34 results on '"Rachel A. Sabol"'
Search Results
2. Drug resistance profiling of a new triple negative breast cancer patient-derived xenograft model
- Author
-
Margarite D. Matossian, Hope E. Burks, Steven Elliott, Van T. Hoang, Annie C. Bowles, Rachel A. Sabol, Bahia Wahba, Muralidharan Anbalagan, Brian Rowan, Mohamed E. Abazeed, Bruce A. Bunnell, Krzysztof Moroz, Lucio Miele, Lyndsay V. Rhodes, Steven D. Jones, Elizabeth C. Martin, Bridgette M. Collins-Burow, and Matthew E. Burow
- Subjects
Triple-negative breast cancer ,Patient-derived xenograft ,Mammosphere ,Chemoresistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Triple-negative breast cancer (TNBC) represents an aggressive subtype with limited therapeutic options. Experimental preclinical models that recapitulate their tumors of origin can accelerate target identification, thereby potentially improving therapeutic efficacy. Patient-derived xenografts (PDXs), due to their genomic and transcriptomic fidelity to the tumors from which they are derived, are poised to improve the preclinical testing of drug-target combinations in translational models. Despite the previous development of breast and TNBC PDX models, those derived from patients with demonstrated health-disparities are lacking. Methods We use an aggressive TNBC PDX model propagated in SCID/Beige mice that was established from an African-American woman, TU-BcX-2 K1, and assess its metastatic potential and drug sensitivities under distinct in vitro conditions. Cellular derivatives of the primary tumor or the PDX were grown in 2D culture conditions or grown in mammospheres 3D culture. Flow cytometry and fluorescence staining was used to quantify cancer stem cell-like populations. qRT-PCR was used to describe the mesenchymal gene signature of the tumor. The sensitivity of TU-BcX-2 K1-derived cells to anti-neoplastic oncology drugs was compared in adherent cells and mammospheres. Drug response was evaluated using a live/dead staining kit and crystal violet staining. Results TU-BcX-2 K1 has a low propensity for metastasis, reflects a mesenchymal state, and contains a large burden of cancer stem cells. We show that TU-BcX-2 K1 cells have differential responses to cytotoxic and targeted therapies in 2D compared to 3D culture conditions insofar as several drug classes conferred sensitivity in 2D but not in 3D culture, or cells grown as mammospheres. Conclusions Here we introduce a new TNBC PDX model and demonstrate the differences in evaluating drug sensitivity in adherent cells compared to mammosphere, or suspension, culture.
- Published
- 2019
- Full Text
- View/download PDF
3. Image-Guided Intensity-Modulated Radiation Therapy for IgG4-Related Ophthalmic Disease
- Author
-
Rachel A. Sabol, Virginia Bubenzer, Krzysztof Moroz, Shams Halat, Audrey Dang, Keith Ferdinand, Angela Traylor, Carol Boyd, and Kendra Harris
- Subjects
Ophthalmology ,RE1-994 - Abstract
Background. IgG4-related ophthalmic disease is a rare, newly recognized entity with high failure rates on first-line therapy of systemic corticosteroids and no other proven management options. Case Presentation. Here, we present the clinical course of a patient with IgG4 ophthalmic disease who achieved a favorable response from radiotherapy. Our patient initially presented with a history of recurrent painful flares of orbital inflammation, a pathologic diagnosis follicular lymphoid hyperplasia from a right lacrimal gland biopsy, and MRI imaging noting expansion of the lateral rectus muscle of the right eye. Initial treatment with dacryoadenectomy and multiple courses of corticosteroids failed to keep his symptoms at bay. Further evaluation revealed florid IgG4 staining. In this context, he was evaluated for image-guided intensity-modulated radiotherapy (IG-IMRT) to the orbit to 20 Gy in 10 fractions. His ophthalmic symptoms resolved. Conclusions. This treatment experience suggests radiotherapy may be a favorable option for symptom relief in patients with IgG4-related ophthalmic disease not controlled by corticosteroids.
