L, and Grant S
- Subjects
- Humans, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Apoptosis, Cell Line, Tumor, bcl-X Protein genetics, Phosphorylation, RNA, Small Interfering metabolism, STAT3 Transcription Factor metabolism, Multiple Myeloma genetics, Multiple Myeloma metabolism, Ataxia Telangiectasia
- Abstract
Purpose: The goal of this study was to characterize the relationship between ATR and STAT3 interactions in human multiple myeloma (MM) cells., Methods: Various MM cell lines, including IL-6-dependent cells were exposed to ATR inhibitors and effects on STAT3 Tyr705 and Ser727 were monitored by WB analysis and ImageStream analysis. Parallel studies examined induction of cell death, STAT3 DNA binding activity, and expression of STAT3 downstream targets (BCL-X
L , MCL-1, c-MYC). Validation was obtained in ATR shRNA knock-down cells, and in cells ectopically expressing BCL-XL , MCL-1, or c-MYC. Analogous studies were performed in primary MM cells and in a MM xenograft model., Results: Multiple pharmacologic ATR inhibitors inhibited STAT3 Tyr705 (but not Ser727) phosphorylation at low uM concentrations and down-regulated BCL-XL , MCL-1, c-MYC in association with cell death induction. Compatible results were observed in ATR shRNA knock-down cells. Cell death induced by ATR inhibitors was significantly attenuated in cells ectopically expressing constitutively active STAT3, BCL-XL , MCL-1, or c-MYC. Concordant results were observed in primary human MM cells and in an in vivo MM xenograft model., Conclusions: Collectively, these findings argue for a non-canonical role for the ATR kinase in STAT3 activation in MM cells, and suggest that STAT3 inactivation contributes to the lethal actions of ATR inhibitors in MM., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
23. Dual mTORC1/2 Inhibition Synergistically Enhances AML Cell Death in Combination with the BCL2 Antagonist Venetoclax.
- Author
-
Satta T, Li L, Chalasani SL, Hu X, Nkwocha J, Sharma K, Kmieciak M, Rahmani M, Zhou L, and Grant S
- Subjects
- Humans, Myeloid Cell Leukemia Sequence 1 Protein genetics, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Mechanistic Target of Rapamycin Complex 1, bcl-2-Associated X Protein genetics, bcl-2-Associated X Protein metabolism, Proto-Oncogene Proteins c-akt, Cell Line, Tumor, Apoptosis, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cell Death, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-bcl-2, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism
- Abstract
Purpose: Acute myelogenous leukemia (AML) is an aggressive disease with a poor outcome. We investigated mechanisms by which the anti-AML activity of ABT-199 (venetoclax) could be potentiated by dual mTORC1/TORC2 inhibition., Experimental Design: Venetoclax/INK128 synergism was assessed in various AML cell lines and primary patient AML samples in vitro. AML cells overexpressing MCL-1, constitutively active AKT, BAK, and/or BAX knockout, and acquired venetoclax resistance were investigated to define mechanisms underlying interactions. The antileukemic efficacy of this regimen was also examined in xenograft and patient-derived xenograft (PDX) models., Results: Combination treatment with venetoclax and INK128 (but not the mTORC1 inhibitor rapamycin) dramatically enhanced cell death in AML cell lines. Synergism was associated with p-AKT and p-4EBP1 downregulation and dependent upon MCL-1 downregulation and BAK/BAX upregulation as MCL-1 overexpression and BAX/BAK knockout abrogated cell death. Constitutive AKT activation opposed synergism between venetoclax and PI3K or AKT inhibitors, but not INK128. Combination treatment also synergistically induced cell death in venetoclax-resistant AML cells. Similar events occurred in primary patient-derived leukemia samples but not normal CD34+ cells. Finally, venetoclax and INK128 co-treatment displayed increased antileukemia effects in in vivo xenograft and PDX models., Conclusions: The venetoclax/INK128 regimen exerts significant antileukemic activity in various preclinical models through mechanisms involving MCL-1 downregulation and BAK/BAX activation, and offers potential advantages over PI3K or AKT inhibitors in cells with constitutive AKT activation. This regimen is active against primary and venetoclax-resistant AML cells, and in in vivo AML models. Further investigation of this strategy appears warranted., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
24. Phase 1 study of belinostat and adavosertib in patients with relapsed or refractory myeloid malignancies.
- Author
-
Shafer D, Kagan AB, Rudek MA, Kmieciak M, Tombes MB, Shrader E, Bandyopadhyay D, Hudson D, Sankala H, Weir C, Lancet JE, and Grant S
- Subjects
- Humans, Animals, Mice, Pyrimidinones therapeutic use, Hydroxamic Acids adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology
- Abstract
Purpose: Belinostat is an intravenous histone deacetylase inhibitor with approval for T-cell lymphomas. Adavosertib is a first in class oral Wee1 inhibitor. Preclinical studies of the combination demonstrated synergy in various human acute myeloid leukemia (AML) lines as well as AML xenograft mouse models., Experimental Design: This was a phase 1 dose-escalation study of belinostat and adavosertib in patients with relapsed/refractory AML and myelodysplastic syndrome (MDS). Patients received both drugs on days 1-5 and 8-12 of a 21-day cycle. Safety and toxicity were monitored throughout the study. Plasma levels of both drugs were measured for pharmacokinetic analysis. Response was determined by standard criteria including bone marrow biopsy., Results: Twenty patients were enrolled and treated at 4 dose levels. A grade 4 cytokine release syndrome at dose level 4 (adavosertib 225 mg/day; belinostat 1000 mg/m
2 ) qualified as a dose-limiting toxicity event. The most common non-hematologic treatment-related adverse events were nausea, vomiting, diarrhea, dysgeusia, and fatigue. No responses were seen. The study was terminated prior to maximum tolerated dose/recommended phase 2 dose determination., Conclusions: The combination of belinostat and adavosertib at the tested dose levels was feasible but without efficacy signals in the relapsed/refractory MDS/AML population., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2023
- Full Text
- View/download PDF
25. Phase 1 study of belinostat (PXD-101) and bortezomib (Velcade, PS-341) in patients with relapsed or refractory acute leukemia and myelodysplastic syndrome.
- Author
-
Holkova B, Shafer D, Yazbeck V, Dave S, Bose P, Tombes MB, Shrader E, Wan W, Bandyopadhyay D, Weir C, Collins EB, Garnett A, Kmieciak M, Roberts JD, Garcia-Manero G, and Grant S
- Subjects
- Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib therapeutic use, Humans, Hydroxamic Acids adverse effects, Sulfonamides, Treatment Outcome, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics
- Abstract
We report the results of a phase 1 dose-escalation study of belinostat and bortezomib in adult patients with acute leukemia or MDS or CML with blast crisis. Thirty-eight patients received IV belinostat days 1-5 and 8-12 with IV bortezomib days 1, 4, 8, and 11 every 21 days. QTc prolongation was the only identified DLT. The RP2Ds were 1.3 mg/m
2 bortezomib and 1000 mg/m2 belinostat. One patient with highly refractory MLL-ENL rearranged biphenotypic AML with multiple karyotypic aberrations had a complete pathologic and karyotypic response. One patient with post-MPN AML remained on study with stable disease (SD) for 32 cycles. Whole-exome sequencing revealed no aberrations in the first patient and a hyper-mutator genotype in the second. Eighteen patients had a best response of SD. We conclude that this treatment strategy is feasible but has limited activity in this population. Nevertheless, the factors that predict exceptional responses to this strategy warrant further investigation.- Published
