58 results on '"Annunziata, Christina M."'
Search Results
2. Overcoming the challenges of drug development in platinumresistant ovarian cancer.
- Author
-
Eskander, Ramez N., Moore, Kathleen N., Monk, Bradley J., Herzog, Thomas J., Annunziata, Christina M., O'Malley, David M., and Coleman, Robert L.
- Subjects
OVARIAN cancer ,DRUG development ,CLINICAL trials ,IMMUNE checkpoint inhibitors ,HEMATOLOGIC malignancies ,GYNECOLOGIC cancer - Abstract
The definition of "platinum-resistant ovarian cancer" has evolved; it now also reflects cancers for which platinum treatment is no longer an option. Standard of care for platinum-resistant ovarian cancer is single-agent, non-platinum chemotherapy with or without bevacizumab, which produces modest response rates, with the greatest benefits achieved using weekly paclitaxel. Several recent phase 3 trials of pretreated patients with prior bevacizumab exposure failed to meet their primary efficacy endpoints, highlighting the challenge in improving clinical outcomes among these patients. Combination treatment with antiangiogenics has improved outcomes, whereas combination strategies with immune checkpoint inhibitors have yielded modest results. Despite extensive translational research, there has been a lack of reliable and established biomarkers that predict treatment response in platinum-resistant ovarian cancer. Additionally, in the platinum-resistant setting, implications for the time between the penultimate dose of platinum therapy and platinum retreatment remain an area of debate. Addressing the unmet need for an effective treatment in the platinum-resistant setting requires thoughtful clinical trial design based on a growing understanding of the disease. Recent cancer drug approvals highlight the value of incorporating molecular phenotypes to better define patients who are more likely to respond to novel therapies. Clinical trials designed per the Gynecologic Cancer InterGroup recommendations--which advocate against relying solely upon the platinum-free interval--will help advance our understanding of recurrent ovarian cancer response where platinum rechallenge in the platinum-resistant setting may be considered. The inclusion of biomarkers in clinical trials will improve patient stratification and potentially demonstrate correlations with biomarker expression and duration of response. With the efficacy of antibody-drug conjugates shown for the treatment of some solid and hematologic cancers, current trials are evaluating the use of various novel conjugates in the setting of platinum-resistant ovarian cancer. Emerging novel treatments coupled with combination trials and Frontiers in Oncology frontiersin. biomarker explorations offer encouraging results for potential strategies to improve response rates and prolong progression-free survival in this population with high unmet need. This review outlines existing data from contemporary clinical trials of patients with platinum-resistant ovarian cancer and suggests historical synthetic benchmarks for non-randomized trials. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Patients' perspectives on prenatal screening results that suggest maternal cancer: A qualitative analysis.
- Author
-
Turriff, Amy, Miner, Skye A., Annunziata, Christina M., and Bianchi, Diana W.
- Abstract
Objective: To explore patient perspectives after receiving non‐invasive prenatal testing (NIPT) results that suggest maternal cancer. Methods: Individuals who received non‐reportable or discordant NIPT results during pregnancy and enrolled in a study were interviewed prior to and after receiving the outcome of their clinical evaluation for cancer. Interviews were independently coded by two researchers and analyzed thematically. Results: Forty‐nine participants were included. Three themes were identified: 1) limited pre‐test awareness of maternal incidental findings caused considerable confusion for participants, whose initial concerns focused on their babies; 2) providers' communication influenced how participants perceived their risk of cancer and the need to be evaluated; and 3) participants perceived value in receiving maternal incidental findings from NIPT despite any stress it caused during their pregnancy. Conclusion: Participants viewed the ability to detect occult malignancy as an added benefit of NIPT and felt strongly that these results should be disclosed. Obstetric providers need to be aware of maternal incidental findings from NIPT, inform pregnant people of the potential to receive these results during pre‐test counseling, and provide accurate and objective information during post‐test counseling. Clinical Trial Registration: Incidental Detection of Maternal Neoplasia Through Non‐Invasive Cell‐Free DNA Analysis (IDENTIFY), a Natural History Study, NCT4049604. Key points: What's already known about this topic?Non‐invasive prenatal screening (NIPT) using cell‐free DNA sequencing of maternal plasma can incidentally detect maternal cancer.A "non‐reportable" test result may be generated when the sequencing results do not conform to an individual laboratory's proprietary analytic algorithm. There are both technical (e.g., insufficient DNA or low fetal fraction) and biological (e.g., vanishing twin, confined placental mosaicism, maternal incidental finding) reasons for this to occur. NIPT results that suggest maternal cancer are often non‐reportable.There is limited prospective evidence to inform patient counseling and medical management of NIPT results that suggest maternal cancer.In the United States, how NIPT laboratories report malignancy suspicious results, what information clinicians disclose to pregnant individuals, and what clinical evaluation is offered varies. What does this study add?This study describes patients' perspectives on receiving NIPT results that suggest maternal cancer.Pre‐ and post‐test counseling recommendations were made based on participants' experiences. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. NF-κB Signaling Modulates miR-452-5p and miR-335-5p Expression to Functionally Decrease Epithelial Ovarian Cancer Progression in Tumor-Initiating Cells.
- Author
-
Kamdar, Rahul D., Harrington, Brittney S., Attar, Emma, Korrapati, Soumya, Shetty, Jyoti, Zhao, Yongmei, Tran, Bao, Wong, Nathan, House, Carrie D., and Annunziata, Christina M.
- Subjects
OVARIAN epithelial cancer ,GENE expression ,MICRORNA ,CANCER invasiveness ,DISEASE relapse - Abstract
Epithelial ovarian cancer (EOC) remains the fifth leading cause of cancer-related death in women worldwide, partly due to the survival of chemoresistant, stem-like tumor-initiating cells (TICs) that promote disease relapse. We previously described a role for the NF-κB pathway in promoting TIC chemoresistance and survival through NF-κB transcription factors (TFs) RelA and RelB, which regulate genes important for the inflammatory response and those associated with cancer, including microRNAs (miRNAs). We hypothesized that NF-κB signaling differentially regulates miRNA expression through RelA and RelB to support TIC persistence. Inducible shRNA was stably expressed in OV90 cells to knockdown RELA or RELB; miR-seq analyses identified differentially expressed miRNAs hsa-miR-452-5p and hsa-miR-335-5p in cells grown in TIC versus adherent conditions. We validated the miR-seq findings via qPCR in TIC or adherent conditions with RELA or RELB knocked-down. We confirmed decreased expression of hsa-miR-452-5p when either RELA or RELB were depleted and increased expression of hsa-miR-335-5p when RELA was depleted. Either inhibiting miR-452-5p or mimicking miR-335-5p functionally decreased the stem-like potential of the TICs. These results highlight a novel role of NF-κB TFs in modulating miRNA expression in EOC cells, thus opening a better understanding toward preventing recurrence of EOC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. SMAC Mimetics Synergistically Cooperate with HDAC Inhibitors Enhancing TNF-α Autocrine Signaling.
- Author
-
Shibuya, Yusuke, Kudo, Kei, Zeligs, Kristen P., Anderson, David, Hernandez, Lidia, Ning, Franklin, Cole, Christopher B., Fergusson, Maria, Kedei, Noemi, Lyons, John, Taylor, Jason, Korrapati, Soumya, and Annunziata, Christina M.
- Subjects
IN vitro studies ,OVARIAN tumors ,INDOLE compounds ,IN vivo studies ,OLIGOPEPTIDES ,CELL physiology ,CANCER relapse ,APOPTOSIS ,SIGNAL peptides ,EARLY detection of cancer ,NF-kappa B ,CELLULAR signal transduction ,GENE expression ,DRUG synergism ,TUMOR necrosis factors ,RESEARCH funding ,HISTONE deacetylase ,CASPASES ,CHEMICAL inhibitors - Abstract
Simple Summary: The combination of a SMAC mimetic with an HDAC inhibitor is a novel and promising strategy for cancer treatment. The HDAC inhibitor mechanistically synergizes with SMAC mimetics by stimulating autocrine TNF-α production. The overexpression of inhibitor of apoptosis (IAP) proteins is strongly related to poor survival of women with ovarian cancer. Recurrent ovarian cancers resist apoptosis due to the dysregulation of IAP proteins. Mechanistically, Second Mitochondrial Activator of Caspases (SMAC) mimetics suppress the functions of IAP proteins to restore apoptotic pathways resulting in tumor death. We previously conducted a phase 2 clinical trial of the single-agent SMAC mimetic birinapant and observed minimal drug response in women with recurrent ovarian cancer despite demonstrating on-target activity. Accordingly, we performed a high-throughput screening matrix to identify synergistic drug combinations with birinapant. SMAC mimetics in combination with an HDAC inhibitor showed remarkable synergy and was, therefore, selected for further evaluation. We show here that this synergy observed both in vitro and in vivo results from multiple convergent pathways to include increased caspase activation, HDAC inhibitor-mediated TNF-α upregulation, and alternative NF-kB signaling. These findings provide a rationale for the integration of SMAC mimetics and HDAC inhibitors in clinical trials for recurrent ovarian cancer where treatment options are still limited. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Identification of the O-Glycan Epitope Targeted by the Anti-Human Carcinoma Monoclonal Antibody (mAb) NEO-201.
- Author
-
Tsang, Kwong Y., Fantini, Massimo, Zaki, Anjum, Mavroukakis, Sharon A., Morelli, Maria Pia, Annunziata, Christina M., and Arlen, Philip M.