- Published
- 2020
- Full Text
- View/download PDF
4. AUA/ASTRO 2022 Guidelines: 'Scrutinized'
- Author
-
Mack Roach III, Pamela W. Coleman, and Rachel A. Sabol
- Subjects
Oncology ,Radiology, Nuclear Medicine and imaging - Published
- 2023
- Full Text
- View/download PDF
5. In-depth characterization of a new patient-derived xenograft model for metaplastic breast carcinoma to identify viable biologic targets and patterns of matrix evolution within rare tumor types
- Author
-
Augusto C. Ochoa, E F Cromwell, H Brown, Elizabeth C. Martin, O Sirenko, Krzysztof Moroz, Matthew E. Burow, A M Ham, Katherine Hebert, S Sinha, Erik F. Flemington, Jovanny Zabaleta, Hope E. Burks, Jacob Bursavich, Margarite D. Matossian, A I Riker, Steven Elliott, Bridgette M. Collins-Burow, Khoa Nguyen, Maryl K. Wright, Gabrielle O. Windsor, Frank H. Lau, Connor T. King, Henri Wathieu, Reza Izadpanah, Melody Baddoo, Madlin Alzoubi, Arnold H. Zea, Tiffany R. Chang, Lucio Miele, Rachel A. Sabol, Bruce A. Bunnell, and J J Savoie
- Subjects
Cancer Research ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,Models, Biological ,Article ,Metastasis ,Circulating tumor cell ,Patient-derived xenograft ,In vivo ,Animals ,Humans ,Medicine ,Triple negative breast cancer ,Metaplastic breast carcinoma ,Triple-negative breast cancer ,Decellularization ,business.industry ,Cancer ,Extracellular matrix ,General Medicine ,Metaplastic Breast Carcinoma ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Oncology ,Cancer research ,Heterografts ,Collagen ,business ,Ex vivo - Abstract
Metaplastic breast carcinoma (MBC) is a rare breast cancer subtype with rapid growth, high rates of metastasis, recurrence and drug resistance, and diverse molecular and histological heterogeneity. Patient-derived xenografts (PDXs) provide a translational tool and physiologically relevant system to evaluate tumor biology of rare subtypes. Here, we provide an in-depth comprehensive characterization of a new PDX model for MBC, TU-BcX-4IC. TU-BcX-4IC is a clinically aggressive tumor exhibiting rapid growth in vivo, spontaneous metastases, and elevated levels of cell-free DNA and circulating tumor cell DNA. Relative chemosensitivity of primary cells derived from TU-BcX-4IC was performed using the National Cancer Institute (NCI) oncology drug set, crystal violet staining, and cytotoxic live/dead immunofluorescence stains in adherent and organoid culture conditions. We employed novel spheroid/organoid incubation methods (Pu·MA system) to demonstrate that TU-BcX-4IC is resistant to paclitaxel. An innovative physiologically relevant system using human adipose tissue was used to evaluate presence of cancer stem cell-like populations ex vivo. Tissue decellularization, cryogenic-scanning electron microscopy imaging and rheometry revealed consistent matrix architecture and stiffness were consistent despite serial transplantation. Matrix-associated gene pathways were essentially unchanged with serial passages, as determined by qPCR and RNA sequencing, suggesting utility of decellularized PDXs for in vitro screens. We determined type V collagen to be present throughout all serial passage of TU-BcX-4IC tumor, suggesting it is required for tumor maintenance and is a potential viable target for MBC. In this study we introduce an innovative and translational model system to study cell–matrix interactions in rare cancer types using higher passage PDX tissue.