- 2021
- Full Text
- View/download PDF
26. The Covalent CDK7 Inhibitor THZ1 Potently Induces Apoptosis in Multiple Myeloma Cells In Vitro and In Vivo .
- Author
-
Zhang Y, Zhou L, Bandyopadhyay D, Sharma K, Allen AJ, Kmieciak M, and Grant S
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis genetics, Bortezomib pharmacology, Bortezomib therapeutic use, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Cyclin-Dependent Kinases genetics, Down-Regulation drug effects, Drug Synergism, Gene Expression Regulation, Neoplastic drug effects, Gene Knockout Techniques, Humans, Inhibitory Concentration 50, Mice, Multiple Myeloma pathology, Myeloid Cell Leukemia Sequence 1 Protein genetics, Oligopeptides pharmacology, Oligopeptides therapeutic use, Phenylenediamines therapeutic use, Proto-Oncogene Proteins c-myc genetics, Pyrimidines therapeutic use, Sulfonamides pharmacology, Sulfonamides therapeutic use, Transcription, Genetic drug effects, Xenograft Model Antitumor Assays, bcl-X Protein genetics, Cyclin-Dependent Kinase-Activating Kinase, Antineoplastic Combined Chemotherapy Protocols pharmacology, Apoptosis drug effects, Cyclin-Dependent Kinases antagonists & inhibitors, Multiple Myeloma drug therapy, Phenylenediamines pharmacology, Pyrimidines pharmacology
- Abstract
Purpose: The goal of this study was to characterize the activity of the covalent CDK7 inhibitor THZ1 in multiple myeloma models., Experimental Design: Multiple myeloma lines were exposed to varying THZ1 concentrations alone or with carfilzomib or ABT-199, after which apoptosis was monitored by flow cytometry, protein expression by Western blot analysis, mRNA by RT-PCR. Analogous studies were performed in cells ectopically expressing c-MYC, MCL-1, or BCL-XL, or CRISPER-Cas CDK7 sgRNA knockout. Primary multiple myeloma cells were exposed to THZ1 ± carfilzomib or ABT-199. In vivo effects of THZ1 were examined in a systemic U266 xenograft model., Results: THZ1 markedly diminished multiple myeloma cell proliferation and survival despite bortezomib or stromal cell resistance in association with G
2 -M arrest, inactivation of CTD RNA Pol II, dephosphorylation of CDKs 7 as well as 1, 2, and 9, and MCL-1, BCL-xL, and c-MYC mRNA or protein downregulation. Ectopic MCL-1, c-MYC, or BCL-XL expression significantly protected cells from THZ1 lethality. Both THZ1 and CRISPR-Cas CDK7 knockout sharply diminished multiple myeloma cell proliferation and significantly increased carfilzomib and ABT-199 lethality. Parallel effects and interactions were observed in primary CD138+ ( N = 22) or primitive multiple myeloma cells (CD138- /CD19+ /CD20+ /CD27+ ; N = 16). THZ1 administration [10 mg/kg i.p. twice daily (BID), 5 days/week] significantly improved survival in a systemic multiple myeloma xenograft model with minimal toxicity and induced similar events observed in vitro , for example, MCL-1 and c-MYC downregulation., Conclusions: THZ1 potently reduces multiple myeloma cell proliferation through transcriptional downregulation of MCL-1, BCL-XL , and c-MYC in vitro and in vivo . It warrants further attention as a therapeutic agent in multiple myeloma., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
27. Correction: Synergistic interactions between PLK1 and HDAC inhibitors in non-Hodgkin's lymphoma cells occur in vitro and in vivo and proceed through multiple mechanisms.
- Author
-
Nguyen T, Parker R, Hawkins E, Holkova B, Yazbeck V, Kolluri A, Kmieciak M, Rahmani M, and Grant S
- Abstract
[This corrects the article DOI: 10.18632/oncotarget.15649.].
- Published
- 2019
- Full Text
- View/download PDF
28. Phase I Study of Sorafenib and Vorinostat in Advanced Hepatocellular Carcinoma.
- Author
-
Gordon SW, McGuire WP 3rd, Shafer DA, Sterling RK, Lee HM, Matherly SC, Roberts JD, Bose P, Tombes MB, Shrader EE, Ryan AA, Kmieciak M, Nguyen T, Deng X, Bandyopadhyay D, Dent P, and Poklepovic AS
- Subjects
- Aged, Chemical and Drug Induced Liver Injury etiology, Drug Eruptions etiology, Female, Humans, Hypokalemia chemically induced, Hypophosphatemia chemically induced, Male, Middle Aged, Sorafenib administration & dosage, Thrombocytopenia chemically induced, Vorinostat administration & dosage, Vorinostat adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy
- Abstract
Objectives: Preclinical data suggest histone deacetylase inhibitors improve the therapeutic index of sorafenib. A phase I study was initiated to establish the recommended phase 2 dose of sorafenib combined with vorinostat in patients with unresectable hepatocellular carcinoma., Materials and Methods: Patients received vorinostat (200 to 400 mg by mouth once daily, 5 of 7 d) and sorafenib at standard or reduced doses (400 mg [cohort A] or 200 mg [cohort B] by mouth twice daily). Patients who received 14 days of vorinostat in cycle 1 were evaluable for dose-limiting toxicity (DLT)., Results: Sixteen patients were treated. Thirteen patients were evaluable for response. Three patients experienced DLTs, 2 in cohort A (grade [gr] 3 hypokalemia; gr 3 maculopapular rash) and 1 in cohort B (gr 3 hepatic failure; gr 3 hypophosphatemia; gr 4 thrombocytopenia). Eleven patients required dose reductions or omissions for non-DLTtoxicity. Ten patients (77%) had stable disease (SD). The median treatment duration was 4.7 months for response-evaluable patients. One patient with SD was on treatment for 29.9 months, and another patient, also with SD, was on treatment for 18.7 months. Another patient electively stopped therapy after 15 months and remains without evidence of progression 3 years later., Conclusions: Although some patients had durable disease control, the addition of vorinostat to sorafenib led to toxicities in most patients, requiring dose modifications that prevented determination of the recommended phase 2 dose. The combination is not recommended for further exploration with this vorinostat schedule in this patient population.
- Published
- 2019
- Full Text
- View/download PDF
29. The IAP antagonist birinapant potentiates bortezomib anti-myeloma activity in vitro and in vivo.
- Author
-
Zhou L, Zhang Y, Leng Y, Dai Y, Kmieciak M, Kramer L, Sharma K, Wang Y, Craun W, and Grant S
- Subjects
- Animals, Antineoplastic Agents pharmacology, Bortezomib pharmacology, Dipeptides pharmacology, Disease Models, Animal, Humans, Indoles pharmacology, Mice, Mice, Inbred NOD, Mice, SCID, Antineoplastic Agents therapeutic use, Bortezomib therapeutic use, Dipeptides therapeutic use, Indoles therapeutic use, Multiple Myeloma drug therapy
- Abstract
Background: Mechanisms by which Smac mimetics (SMs) interact with proteasome inhibitors (e.g., bortezomib) are largely unknown, particularly in multiple myeloma (MM), a disease in which bortezomib represents a mainstay of therapy., Methods: Interactions between the clinically relevant IAP (inhibitor of apoptosis protein) antagonist birinapant (TL32711) and the proteasome inhibitor bortezomib were investigated in multiple myeloma (MM) cell lines and primary cells, as well as in vivo models. Induction of apoptosis and changes in gene and protein expression were monitored using MM cell lines and confirmed in primary MM cell populations. Genetically modified cells (e.g., exhibiting shRNA knockdown or ectopic expression) were employed to evaluate the functional significance of birinapant/bortezomib-induced changes in protein levels. A MM xenograft model was used to evaluate the in vivo activity of the birinapant/bortezomib regimen., Results: Birinapant and bortezomib synergistically induced apoptosis in diverse cell lines, including bortezomib-resistant cells (PS-R). The regimen robustly downregulated cIAP1/2 but not the canonical NF-κB pathway, reflected by p65 phosphorylation and nuclear accumulation. In contrast, the bortezomib/birinapant regimen upregulated TRAF3, downregulated TRAF2, and diminished p52 processing and BCL-X
L expression, consistent with disruption of the non-canonical NF-κB pathway. TRAF3 knockdown, ectopic TRAF2, or BCL-XL expression significantly diminished birinapant/bortezomib toxicity. The regimen sharply increased extrinsic apoptotic pathway activation, and cells expressing dominant-negative FADD or caspase-8 displayed markedly reduced birinapant/bortezomib sensitivity. Primary CD138+ (n = 43) and primitive MM populations (CD138- /19+ /20+ /27+ ; n = 31) but not normal CD34+ cells exhibited significantly enhanced toxicity with combined treatment (P < 0.0001). The regimen was also fully active in the presence of HS-5 stromal cells or growth factors (e.g., IL-6 and VEGF). Finally, the regimen was well tolerated and significantly increased survival (P < 0.05 and P < 0.001) compared to single agents in a MM xenograft model. Combined treatment also downregulated cIAP1/2 and p52 while increasing PARP cleavage in MM cells in vivo., Conclusions: Our data suggest that birinapant and bortezomib interact synergistically in MM cells, including those resistant to bortezomib, through inactivation of the non-canonical NF-κB and activation of the extrinsic apoptotic pathway both in vitro and in vivo. They also argue that a strategy combining cIAP antagonists and proteasome inhibitors warrants attention in MM.- Published
- 2019
- Full Text
- View/download PDF
30. Homoharringtonine interacts synergistically with bortezomib in NHL cells through MCL-1 and NOXA-dependent mechanisms.