- Subjects
CANCER chemotherapy ,THERAPEUTIC use of monoclonal antibodies ,POLYSACCHARIDES ,PANCREATIC tumors ,TISSUE arrays ,GLYCOSYLATION ,THREONINE ,NEUTROPHILS ,SERINE ,CELL lines ,ANTIGENS - Abstract
Simple Summary: Glycosylation is an important post-translational modification made on mammalian proteins and lipids. In cancer cells, the disruption of several glycosylation patterns, such as the O-glycosylation, has been observed. The expression of incomplete/truncated O-glycans in cancer cells occurs in both solid and liquid tumors and is correlated with poor prognosis and tumor progression. The employment of monoclonal antibodies (mAbs) targeting truncated O-glycans in cancer cells could serve as an effective strategy to counteract tumor growth. In previous studies, we reported that the IgG1-humanized mAb NEO-201 binds specifically to tumor-associated variants of CEACAM5 and CEACAM6 expressed by colon, ovarian, pancreatic, non-small cell lung, head and neck, cervical, uterine and breast cancers but is not reactive against most normal tissues. Since CEACAMs are highly glycosylated proteins, in this article, we evaluated whether the epitope recognized by NEO-201 is an O-glycan. This study demonstrated that NEO-201 binds to core 1 O-glycans and targets and kills cancer cells expressing core 1 and extended core 1 O-glycans. Usually, GalNAc residue can be added on to threonine and serine to form O-glycans, suggesting that NEO-201 binds to core 1 and extended core 1 O-glycans attached to any protein carrying amino acid regions containing serine and threonine Truncated O-glycans expressed in cancer cells support tumor progression, and they may serve as potential targets to improve the monitoring and treatment of cancers. Previously, we reported that NEO-201 binds to several tumors expressing tumor-associated CEACAM5 and CEACAM6 variants but does not bind to those expressed in healthy tissues. This specific binding may be associated with the presence of truncated O-glycans attached on the protein sequence of these variants. To evaluate the glycosylation pattern targeted by NEO-201 we performed an O-glycan array consisting of 94 O-glycans. O-glycan profiles were elucidated from the human pancreatic cancer cell line CFPAC-1, human hematological neoplastic cells (HL-60, U937, K562) and human neutrophils. The O-glycan array analysis showed that NEO-201 interacts with core 1-4 O-glycans and that the binding to a specific core 1 O-glycan was the strongest. The O-glycan profiling of the NEO-201-reactive cells CFPAC-1, HL-60, U937 and human neutrophils showed that cells recognized by NEO-201 express mostly core 1 and/or extended core 1 O-glycans. In addition, NEO-201 mediates antibody-dependent cell-mediated cytotoxicity (ADCC) against tumor cells expressing core 1 or extended core 1 O-glycan profiles. These results demonstrated that NEO-201 binds to core 1 and extended core 1 O-glycans expressed in its target cells. Since GalNAc residue can be added onto threonine and serine to form O-glycans, it is very likely that NEO-201 recognizes these O-glycans attached to any protein with amino acid regions containing serine and threonine. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Development and Characterization of an Anti-Cancer Monoclonal Antibody for Treatment of Human Carcinomas.
- Author
-
Tsang, Kwong yok, Fantini, Massimo, Mavroukakis, Sharon A., Zaki, Anjum, Annunziata, Christina M., and Arlen, Philip M.
- Subjects
THERAPEUTIC use of monoclonal antibodies ,PROGRAMMED cell death 1 receptors ,CLINICAL drug trials ,IMMUNE checkpoint inhibitors ,COMPLEMENT (Immunology) ,KILLER cells ,CELLULAR signal transduction ,CELL adhesion molecules ,TUMORS ,DRUG development ,CELL lines ,IMMUNOTHERAPY ,PHARMACODYNAMICS - Abstract
Simple Summary: Cancers can grow and spread to different parts of the body. The aim of immunotherapy is to stimulate the immune system to eliminate cancer cells in a selective manner. Tumor-targeting monoclonal antibodies (mAb) can be used as immunotherapy to stimulate innate antitumor immunity. In this review, we have described the development and characterization of an anti-cancers mAb NEO-201. NEO-201 is an IgG1 humanized mAb that binds specifically to tumor-associated variants of CEACAM-5 and CEACAM-6 expressed by colon, ovarian, pancreatic, non-small cell lung, head and neck, cervical, uterine and breast cancers, but is not reactive against most normal tissues. The peculiarity of NEO-201 is its ability to counteract tumor growth through different mechanisms. NEO-201 engages components of immune system to kill tumor cells expressing its target via antibody-dependent cell-mediated cytotoxicity and complement dependent cytotoxicity. NEO-201 can also indirectly enhance anti-cancer activity through the blockade of the interaction between CEACAM-5 expressed on tumor cells and CEACAM-1 expressed on natural killer cells to reverse CEACAM-1-dependent inhibition of NK cytotoxicity or through its binding to human regulatory T cells. The specificity of NEO-201 in recognizing suppressive regulatory T cells provides the basis for combination cancer immunotherapy with checkpoint inhibitors targeting the PD-1/PD-L1 pathway. NEO-201 is an IgG1 humanized monoclonal antibody (mAb) that binds to tumor-associated variants of carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-5 and CEACAM-6. NEO-201 reacts to colon, ovarian, pancreatic, non-small cell lung, head and neck, cervical, uterine and breast cancers, but is not reactive against most normal tissues. NEO-201 can kill tumor cells via antibody-dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC) to directly kill tumor cells expressing its target. We explored indirect mechanisms of its action that may enhance immune tumor killing. NEO-201 can block the interaction between CEACAM-5 expressed on tumor cells and CEACAM-1 expressed on natural killer (NK) cells to reverse CEACAM-1-dependent inhibition of NK cytotoxicity. Previous studies have demonstrated safety/tolerability in non-human primates, and in a first in human phase 1 clinical trial at the National Cancer Institute (NCI). In addition, preclinical studies have demonstrated that NEO-201 can bind to human regulatory T (Treg) cells. The specificity of NEO-201 in recognizing suppressive Treg cells provides the basis for combination cancer immunotherapy with checkpoint inhibitors targeting the PD-1/PD-L1 pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Another wrinkle with age: Aged collagen and intra‐peritoneal metastasis of ovarian cancer.
- Author
-
Harper, Elizabeth I., Hilliard, Tyvette S., Sheedy, Emma F., Carey, Preston, Wilkinson, Paul, Siroky, Michael D., Yang, Jing, Agadi, Elizabeth, Leonard, Annemarie K., Low, Ethan, Liu, Yueying, Biragyn, Arya, Annunziata, Christina M., and Stack, Mary Sharon
- Published
- 2022
- Full Text
- View/download PDF
9. Disulfiram Transcends ALDH Inhibitory Activity When Targeting Ovarian Cancer Tumor-Initiating Cells.
- Author
-
Caminear, Michael W., Harrington, Brittney S., Kamdar, Rahul D., Kruhlak, Michael J., and Annunziata, Christina M.
- Subjects
TIC disorders ,DISULFIRAM ,OVARIAN cancer ,OVARIAN epithelial cancer ,CANCER cells ,DISEASE relapse - Abstract
Epithelial ovarian cancer (EOC) is a global health burden and remains the fifth leading cause of cancer related death in women worldwide with the poorest five-year survival rate of the gynecological malignancies. EOC recurrence is considered to be driven by the survival of chemoresistant, stem-like tumor-initiating cells (TICs). We previously showed that disulfiram, an ALDH inhibitor, effectively targeted TICs compared to adherent EOC cells in terms of viability, spheroid formation, oxidative stress and also prevented relapse in an in vivo model of EOC. In this study we sought to determine whether specific targeting of ALDH isoenzyme ALDH1A1 would provide similar benefit to broader pathway inhibition by disulfiram. NCT-505 and NCT-506 are isoenzyme-specific ALDH1A1 inhibitors whose activity was compared to the effects of disulfiram. Following treatment with both the NCTs and disulfiram, the viability of TICs versus adherent cells, sphere formation, and cell death in our in vitro relapse model were measured and compared in EOC cell lines. We found that disulfiram decreased the viability of TICs significantly more effectively versus adherent cells, while no consistent trend was observed when the cells were treated with the NCTs. Disulfiram also affected the expression of proteins associated with NFκB signaling. Comparison of disulfiram to the direct targeting of ALDH1A1 with the NCTs suggests that the broader cellular effects of disulfiram are more suitable as a therapeutic to eradicate TICs from tumors and prevent EOC relapse. In addition to providing insight into a fitting treatment for TICs, the comparison of disulfiram to NCT-505 and -506 has increased our understanding of the mechanism of action of disulfiram. Further elucidation of the mechanism of disulfiram has the potential to reveal additional targets to treat EOC TICs and prevent disease recurrence. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. Smac-mimetic enhances antitumor effect of standard chemotherapy in ovarian cancer models via Caspase 8-independent mechanism.
- Author
-
Hernandez, Lidia F., Dull, Angie B., Korrapati, Soumya, and Annunziata, Christina M.
- Published
- 2021
- Full Text
- View/download PDF
11. Driving Immune Responses in the Ovarian Tumor Microenvironment.
- Author
-
Ning, Franklin, Cole, Christopher B., and Annunziata, Christina M.
- Subjects
TUMOR microenvironment ,IMMUNE response ,OVARIAN tumors ,IMMUNE checkpoint inhibitors ,SUPPRESSOR cells ,OVARIAN cancer - Abstract
Ovarian cancer is the leading cause of death among gynecological neoplasms, with an estimated 14,000 deaths in 2019. First-line treatment options center around a taxane and platinum-based chemotherapy regimen. However, many patients often have recurrence due to late stage diagnoses and acquired chemo-resistance. Recent approvals for bevacizumab and poly (ADP-ribose) polymerase inhibitors have improved treatment options but effective treatments are still limited in the recurrent setting. Immunotherapy has seen significant success in hematological and solid malignancies. However, effectiveness has been limited in ovarian cancer. This may be due to a highly immunosuppressive tumor microenvironment and a lack of tumor-specific antigens. Certain immune cell subsets, such as regulatory T cells and tumor-associated macrophages, have been implicated in ovarian cancer. Consequently, therapies augmenting the immune response, such as immune checkpoint inhibitors and dendritic cell vaccines, may be unable to properly enact their effector functions. A better understanding of the various interactions among immune cell subsets in the peritoneal microenvironment is necessary to develop efficacious therapies. This review will discuss various cell subsets in the ovarian tumor microenvironment, current immunotherapy modalities to target or augment these immune subsets, and treatment challenges. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. Pilot Study Assessing Tolerability and Metabolic Effects of Metformin in Patients With Li-Fraumeni Syndrome.