- Published
- 2021
- Full Text
- View/download PDF
6. Editorial Comment
- Author
-
Rachel A. Sabol and Mack Roach
- Subjects
Urology - Published
- 2022
- Full Text
- View/download PDF
7. A novel screening approach comparing kinase activity of small molecule inhibitors with similar molecular structures and distinct biologic effects in triple-negative breast cancer to identify targetable signaling pathways
- Author
-
Nirav Kapadia, Thomas J. Yan, Van T. Hoang, Steven Elliott, Margarite D. Matossian, David H. Drewry, Douglas B. Rusch, Fang Fang, Deniz A. Ucar, Lucio Miele, Khoa Nguyen, Carrow I. Wells, Hope E. Burks, William J. Zuercher, Jovanny Zabaleta, Matthew E. Burow, Tiffany R. Chang, Rachel A. Sabol, Bruce A. Bunnell, Kenneth P. Nephew, Aaron Buechlein, Gabrielle O. Windsor, and Bridgette M. Collins-Burow
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,Apoptosis ,Triple Negative Breast Neoplasms ,Article ,Metastasis ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,Medicine ,Pharmacology (medical) ,Epithelial–mesenchymal transition ,Phosphorylation ,Kinase activity ,Protein Kinase Inhibitors ,Triple-negative breast cancer ,Cell Proliferation ,Pharmacology ,Molecular Structure ,business.industry ,Kinase ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Female ,Signal transduction ,business ,Signal Transduction - Abstract
Breast cancer affects women globally; the majority of breast cancer-related mortalities are due to metastasis. Acquisition of a mesenchymal phenotype has been implicated in the progression of breast cancer cells to an invasive, metastatic state. Triple-negative breast cancer (TNBC) subtypes have high rates of metastases, recurrence, and have poorer prognoses compared to other breast cancer types, partially due to lack of commonly targeted receptors. Kinases have diverse and pivotal functions in metastasis in TNBC, and discovery of new kinase targets for TNBC is warranted. We previously used a screening approach to identify intermediate-synthesis nonpotent, nonselective small-molecule inhibitors from the Published Kinase Inhibitor Set that reversed the mesenchymal phenotype in TNBC cells. Two of these inhibitors (GSK346294A and GSK448459A) are structurally similar, but have unique kinase activity profiles and exhibited differential biologic effects on TNBC cells, specifically on epithelial-to-mesenchymal transition (EMT). Here, we further interrogate these effects and compare activity of these inhibitors on transwell migration, gene (qRT-PCR) and protein (western blot) expressions, and cancer stem cell-like behavior. We incorporated translational patient-derived xenograft models in these studies, and we focused on the lead inhibitor hit, GSK346294A, to demonstrate the utility of our comparative analysis as a screening modality to identify novel kinase targets and signaling pathways to pursue in TNBC. This study introduces a new method for discovering novel kinase targets that reverse the EMT phenotype; this screening approach can be applied to all cancer types and is not limited to breast cancer.
- Published
- 2020
- Full Text
- View/download PDF
8. Acellular Biologic Nipple–Areolar Complex Graft: In Vivo Murine and Nonhuman Primate Host Response Evaluation
- Author
-
Brooke Grasperge, Vince C. Caronna, Bruce A. Bunnell, Joshua Helm, Nicholas C. Pashos, Abigail E. Chaffin, Scott Sullivan, Rachel A. Sabol, William Heim, David M. Graham, Elizabeth C. Martin, Ben O'Donnell, Brian J. Burkett, Kristin S. Miller, and Annie C. Bowles
- Subjects
Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,0206 medical engineering ,Biomedical Engineering ,Bioengineering ,02 engineering and technology ,Biochemistry ,Biomaterials ,Neovascularization ,Extracellular matrix ,03 medical and health sciences ,Breast cancer ,Tissue engineering ,In vivo ,medicine ,030304 developmental biology ,0303 health sciences ,Decellularization ,business.industry ,medicine.disease ,020601 biomedical engineering ,medicine.symptom ,Breast reconstruction ,business ,Mastectomy - Abstract