- Author
-
Nguyen T, Parker R, Zhang Y, Hawkins E, Kmieciak M, Craun W, and Grant S
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Down-Regulation drug effects, Drug Synergism, Humans, Lymphoma, Large B-Cell, Diffuse metabolism, Lymphoma, Mantle-Cell metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Proteasome Inhibitors pharmacology, Signal Transduction drug effects, Up-Regulation drug effects, Xenograft Model Antitumor Assays, Bortezomib pharmacology, Homoharringtonine pharmacology, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Mantle-Cell drug therapy, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism
- Abstract
Background: Interactions between the protein synthesis inhibitor homoharringtonine (HHT) and the proteasome inhibitor bortezomib were investigated in DLBCL and mantle cell lymphoma cells (MCL)., Methods: Various DLBCL and MCL cells were exposed to HHT and bortezomib alone or together after which apoptosis and signaling pathway perturbations were monitored by flow cytometry and Western blot analysis. Xenograft mouse models were used to assess tumor growth and animal survival., Results: HHT and bortezomib co-administration synergistically induced apoptosis in GC-, ABC- and double-hit DLBCL cells. Similar interactions were observed in MCL cells and in primary lymphoma cells. HHT/bortezomib co-administration diminished binding of MCL-1 to both BAK and NOXA. Knock-down of NOXA significantly diminished lethality whereas MCL-1 knock-down or ectopic NOXA expression increased cell death. Notably, HHT/bortezomib lethality was dramatically reduced in BAK knockout or knockdown cells. Finally, HHT/bortezomib co-administration significantly improved survival compared to single agents in GC- and ABC- xenograft models while exhibiting little toxicity., Conclusions: These findings indicate that HHT and bortezomib cooperate to kill DLBCL and MCL cells through a process involving MCL-1 down-regulation, NOXA up-regulation, and BAK activation. They also suggest that a strategy combining HHT with bortezomib warrants attention in DLBCL and MCL.
- Published
- 2018
- Full Text
- View/download PDF
31. A Phase II Trial of Bortezomib and Vorinostat in Mantle Cell Lymphoma and Diffuse Large B-cell Lymphoma.
- Author
-
Yazbeck V, Shafer D, Perkins EB, Coppola D, Sokol L, Richards KL, Shea T, Ruan J, Parekh S, Strair R, Flowers C, Morgan D, Kmieciak M, Bose P, Kimball A, Badros AZ, Baz R, Lin HY, Zhao X, Reich RR, Tombes MB, Shrader E, Sankala H, Roberts JD, Sullivan D, Grant S, and Holkova B
- Subjects
- Adult, Aged, Aged, 80 and over, Bortezomib administration & dosage, Female, Follow-Up Studies, Humans, Lymphoma, Large B-Cell, Diffuse pathology, Lymphoma, Mantle-Cell pathology, Male, Middle Aged, Neoplasm Recurrence, Local pathology, Prognosis, Prospective Studies, Survival Rate, Vorinostat administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Drug Resistance, Neoplasm drug effects, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Mantle-Cell drug therapy, Neoplasm Recurrence, Local drug therapy, Salvage Therapy
- Abstract
Background: The proteasome inhibitor bortezomib has demonstrated marked preclinical activity when combined with the histone deacetylase inhibitor vorinostat in leukemia, multiple myeloma, and mantle cell lymphoma (MCL) cells. The present study evaluated the efficacy and safety of the combination in patients with relapsed or refractory MCL and diffuse large B-cell lymphoma (DLBCL)., Patients and Methods: The present multicenter, nonrandomized phase II trial used a Simon 2-stage design with 3 cohorts: cohort A, MCL with no previous bortezomib (including untreated MCL); cohort B, MCL with previous bortezomib; and cohort C, relapsed or refractory DLBCL with no previous bortezomib. Vorinostat (400 mg) was administered orally on days 1 to 5 and 8 to 12 before bortezomib (1.3 mg/m
2 ), which was administered intravenously on days 1, 4, 8, and 11 of each 21-day cycle., Results: For the 65 treated patients (22 in cohort A, 4 in cohort B, and 39 in cohort C), the overall response rate was 31.8%, 0%, and 7.7%, respectively. The median progression-free survival was 7.6 months for cohort A and 1.8 months for cohort C. In cohort A, 7 patients had a partial response (PRs), 5 had stable disease (SD), 7 had progressive disease (PD), 1 was not assessed, and 2 were not evaluable. In cohort B, 2 had SD and 2 had PD. In cohort C, 3 had a PR, 8 had SD, 23 had PD, and 5 were not assessed. Baseline NF-κB activation, measured as nuclear RelA by immunohistochemistry, did not correlate with clinical response., Conclusion: The combination of bortezomib and vorinostat is safe and has modest activity in MCL and limited activity in DLBCL., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
32. Cotargeting BCL-2 and PI3K Induces BAX-Dependent Mitochondrial Apoptosis in AML Cells.
- Author
-
Rahmani M, Nkwocha J, Hawkins E, Pei X, Parker RE, Kmieciak M, Leverson JD, Sampath D, Ferreira-Gonzalez A, and Grant S
- Subjects
- Animals, Apoptosis drug effects, Apoptosis Regulatory Proteins metabolism, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cell Line, Tumor, Down-Regulation drug effects, Heterografts, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Mitochondria drug effects, Pyrimidines pharmacology, Sulfonamides pharmacology, U937 Cells, Apoptosis physiology, Leukemia, Myeloid, Acute metabolism, Mitochondria metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, bcl-2-Associated X Protein metabolism
- Abstract
Inhibitors targeting BCL-2 apoptotic proteins have significant potential for the treatment of acute myeloid leukemia (AML); however, complete responses are observed in only 20% of patients, suggesting that targeting BCL-2 alone is insufficient to yield durable responses. Here, we assessed the efficacy of coadministration of the PI3K/mTOR inhibitor GDC-0980 or the p110β-sparing PI3K inhibitor taselisib with the selective BCL-2 antagonist venetoclax in AML cells. Tetracycline-inducible downregulation of BCL-2 significantly sensitized MV4-11 and MOLM-13 AML cells to PI3K inhibition. Venetoclax/GDC-0980 coadministration induced rapid and pronounced BAX mitochondrial translocation, cytochrome c release, and apoptosis in various AML cell lines in association with AKT/mTOR inactivation and MCL-1 downregulation; ectopic expression of MCL-1 significantly protected cells from this regimen. Combined treatment was also effective against primary AML blasts from 17 patients, including those bearing various genetic abnormalities. Venetoclax/GDC-0980 markedly induced apoptosis in primitive CD34
+ /38- /123+ AML cell populations but not in normal hematopoietic progenitor CD34+ cells. The regimen was also active against AML cells displaying intrinsic or acquired venetoclax resistance or tumor microenvironment-associated resistance. Either combinatorial treatment markedly reduced AML growth and prolonged survival in a systemic AML xenograft mouse model and diminished AML growth in two patient-derived xenograft models. Venetoclax/GDC-0980 activity was partially diminished in BAK-/- cells and failed to induce apoptosis in BAX-/- and BAX-/- BAK-/- cells, whereas BIM-/- cells were fully sensitive. Similar results were observed with venetoclax alone in in vitro and in vivo systemic xenograft models. Collectively, these studies demonstrate that venetoclax/GDC-0980 exhibits potent anti-AML activity primarily through BAX and, to a lesser extent, BAK. These findings argue that dual BCL-2 and PI3K inhibition warrants further evaluation in AML. Significance: Combined treatment with clinically relevant PI3K and BCL-2 inhibitors may prove effective in the treatment of acute myeloid leukemia. Cancer Res; 78(11); 3075-86. ©2018 AACR ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
33. Flavopiridol enhances ABT-199 sensitivity in unfavourable-risk multiple myeloma cells in vitro and in vivo.
- Author
-
Zhou L, Zhang Y, Sampath D, Leverson J, Dai Y, Kmieciak M, Nguyen M, Orlowski RZ, and Grant S
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bcl-2-Like Protein 11 genetics, Bcl-2-Like Protein 11 metabolism, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Cell Line, Tumor, Cyclin-Dependent Kinase 9 antagonists & inhibitors, Cyclin-Dependent Kinase 9 genetics, Cyclin-Dependent Kinase 9 metabolism, Down-Regulation drug effects, Drug Resistance, Neoplasm, Drug Synergism, Flavonoids administration & dosage, Gene Knockdown Techniques, Humans, Mice, Myeloid Cell Leukemia Sequence 1 Protein genetics, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Neoplasm Transplantation, Piperidines administration & dosage, Primary Cell Culture, Proto-Oncogene Proteins c-bcl-2 metabolism, Risk Assessment, Sulfonamides administration & dosage, Up-Regulation drug effects, Antineoplastic Agents pharmacology, Apoptosis drug effects, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Flavonoids pharmacology, Multiple Myeloma drug therapy, Piperidines pharmacology, Sulfonamides pharmacology
- Abstract
Background: The BCL-2-specific BH3-mimetic ABT-199 (venetoclax) has been reported to be principally active against favourable-risk multiple myeloma (MM) cells, prompting efforts to extend its activity to include more resistant, higher-risk MM subsets., Methods: Effects of the CDK9 inhibitor flavopiridol (FP; alvocidib) on responses to ABT-199 were examined in MM cells. Cell death and protein expression were evaluated by western blot and immunofluorescence. Xenograft models were used to study combination effects in vivo., Results: FP synergistically increased ABT-199 lethality in both ABT-199-sensitive and insensitive MM cells. FP blocked CDK9 activation/positive transcription elongation factor B phosphorylation, downregulated MCL-1, increased BCL-2/MCL-1 ratios, and upregulated BIM. MCL-1 ectopic expression or knockdown in MM cells significantly diminished or increased ABT-199 sensitivity, respectively. CDK9 knockdown triggered MCL-1 downregulation and increased ABT-199 activity, whereas BIM knockdown significantly reduced FP/ABT-199 lethality. FP also enhanced ABT-199 lethality in unfavourable prognosis primary MM cells. HS-5 cell co-culture failed to protect MM cells from the FP/ABT-199 regimen, suggesting circumvention of microenvironmental signals. Finally, FP/ABT-199 significantly increased survival in systemic xenograft and immune-competent MM models while exhibiting minimal toxicity., Conclusions: These findings argue that CDK9 inhibitors, for example, FP may increase the antimyeloma activity of ABT-199, including in unfavourable-risk MM minimally responsive to ABT-199 alone.