- Author
-
Walcott, Farzana L, Wang, Ping-Yuan, Bryla, Christine M, Huffstutler, Rebecca D, Singh, Neha, Pollak, Michael N, Khincha, Payal P, Savage, Sharon A, Mai, Phuong L, Dodd, Kevin W, Hwang, Paul M, Fojo, Antonio T, and Annunziata, Christina M
- Subjects
METFORMIN ,LI-Fraumeni syndrome ,OXIDATIVE phosphorylation ,DRUG metabolism ,DRUG tolerance ,MEDICATION safety - Abstract
Background Li-Fraumeni syndrome (LFS) is a highly penetrant autosomal dominant cancer predisposition disorder caused by germline TP53 pathogenic variants. Patients with LFS have increased oxidative phosphorylation capacity in skeletal muscle and oxidative stress in blood. Metformin inhibits oxidative phosphorylation, reducing available energy for cancer cell proliferation and decreasing production of reactive oxygen species that cause DNA damage. Thus, metformin may provide pharmacologic risk reduction for cancer in patients with LFS, but its safety in nondiabetic patients with germline TP53 pathogenic variants has not been documented. Methods This study assessed safety and tolerability of metformin in nondiabetic LFS patients and measured changes in metabolic profiles. Adult patients with LFS and germline TP53 variant received 14 weeks of metformin. Blood samples were obtained for measurement of serum insulin-like growth factor–1, insulin, and insulin-like growth factor binding protein 3. Hepatic mitochondrial function was assessed with fasting exhaled CO
2 after ingestion of13 C-labeled methionine. Changes in serum metabolome were measured. All statistical tests were 2-sided. Results We enrolled 26 participants: 20 females and 6 males. The most common adverse events were diarrhea (50.0%) and nausea (46.2%). Lactic acidosis did not occur, and there were no changes in fasting glucose. Cumulative mean13 C exhalation was statistically significantly suppressed by metformin (P =.001). Mean levels of insulin-like growth factor binding protein 3 and insulin-like growth factor-1 were statistically significantly lowered (P =.02). Lipid metabolites and branched-chain amino acids accumulated. Conclusions Metformin was safe and tolerable in patients with LFS. It suppressed hepatic mitochondrial function as expected in these individuals. This study adds to the rationale for development of a pharmacologic risk-reduction clinical trial of metformin in LFS. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
13. Rapid image deconvolution and multiview fusion for optical microscopy.
- Author
-
Guo, Min, Li, Yue, Su, Yijun, Lambert, Talley, Nogare, Damian Dalle, Moyle, Mark W., Duncan, Leighton H., Ikegami, Richard, Santella, Anthony, Rey-Suarez, Ivan, Green, Daniel, Beiriger, Anastasia, Chen, Jiji, Vishwasrao, Harshad, Ganesan, Sundar, Prince, Victoria, Waters, Jennifer C., Annunziata, Christina M., Hafner, Markus, and Mohler, William A.
- Abstract
The contrast and resolution of images obtained with optical microscopes can be improved by deconvolution and computational fusion of multiple views of the same sample, but these methods are computationally expensive for large datasets. Here we describe theoretical and practical advances in algorithm and software design that result in image processing times that are tenfold to several thousand fold faster than with previous methods. First, we show that an 'unmatched back projector' accelerates deconvolution relative to the classic Richardson–Lucy algorithm by at least tenfold. Second, three-dimensional image-based registration with a graphics processing unit enhances processing speed 10- to 100-fold over CPU processing. Third, deep learning can provide further acceleration, particularly for deconvolution with spatially varying point spread functions. We illustrate our methods from the subcellular to millimeter spatial scale on diverse samples, including single cells, embryos and cleared tissue. Finally, we show performance enhancement on recently developed microscopes that have improved spatial resolution, including dual-view cleared-tissue light-sheet microscopes and reflective lattice light-sheet microscopes. Microscopy datasets are processed orders-of-magnitude faster with improved algorithms and deep learning. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. Leveraging locus-specific epigenetic heterogeneity to improve the performance of blood-based DNA methylation biomarkers.
- Author
-
Miller, Brendan F., Pisanic II, Thomas R., Margolin, Gennady, Petrykowska, Hanna M., Athamanolap, Pornpat, Goncearenco, Alexander, Osei-Tutu, Akosua, Annunziata, Christina M., Wang, Tza-Huei, and Elnitski, Laura
- Subjects
CELL-free DNA ,DNA methylation ,OVARIAN epithelial cancer ,EPIGENOMICS - Abstract
Background: Variation in intercellular methylation patterns can complicate the use of methylation biomarkers for clinical diagnostic applications such as blood-based cancer testing. Here, we describe development and validation of a methylation density binary classification method called EpiClass (available for download at https://github.com/Elnitskilab/EpiClass) that can be used to predict and optimize the performance of methylation biomarkers, particularly in challenging, heterogeneous samples such as liquid biopsies. This approach is based upon leveraging statistical differences in single-molecule sample methylation density distributions to identify ideal thresholds for sample classification. Results: We developed and tested the classifier using reduced representation bisulfite sequencing (RRBS) data derived from ovarian carcinoma tissue DNA and controls. We used these data to perform in silico simulations using methylation density profiles from individual epiallelic copies of ZNF154, a genomic locus known to be recurrently methylated in numerous cancer types. From these profiles, we predicted the performance of the classifier in liquid biopsies for the detection of epithelial ovarian carcinomas (EOC). In silico analysis indicated that EpiClass could be leveraged to better identify cancer-positive liquid biopsy samples by implementing precise thresholds with respect to methylation density profiles derived from circulating cell-free DNA (cfDNA) analysis. These predictions were confirmed experimentally using DREAMing to perform digital methylation density analysis on a cohort of low volume (1-ml) plasma samples obtained from 26 EOC-positive and 41 cancer-free women. EpiClass performance was then validated in an independent cohort of 24 plasma specimens, derived from a longitudinal study of 8 EOC-positive women, and 12 plasma specimens derived from 12 healthy women, respectively, attaining a sensitivity/specificity of 91.7%/100.0%. Direct comparison of CA-125 measurements with EpiClass demonstrated that EpiClass was able to better identify EOC-positive women than standard CA-125 assessment. Finally, we used independent whole genome bisulfite sequencing (WGBS) datasets to demonstrate that EpiClass can also identify other cancer types as well or better than alternative methylation-based classifiers. Conclusions: Our results indicate that assessment of intramolecular methylation density distributions calculated from cfDNA facilitates the use of methylation biomarkers for diagnostic applications. Furthermore, we demonstrated that EpiClass analysis of ZNF154 methylation was able to outperform CA-125 in the detection of etiologically diverse ovarian carcinomas, indicating broad utility of ZNF154 for use as a biomarker of ovarian cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
15. Evaluation of the Anti-Tumor Activity of the Humanized Monoclonal Antibody NEO-201 in Preclinical Models of Ovarian Cancer.
- Author
-
Zeligs, Kristen P., Morelli, Maria Pia, David, Justin M., Neuman, Monica, Hernandez, Lidia, Hewitt, Stephen, Ozaki, Michelle, Osei-Tutu, Akosua, Anderson, David, Andresson, Thorkell, Das, Sudipto, Lack, Justin, Abdelmaksoud, Abdalla, Fantini, Massimo, Arlen, Philip M., Tsang, Kwong Y., and Annunziata, Christina M.
- Subjects
ANIMAL models in research ,ANTIBODY-dependent cell cytotoxicity ,MONOCLONAL antibodies ,CELL adhesion molecules ,OVARIAN cancer ,TUMOR antigens - Abstract
Purpose: Despite high initial response rates with cytoreductive surgery, conventional chemotherapy and the incorporation of biologic agents, ovarian cancer patients often relapse and die from their disease. New approaches are needed to improve patient outcomes. This study was designed to evaluate the antitumor activity of NEO-201 monoclonal antibody (mAb) in preclinical models of ovarian cancer where the NEO-201 target is highly expressed. Experimental Design: Functional analysis of NEO-201 against tumor cell lines was performed by antibody-dependent cellular cytotoxicity (ADCC) assays. Binding of NEO-201 to tumor tissues and cell lines were determined by immunohistochemistry (IHC) and flow cytometry, respectively. Further characterization of the antigen recognized by NEO-201 was performed by mass spectrometry. Ovarian cancer models were used to evaluate the anti-tumor activity of NEO-201 in vivo. NEO-201 at a concentration of 250 g/mouse was injected intraperitoneally (IP) on days 1, 4, and 8. Human PBMCs were injected IP simultaneously as effector cells. Results: Both IHC and flow cytometry revealed that NEO-201 binds prominently to the colon, pancreatic, and mucinous ovarian cancer tissues and cell lines. Immunoprecipitation of the antigen recognized by NEO-201 was performed in human ovarian, colon, and pancreatic cancer cell lines. From these screening, carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) and CEACAM6 were identified as the most likely targets of NEO-201. Our results confirmed that NEO-201 binds different types of cancers; the binding is highly selective for the tumor cells without cross reactivity with the surrounding healthy tissue. Functional analysis revealed that NEO-201 mediates ADCC killing against human ovarian and colorectal carcinoma cell lines in vitro. In addition, NEO-201 inhibited tumor growth in the presence of activated human PBMCs in orthotopic mouse models of both primary and metastatic ovarian cancer. Importantly, NEO-201 prolonged survival of tumor-bearing mice. Conclusions: These data suggested that NEO-201 has an antitumor activity against tumor cells expressing its antigen. Targeting an antigen expressed in tumors, but not in normal tissues, allows patient selection for optimal treatment. These findings strongly indicate that NEO-201 warrants clinical testing as both a novel therapeutic and diagnostic agent for treatment of ovarian carcinomas. A first in human clinical trial evaluating NEO-201 in adults with chemo-resistant solid tumors is ongoing at the NIH clinical Center. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
16. The Monoclonal Antibody NEO-201 Enhances Natural Killer Cell Cytotoxicity Against Tumor Cells Through Blockade of the Inhibitory CEACAM5/CEACAM1 Immune Checkpoint Pathway.
- Author
-
Fantini, Massimo, David, Justin M., Annunziata, Christina M., Morelli, Maria Pia, Arlen, Phillip M., and Tsang, Kwong Y.
- Published
- 2020
- Full Text
- View/download PDF
17. A phase I study of the PD-L1 inhibitor, durvalumab, in combination with a PARP inhibitor, olaparib, and a VEGFR1--3 inhibitor, cediranib, in recurrent women's cancers with biomarker analyses.
- Author
-
Zimmer, Alexandra S., Nichols, Erin, Cimino-Mathews, Ashley, Peer, Cody, Liang Cao, Min-Jung Lee, Kohn, Elise C., Annunziata, Christina M., Lipkowitz, Stanley, Trepel, Jane B., Sharma, Rajni, Mikkilineni, Lekha, Gatti-Mays, Margaret, Figg, William D., Houston, Nicole D., and Jung-Min Lee
- Subjects
OLAPARIB ,TRIPLE-negative breast cancer ,CANCER patients ,POLY(ADP-ribose) polymerase ,PROGRAMMED death-ligand 1 ,ECULIZUMAB ,IPILIMUMAB ,LYMPHOPENIA - Abstract
Background: Strategies to improve activity of immune checkpoint inhibitors are needed. We hypothesized enhanced DNA damage by olaparib, a PARP inhibitor, and reduced VEGF signaling by cediranib, a VEGFR1--3 inhibitor, would complement anti-tumor activity of durvalumab, a PD-L1 inhibitor, and the 3-drug combination would be tolerable. Methods: This phase 1 study tested the 3-drug combination in a 3 + 3 dose escalation. Cediranib was taken intermittently (5 days on/2 days off) at 15 or 20 mg (dose levels 1 and 2, respectively) with durvalumab 1500 mg IV every 4 weeks, and olaparib tablets 300 mg twice daily. The primary end point was the recommended phase 2 dose (RP2D). Response rate, pharmacokinetic (PK), and correlative analyses were secondary endpoints. Results: Nine patients (7 ovarian/1 endometrial/1 triple negative breast cancers, median 3 prior therapies [2-6]) were treated. Grade 3/4 adverse events include hypertension (1/9), anemia (1/9) and lymphopenia (3/9). No patients experienced dose limiting toxicities. The RP2D is cediranib, 20 mg (5 days on/2 days off) with full doses of durvalumab and olaparib. Four patients had partial responses (44%) and 3 had stable disease lasting ≥6 months, yielding a 67% clinical benefit rate. No significant effects on olaparib or cediranib PK parameters from the presence of durvalumab, or the co-administration of cediranib or olaparib were identified. Tumoral PD-L1 expression correlated with clinical benefit but cytokines and peripheral immune subsets did not. Conclusions: The RP2D is tolerable and has preliminary activity in recurrent women's cancers. A phase 2 expansion study is now enrolling for recurrent ovarian cancer patients. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
18. Patients with BRCA mutated ovarian cancer may have fewer circulating MDSC and more peripheral CD8+ T cells compared with women with BRCA wild-type disease during the early disease course.