There are more than 3 million breast cancer survivors living in the United States of which a significant number have undergone mastectomy followed by breast and nipple-areolar complex (NAC) reconstruction. Current strategies for NAC reconstruction are dependent on nonliving or nonpermanent techniques, including tattooing, nipple prosthetics, or surgical nipple-like structures. Described herein is a tissue engineering approach demonstrating the feasibility of an allogeneic acellular graft for nipple reconstruction. Nonhuman primate (NHP)-derived NAC tissues were decellularized and their extracellular matrix components analyzed by both proteomic and histological analyses. Decellularized NHP nipple tissue showed the removal of intact cells and greatly diminished profiles for intracellular proteins, as compared with intact NHP nipple tissue. We further evaluated the biocompatibility of decellularized grafts and their potential to support host-mediated neovascularization against commercially available acellular dermal grafts by performing in vivo studies in a murine model. A follow-up NHP pilot study evaluated the host-mediated neovascularization and re-epithelialization of onlay engrafted decellularized NAC grafts. The murine model revealed greater neovascularization in the decellularized NAC than in the commercially available control grafts, with no observed biocompatibility issues. The in vivo NHP model confirmed that the decellularized NAC grafts encourage neovascularization as well as re-epithelialization. These results support the concept that a biologically derived acellular nipple graft is a feasible approach for nipple reconstruction, supporting neovascularization in the absence of adverse systemic responses. Impact statement Currently, women in the United States most often undergo a mastectomy, followed by reconstruction, after being diagnosed with breast cancer. These breast cancer survivors are often left with nipple-areolar complex (NAC) reconstructions that are subsatisfactory, nonliving, and/or nonpermanent. Utilizing an acellular biologically derived whole NAC graft would allow these patients a living and permanent tissue engineering solution to nipple reconstruction.
- Published
- 2020
- Full Text
- View/download PDF
9. TrxR1, Gsr, and oxidative stress determine hepatocellular carcinoma malignancy
- Author
-
Emily A. Talago, Luke O. Brandenberger, Michael R. McLoughlin, Rachel A. Sabol, Elias S.J. Arnér, Sonya V. Iverson, Pushya Krishna, David J. Orlicky, Sofi Eriksson, Colin T. Shearn, Colin G. Miller, Brian Bothner, Ian Cavigli, Volkan I. Sayin, Edward E. Schmidt, Jean A. Kundert, Thales Papagiannakopoulos, Joshua Heinemann, and Justin R. Prigge
- Subjects
Male ,Thioredoxin Reductase 1 ,Carcinoma, Hepatocellular ,NF-E2-Related Factor 2 ,DNA damage ,Glutathione reductase ,Malignancy ,medicine.disease_cause ,Antioxidants ,Mice ,medicine ,Animals ,Carcinogen ,Multidisciplinary ,business.industry ,Liver Neoplasms ,medicine.disease ,Glutathione ,Oxidative Stress ,Glutathione Reductase ,PNAS Plus ,Gene Expression Regulation ,Liver ,Hepatocellular carcinoma ,Cancer cell ,Disease Progression ,Hepatocytes ,Metabolome ,Cancer research ,business ,Liver cancer ,Oxidation-Reduction ,Oxidative stress ,DNA Damage - Abstract
Thioredoxin reductase-1 (TrxR1)-, glutathione reductase (Gsr)-, and Nrf2 transcription factor-driven antioxidant systems form an integrated network that combats potentially carcinogenic oxidative damage yet also protects cancer cells from oxidative death. Here we show that although unchallenged wild-type (WT), TrxR1-null, or Gsr-null mouse livers exhibited similarly low DNA damage indices, these were 100-fold higher in unchallenged TrxR1/Gsr–double-null livers. Notwithstanding, spontaneous cancer rates remained surprisingly low in TrxR1/Gsr-null livers. All genotypes, including TrxR1/Gsr-null, were susceptible to N -diethylnitrosamine (DEN)-induced liver cancer, indicating that loss of these antioxidant systems did not prevent cancer cell survival. Interestingly, however, following DEN treatment, TrxR1-null livers developed threefold fewer tumors compared with WT livers. Disruption of TrxR1 in a marked subset of DEN-initiated cancer cells had no effect on their subsequent contributions to tumors, suggesting that TrxR1-disruption does not affect cancer progression under normal care, but does decrease the frequency of DEN-induced cancer initiation. Consistent with this idea, TrxR1-null livers showed altered basal and DEN-exposed metabolomic profiles compared with WT livers. To examine how oxidative stress influenced cancer progression, we compared DEN-induced cancer malignancy under chronically low oxidative stress (TrxR1-null, standard care) vs. elevated oxidative stress (TrxR1/Gsr-null livers, standard care or phenobarbital-exposed TrxR1-null livers). In both cases, elevated oxidative stress was correlated with significantly increased malignancy. Finally, although TrxR1-null and TrxR1/Gsr-null livers showed strong Nrf2 activity in noncancerous hepatocytes, there was no correlation between malignancy and Nrf2 expression within tumors across genotypes. We conclude that TrxR1, Gsr, Nrf2, and oxidative stress are major determinants of liver cancer but in a complex, context-dependent manner.