- Published
- 2018
- Full Text
- View/download PDF
34. Randomized study of doxorubicin-based chemotherapy regimens, with and without sildenafil, with analysis of intermediate cardiac markers.
- Author
-
Poklepovic A, Qu Y, Dickinson M, Kontos MC, Kmieciak M, Schultz E, Bandopadhyay D, Deng X, and Kukreja RC
- Abstract
Background: Doxorubicin chemotherapy is used across a range of adult and pediatric malignancies. Cardiac toxicity is common, and dysfunction develops over time in many patients. Biomarkers used for predicting late cardiac dysfunction following doxorubicin exposure have shown promise. Preclinical studies have demonstrated potential cardioprotective effects of sildenafil., Methods: We sought to confirm the safety of adding sildenafil to doxorubicin-based chemotherapy and assess N-terminal Pro-Brain Natriuretic Peptide (NT-proBNP) and high sensitivity cardiac troponin I (hsTnI) as early markers of anthracycline-induced cardiotoxicity. We randomized 27 patients (ages 31-77, 92.3% female) receiving doxorubicin chemotherapy using a blocked randomization scheme with randomly permuted block sizes to receive standard chemotherapy alone or with the addition of sildenafil. The study was not blinded. Sildenafil was dosed at 100 mg by mouth daily during therapy; patients took sildenafil three times daily on the day of doxorubicin. Doxorubicin dosing and schedule were dependent on the treatment regimen. Echocardiography was obtained prior to initiation of treatment and routinely thereafter up to 4 years. NT-proBNP and hsTnI were obtained with each cycle before, 1-3 h after, and 24 h after doxorubicin., Results: Fourteen patients were randomized to receive standard doxorubicin chemotherapy alone (14 treated and analyzed), while 13 patients were randomized to the experimental doxorubicin and sildenafil arm (10 treated and analyzed). No toxicity signal was seen with the addition of sildenafil to doxorubicin-based regimens. There was no statistical difference between the treatment arms in relation to the change of mean left ventricular ejection fraction (LVEF) between the first and last evaluation. In both arms, hsTnI levels increased over time; however, elevated hsTnI was not associated with declines in LVEF., Conclusion: Adding sildenafil was safe, but did not offer cardioprotection following doxorubicin treatment. Increases in hsTnI levels were observed over time, but elevations during therapy did not correlate with subsequent decreases in LVEF., Trial Registration: This clinical trial (NCT01375699) was registered at www.clinicaltrials.gov on June 17, 2011., Competing Interests: Competing interests The authors declare that they have no competing interests.
- Published
- 2018
- Full Text
- View/download PDF
35. A phase 1 study of bortezomib and romidepsin in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma, indolent B-cell lymphoma, peripheral T-cell lymphoma, or cutaneous T-cell lymphoma.
- Author
-
Holkova B, Yazbeck V, Kmieciak M, Bose P, Ma S, Kimball A, Tombes MB, Shrader E, Wan W, Weir-Wiggins C, Singh A, Hogan KT, Conine S, Sankala H, Roberts JD, Shea TC, and Grant S
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib administration & dosage, Combined Modality Therapy, Depsipeptides administration & dosage, Female, Humans, Leukemia, Lymphocytic, Chronic, B-Cell diagnosis, Lymphoma, T-Cell, Cutaneous diagnosis, Lymphoma, T-Cell, Peripheral diagnosis, Male, Maximum Tolerated Dose, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Lymphoma, T-Cell, Cutaneous drug therapy, Lymphoma, T-Cell, Peripheral drug therapy
- Abstract
A phase 1 study was conducted to determine the dose-limiting toxicities and maximum-tolerated dose (MTD) for bortezomib followed by romidepsin on days 1, 8, and 15 in patients with relapsed/refractory CLL/SLL or B- or T-cell lymphoma. Eighteen treated patients were evaluable for response. The MTD was 1.3 mg/m
2 bortezomib and 10 mg/m2 romidepsin; median treatment duration was 3 cycles at this dose. The dose-limiting toxicities were grade 3 fatigue, vomiting, and chills. Two patients had partial responses, one lasting >2 years, 8 had stable disease, and 8 had progressive disease. The median duration of stable disease was 3.5 cycles. Correlative studies examining expression of NF-кB, XIAP, Bcl-xL, and Bim yielded variable results. The safety profile was consistent with that reported for single-agent bortezomib and romidepsin. This regimen has modest activity in heavily pretreated patients with relapsed/refractory CLL or B- or T-cell lymphoma. NCT00963274.- Published
- 2017
- Full Text
- View/download PDF
36. Synergistic interactions between PLK1 and HDAC inhibitors in non-Hodgkin's lymphoma cells occur in vitro and in vivo and proceed through multiple mechanisms.
- Author
-
Nguyen T, Parker R, Hawkins E, Holkova B, Yazbeck V, Kolluri A, Kmieciak M, Rahmani M, and Grant S
- Subjects
- Animals, Caspases metabolism, Cell Cycle Checkpoints drug effects, Cell Cycle Proteins genetics, Cell Line, Tumor, Cell Proliferation, DNA Damage drug effects, Disease Models, Animal, Drug Synergism, Gene Expression Regulation, Neoplastic drug effects, Gene Knockdown Techniques, Genes, Lethal, Genes, myc, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells metabolism, Histones metabolism, Humans, Hydroxamic Acids pharmacology, Lymphoma, Non-Hodgkin drug therapy, Lymphoma, Non-Hodgkin metabolism, Lymphoma, Non-Hodgkin mortality, Lymphoma, Non-Hodgkin pathology, Mice, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins genetics, Pteridines pharmacology, Sulfonamides pharmacology, Xenograft Model Antitumor Assays, Polo-Like Kinase 1, Cell Cycle Proteins antagonists & inhibitors, Histone Deacetylase Inhibitors pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors
- Abstract
Interactions between the polo-like kinase 1 (PLK1) inhibitor volasertib and the histone deacetylase inhibitor (HDACI) belinostat were examined in diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL) cells in vitro and in vivo. Exposure of DLBCL cells to very low concentrations of volasertib in combination with belinostat synergistically increased cell death (apoptosis). Similar interactions occurred in GC-, ABC-, double-hit DLBCL cells, MCL cells, bortezomib-resistant cells and primary lymphoma cells. Co-exposure to volasertib/belinostat induced a marked increase in M-phase arrest, phospho-histone H3, mitotic errors, cell death in M-phase, and DNA damage. Belinostat diminished c-Myc mRNA and protein expression, an effect significantly enhanced by volasertib co-exposure. c-Myc knock-down increased DNA damage and cell death in response to volasertib, arguing that c-Myc down-regulation plays a functional role in the lethality of this regimen. Notably, PLK1 knock-down in DLBCL cells significantly increased belinostat-induced M-phase accumulation, phospho-histone H3, γH2AX, and cell death. Co-administration of volasertib and belinostat dramatically reduced tumor growth in an ABC-DLBCL flank model (U2932) and a systemic double-hit lymphoma model (OCI-Ly18), accompanied by a pronounced increase in survival without significant weight loss or other toxicities. Together, these findings indicate that PLK1/HDAC inhibition warrants attention as a therapeutic strategy in NHL.