- Author
-
Lee, Jung-Min, Botesteanu, Dana-Adriana, Tomita, Yusuke, Yuno, Akira, Lee, Min-Jung, Kohn, Elise C., Annunziata, Christina M., Matulonis, Ursula, MacDonald, Lauren A., Nair, Jayakumar R., Macneill, Kimberley M., and Trepel, Jane B.
- Subjects
T cells ,SUPPRESSOR cells ,DISEASE progression ,BLOOD cells ,APOPTOSIS - Abstract
Immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) are associated with immunologic tolerance and poor prognosis in ovarian cancer (OvCa). We hypothesized that women with germline BRCA1 and BRCA2 mutation-associated (gBRCAm) OvCa would have fewer circulating immunosuppressive immune cells compared to those with BRCA wild-type (BRCAwt) disease during their early disease course (<5 years post-diagnosis) where gBRCAm is a favorable prognostic factor. We collected and viably froze peripheral blood mononuclear cells (PBMCs) from patients with recurrent OvCa olaparib clinical trials (NCT01445418/NCT01237067). Immune subset analyses were performed using flow cytometry for Tregs, exhausted CD8
+ T cells, monocytes and MDSCs. Functional marker expression, including cytotoxic T lymphocyte-associated protein 4 (CTLA-4), T cell immunoglobulin and mucin domain 3 (TIM-3) and programmed cell death protein 1 (PD-1) was evaluated. Data were analyzed using FlowJo. Pretreatment PBMCs were collected from 41 patients (16 gBRCAm/25 BRCAwt). The percentage of MDSCs among viable CD45+ PBMC was lower in gBRCAm OvCa compared with BRCAwt OvCa (median 0.565 vs. 0.93%, P=0.0086) but this difference was not seen in those women >5 years post-diagnosis. CD8+ T cells among viable CD45+ PBMCs and CTLA-4+ /CD8+ T cells were higher in gBRCAm carriers than patients with BRCAwt, in particular for those <5 years post-diagnosis (median 20.4 vs. 9.78%, P=0.031 and median MFI 0.19 vs. 0.22, P=0.0074, respectively). TIM-3 expression on Tregs was associated with poor progression-free survival, independent of gBRCAm status (P<0.001). Our pilot data suggested that patients with gBRCAm OvCa may have fewer circulating MDSCs but higher CD8+ T cells in PBMCs during their early disease course. This may contribute to the observed survival benefit for these women in their first post-diagnosis decade. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
19. An IL-15 Superagonist, ALT-803, Enhances Antibody-Dependent Cell-Mediated Cytotoxicity Elicited by the Monoclonal Antibody NEO-201 Against Human Carcinoma Cells.
- Author
-
Fantini, Massimo, David, Justin M., Wong, Hing C., Annunziata, Christina M., Arlen, Philip M., and Tsang, Kwong Y.
- Published
- 2019
- Full Text
- View/download PDF
20. Efficacy and Safety of Avelumab for Patients With Recurrent or Refractory Ovarian Cancer: Phase 1b Results From the JAVELIN Solid Tumor Trial.
- Author
-
Disis, Mary L., Taylor, Matthew H., Kelly, Karen, Beck, J. Thaddeus, Gordon, Michael, Moore, Kathleen M., Patel, Manish R., Chaves, Jorge, Park, Haeseong, Mita, Alain C., Hamilton, Erika P., Annunziata, Christina M., Grote, Hans Juergen, von Heydebreck, Anja, Grewal, Jaspreet, Chand, Vikram, and Gulley, James L.
- Published
- 2019
- Full Text
- View/download PDF
21. A Single-Arm, Open-Label Phase II Study of ONC201 in Recurrent/Refractory Metastatic Breast Cancer and Advanced Endometrial Carcinoma.
- Author
-
Atkins, Sarah L P, Greer, Yoshimi Endo, Jenkins, Sarah, Gatti-Mays, Margaret E, Houston, Nicole, Lee, Sunmin, Lee, Min-Jung, Rastogi, Shraddha, Sato, Nahoko, Burks, Christina, Annunziata, Christina M, Lee, Jung-Min, Nagashima, Kunio, Trepel, Jane B, Lipkowitz, Stanley, and Zimmer, Alexandra S
- Subjects
MITOCHONDRIAL pathology ,THERAPEUTIC use of antineoplastic agents ,DISEASE progression ,CLINICAL trials ,BIOPSY ,CANCER relapse ,METASTASIS ,TREATMENT effectiveness ,ENDOMETRIAL tumors ,DESCRIPTIVE statistics ,RESEARCH funding ,DRUG side effects ,BREAST tumors ,CARRIER proteins ,LONGITUDINAL method - Abstract
Background ONC201 is a small molecule that can cause nonapoptotic cell death through loss of mitochondrial function. Results from the phase I/II trials of ONC201 in patients with refractory solid tumors demonstrated tumor responses and prolonged stable disease in some patients. Methods This single-arm, open-label, phase II clinical trial evaluated the efficacy of ONC201 at the recommended phase II dose (RP2D) in patients with recurrent or refractory metastatic breast or endometrial cancer. Fresh tissue biopsies and blood were collected at baseline and at cycle 2 day 2 for correlative studies. Results Twenty-two patients were enrolled; 10 patients with endometrial cancer, 7 patients with hormone receptor–positive breast cancer, and 5 patients with triple-negative breast cancer. The overall response rate was 0%, and the clinical benefit rate, defined by complete response (CR) + partial response (PR) + stable disease (SD), was 27% (n = 3/11). All patients experienced an adverse event (AE), which was primarily low grade. Grade 3 AEs occurred in 4 patients; no grade 4 AEs occurred. Tumor biopsies did not show that ONC201 consistently induced mitochondrial damage or alterations in tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or the TRAIL death receptors. ONC201 treatment caused alterations in peripheral immune cell subsets. Conclusion ONC201 monotherapy did not induce objective responses in recurrent or refractory metastatic breast or endometrial cancer at the RP2D dose of 625 mg weekly but had an acceptable safety profile (ClinicalTrials.gov Identifier: NCT03394027). [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. Germline and Somatic Tumor Testing in Epithelial Ovarian Cancer: ASCO Guideline Summary.
- Author
-
Konstantinopoulos, Panagiotis A., Lacchetti, Christina, and Annunziata, Christina M.
- Subjects
MEDICAL protocols ,GENETIC mutation ,GENOMICS ,EARLY detection of cancer ,OVARIAN epithelial cancer - Abstract
The article provides recommendations on genetic and tumor testing for women diagnosed with epithelial ovarian cancer based on available evidence and expert consensus.
- Published
- 2020
- Full Text
- View/download PDF
23. IΚΚε cooperates with either MEK or non-canonical NF-kB driving growth of triple-negative breast cancer cells in different contexts.
- Author
-
House, Carrie D., Grajales, Valentina, Ozaki, Michelle, Jordan, Elizabeth, Wubneh, Helmae, Kimble, Danielle C., James, Jana M., Kim, Marianne K., and Annunziata, Christina M.
- Subjects
BREAST cancer ,CANCER cells ,ANOIKIS ,RNA interference ,MITOGEN-activated protein kinases - Abstract
Background: Metastatic breast cancer carries a poor prognosis despite the success of newly targeted therapies. Treatment options remain especially limited for the subtype of triple negative breast cancer (TNBC). Several signaling pathways, including NF-κB, are altered in TNBC, and the complexity of this disease implies multi-faceted pathway interactions. Given that IKKε behaves as an oncogene in breast cancer, we hypothesized that IKKε regulates NF-κB signaling to control diverse oncogenic functions in TNBC.Methods: Vector expression and RNA interference were used to investigate the functional role of IKKε in triple-negative breast cancer cells. Viability, protein expression, NF-κB binding activity, invasion, anoikis, and spheroid formation were examined in cells expressing high or low levels of IKKε, in conjunction with p52 RNA interference or MEK inhibition.Results: This study found that non-canonical NF-κB p52 levels are inversely proportional to ΙΚΚε, and growth of TNBC cells in anchorage supportive, high-attachment conditions requires IKKε and activated MEK. Growth of these cells in anchorage resistant conditions requires IKKε and activated MEK or p52. In this model, IKKε and MEK cooperate to support overall viability whereas the p52 transcription factor is only required for viability in low attachment conditions, underscoring the contrasting roles of these proteins.Conclusions: This study illustrates the diverse functions of IKKε in TNBC and highlights the adaptability of NF-κB signaling in maintaining cancer cell survival under different growth conditions. A better understanding of the diversity of NF-κB signaling may ultimately improve the development of novel therapeutic regimens for TNBC. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
24. Preclinical Characterization of a Novel Monoclonal Antibody NEO-201 for the Treatment of Human Carcinomas.
- Author
-
Fantini, Massimo, David, Justin M., Saric, Olga, Dubeykovskiy, Alexander, Cui, Yongzhi, Mavroukakis, Sharon A., Bristol, Andrew, Annunziata, Christina M., Tsang, Kwong Y., and Arlen, Philip M.
- Subjects
THERAPEUTIC use of monoclonal antibodies ,COLON cancer treatment ,DRUG side effects - Abstract
NEO-201 is a novel humanized IgG1 monoclonal antibody that was derived from an immunogenic preparation of tumor-associated antigens from pooled allogeneic colon tumor tissue extracts. It was found to react against a variety of cultured human carcinoma cell lines and was highly reactive against the majority of tumor tissues from many different carcinomas, including colon, pancreatic, stomach, lung, and breast cancers. NEO-201 also exhibited tumor specificity, as the majority of normal tissues were not recognized by this antibody. Functional assays revealed that treatment with NEO-201 is capable of mediating both antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against tumor cells. Furthermore, the growth of human pancreatic xenograft tumors in vivo was largely attenuated by treatment with NEO-201 both alone and in combination with human peripheral blood mononuclear cells as an effector cell source for ADCC. In vivo biodistribution studies in human tumor xenograft-bearing mice revealed that NEO-201 preferentially accumulates in the tumor but not organ tissue. Finally, a single-dose toxicity study in non-human primates demonstrated safety and tolerability of NEO-201, as a transient decrease in circulating neutrophils was the only related adverse effect observed. These findings indicate that NEO-201 warrants clinical testing as both a novel diagnostic and therapeutic agent for the treatment of a broad variety of carcinomas. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
25. SARC006: Phase II Trial of Chemotherapy in Sporadic and Neurofibromatosis Type 1 Associated Chemotherapy-Naive Malignant Peripheral Nerve Sheath Tumors.