- Published
- 2019
- Full Text
- View/download PDF
10. Image-Guided Intensity-Modulated Radiation Therapy for IgG4-Related Ophthalmic Disease
- Author
-
Keith C. Ferdinand, Shams Halat, Audrey Dang, Carol Boyd, Angela Traylor, Virginia Bubenzer, Krzysztof Moroz, Rachel A. Sabol, and Kendra Harris
- Subjects
030203 arthritis & rheumatology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,Lateral rectus muscle ,Context (language use) ,Case Report ,General Medicine ,Intensity-modulated radiation therapy ,RE1-994 ,Lymphoid hyperplasia ,Radiation therapy ,03 medical and health sciences ,Ophthalmology ,0302 clinical medicine ,medicine.anatomical_structure ,Biopsy ,030221 ophthalmology & optometry ,medicine ,Radiology ,medicine.symptom ,Ophthalmic disease ,business ,Orbit (anatomy) - Abstract
Background. IgG4-related ophthalmic disease is a rare, newly recognized entity with high failure rates on first-line therapy of systemic corticosteroids and no other proven management options. Case Presentation. Here, we present the clinical course of a patient with IgG4 ophthalmic disease who achieved a favorable response from radiotherapy. Our patient initially presented with a history of recurrent painful flares of orbital inflammation, a pathologic diagnosis follicular lymphoid hyperplasia from a right lacrimal gland biopsy, and MRI imaging noting expansion of the lateral rectus muscle of the right eye. Initial treatment with dacryoadenectomy and multiple courses of corticosteroids failed to keep his symptoms at bay. Further evaluation revealed florid IgG4 staining. In this context, he was evaluated for image-guided intensity-modulated radiotherapy (IG-IMRT) to the orbit to 20 Gy in 10 fractions. His ophthalmic symptoms resolved. Conclusions. This treatment experience suggests radiotherapy may be a favorable option for symptom relief in patients with IgG4-related ophthalmic disease not controlled by corticosteroids.