- Published
- 2017
- Full Text
- View/download PDF
37. Phase I study of pemetrexed with sorafenib in advanced solid tumors.
- Author
-
Poklepovic A, Gordon S, Shafer DA, Roberts JD, Bose P, Geyer CE Jr, McGuire WP, Tombes MB, Shrader E, Strickler K, Quigley M, Wan W, Kmieciak M, Massey HD, Booth L, Moran RG, and Dent P
- Subjects
- Adult, Aged, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor, Cohort Studies, Female, Humans, Inflammation, Male, Maximum Tolerated Dose, Middle Aged, Niacinamide administration & dosage, PTEN Phosphohydrolase metabolism, Sorafenib, Treatment Outcome, Triple Negative Breast Neoplasms drug therapy, Neoplasms drug therapy, Niacinamide analogs & derivatives, Pemetrexed administration & dosage, Phenylurea Compounds administration & dosage
- Abstract
Purpose: To determine if combination treatment with pemetrexed and sorafenib is safe and tolerable in patients with advanced solid tumors., Results: Thirty-seven patients were enrolled and 36 patients were treated (24 in cohort A; 12 in cohort B). The cohort A dose schedule resulted in problematic cumulative toxicity, while the cohort B dose schedule was found to be more tolerable. The maximum tolerated dose (MTD) was pemetrexed 750 mg/m2 every 14 days with oral sorafenib 400 mg given twice daily on days 1-5. Because dosing delays and modifications were associated with the MTD, the recommended phase II dose was declared to be pemetrexed 500 mg/m2 every 14 days with oral sorafenib 400 mg given twice daily on days 1-5. Thirty-three patients were evaluated for antitumor activity. One complete response and 4 partial responses were observed (15% overall response rate). Stable disease was seen in 15 patients (45%). Four patients had a continued response at 6 months, including 2 of 5 patients with triple-negative breast cancer., Experimental Design: A phase I trial employing a standard 3 + 3 design was conducted in patients with advanced solid tumors. Cohort A involved a novel dose escalation schema exploring doses of pemetrexed every 14 days with continuous sorafenib. Cohort B involved a modified schedule of sorafenib dosing on days 1-5 of each 14-day pemetrexed cycle. Radiographic assessments were conducted every 8 weeks., Conclusions: Pemetrexed and intermittent sorafenib therapy is a safe and tolerable combination for patients, with promising activity seen in patients with breast cancer., Competing Interests: The authors have no conflicts of interest to disclose.
- Published
- 2016
- Full Text
- View/download PDF
38. The NAE inhibitor pevonedistat interacts with the HDAC inhibitor belinostat to target AML cells by disrupting the DDR.
- Author
-
Zhou L, Chen S, Zhang Y, Kmieciak M, Leng Y, Li L, Lin H, Rizzo KA, Dumur CI, Ferreira-Gonzalez A, Rahmani M, Povirk L, Chalasani S, Berger AJ, Dai Y, and Grant S
- Subjects
- Animals, Apoptosis drug effects, Bcl-2-Like Protein 11 antagonists & inhibitors, Bcl-2-Like Protein 11 genetics, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins genetics, Cells, Cultured, Checkpoint Kinase 1 antagonists & inhibitors, Checkpoint Kinase 1 genetics, Cyclopentanes pharmacology, Drug Synergism, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxamic Acids pharmacology, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute pathology, Mice, Myelodysplastic Syndromes pathology, NF-kappa B metabolism, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins genetics, Protein Processing, Post-Translational drug effects, Protein-Tyrosine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases genetics, Pyrimidines pharmacology, RNA Interference, RNA, Small Interfering genetics, S Phase Cell Cycle Checkpoints drug effects, Sulfonamides pharmacology, U937 Cells, Xenograft Model Antitumor Assays, Cyclopentanes therapeutic use, DNA Damage, DNA Repair drug effects, Histone Deacetylase Inhibitors therapeutic use, Hydroxamic Acids therapeutic use, Leukemia, Myeloid, Acute drug therapy, Molecular Targeted Therapy, Myelodysplastic Syndromes drug therapy, Pyrimidines therapeutic use, Sulfonamides therapeutic use, Ubiquitin-Activating Enzymes antagonists & inhibitors
- Abstract
Two classes of novel agents, NEDD8-activating enzyme (NAE) and histone deacetylase (HDAC) inhibitors, have shown single-agent activity in acute myelogenous leukemia (AML)/myelodysplastic syndrome (MDS). Here we examined mechanisms underlying interactions between the NAE inhibitor pevonedistat (MLN4924) and the approved HDAC inhibitor belinostat in AML/MDS cells. MLN4924/belinostat coadministration synergistically induced AML cell apoptosis with or without p53 deficiency or FLT3-internal tandem duplication (ITD), whereas p53 short hairpin RNA (shRNA) knockdown or enforced FLT3-ITD expression significantly sensitized cells to the regimen. MLN4924 blocked belinostat-induced antiapoptotic gene expression through nuclear factor-κB inactivation. Each agent upregulated Bim, and Bim knockdown significantly attenuated apoptosis. Microarrays revealed distinct DNA damage response (DDR) genetic profiles between individual vs combined MLN4924/belinostat exposure. Whereas belinostat abrogated the MLN4924-activated intra-S checkpoint through Chk1 and Wee1 inhibition/downregulation, cotreatment downregulated multiple homologous recombination and nonhomologous end-joining repair proteins, triggering robust double-stranded breaks, chromatin pulverization, and apoptosis. Consistently, Chk1 or Wee1 shRNA knockdown significantly sensitized AML cells to MLN4924. MLN4924/belinostat displayed activity against primary AML or MDS cells, including those carrying next-generation sequencing-defined poor-prognostic cancer hotspot mutations, and CD34(+)/CD38(-)/CD123(+) populations, but not normal CD34(+) progenitors. Finally, combined treatment markedly reduced tumor burden and significantly prolonged animal survival (P < .0001) in AML xenograft models with negligible toxicity, accompanied by pharmacodynamic effects observed in vitro. Collectively, these findings argue that MLN4924 and belinostat interact synergistically by reciprocally disabling the DDR in AML/MDS cells. This strategy warrants further consideration in AML/MDS, particularly in disease with unfavorable genetic aberrations., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
39. A Phase II Trial of AZD6244 (Selumetinib, ARRY-142886), an Oral MEK1/2 Inhibitor, in Relapsed/Refractory Multiple Myeloma.
- Author
-
Holkova B, Zingone A, Kmieciak M, Bose P, Badros AZ, Voorhees PM, Baz R, Korde N, Lin HY, Chen JQ, Herrmann M, Xi L, Raffeld M, Zhao X, Wan W, Tombes MB, Shrader E, Weir-Wiggins C, Sankala H, Hogan KT, Doyle A, Annunziata CM, Wellons M, Roberts JD, Sullivan D, Landgren O, and Grant S
- Subjects
- Adult, Aged, Aged, 80 and over, Benzimidazoles adverse effects, Benzimidazoles pharmacokinetics, Disease-Free Survival, Female, Humans, Kaplan-Meier Estimate, MAP Kinase Kinase Kinases antagonists & inhibitors, MAP Kinase Kinase Kinases genetics, Male, Middle Aged, Multiple Myeloma genetics, Multiple Myeloma pathology, Neoplasm Recurrence, Local pathology, Proto-Oncogene Proteins p21(ras) genetics, Benzimidazoles administration & dosage, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy
- Abstract
Purpose: AZD6244 is a MEK1/2 inhibitor with significant preclinical activity in multiple myeloma cells. This phase II study used a two-stage Simon design to determine the AZD6244 response rate in patients with relapsed or refractory multiple myeloma., Experimental Design: AZD6244 (75 mg) was administered orally, twice a day, continuously for 28-day cycles. Response was evaluated after three cycles., Results: Thirty-six patients received therapy. The median age was 65 years (range: 43-81) and the median number of prior therapies was 5 (range: 2-11). The most common grade 3 and 4 toxicities included anemia, neutropenia, thrombocytopenia, diarrhea, and fatigue. Three deaths occurred possibly related to AZD6244 (2 due to sepsis, 1 due to acute kidney injury). After AZD6244 discontinuation, three additional deaths occurred due to disease progression. The response rate (CR + PR) was 5.6% with a mean duration of response of 4.95 months and median progression-free survival time of 3.52 months. One patient had a very good partial response (VGPR), 1 patient had a partial response, 17 patients had stable disease, 13 patients had progressive disease, and 4 patients could not be assessed for response. Pharmacodynamic studies revealed variable effects on bone marrow CD138(+) cell MEK1/2 and ERK1/2 phosphorylation. The best clinical response, a prolonged VGPR, occurred in a patient with an MMSET translocation., Conclusions: Single-agent AZD6244 was tolerable and had minimal activity in this heavily pretreated population., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
40. Phase 1 trial of carfilzomib (PR-171) in combination with vorinostat (SAHA) in patients with relapsed or refractory B-cell lymphomas.