- Author
-
Higham, Christine S., Steinberg, Seth M., Dombi, Eva, Perry, Arie, Helman, Lee J., Schuetze, Scott M., Ludwig, Joseph A., Staddon, Arthur, Milhem, Mohammed M., Rushing, Daniel, Jones, Robin L., Livingston, Michael, Goldman, Stewart, Moertel, Christopher, Wagner, Lars, Janhofer, David, Annunziata, Christina M., Reinke, Denise, Long, Lauren, and Viskochil, David
- Subjects
CANCER chemotherapy ,CLINICAL trials ,COMBINED modality therapy ,DOXORUBICIN ,ETOPOSIDE ,NERVOUS system tumors ,TREATMENT effectiveness ,NEUROFIBROMATOSIS 1 ,IFOSFAMIDE - Abstract
Background. Worse chemotherapy response for neurofibromatosis type 1- (NF1-) associated compared to sporadic malignant peripheral nerve sheath tumors (MPNST) has been reported. Methods. We evaluated the objective response (OR) rate of patients with AJCC Stage III/IV chemotherapy-naive NF1 MPNST versus sporadic MPNST after 4 cycles of neoadjuvant chemotherapy, 2 cycles of ifosfamide/doxorubicin, and 2 cycles of ifosfamide/etoposide. A Simon optimal two-stage design was used (target response rate 40%). Results. 34 NF1 (median age 33 years) and 14 sporadic (median age 40 years) MPNST patients enrolled. Five of 28 (17.9%) evaluable NF1 MPNST patients had a partial response (PR), as did 4 of 9 (44.4%) patients with sporadic MPNST. Stable disease (SD) was achieved in 22 NF1 and 4 sporadic MPNST patients. In both strata, results in the initial stages met criteria for expansion of enrollment. Only 1 additional PR was observed in the expanded NF1 stratum. Enrollment was slower than expected and the trial closed before full accrual. Conclusions. This trial was not powered to detect differences in response rates between NF1 and sporadic MPNST. While the OR rate was lower in NF1 compared to sporadic MPNST, qualitative responses were similar, and disease stabilization was achieved in most patients. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
26. Evidence for a Mesothelial Origin of Body Cavity Effusion Lymphomas.
- Author
-
Sanchez-Martin, David, Uldrick, Thomas S., Kwak, Hyeongil, Ohnuki, Hidetaka, Polizzotto, Mark N., Annunziata, Christina M., Raffeld, Mark, Wyvill, Kathleen M., Aleman, Karen, Victoria Wang, Marshall, Vickie A., Whitby, Denise, Yarchoan, Robert, Tosato, Giovanna, and Wang, Victoria
- Subjects
LYMPHOMAS ,LYMPHATIC cancer -- Etiology ,KAPOSI'S sarcoma-associated herpesvirus ,EPSTEIN-Barr virus ,BODY cavities ,MIXED infections ,POLYMERASE chain reaction ,LABORATORY mice - Abstract
Background: Primary effusion lymphoma (PEL) is a Kaposi's sarcoma herpes virus (KSHV)-induced lymphoma that typically arises in body cavities of HIV-infected patients. PEL cells are often co-infected with Epstein-Barr virus (EBV). "PEL-like" lymphoma is a KSHV-unrelated lymphoma that arises in body cavities of HIV-negative patients. "PEL-like" lymphoma is sometimes EBV positive. The derivation of PEL/"PEL-like" cells is unclear.Methods: Mesothelial cells were cultured from body cavity effusions of 23 patients. Cell proliferation, cytokine secretion, marker phenotypes, KSHV/EBV infection, and clonality were evaluated by standard methods. Gene expression was measured by quantitative polymerase chain reaction and immunoblotting. A mouse model of PEL (3 mice/group) was used to evaluate tumorigenicity.Results: We found that the mesothelia derived from six effusions of HIV-infected patients with PEL or other KSHV-associated diseases contained rare KSHV + or EBV + mesothelial cells. After extended culture (16-17 weeks), some mesothelial cells underwent a trans-differentiation process, generating lymphoid-type CD45 + /B220 + , CD5 + , CD27 + , CD43 + , CD11c + , and CD3 - cells resembling "B1-cells," most commonly found in mouse body cavities. These "B1-like" cells were short lived. However, long-term KSHV + EBV - and EBV + KSHV - clonal cell lines emerged from mesothelial cultures from two patients that were clonally distinct from the monoclonal or polyclonal B-cell populations found in the patients' original effusions.Conclusions: Mesothelial-to-lymphoid transformation is a newly identified in vitro process that generates "B1-like" cells and is associated with the emergence of long-lived KSHV or EBV-infected cell lines in KSHV-infected patients. These results identify mesothelial cultures as a source of PEL cells and lymphoid cells in humans. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
27. Population pharmacokinetic analyses of the effect of carboplatin pretreatment on olaparib in recurrent or refractory women's cancers.
- Author
-
Peer, Cody, Lee, Jung-Min, Roth, Jeffrey, Rodgers, Louis, Nguyen, Jeffers, Annunziata, Christina, Minasian, Lori, Kohn, Elise, Figg, William, Peer, Cody J, Annunziata, Christina M, Kohn, Elise C, and Figg, William D
- Subjects
CARBOPLATIN ,CANCER in women ,COMPUTER simulation ,PHARMACOKINETICS ,DRUG interactions ,THERAPEUTICS - Abstract
Purpose: Combining olaparib with carboplatin was recently shown to be active in both BRCA and non-BRCA mutant cancers in a recent phase I/Ib combination trial. The optimal drug sequence recommended was carboplatin 1-day before olaparib. However, carboplatin pre-treatment induced a ~50% faster olaparib clearance.Methods: To further explore this drug interaction, a population pharmacokinetic (PK) model was designed that included a lag time parameter, a second absorption compartment from tablet formulation, a single distribution/elimination compartment, and covariance among the clearance and volume parameters.Results: Clearance (6.8 L/h) and volume (33 L) estimates were comparable with literature. The only significant covariate was the presence of carboplatin on olaparib clearance, consistent with published noncompartmental PK and in vitro data.Conclusions: Simulations predicted lower steady-state peak/trough olaparib exposure through 24-36 h post carboplatin pre-treatment, but this effect was lost by day 2 and thus no dose adjustment is recommended. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
28. Identification of therapeutic targets applicable to clinical strategies in ovarian cancer.
- Author
-
Kim, Marianne K., Caplen, Natasha, Chakka, Sirisha, Hernandez, Lidia, House, Carrie, Pongas, Georgios, Jordan, Elizabeth, and Annunziata, Christina M.
- Subjects
SMALL interfering RNA ,OVARIAN cancer diagnosis ,OVARIAN cancer treatment ,LAPATINIB ,CANCER chemotherapy ,CELL-mediated cytotoxicity ,THERAPEUTICS ,FLOW cytometry ,OVARIAN tumors ,RNA ,WESTERN immunoblotting ,DNA-binding proteins ,GENE expression profiling ,SEQUENCE analysis - Abstract
Background: shRNA-mediated lethality screening is a useful tool to identify essential targets in cancer biology. Ovarian cancer (OC) is extremely heterogeneous and most cases are advanced stages at diagnosis. OC has a high response rate initially, but becomes resistant to standard chemotherapy. We previously employed high throughput global shRNA sensitization screens to identify NF-kB related pathways. Here, we re-analyzed our previous shRNA screens in an unbiased manner to identify clinically applicable molecular targets.Methods: We proceeded with siRNA lethality screening using the top 55 genes in an expanded set of 6 OC cell lines. We investigated clinical relevance of candidate targets in The Cancer Genome Atlas OC dataset. To move these findings towards the clinic, we chose four pharmacological inhibitors to recapitulate the top siRNA effects: Oxozeaenol (for MAP3K7/TAK1), BI6727 (PLK1), MK1775 (WEE1), and Lapatinib (ERBB2). Cytotoxic effects were measured by cellular viability assay, as single agents and in 2-way combinations. Co-treatments were evaluated in either sequential or simultaneous exposure to drug for short term and extended periods to simulate different treatment strategies.Results: Loss-of-function shRNA screens followed by short-term siRNA validation screens identified therapeutic targets in OC cells. Candidate genes were dysregulated in a subset of TCGA OCs although the alterations of these genes showed no statistical significance to overall survival. Pharmacological inhibitors such as Oxozeaenol, BI6727, and MK1775 showed cytotoxic effects in OC cells regardless of cisplatin responsiveness, while all OC cells tested were cytostatic to Lapatinib. Co-treatment with BI6727 and MK1775 at sub-lethal concentrations was equally potent to BI6727 alone at lethal concentrations without cellular re-growth after the drugs were washed off, suggesting the co-inhibition at reduced dosages may be more efficacious than maximal single-agent cytotoxic concentrations.Conclusions: Loss-of-function screen followed by in vitro target validation using chemical inhibitors identified clinically relevant targets. This approach has the potential to systematically refine therapeutic strategies in OC. These molecular target-driven strategies may provide additional therapeutic options for women whose tumors have become refractory to standard chemotherapy. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
29. Pharmacodynamic markers and clinical results from the phase 2 study of the SMAC mimetic birinapant in women with relapsed platinum-resistant or -refractory epithelial ovarian cancer.