- Published
- 2020
11. Acellular Biologic Nipple-Areolar Complex Graft
- Author
-
Nicholas C, Pashos, David M, Graham, Brian J, Burkett, Ben, O'Donnell, Rachel A, Sabol, Joshua, Helm, Elizabeth C, Martin, Annie C, Bowles, William M, Heim, Vince C, Caronna, Kristin S, Miller, Brooke, Grasperge, Scott, Sullivan, Abigail E, Chaffin, and Bruce A, Bunnell
- Subjects
Proteomics ,Biological Products ,Mice ,Mammaplasty ,Nipples ,Animals ,Breast Neoplasms ,Female ,Pilot Projects ,Original Articles ,Plastic Surgery Procedures ,Macaca mulatta ,Mastectomy - Abstract
There are more than 3 million breast cancer survivors living in the United States of which a significant number have undergone mastectomy followed by breast and nipple–areolar complex (NAC) reconstruction. Current strategies for NAC reconstruction are dependent on nonliving or nonpermanent techniques, including tattooing, nipple prosthetics, or surgical nipple-like structures. Described herein is a tissue engineering approach demonstrating the feasibility of an allogeneic acellular graft for nipple reconstruction. Nonhuman primate (NHP)-derived NAC tissues were decellularized and their extracellular matrix components analyzed by both proteomic and histological analyses. Decellularized NHP nipple tissue showed the removal of intact cells and greatly diminished profiles for intracellular proteins, as compared with intact NHP nipple tissue. We further evaluated the biocompatibility of decellularized grafts and their potential to support host-mediated neovascularization against commercially available acellular dermal grafts by performing in vivo studies in a murine model. A follow-up NHP pilot study evaluated the host-mediated neovascularization and re-epithelialization of onlay engrafted decellularized NAC grafts. The murine model revealed greater neovascularization in the decellularized NAC than in the commercially available control grafts, with no observed biocompatibility issues. The in vivo NHP model confirmed that the decellularized NAC grafts encourage neovascularization as well as re-epithelialization. These results support the concept that a biologically derived acellular nipple graft is a feasible approach for nipple reconstruction, supporting neovascularization in the absence of adverse systemic responses. IMPACT STATEMENT: Currently, women in the United States most often undergo a mastectomy, followed by reconstruction, after being diagnosed with breast cancer. These breast cancer survivors are often left with nipple–areolar complex (NAC) reconstructions that are subsatisfactory, nonliving, and/or nonpermanent. Utilizing an acellular biologically derived whole NAC graft would allow these patients a living and permanent tissue engineering solution to nipple reconstruction.
- Published
- 2020
12. Evaluation of deacetylase inhibition in metaplastic breast carcinoma using multiple derivations of preclinical models of a new patient-derived tumor
- Author
-
Deniz A. Ucar, Elizabeth C. Martin, Bridgette M. Collins-Burow, Matthew E. Burow, Sukhmani Gill, Hope E. Burks, Adam I. Riker, Henri Wathieu, Lucio Miele, Steven Elliott, Bruce A. Bunnell, Margarite D. Matossian, Rachel A. Sabol, Luis Del Valle, Jovanny Zabaleta, and Tiffany C. Chang
- Subjects
Patient-Specific Modeling ,Lung Neoplasms ,Cancer Treatment ,Gene Expression ,Cell Cycle Proteins ,Cell morphology ,Lung and Intrathoracic Tumors ,Metastasis ,Romidepsin ,Mice ,chemistry.chemical_compound ,Circulating tumor cell ,Cell Movement ,Depsipeptides ,Panobinostat ,Breast Tumors ,Basic Cancer Research ,Medicine and Health Sciences ,education.field_of_study ,Multidisciplinary ,Liver Diseases ,Drug Synergism ,Middle Aged ,Metaplastic Breast Carcinoma ,Neoplastic Cells, Circulating ,Gene Expression Regulation, Neoplastic ,Oncology ,Medicine ,Female ,medicine.drug ,Research Article ,Epithelial-Mesenchymal Transition ,Cell Survival ,Science ,Population ,Breast Neoplasms ,Gastroenterology and Hepatology ,Biology ,Cell Line, Tumor ,Breast Cancer ,Gastrointestinal Tumors ,medicine ,Genetics ,Animals ,Humans ,education ,Protein Kinase Inhibitors ,Colorectal Cancer ,Carcinoma ,Correction ,Cancer ,Cancers and Neoplasms ,Biology and Life Sciences ,Hepatocellular Carcinoma ,medicine.disease ,Histone Deacetylase Inhibitors ,chemistry ,Cancer research - Abstract