- Author
-
Holkova B, Kmieciak M, Bose P, Yazbeck VY, Barr PM, Tombes MB, Shrader E, Weir-Wiggins C, Rollins AD, Cebula EM, Pierce E, Herr M, Sankala H, Hogan KT, Wan W, Feng C, Peterson DR, Fisher RI, Grant S, and Friedberg JW
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Biomarkers, Combined Modality Therapy, Cytokines blood, Drug Monitoring, Drug Resistance, Neoplasm, Female, Humans, Hydroxamic Acids administration & dosage, Lymphoma, B-Cell blood, Male, Middle Aged, Neoplasm Recurrence, Local, Oligopeptides administration & dosage, Retreatment, Treatment Outcome, Vorinostat, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Lymphoma, B-Cell diagnosis, Lymphoma, B-Cell drug therapy
- Abstract
A phase 1 study with carfilzomib and vorinostat was conducted in 20 B-cell lymphoma patients. Vorinostat was given orally twice daily on days 1, 2, 3, 8, 9, 10, 15, 16, and 17 followed by carfilzomib (given as a 30-min infusion) on days 1, 2, 8, 9, 15, and 16. A treatment cycle was 28 days. Dose escalation initially followed a standard 3 + 3 design, but adapted a more conservative accrual rule following dose de-escalation. The maximum tolerated dose was 20 mg/m2 carfilzomib and 100 mg vorinostat (twice daily). The dose-limiting toxicities were grade 3 pneumonitis, hyponatremia, and febrile neutropenia. One patient had a partial response and two patients had stable disease. Correlative studies showed a decrease in NF-κB activation and an increase in Bim levels in some patients, but these changes did not correlate with clinical response.
- Published
- 2016
- Full Text
- View/download PDF
41. Synergism between bosutinib (SKI-606) and the Chk1 inhibitor (PF-00477736) in highly imatinib-resistant BCR/ABL⁺ leukemia cells.
- Author
-
Nguyen T, Hawkins E, Kolluri A, Kmieciak M, Park H, Lin H, and Grant S
- Subjects
- Amino Acid Substitution, Animals, Checkpoint Kinase 1, Drug Resistance, Neoplasm genetics, Drug Synergism, Fusion Proteins, bcr-abl antagonists & inhibitors, Fusion Proteins, bcr-abl genetics, Humans, Imatinib Mesylate, K562 Cells, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Leukemia, Lymphocytic, Chronic, B-Cell pathology, MAP Kinase Kinase 1 genetics, MAP Kinase Kinase 1 metabolism, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System genetics, Mice, Mice, Nude, Mitogen-Activated Protein Kinase 1 genetics, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 genetics, Mitogen-Activated Protein Kinase 3 metabolism, Mutation, Missense, Myeloid Cell Leukemia Sequence 1 Protein, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Xenograft Model Antitumor Assays, Aniline Compounds agonists, Aniline Compounds pharmacology, Antineoplastic Agents pharmacology, Benzamides, Benzodiazepinones agonists, Benzodiazepinones pharmacology, Drug Resistance, Neoplasm drug effects, Fusion Proteins, bcr-abl metabolism, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Nitriles agonists, Nitriles pharmacology, Piperazines, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Kinase Inhibitors, Protein Kinases metabolism, Pyrazoles agonists, Pyrazoles pharmacology, Pyrimidines, Quinolines agonists, Quinolines pharmacology
- Abstract
Interactions between the dual BCR/ABL and Src inhibitor bosutinib and the Chk1 inhibitor PF-00477736 were examined in BCR/ABL(+) leukemia cells, particularly imatinib-resistant cells, including those with the T315I mutation. Bosutinib blocked PF-00477736-induced ERK1/2 activation and sharply increased apoptosis in association with Mcl-1 inhibition, p34(cdc2) dephosphorylation, BimEL up-regulation, and DNA damage in imatinib-resistant CML or Ph(+) ALL cell lines. Inhibition of Src or MEK1 by shRNA significantly enhanced PF-0047736 lethality. Bosutinib/PF-00477736 co-treatment also potentiated cell death in CD34(+) CML patient samples, including dasatinib-resistant blast crisis cells exhibiting both T315I and E355G mutations, but was minimally toxic to normal CD34(+) cells. Finally, combined in vivo treatment significantly suppressed BaF3/T315I tumor growth and prolonged survival in an allogeneic mouse model. Together, these findings suggest that this targeted combination strategy warrants attention in IM-resistant CML or Ph(+) ALL., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
42. Phase I trial of bortezomib (PS-341; NSC 681239) and "nonhybrid" (bolus) infusion schedule of alvocidib (flavopiridol; NSC 649890) in patients with recurrent or refractory indolent B-cell neoplasms.
- Author
-
Holkova B, Kmieciak M, Perkins EB, Bose P, Baz RC, Roodman GD, Stuart RK, Ramakrishnan V, Wan W, Peer CJ, Dawson J, Kang L, Honeycutt C, Tombes MB, Shrader E, Weir-Wiggins C, Wellons M, Sankala H, Hogan KT, Colevas AD, Doyle LA, Figg WD, Coppola D, Roberts JD, Sullivan D, and Grant S
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Boronic Acids administration & dosage, Boronic Acids pharmacokinetics, Bortezomib, Combined Modality Therapy, Drug Administration Schedule, Drug Monitoring, Female, Flavonoids administration & dosage, Flavonoids pharmacokinetics, Humans, Lymphoproliferative Disorders diagnosis, Male, Middle Aged, Piperidines administration & dosage, Piperidines pharmacokinetics, Pyrazines administration & dosage, Pyrazines pharmacokinetics, Recurrence, Retreatment, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, B-Lymphocytes pathology, Lymphoproliferative Disorders drug therapy, Lymphoproliferative Disorders pathology
- Abstract
Purpose: This phase I study was conducted to determine the dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) for the combination of bortezomib and alvocidib in patients with B-cell malignancies (multiple myeloma, indolent lymphoma, Waldenstrom macroglobulinemia, and mantle cell lymphoma)., Experimental Design: Patients received bortezomib (intravenous push), followed by alvocidib (1-hour infusion), on days 1, 4, 8, and 11 of a 21-day treatment cycle. Patients experiencing responses or stable disease continued on treatment at the investigator's discretion. A standard 3+3 dose-escalation design was used to identify the MTD based on DLTs, and pharmacokinetic and pharmacodynamic studies were conducted., Results: A total of 44 patients were enrolled, with 39 patients assessed for response. The MTD was established as 1.3 mg/m(2) for bortezomib and 40 mg/m(2) for alvocidib. The most common hematologic toxicities included leukopenia, lymphopenia, neutropenia, and thrombocytopenia. The most common nonhematologic toxicities included diarrhea, fatigue, and sensory neuropathy. Three complete remissions (8%) and 10 partial remissions (26%) were observed for a total response rate of 33%. Pharmacokinetic findings with the current dosing regimen were consistent with the comparable literature and the hybrid dosing regimen. Pharmacodynamic study results did not correlate with clinical responses., Conclusions: The combination of bortezomib and alvocidib is tolerable, and an MTD has been established for this schedule. The regimen appears to be efficacious in patients with relapsed/refractory multiple myeloma or indolent non-Hodgkin lymphoma. As the nonhybrid regimen is less cumbersome than the previous hybrid dosing schedule regimen, the current schedule is recommended for successor studies., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
43. A Bim-targeting strategy overcomes adaptive bortezomib resistance in myeloma through a novel link between autophagy and apoptosis.
- Author
-
Chen S, Zhang Y, Zhou L, Leng Y, Lin H, Kmieciak M, Pei XY, Jones R, Orlowski RZ, Dai Y, and Grant S
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Apoptosis genetics, Apoptosis Regulatory Proteins antagonists & inhibitors, Apoptosis Regulatory Proteins genetics, Autophagy drug effects, Autophagy genetics, Bcl-2-Like Protein 11, Boronic Acids pharmacology, Bortezomib, Cell Line, Tumor, Cells, Cultured, Drug Resistance, Neoplasm drug effects, Fluorescent Antibody Technique, Histone Deacetylase Inhibitors administration & dosage, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxamic Acids administration & dosage, Hydroxamic Acids pharmacology, Immunoblotting, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Pyrazines pharmacology, RNA Interference, Tumor Burden drug effects, Tumor Burden genetics, Xenograft Model Antitumor Assays, Apoptosis physiology, Apoptosis Regulatory Proteins metabolism, Autophagy physiology, Membrane Proteins metabolism, Multiple Myeloma metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Bim contributes to resistance to various standard and novel agents. Here we demonstrate that Bim plays a functional role in bortezomib resistance in multiple myeloma (MM) cells and that targeting Bim by combining histone deacetylase inhibitors (HDACIs) with BH3 mimetics (eg, ABT-737) overcomes bortezomib resistance. BH3-only protein profiling revealed high Bim levels (Bim(hi)) in most MM cell lines and primary CD138(+) MM samples. Whereas short hairpin RNA Bim knockdown conferred bortezomib resistance in Bim(hi) cells, adaptive bortezomib-resistant cells displayed marked Bim downregulation. HDACI upregulated Bim and, when combined with ABT-737, which released Bim from Bcl-2/Bcl-xL, potently killed bortezomib-resistant cells. These events were correlated with Bim-associated autophagy attenuation, whereas Bim knockdown sharply increased autophagy in Bim(hi) cells. In Bim(low) cells, autophagy disruption by chloroquine (CQ) was required for HDACI/ABT-737 to induce Bim expression and lethality. CQ also further enhanced HDACI/ABT-737 lethality in bortezomib-resistant cells. Finally, HDACI failed to diminish autophagy or potentiate ABT-737-induced apoptosis in bim(-/-) mouse embryonic fibroblasts. Thus, Bim deficiency represents a novel mechanism of adaptive bortezomib resistance in MM cells, and Bim-targeting strategies combining HDACIs (which upregulate Bim) and BH3 mimetics (which unleash Bim from antiapoptotic proteins) overcomes such resistance, in part by disabling cytoprotective autophagy., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