- Author
-
Noonan, Anne M., Bunch, Kristen P., Chen, Jin‐Qiu, Herrmann, Michelle A., Lee, Jung‐Min, Kohn, Elise C., O'Sullivan, Ciara C., Jordan, Elizabeth, Houston, Nicole, Takebe, Naoko, Kinders, Robert J., Cao, Liang, Peer, Cody J., Figg, W. Douglas, Annunziata, Christina M., Chen, Jin-Qiu, and Lee, Jung-Min
- Subjects
OVARIAN cancer ,OVARIAN cancer treatment ,APOPTOSIS ,KILLER cells ,PHARMACODYNAMICS ,PROTEIN metabolism ,PLATINUM compounds ,ENZYME metabolism ,ADENOCARCINOMA ,ANTINEOPLASTIC agents ,B cells ,CLINICAL trials ,COMPARATIVE studies ,DRUG resistance in cancer cells ,EPITHELIAL cell tumors ,RESEARCH methodology ,MEDICAL cooperation ,OLIGOPEPTIDES ,OVARIAN tumors ,PROGNOSIS ,PROTEINS ,RESEARCH ,RESEARCH funding ,T cells ,TUMORS ,ENDOMETRIAL tumors ,EVALUATION research ,TREATMENT effectiveness ,LYMPHOPENIA ,INDOLE compounds ,SIGNAL peptides ,LYMPHOCYTE count ,MONONUCLEAR leukocytes ,THERAPEUTICS - Abstract
Background: Inhibitors of apoptosis proteins (IAPs) are key regulators of apoptosis and are frequently dysregulated in ovarian cancer. It was hypothesized that blocking IAPs with birinapant would increase tumor cell death and result in objective responses for women with platinum-refractory and -resistant ovarian cancer.Methods: In this phase 2, Cancer Therapy Evaluation Program-sponsored study, patients received birinapant at 47 mg/m(2) on days 1, 8, and 15 of 28-day cycles. Pharmacokinetics were obtained during cycle 1. Plasma, peripheral blood mononuclear cells (PBMCs), and percutaneous tumor biopsy samples were collected before cycle 1 and after 6 weeks. The primary endpoint was an objective response or progression-free survival lasting greater than 6 months in a mini-max design.Results: Eleven patients received birinapant; after this, accrual was terminated for lack of a clinical benefit. Birinapant was well tolerated, with predominantly grade 2 adverse events and 1 case of grade 3 lymphopenia. Pretreatment biopsy samples and PBMCs were collected; paired posttreatment biopsy samples and PBMCs were collected from 7 and 10 patients, respectively. There was consistent downregulation of cellular inhibitor of apoptosis protein 1 in tumors (P = .016) and PBMCs (P < .01). Procaspase 3 also decreased in tumors (P = .031) and PBMCs (P < .01); cleaved caspase 3 colocalized with H2A histone family member X (γ-H2AX) in tumors after birinapant exposure. Peripheral T and B cells decreased significantly after treatment, but natural killer cells did not (P = .04, P = .05, and P = .43, respectively).Conclusions: Birinapant shows consistent target suppression in vivo without single-agent antitumor activity in this small population. Single-agent pharmacodynamics are necessary to understand the drug's mechanism of action and set the stage for rational combination therapy. Preclinical studies are ongoing to identify optimal synergistic combinations for future clinical trials. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
30. The role of reproductive hormones in epithelial ovarian carcinogenesis.
- Author
-
Gharwan, Helen, Bunch, Kristen P., and Annunziata, Christina M.
- Subjects
OVARIAN cancer ,SEX hormones ,IMMUNE system ,DRUG development ,CANCER invasiveness ,EPITHELIAL cells - Abstract
Epithelial ovarian cancer comprises ~85% of all ovarian cancer cases. Despite acceptance regarding the influence of reproductive hormones on ovarian cancer risk and considerable advances in the understanding of epithelial ovarian carcinogenesis on a molecular level, complete understanding of the biologic processes underlying malignant transformation of ovarian surface epithelium is lacking. Various hypotheses have been proposed over the past several decades to explain the etiology of the disease. The role of reproductive hormones in epithelial ovarian carcinogenesis remains a key topic of research. Primary questions in the field of ovarian cancer biology center on its developmental cell of origin, the positive and negative effects of each class of hormones on ovarian cancer initiation and progression, and the role of the immune system in the ovarian cancer microenvironment. The development of the female reproductive tract is dictated by the hormonal milieu during embryogenesis. Intensive research efforts have revealed that ovarian cancer is a heterogenous disease that may develop from multiple extra-ovarian tissues, including both Müllerian (fallopian tubes, endometrium) and non-Müllerian structures (gastrointestinal tissue), contributing to its heterogeneity and distinct histologic subtypes. The mechanism underlying ovarian localization, however, remains unclear. Here, we discuss the role of reproductive hormones in influencing the immune system and tipping the balance against or in favor of developing ovarian cancer. We comment on animal models that are critical for experimentally validating existing hypotheses in key areas of endocrine research and useful for preclinical drug development. Finally, we address emerging therapeutic trends directed against ovarian cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
31. Molecular Targets.
- Author
-
Annunziata, Christina M. and Dennis, Phillip A.
- Published
- 2014
- Full Text
- View/download PDF
32. Are beta-blockers on the therapeutic horizon for ovarian cancer treatment?
- Author
-
Bunch, Kristen P. and Annunziata, Christina M.
- Subjects
ADRENERGIC beta blockers ,OVARIAN tumors - Abstract
Widely used medications for other disease processes, such as beta‐blockers used for the management of hypertension, may provide therapeutic potential in patients with cancer, perhaps by altering the tumor microenvironment. The article by Watkins et al in the current issue performed a comprehensive review of beta‐blocker use in women with ovarian cancer, and identified nonselective beta‐blockers associated with longer overall survival. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
33. Topotecan synergizes with CHEK1 (CHK1) inhibitor to induce apoptosis in ovarian cancer cells.
- Author
-
Kim, Marianne K., James, Jana, and Annunziata, Christina M.
- Subjects
OVARIAN cancer treatment ,TOPOTECAN ,APOPTOSIS ,DRUG synergism ,ENZYME inhibitors ,WOMEN'S health ,THERAPEUTICS ,CHECKPOINT kinase 1 - Abstract
Background: Topotecan (TPT) is a therapeutic option for women with platinum-resistant or -refractory ovarian cancer. However, the dose-limiting toxicity of TPT is myelosuppression. This led us to seek a combination treatment to augment TPT anti-cancer activity in a cancer-targeted manner. Ovarian serous cancers, a major subtype, show dysregulated DNA repair pathway and often display a high level of CHEK1 (CHK1), a cell cycle regulator and DNA damage sensor. CHEK1 inhibitors are a novel approach to treatment, and have been used as single agents or in combination chemotherapy in many cancers. Methods: We evaluated the cellular effects of TPT in a panel of high grade serous (HGS) and non-HGS ovarian cancer cells. We then determined IC50s of TPT in the absence and presence of CHEK1 inhibitor, PF477736. Synergism between TPT and PF477736 was calculated based on cellular viability assays. Cytotoxic effect of the combined treatment was compared with apoptotic activities by Caspase3/7 activity assay and Western blotting of cleaved-PARP1 and γH2AX. Results: Non-HGS ovarian cancer cells were generally more sensitive to TPT treatment compared to HGS ovarian cancer cells. When combined with CHEK1 inhibitor, TPT potently and synergistically inhibited the proliferation of HGS ovarian cancer cells. This dramatic synergism in cellular toxicity was consistent with increases in markers of apoptosis. Conclusions: Our findings suggest that the addition of CHEK1 inhibitor increases the response of ovarian cancer cells to TPT. Furthermore, reduced dosages of both drugs achieved maximal cytotoxic effects by combining TPT with CHEK1 inhibitor. This strategy would potentially minimize side effects of the drugs for extended clinical benefit. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
34. The small molecule NSC676914A is cytotoxic and differentially affects NFκB signaling in ovarian cancer cells and HEK293 cells.
- Author
-
Sagher, Ethan, Hernandez, Lidia, Heywood, Callee, Pauly, Gary T., Young, Matthew R., Schneider, Joel, Colburn, Nancy H., and Annunziata, Christina M.
- Subjects
CYTOTOXIC T cells ,OVARIAN cancer ,CARCINOGENS ,REGENERATION (Biology) ,EMBRYOLOGY - Abstract
Background The small molecule NSC676914A was previously identified as an NF-κB inhibitor in TPAstimulated HEK293 cells (Mol Can Ther 8:571-581, 2009). We hypothesized that this effect would also be seen in ovarian cancer cells, and serve as its mechanism of cytotoxicity. OVCAR3 and HEK293 cell lines stably containing a NF-κB luciferase reporter gene were generated. Methods Levels of NF-κB activity were assessed by luciferase reporter assays, after stimulation with LPA, LPS, TPA, and TNFα, in the presence or absence of a known NF-κB inhibitor or NSC676914A, and cytotoxicity was measured. Results NSC676914A was toxic to both OVCAR3 and HEK293 cells. We also investigated the cytotoxicity of NSC676914A on a panel of lymphoma cell lines with well characterized mutations previously shown to determine sensitivity or resistance to NF-κB inhibition. The compound did not show predicted patterns of effects on NF-κB activity in either lymphoma, ovarian or HEK293 cell lines. In HEK293 cells, the small molecule inhibited NF-κB when cells were stimulated, while in OVCAR3 cells it only partially inhibited NF-κB. Interestingly, we observed rescue of cell death with ROS inhibition. Conclusions The current study suggests that the effect of NSC676914A on NF-κB depends on cell type and the manner in which the pathway is stimulated. Furthermore, as it is similarly toxic to lymphoma, OVCAR3 and HEK293 cells, NSC676914A shows promising NF-κBindependent anti-cancer activity in ovarian tumor cells. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
35. Phase l/lb Study of Olaparib and Carboplatin in BRCA1 or BRCA2 Mutation-Associated Breast or Ovarian Cancer With Biomarker Analyses.
- Author
-
Jung-Min Lee, Hays, John L., Annunziata, Christina M., Noonan, Anne M., Minasian, Lori, Zujewski, Jo Anne, Minshu Yu, Gordon, Nicolas, Jiuping Ji, Sissung, Tristan M., Figg, William D., Azad, Nilofer, Wood, Bradford J., Doroshow, James, and Kohn, Elise C.