Metaplastic breast carcinoma (MBC) is a clinically aggressive and rare subtype of breast cancer, with similar features to basal-like breast cancers. Due rapid growth rates and characteristic heterogeneity, MBC is often unresponsive to standard chemotherapies; and novel targeted therapeutic discovery is urgently needed. Histone deacetylase inhibitors (DACi) suppress tumor growth and metastasis through regulation of the epithelial-to-mesenchymal transition axis in various cancers, including basal-like breast cancers.We utilized a new MBC patient-derived xenograft (PDX) to examine the effect of DACi therapy on MBC. Cell morphology, cell cycle-associated gene expressions, transwell migration, and metastasis were evaluated in patient-derived cells and tumors after treatment with romidepsin and panobinostat. Derivations of our PDX model, including cells, spheres, organoids, explants, andin vivoimplanted tumors were treated. Finally, we tested the effects of combining DACi with approved chemotherapeutics on relative cell biomass.DACi significantly suppressed the total number of lung metastasisin vivousing our PDX model, suggesting a role for DACi in preventing circulating tumor cells from seeding distal tissue sites. These data were supported by our findings that DACi reduced cell migration, populations, and expression of mesenchymal-associated genes. While DACi treatment did affect cell cycle-regulating genesin vitro,tumor growth was not affected compared to controls. Importantly, gene expression results varied depending on the cellular or tumor system used, emphasizing the importance of using multiple derivations of cancer models in preclinical therapeutic discovery research. Furthermore, DACi sensitized and produced a synergistic effect with approved oncology therapeutics on inherently resistant MBC.This study introduced a role for DACi in suppressing the migratory and mesenchymal phenotype of MBC cells through regulation of the epithelial-mesenchymal transition axis and suppression of the CTC population. Preliminary evidence that DACi treatment in combination with MEK1/2 inhibitors exerts a synergistic effect on MBC cells was also demonstrated.
- Published
- 2020
13. Panobinostat suppresses the mesenchymal phenotype in a novel claudin-low triple negative patient-derived breast cancer model
- Author
-
Rachel A. Sabol, Elizabeth C. Martin, Steven Elliott, Hope E. Burks, Bruce A. Bunnell, Annie C. Bowles, Matthew E. Burow, Bridgette M. Collins-Burow, Margarite D. Matossian, and Van T. Hoang
- Subjects
0301 basic medicine ,Cancer Research ,patient-derived xenograft ,medicine.drug_class ,extracellular matrix ,Metastasis ,claudin-low ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Panobinostat ,Medicine ,histone deacetylase inhibitor ,Triple-negative breast cancer ,business.industry ,Histone deacetylase inhibitor ,Claudin-Low ,medicine.disease ,3. Good health ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,triple-negative breast cancer ,Cancer research ,Histone deacetylase ,business ,Ex vivo ,Research Paper - Abstract
Claudin-low triple negative breast cancer (CL-TNBC) is a clinically aggressive molecular TNBC subtype characterized by a propensity to metastasize, recur and acquire chemoresistance. CL-TNBC has a diverse intra- and extracellular composition and microenvironment, and currently there are no clinically approved targeted therapies. Histone deacetylase inhibitors (HDACi) have been investigated as therapeutic agents targeting invasive TNBC phenotypes. However, further studies are required to evaluate HDAC inhibition in CL-TNBC. Here, we utilize a novel CL- TNBC patient-derived xenograft model to study the various and diverse therapeutic potential targets within CL-TNBC tumors. To evaluate effects of the pan-HDACi panobinostat on metastasis and the mesenchymal phenotype of CL-TNBC, we utilize immunohistochemistry staining and qRT-PCR in in vitro, ex vivo and in vivo studies. Further, we evaluate pan-HDAC inhibition on stem-like subpopulations using 3D mammosphere culture techniques and quantification. Finally, we show that pan- HDACi suppresses collagen expression in CL-TNBC. In this study, we provide evidence that pan-HDAC inhibition has effects on various components of the CL-TNBC subtype, and we demonstrate the potential of our novel CL-TNBC PDX model in therapeutic discovery research.