44. IFN-γ Rα is a key determinant of CD8+ T cell-mediated tumor elimination or tumor escape and relapse in FVB mouse.
- Author
-
Kmieciak M, Payne KK, Wang XY, and Manjili MH
- Subjects
- Animals, Apoptosis drug effects, CD24 Antigen metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, Female, Gene Expression, Hyaluronan Receptors metabolism, Interferon-gamma pharmacology, Mammary Neoplasms, Experimental, Mice, Neoplasm Recurrence, Local, Neoplasms genetics, Neoplasms pathology, Neoplastic Stem Cells metabolism, Neuregulin-1 immunology, Receptors, Interferon genetics, Tumor Burden drug effects, Interferon gamma Receptor, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Neoplasms immunology, Neoplasms metabolism, Receptors, Interferon metabolism, Tumor Escape genetics
- Abstract
During the past decade, the dual function of the immune system in tumor inhibition and tumor progression has become appreciated. We have previously reported that neu-specific T cells can induce rejection of neu positive mouse mammary carcinoma (MMC) and also facilitate tumor relapse by inducing neu antigen loss and epithelial to mesenchymal transition (EMT). Here, we sought to determine the mechanism by which CD8+ T cells either eliminate the tumor, or maintain tumor cells in a dormant state and eventually facilitate tumor relapse. We show that tumor cells that express high levels of IFN-γ Rα are eliminated by CD8+ T cells. In contrast, tumor cells that express low levels of IFN-γ Rα do not die but remain dormant and quiescent in the presence of IFN-γ producing CD8+ T cells until they hide themselves from the adaptive immune system by losing the tumor antigen, neu. Relapsed tumor cells show CD44+CD24- phenotype with higher rates of tumorigenesis, in vivo. Acquisition of CD44+CD24- phenotype in relapsed tumors was not solely due to Darwinian selection. Our data suggest that tumor cells control the outcome of tumor immune surveillance through modulation of the expression of IFN-γ Rα.
- Published
- 2013
- Full Text
- View/download PDF
45. Multi-therapeutic potential of autoantibodies induced by immune complexes trapped on follicular dendritic cells.
- Author
-
El Shikh ME, Kmieciak M, Manjili MH, Szakal AK, Pitzalis C, and Tew JG
- Subjects
- Animals, Antibodies, Neutralizing blood, Humans, Inhibitory Concentration 50, Mice, Inbred BALB C, Antigen-Antibody Complex immunology, Antigen-Antibody Complex metabolism, Autoantibodies immunology, Dendritic Cells, Follicular immunology
- Abstract
Induction of autoantibodies (autoAbs) targeting disease drivers / mediators is emerging as a potential immunotherapeutic strategy. Auto-immune complex (IC)-retaining follicular dendritic cells (FDCs) critically regulate pathogenic autoAb production in autoreactive germinal centers (GCs); however, their ability to induce potentially therapeutic autoAbs has not been explored. We hypothesized that deliberate display of clinically targeted antigens (Ags) in the form of ICs on FDC membranes induces target-specific autoreactive GCs and autoAbs that may be exploited therapeutically. To test our hypothesis, three therapeutically relevant Ags: TNF-α, HER2/neu and IgE, were investigated. Our results indicated that TNF-α-, HER2/neu- and IgE-specific autoAbs associated with strong GC reactions were induced by TNF-α-, HER2/neu- and IgE-IC retention on FDCs. Moreover, the induced anti-TNF-α autoAbs neutralized mouse and human TNF-α with half maximal Inhibitory Concentration (IC₅₀) of 7.1 and 1.6 nM respectively. In addition, we demonstrated that FDC-induced Ab production could be non-specifically inhibited by the IgG-specific Endo-S that accessed the light zones of GCs and interfered with FDC-IC retention. In conclusion, the ability of FDCs to productively present autoAgs raises the potential for a novel immunotherapeutic platform targeting mediators of autoimmune disorders, allergic diseases, and Ab responsive cancers.
- Published
- 2013
- Full Text
- View/download PDF
46. The novel Chk1 inhibitor MK-8776 sensitizes human leukemia cells to HDAC inhibitors by targeting the intra-S checkpoint and DNA replication and repair.
- Author
-
Dai Y, Chen S, Kmieciak M, Zhou L, Lin H, Pei XY, and Grant S
- Subjects
- Antineoplastic Combined Chemotherapy Protocols administration & dosage, Bone Marrow Cells pathology, Cells, Cultured, Checkpoint Kinase 1, DNA Repair drug effects, DNA Replication drug effects, Gene Expression Regulation, Leukemic, Histone Deacetylases biosynthesis, Histone Deacetylases metabolism, Humans, Hydroxamic Acids administration & dosage, Leukemia metabolism, Leukemia pathology, Protein Kinases chemistry, Pyrazoles administration & dosage, Pyrimidines administration & dosage, S Phase Cell Cycle Checkpoints drug effects, Tumor Suppressor Protein p53 antagonists & inhibitors, Vorinostat, Apoptosis drug effects, Histone Deacetylase Inhibitors administration & dosage, Leukemia drug therapy, Protein Kinases metabolism
- Abstract
Interactions between the novel Chk1 inhibitor MK-8776 and the histone deacetylase (HDAC) inhibitor (HDACI) vorinostat were examined in human leukemia cells harboring wild-type (wt) or deficient p53. MK-8776 synergistically potentiated vorinostat-mediated apoptosis in various p53-wt or -deficient leukemia cell lines, whereas p53 knockdown by short hairpin RNA (shRNA) sensitized p53-wt cells to lethality of this regimen. Leukemia cell lines carrying FLT3-ITD were also sensitive to the MK-8776/vorinostat regimen. Synergistic interactions were associated with inhibition of Chk1 activity, interference with the intra-S-phase checkpoint, disruption of DNA replication, and downregulation of proteins involved in DNA replication (e.g., Cdt1) and repair (e.g., CtIP and BRCA1), resulting in sharp increases in DNA damage, reflected by enhanced γ-H2A.X formation, and apoptosis. Moreover, leukemia cells expressing kinase-dead Chk1 (D130A) or Chk1 shRNA were significantly more sensitive to HDACIs compared with their wt counterparts and displayed downregulation of CtIP and BRCA1 phosphorylation following HDACI exposure. Finally, the MK-8776/vorinostat regimen was active in primary acute myelogenous leukemia (AML) blasts, particularly against the CD34(+)/CD38(-)/CD123(+) population enriched for leukemia-initiating cells. In contrast, identical regimens were relatively sparing toward normal cord blood CD34(+) cells. Together, these findings indicate that the novel Chk1 inhibitor MK-8776 markedly potentiates HDACI lethality in leukemia cells displaying various genetic backgrounds through mechanisms involving disruption of the intra-S checkpoint, DNA replication, and DNA repair. They also argue that leukemic cells, including those bearing oncogenic mutations associated with poor prognosis, for example, p53 deletion/mutation or FLT3-ITD, may also be susceptible to this strategy., (©2013 AACR)
- Published
- 2013
- Full Text
- View/download PDF
47. A phase I trial of vorinostat and alvocidib in patients with relapsed, refractory, or poor prognosis acute leukemia, or refractory anemia with excess blasts-2.