- Subjects
OVARIAN cancer ,GERM cells ,BRCA genes ,CARBOPLATIN ,BIOMARKERS ,PROTEOMICS ,APOPTOSIS - Abstract
Background Olaparib has single-agent activity against breast/ovarian cancer (BrCa/OvCa) in germline BRCA1 or BRCA2 mutation carriers (gBRCAm). We hypothesized addition of olaparib to carboplatin can be administered safely and yield preliminary clinical activity. Methods Eligible patients had measurable or evaluable disease, gBRCAm, and good end-organ function. A 3 + 3 dose escalation tested daily oral capsule olaparib (100 or 200mg every 12 hours; dose levell or 2) with carboplatin area under the curve (AUC) on day 8 (AUC3 day 8), then every 21 days. For dose levels 3 to 6, patients were given olaparib days 1 to 7 at 200 and 400 mg every 12 hours, with carboplatin AUC3 to 5 on day 1 or 2 every 21 days; a maximum of eight combination cycles were permitted, after which daily maintenance of olaparib 400 mg every12 hours continued until progression. Dose-limiting toxicity was defined in the first two cycles. Peripheral blood mononuclear cells were collected for polymorphism analysis and polyADP-ribose incorporation. Paired tumor biopsies (before/after cycle 1) were obtained for biomarker proteomics and apoptosis endpoints. Results Forty-five women (37 OvCa/8 BrCa) were treated. Dose-limiting toxicity was not reached on the intermittent schedule. Expansion proceeded with olaparib 400 mg every 12 hours on days 1 to 7/carboplatin AUC5. Grade 3/4 adverse events included neutropenia (42.2%), thrombocytopenia (20.0%), and anemia (15.6%). Responses included 1 complete response (1 BrCa; 23 months) and 21 partial responses (50.0%; 15 OvCa; 6 BrCa; median = 16 [4 to > 45] in OvCa and 10 [6 to > 40] months in BrCa). Proteomic analysis suggests high pretreatment pS209-elF4E and FOX03a correlated with duration of response (two-sided P < .001; Pearson's R² = 0.94). Conclusions Olaparib capsules 400 mg every 12 hours on days 1 to 7/carboplatin AUC5 is safe and has activity in gBRCAm BrCa/OvCa patients. Exploratory translational studies indicate pretreatment tissue FOX03a expression may be predictive for response to therapy, requiring prospective validation. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
36. Pathway-Specific Engineered Mouse Allograft Models Functionally Recapitulate Human Serous Epithelial Ovarian Cancer.
- Author
-
Szabova, Ludmila, Bupp, Sujata, Kamal, Muhaymin, Householder, Deborah B., Hernandez, Lidia, Schlomer, Jerome J., Baran, Maureen L., Yi, Ming, Stephens, Robert M., Annunziata, Christina M., Martin, Philip L., Van Dyke, Terry A., Ohler, Zoe Weaver, and Difilippantonio, Simone
- Subjects
CARCINOMA ,LABORATORY mice ,HOMOGRAFTS ,OVARIAN cancer diagnosis ,CANCER-related mortality ,OVARIAN cancer treatment - Abstract
The high mortality rate from ovarian cancers can be attributed to late-stage diagnosis and lack of effective treatment. Despite enormous effort to develop better targeted therapies, platinum-based chemotherapy still remains the standard of care for ovarian cancer patients, and resistance occurs at a high rate. One of the rate limiting factors for translation of new drug discoveries into clinical treatments has been the lack of suitable preclinical cancer models with high predictive value. We previously generated genetically engineered mouse (GEM) models based on perturbation of Tp53 and Rb with or without Brca1 or Brca2 that develop serous epithelial ovarian cancer (SEOC) closely resembling the human disease on histologic and molecular levels. Here, we describe an adaptation of these GEM models to orthotopic allografts that uniformly develop tumors with short latency and are ideally suited for routine preclinical studies. Ovarian tumors deficient in Brca1 respond to treatment with cisplatin and olaparib, a PARP inhibitor, whereas Brca1-wild type tumors are non-responsive to treatment, recapitulating the relative sensitivities observed in patients. These mouse models provide the opportunity for evaluation of effective therapeutics, including prediction of differential responses in Brca1-wild type and Brca1–deficient tumors and development of relevant biomarkers. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
37. Phase 1, open-label study of MEDI-547 in patients with relapsed or refractory solid tumors.
- Author
-
Annunziata, Christina M., Kohn, Elise C., LoRusso, Patricia, Houston, Nicole D., Coleman, Robert L., Buzoianu, Manuela, Robbie, Gabriel, and Lechleider, Robert
- Published
- 2013
- Full Text
- View/download PDF
38. Targeting the proteasome with bortezomib in multiple myeloma: update on therapeutic benefit as an upfront single agent, induction regimen for stem-cell transplantation and as maintenance therapy.
- Author
-
Driscoll JJ, Burris J, Annunziata CM, Driscoll, James J, Burris, Jason, and Annunziata, Christina M
- Published
- 2012
- Full Text
- View/download PDF
39. Nuclear factor kappaB transcription factors are coexpressed and convey a poor outcome in ovarian cancer.
- Author
-
Annunziata CM, Stavnes HT, Kleinberg L, Berner A, Hernandez LF, Birrer MJ, Steinberg SM, Davidson B, Kohn EC, Annunziata, Christina M, Stavnes, Helene Tuft, Kleinberg, Lilach, Berner, Aasmund, Hernandez, Lidia F, Birrer, Michael J, Steinberg, Seth M, Davidson, Ben, and Kohn, Elise C
- Abstract
Background: Recent work has suggested a role for nuclear factor kappaB (NF-kappaB) in the propagation of ovarian cancer cell lines, but the significance and mechanism of NF-kappaB in ovarian cancer is unknown. The authors hypothesized that the NF-kappaB pathway is over activated in aggressive ovarian cancers.Methods: The levels of 3 NF-kappaB transcription factors, the activating inhibitors of NF-kappaB (IkappaB) kinases, and the NF-kappaB target matrix metalloproteinase 9 (MMP9) were assessed by immunohistochemistry in specimens of ovarian cancer that were obtained at diagnosis from a cohort of 33 patients who subsequently received combined paclitaxel, cisplatin, and cyclophosphamide. Associations were made between NF-kappaB pathway proteins and outcome. The validation of coexpression was performed at the gene level in 2 independently collected cohorts of 185 and 153 ovarian cancers.Results: The presence of NF-kappaB proteins in newly diagnosed advanced ovarian cancers was established, and a potential association with overall survival was identified. Transcription factors p65 and v-rel reticuloendotheliosis viral oncogene homolog B (RelB) were coexpressed with IkappaB kinase alpha, 1 component of a key trimolecular regulatory complex. Coexpression of the NF-kappaB machinery suggested activity of NF-kappaB signaling in these ovarian tumors. A significant association of p50 with poor overall survival was observed (P = .02). MMP9 expression had the opposite association, in which patients who had tumors without MMP9 staining had the poorest prognosis (P = .01), and this association held true at the gene expression level in an independently collected cohort of 185 ovarian cancers.Conclusions: The deregulation of NF-kappaB activity may influence outcome in women who receive standard therapy for advanced ovarian cancer. Modification of the NF-kappaB pathway may present an opportunity to improve outcome in the subset of women who have pathway activity. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
40. Nuclear Factor κB Transcription Factors Are Coexpressed and Convey a Poor Outcome in Ovarian Cancer.
- Author
-
Annunziata, Christina M., Stavnes, Helene Tuft, Kleinberg, Lilach, Berner, Aasmund, Hernandez, Lidia F., Birrer, Michael J., Steinberg, Seth M., Davidson, Ben, and Kohn, Elise C.
- Subjects
NF-kappa B ,OVARIAN cancer ,CANCER cells ,CELL lines ,TRANSCRIPTION factors - Abstract
The article details a study which examined the role of nuclear factor κB (NF-κB) in the propagation of ovarian cancer cell lines. Study authors assessed the levels of 3 NF-κB transcription factors, the activating inhibitors of NF-κB kinases, and the NF-κB target matrix metalloproteinase 9 (NMP9). They found that NF-κB proteins are present in newly diagnosed advanced ovarian cancers.
- Published
- 2010
- Full Text
- View/download PDF
41. Lack of Reliability of CA125 Response Criteria With Anti-VEGF Molecularly Targeted Therapy.
- Author
-
Azad, Nilofer S., Annunziata, Christina M., Steinberg, Seth M., Minasian, Lori, Premkumar, Ahalya, Chow, Catherine, Kotz, Herbert L., and Kohn, Elise C.
- Subjects
ANTIGENS ,OVARIAN cancer ,CANCER treatment ,ANTINEOPLASTIC agents ,BEVACIZUMAB - Abstract
The article discusses a study on the use of CA125 to predict disease behavior in patients receiving sorafenib, a Raf-kinase/VEGFR2 inhibitor, and bevacizumab. CA125 is capable of indicating epithelial ovarian cancer (EOC) response and can be used to monitor patients treated with cytotoxic chemotherapy. Key findings indicated that CA125 changes may not correspond to imaging response criteria for EOC patients who are receiving sorafenib and bevacizumab.
- Published
- 2008
- Full Text
- View/download PDF
42. ECT in a patient with a deep brain-stimulating electrode in place.
- Author
-
Moscarillo, Frank M., Annunziata, Christina M., Moscarillo, F M, and Annunziata, C M
- Published
- 2000
- Full Text
- View/download PDF
43. Matrix Drug Screen Identifies Synergistic Drug Combinations to Augment SMAC Mimetic Activity in Ovarian Cancer.
- Author
-
Noonan, Anne M., Cousins, Amanda, Anderson, David, Zeligs, Kristen P., Bunch, Kristen, Hernandez, Lidia, Shibuya, Yusuke, Goldlust, Ian S., Guha, Rajarshi, Ferrer, Marc, Thomas, Craig J., and Annunziata, Christina M.
- Subjects
ANIMAL experimentation ,ANTINEOPLASTIC agents ,APOPTOSIS ,CANCER relapse ,CELL lines ,CELLULAR signal transduction ,CYTOPLASM ,DRUG synergism ,MICE ,OVARIAN tumors ,TUMOR necrosis factors ,DOCETAXEL ,DNA-binding proteins ,CASPASES ,INVESTIGATIONAL drugs ,IN vitro studies ,PHARMACODYNAMICS - Abstract
Simple Summary: Recurrent ovarian cancer is difficult to treat due to the development of chemotherapy resistance. This resistance develops through multiple mechanisms to include the avoidance of cell death by cancer cells. Prior studies have shown birinapant, a second mitochondrial activator of caspases (SMAC) mimetic drug, to be promising in overcoming this acquired resistance. Despite good tolerability, however, therapy with single-agent birinapant exhibited minimal anti-cancer activity in women with recurrent ovarian cancer. By using a high-throughput drug screen we were able to identify potential therapeutic agents that augment birinapant activity, with docetaxel emerging favorably due to its marked synergy and known utility in the recurrent ovarian cancer setting. We showed that this synergy is the result of several complementary molecular pathways and hope to highlight the promising potential of this therapeutic drug combination for clinical testing where treatment options are often limited. Inhibitor of apoptosis (IAP) proteins are frequently upregulated in ovarian cancer, resulting in the evasion of apoptosis and enhanced cellular survival. Birinapant, a synthetic second mitochondrial activator of caspases (SMAC) mimetic, suppresses the functions of IAP proteins in order to enhance apoptotic pathways and facilitate tumor death. Despite on-target activity, however, pre-clinical trials of single-agent birinapant have exhibited minimal activity in the recurrent ovarian cancer setting. To augment the therapeutic potential of birinapant, we utilized a high-throughput screening matrix to identify synergistic drug combinations. Of those combinations identified, birinapant plus docetaxel was selected for further evaluation, given its remarkable synergy both in vitro and in vivo. We showed that this synergy results from multiple convergent pathways to include increased caspase activation, docetaxel-mediated TNF-α upregulation, alternative NF-kB signaling, and birinapant-induced microtubule stabilization. These findings provide a rationale for the integration of birinapant and docetaxel in a phase 2 clinical trial for recurrent ovarian cancer where treatment options are often limited and minimally effective. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
44. Uterine Papillary Serous Carcinoma: A New Paradigm for Treatment?
- Author
-
Annunziata, Christina M. and Kotz, Herbert L.