- Author
-
Holkova B, Supko JG, Ames MM, Reid JM, Shapiro GI, Perkins EB, Ramakrishnan V, Tombes MB, Honeycutt C, McGovern RM, Kmieciak M, Shrader E, Wellons MD, Sankala H, Doyle A, Wright J, Roberts JD, and Grant S
- Subjects
- Acute Disease, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Female, Flavonoids administration & dosage, Humans, Hydroxamic Acids administration & dosage, Leukemia diagnosis, Leukemia metabolism, Male, Maximum Tolerated Dose, Middle Aged, Myeloid Cell Leukemia Sequence 1 Protein, Piperidines administration & dosage, Prognosis, Proto-Oncogene Proteins c-bcl-2 metabolism, RNA Polymerase II metabolism, Recurrence, Treatment Outcome, Vorinostat, Young Adult, Anemia, Refractory, with Excess of Blasts drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia drug therapy
- Abstract
Purpose: This phase I study was conducted to identify the maximum-tolerated dose (MTD) of alvocidib when combined with vorinostat in patients with relapsed, refractory, or poor prognosis acute leukemia, or refractory anemia with excess blasts-2. Secondary objectives included investigating the pharmacokinetic and pharmacodynamic effects of the combination., Experimental Design: Patients received vorinostat (200 mg orally, three times a day, for 14 days) on a 21-day cycle, combined with 2 different alvocidib administration schedules: a 1-hour intravenous infusion, daily × 5; or a 30-minute loading infusion followed by a 4-hour maintenance infusion, weekly × 2. The alvocidib dose was escalated using a standard 3+3 design., Results: Twenty-eight patients were enrolled and treated. The alvocidib MTD was 20 mg/m(2) (30-minute loading infusion) followed by 20 mg/m(2) (4-hour maintenance infusion) on days one and eight, in combination with vorinostat. The most frequently encountered toxicities were cytopenias, fatigue, hyperglycemia, hypokalemia, hypophosphatemia, and QT prolongation. Dose-limiting toxicities (DLT) were cardiac arrhythmia-atrial fibrillation and QT prolongation. No objective responses were achieved although 13 of 26 evaluable patients exhibited stable disease. Alvocidib seemed to alter vorinostat pharmacokinetics, whereas alvocidib pharmacokinetics were unaffected by vorinostat. Ex vivo exposure of leukemia cells to plasma obtained from patients after alvocidib treatment blocked vorinostat-mediated p21(CIP1) induction and downregulated Mcl-1 and p-RNA Pol II for some specimens, although parallel in vivo bone marrow responses were infrequent., Conclusions: Alvocidib combined with vorinostat is well tolerated. Although disease stabilization occurred in some heavily pretreated patients, objective responses were not obtained with these schedules., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
48. Epigenetic induction of adaptive immune response in multiple myeloma: sequential azacitidine and lenalidomide generate cancer testis antigen-specific cellular immunity.
- Author
-
Toor AA, Payne KK, Chung HM, Sabo RT, Hazlett AF, Kmieciak M, Sanford K, Williams DC, Clark WB, Roberts CH, McCarty JM, and Manjili MH
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Azacitidine administration & dosage, Boronic Acids administration & dosage, Bortezomib, Dexamethasone administration & dosage, Doxorubicin administration & dosage, Drug Administration Schedule, Female, Humans, Lenalidomide, Male, Middle Aged, Multiple Myeloma drug therapy, Multiple Myeloma surgery, Plasma Cells immunology, Prospective Studies, Pyrazines administration & dosage, Thalidomide administration & dosage, Thalidomide analogs & derivatives, Transplantation, Autologous, Antigens, Neoplasm immunology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Azacitidine pharmacology, DNA Methylation drug effects, Epigenesis, Genetic drug effects, Immunity, Cellular drug effects, Lymphocyte Transfusion, Multiple Myeloma immunology, Peripheral Blood Stem Cell Transplantation, T-Cell Antigen Receptor Specificity drug effects
- Abstract
Patients with multiple myeloma (MM) undergoing high dose therapy and autologous stem cell transplantation (SCT) remain at risk for disease progression. Induction of the expression of highly immunogenic cancer testis antigens (CTA) in malignant plasma cells in MM patients may trigger a protective immune response following SCT. We initiated a phase II clinical trial of the DNA hypomethylating agent, azacitidine (Aza) administered sequentially with lenalidomide (Rev) in patients with MM. Three cycles of Aza and Rev were administered and autologous lymphocytes were collected following the 2nd and 3rd cycles of Aza-Rev and cryopreserved. Subsequent stem cell mobilization was followed by high-dose melphalan and SCT. Autologous lymphocyte infusion (ALI) was performed in the second month following transplantation. Fourteen patients have completed the investigational therapy; autologous lymphocytes were collected from all of the patients. Thirteen patients have successfully completed SCT and 11 have undergone ALI. Six patients tested have demonstrated CTA up-regulation in either unfractionated bone marrow (n = 4) or CD138+ cells (n = 2). CTA (CTAG1B)-specific T cell response has been observed in all three patients tested and persists following SCT. Epigenetic induction of an adaptive immune response to cancer testis antigens is safe and feasible in MM patients undergoing SCT., (© 2012 Blackwell Publishing Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
49. Distinct oligoclonal T cells are associated with graft versus host disease after stem-cell transplantation.
- Author
-
Berrie JL, Kmieciak M, Sabo RT, Roberts CH, Idowu MO, Mallory K, Chung HM, McCarty JM, Borrelli CA, Detwiler MM, Kazim AL, Toor AA, and Manjili MH
- Subjects
- Adult, Aged, DNA genetics, Female, Follow-Up Studies, Gene Expression, Graft vs Host Disease etiology, Graft vs Host Disease pathology, Hematologic Neoplasms surgery, Humans, Male, Middle Aged, Receptors, Antigen, T-Cell, alpha-beta biosynthesis, Receptors, Antigen, T-Cell, alpha-beta genetics, Reverse Transcriptase Polymerase Chain Reaction, T-Lymphocytes pathology, Graft vs Host Disease immunology, Receptors, Antigen, T-Cell, alpha-beta immunology, Stem Cell Transplantation adverse effects, T-Lymphocytes immunology
- Abstract
Background: In patients with hematologic malignancies who receive stem-cell transplantation, donors' T cells can recognize minor histocompatibility antigens on recipient cells and generate an objective response against the tumor. However, a major side effect of such therapy is graft-versus-host disease (GVHD). The purpose of this study was to characterize distinct T-cell clones that were frequently and exclusively involved in GVHD or graft-versus-tumor (GVT) effects., Methods: We hypothesized that distinct GVHD-associated T-cell clones can be identified during the disease progression. To test this, we conducted comparative analysis of T-cell receptor (TCR) Vβs in donor-recipient pairs of patients with GVHD versus those with GVHD-free and relapse-free survival using quantitative reverse-transcriptase polymerase chain reaction and spectratyping analyses., Results: We identified three sets of T-cell clones that were either frequently involved in GVHD (TCR Vβ4, 11, and 23) or GVT effect (TCR Vβ9, 16, and 20), or were increased at the time of GVHD and GVT effects in a patient-specific manner (TCR Vβ2, 3, 7, 12, 15, and 17). Spectratyping analysis showed restricted clonality of the identified TCR Vβs. Polymerase chain reaction analysis also confirmed the presence of GVHD-associated T-cell clones at the site of the disease., Conclusions: These data suggest that GVHD- and GVT-associated clones can be distinguished by molecular analysis of TCR Vβ to develop targeted therapy for GVHD.
- Published
- 2012
- Full Text
- View/download PDF
50. CD44(+)/CD24(-/low) cancer stem/progenitor cells are more abundant in triple-negative invasive breast carcinoma phenotype and are associated with poor outcome.
- Author
-
Idowu MO, Kmieciak M, Dumur C, Burton RS, Grimes MM, Powers CN, and Manjili MH
- Subjects
- Adult, Aged, Animals, Biomarkers, Tumor metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, CD24 Antigen genetics, Carcinoma, Ductal, Breast genetics, Carcinoma, Ductal, Breast metabolism, Cell Line, Tumor, Disease Models, Animal, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Hyaluronan Receptors genetics, Mice, Mice, Transgenic, Middle Aged, Neoplasm Staging, Neoplastic Stem Cells metabolism, Oligonucleotide Array Sequence Analysis, Breast Neoplasms diagnosis, CD24 Antigen metabolism, Carcinoma, Ductal, Breast diagnosis, Hyaluronan Receptors metabolism, Neoplastic Stem Cells pathology, Receptor, ErbB-2 metabolism, Receptors, Steroid metabolism
- Abstract
Women classified as having triple-negative tumors have a poor prognosis. The importance of CD44(+)/CD24(-/low) (stem/progenitor cell-phenotype) in breast cancer patients has also been appreciated. However, correlation between triple negativity and CD44(+)/CD24(-/low) with tumor recurrence remains elusive. In the present study, we evaluated tumor specimens of 50 breast cancer patients with known hormone receptor status for whom we had follow-up information and outcome data available, and performed immunohistochemistry analysis to determine CD44 and CD24 expression. Gene expression arrays were also independently performed on 52 breast cancer specimens with banked frozen tissue. Lastly, we used FVBN202 transgenic mouse model of breast carcinoma and determined the hormone receptor status, the proportion of CD44(+)/CD24(-/low) breast cancer stem-like cells, and the behavior of the tumor. We determined that patients with triple-negative tumors had significantly higher incidence of recurrence or distant metastasis associated with increased frequency of breast cancer stem cell phenotypes compared with those with non-triple-negative tumors. Preclinical studies in FVBN202 transgenic mice confirmed these findings by showing that relapsed tumors were triple negative and had significantly higher frequency of breast cancer stem cells compared with their related primary tumors. Unlike non-triple-negative primary tumors, relapsed triple-negative tumors were tumorigenic at low doses when inoculated into FVBN202 transgenic mice. These findings suggest that CD44(+)/CD24(-/low) breast cancer stem-like cells play an important role in the clinical behavior of triple-negative breast cancer and that development of therapeutic targets directed to breast cancer stem-like cells may lead to reduction in the aggressiveness of triple-negative breast cancers., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.