- Subjects
MEDICAL research ,CANCER patients ,UTERINE fibroids ,SURGERY ,DRUG therapy ,RADIATION ,CANCER - Abstract
In this article, the author reflects on the study that evaluates patients with stage 1 uterine papillary serous carcinoma (UPSC) who were treated with surgery. It argues that the study analysis was not powered to determine whether the addition of pelvic radiation to chemotherapy resulted in improvements in recurrence and survival outcomes. It suggests that personalized therapy will be achieved only after better biochemical characterization to define the molecular progression of the disease.
- Published
- 2009
- Full Text
- View/download PDF
45. Drugs Targeting Tumor-Initiating Cells Prolong Survival in a Post-Surgery, Post-Chemotherapy Ovarian Cancer Relapse Model.
- Author
-
Harrington, Brittney S., Ozaki, Michelle K., Caminear, Michael W., Hernandez, Lidia F., Jordan, Elizabeth, Kalinowski, Nicholas J., Goldlust, Ian S., Guha, Rajarshi, Ferrer, Marc, Thomas, Craig, Shetty, Jyoti, Tran, Bao, Wong, Nathan, House, Carrie D., and Annunziata, Christina M.
- Subjects
DISEASE risk factors ,MORTALITY risk factors ,REACTIVE oxygen species ,ANTINEOPLASTIC agents ,CANCER chemotherapy ,CANCER relapse ,CELL death ,CELL lines ,CELLULAR signal transduction ,COMPARATIVE studies ,DISULFIRAM ,OVARIAN tumors ,PHOSPHORYLATION ,RNA ,SURVIVAL ,OXIDATIVE stress ,CELL survival ,CARBOPLATIN ,SEQUENCE analysis ,IN vitro studies ,IN vivo studies ,PHARMACODYNAMICS - Abstract
Disease recurrence is the major cause of morbidity and mortality of ovarian cancer (OC). In terms of maintenance therapies after platinum-based chemotherapy, PARP inhibitors significantly improve the overall survival of patients with BRCA mutations but is of little benefit to patients without homologous recombination deficiency (HRD). The stem-like tumor-initiating cell (TIC) population within OC tumors are thought to contribute to disease recurrence and chemoresistance. Therefore, there is a need to identify drugs that target TICs to prevent relapse in OC without HRD. RNA sequencing analysis of OC cells grown in TIC conditions revealed a strong enrichment of genes involved in drug metabolism, oxidative phosphorylation and reactive oxygen species (ROS) pathways. Concurrently, a high-throughput drug screen identified drugs that showed efficacy against OC cells grown as TICs compared to adherent cells. Four drugs were chosen that affected drug metabolism and ROS response: disulfiram, bardoxolone methyl, elesclomol and salinomycin. The drugs were tested in vitro for effects on viability, sphere formation and markers of stemness CD133 and ALDH in TICs compared to adherent cells. The compounds promoted ROS accumulation and oxidative stress and disulfiram, elesclomol and salinomycin increased cell death following carboplatin treatment compared to carboplatin alone. Disulfiram and salinomycin were effective in a post-surgery, post-chemotherapy OC relapse model in vivo, demonstrating that enhancing oxidative stress in TICs can prevent OC recurrence. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
46. Clinical Trials in Gynecologic Oncology: Past, Present, and Future.
- Author
-
Annunziata, Christina M. and Kohn, Elise C.
- Published
- 2018
- Full Text
- View/download PDF
47. NF-κB Signaling in Ovarian Cancer.
- Author
-
Harrington, Brittney S. and Annunziata, Christina M.
- Subjects
CELL lines ,CELLULAR signal transduction ,IMMUNOLOGICAL adjuvants ,METASTASIS ,OVARIAN tumors ,STEM cells ,PHENOTYPES ,DNA-binding proteins ,DISEASE progression ,CELL survival - Abstract
The NF-κB signaling pathway is a master and commander in ovarian cancer (OC) that promotes chemoresistance, cancer stem cell maintenance, metastasis and immune evasion. Many signaling pathways are dysregulated in OC and can activate NF-κB signaling through canonical or non-canonical pathways which have both overlapping and distinct roles in tumor progression. The activation of canonical NF-κB signaling has been well established for anti-apoptotic and immunomodulatory functions in response to the tumor microenvironment and the non-canonical pathway in cancer stem cell maintenance and tumor re-initiation. NF-κB activity in OC cells helps to create an immune-evasive environment and to attract infiltrating immune cells with tumor-promoting phenotypes, which in turn, drive constitutive NF-κB activation in OC cells to promote cell survival and metastasis. For these reasons, NF-κB is an attractive target in OC, but current strategies are limited and broad inhibition of this major signaling pathway in normal physiological and immunological functions may produce unwanted side effects. There are some promising pre-clinical outcomes from developing research to target and inhibit NF-κB only in the tumor-reinitiating cancer cell population of OC and concurrently activate canonical NF-κB signaling in immune cells to promote anti-tumor immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
48. A phase I study of the PD-L1 inhibitor, durvalumab, in combination with a PARP inhibitor, olaparib, and a VEGFR1–3 inhibitor, cediranib, in recurrent women's cancers with biomarker analyses.
- Author
-
Zimmer, Alexandra S., Nichols, Erin, Cimino-Mathews, Ashley, Peer, Cody, Cao, Liang, Lee, Min-Jung, Kohn, Elise C., Annunziata, Christina M., Lipkowitz, Stanley, Trepel, Jane B., Sharma, Rajni, Mikkilineni, Lekha, Gatti-Mays, Margaret, Figg, William D., Houston, Nicole D., and Lee, Jung-Min
- Subjects
TRIPLE-negative breast cancer ,CANCER ,OVARIAN cancer ,VASCULAR endothelial growth factors ,IPILIMUMAB ,DNA damage - Abstract
Background: Strategies to improve activity of immune checkpoint inhibitors are needed. We hypothesized enhanced DNA damage by olaparib, a PARP inhibitor, and reduced VEGF signaling by cediranib, a VEGFR1–3 inhibitor, would complement anti-tumor activity of durvalumab, a PD-L1 inhibitor, and the 3-drug combination would be tolerable. Methods: This phase 1 study tested the 3-drug combination in a 3 + 3 dose escalation. Cediranib was taken intermittently (5 days on/2 days off) at 15 or 20 mg (dose levels 1 and 2, respectively) with durvalumab 1500 mg IV every 4 weeks, and olaparib tablets 300 mg twice daily. The primary end point was the recommended phase 2 dose (RP2D). Response rate, pharmacokinetic (PK), and correlative analyses were secondary endpoints. Results: Nine patients (7 ovarian/1 endometrial/1 triple negative breast cancers, median 3 prior therapies [2–6]) were treated. Grade 3/4 adverse events include hypertension (1/9), anemia (1/9) and lymphopenia (3/9). No patients experienced dose limiting toxicities. The RP2D is cediranib, 20 mg (5 days on/2 days off) with full doses of durvalumab and olaparib. Four patients had partial responses (44%) and 3 had stable disease lasting ≥6 months, yielding a 67% clinical benefit rate. No significant effects on olaparib or cediranib PK parameters from the presence of durvalumab, or the co-administration of cediranib or olaparib were identified. Tumoral PD-L1 expression correlated with clinical benefit but cytokines and peripheral immune subsets did not. Conclusions: The RP2D is tolerable and has preliminary activity in recurrent women's cancers. A phase 2 expansion study is now enrolling for recurrent ovarian cancer patients. Trial registration: ClinicalTrials.gov identifier: NCT02484404. Registered June 29, 2015. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
49. Production of a cellular product consisting of monocytes stimulated with Sylatron® (Peginterferon alfa-2b) and Actimmune® (Interferon gamma-1b) for human use.
- Author
-
Green, Daniel S., Nunes, Ana T., Tosh, Kevin W., David-Ocampo, Virginia, Fellowes, Vicki S., Ren, Jiaqiang, Jin, Jianjian, Frodigh, Sue-Ellen, Pham, Chauha, Procter, Jolynn, Tran, Celina, Ekwede, Irene, Khuu, Hanh, Stroncek, David F., Highfill, Steven L., Zoon, Kathryn C., and Annunziata, Christina M.
- Subjects
CD14 antigen ,CRYOPRESERVATION of organs, tissues, etc. ,MONOCYTES - Abstract
Background: Monocytes are myeloid cells that reside in the blood and bone marrow and respond to inflammation. At the site of inflammation, monocytes express cytokines and chemokines. Monocytes have been shown to be cytotoxic to tumor cells in the presence of pro-inflammatory cytokines such as Interferon Alpha, Interferon Gamma, and IL-6. We have previously shown that monocytes stimulated with both interferons (IFNs) results in synergistic killing of ovarian cancer cells. We translated these observations to an ongoing clinical trial using adoptive cell transfer of autologous monocytes stimulated ex vivo with IFNs and infused into the peritoneal cavity of patients with advanced, chemotherapy resistant, ovarian cancer. Here we describe the optimization of the monocyte elutriation protocol and a cryopreservation protocol of the monocytes isolated from peripheral blood.Methods: Counter flow elutriation was performed on healthy donors or women with ovarian cancer. The monocyte-containing, RO-fraction was assessed for total monocyte number, purity, viability, and cytotoxicity with and without a cryopreservation step. All five fractions obtained from the elutriation procedure were also assessed by flow cytometry to measure the percent of immune cell subsets in each fraction.Results: Both iterative monocyte isolation using counter flow elutriation or cryopreservation following counter flow elutriation can yield over 2 billion monocytes for each donor with high purity. We also show that the monocytes are stable, viable, and retain cytotoxic functions when cultured with IFNs.Conclusion: Large scale isolation of monocytes from both healthy donors and patients with advanced, chemotherapy resistant ovarian cancer, can be achieved with high total number of monocytes. These monocytes can be cryopreserved and maintain viability and cytotoxic function. All of the elutriated cell fractions contain ample immune cells which could be used for other cell therapy-based applications. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
50. Editorial: Inhibiting PARP as a Strategic Target in Cancer.
- Author
-
Zorn, Kristin K. and Annunziata, Christina M.
- Subjects
DNA polymerases ,ENZYME inhibitors ,CANCER treatment - Abstract
The author reflects on the Procyclic acidic repetitive protein (PARP) inhibitors for cancer therapy.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.