- Published
- 2018
- Full Text
- View/download PDF
14. Melanoma complicating treatment with natalizumab for multiple sclerosis: A report from the Southern Network on Adverse Reactions (SONAR)
- Author
-
Paul Ray, LeAnn B. Norris, Paul R. Yarnold, William J. M. Hrushesky, Charles L. Bennett, Joseph R. Berger, Robert C. Kane, Oliver Sartor, Rachel A. Sabol, Peter Georgantopoulos, Dennis W. Raisch, Laura Rose Bobolts, Akida Lebby, Zaina P. Qureshi, and Virginia Noxon
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Multiple Sclerosis ,Advisory committee ,03 medical and health sciences ,Adverse Event Reporting System ,natalizumab ,0302 clinical medicine ,Natalizumab ,medicine ,Humans ,Immunologic Factors ,Radiology, Nuclear Medicine and imaging ,030212 general & internal medicine ,Melanoma ,Original Research ,Safety surveillance ,business.industry ,Multiple sclerosis ,Network on ,Progressive multifocal leukoencephalopathy ,Clinical Cancer Research ,Neoplasms, Second Primary ,medicine.disease ,Dermatology ,3. Good health ,Surgery ,Oncology ,Female ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
A 43‐year‐old female with multiple sclerosis developed urethral melanoma. The only potential risk factor was treatment with natalizumab, a humanized monoclonal antibody against α4 integrins. To investigate the risk‐exposure relationship, we reviewed this case, all other published cases, and cases of natalizumab‐associated melanoma reported to regulatory agencies. Data sources included the Food and Drug Administration's (FDA) Adverse Event Reporting System (FAERS) (2004–2014), a FDA Advisory Committee Meeting Report, and peer‐reviewed publications. In the United States, the manufacturer maintains an FDA‐mandated Tysabri Safety Surveillance Program (part of the Tysabri Outcomes Unified Commitment to Health (TOUCH)) of natalizumab‐treated patients. We statistically compared reporting completeness for natalizumab‐associated melanoma cases in FAERs for which information was obtained entirely from the TOUCH program versus cases where FAERS information was supplemented by TOUCH program information. FAERS included 137 natalizumab‐associated melanoma reports in patients with multiple sclerosis. Median age at melanoma diagnosis was 45 years (range: 21–74 years). Changes in preexisting nevi occurred in 16%, history of cutaneous nevi occurred in 22%, diagnosis within 2 years of beginning natalizumab occurred in 34%, and 74% had primary surgical treatment. Among seven natalizumab‐treated MS patients who developed biopsy‐confirmed melanoma on treatment and reported in the literature, median age at diagnosis was 41 years (range: 38–48 years); and the melanoma diagnosis occurred following a median of 12 natalizumab doses (range: 1–77 doses). A history of mole or nevi was noted in four patients and a history of prior melanoma was noted in one patient. Completeness scores for reports were significantly lower for FAERS cases reported from the TOUCH program versus FAERS cases supplemented by TOUCH information (median score of 2 vs. 4 items out of 8‐possible items, P
- Published
- 2017
- Full Text
- View/download PDF
15. Drug resistance profiling of a new triple negative breast cancer patient-derived xenograft model
- Author
-
Steven Elliott, Krzysztof Moroz, Van T. Hoang, Lucio Miele, Elizabeth C. Martin, Bridgette M. Collins-Burow, Annie C. Bowles, Steven D. Jones, Muralidharan Anbalagan, Rachel A. Sabol, Mohamed E. Abazeed, Hope E. Burks, Brian G. Rowan, Bruce A. Bunnell, Lyndsay V. Rhodes, Bahia M. Wahba, Matthew E. Burow, and Margarite D. Matossian
- Subjects
0301 basic medicine ,Cancer Research ,Fluorescent Antibody Technique ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,Biology ,lcsh:RC254-282 ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Triple-negative breast cancer ,Patient-derived xenograft ,Cancer stem cell ,Surgical oncology ,Cell Line, Tumor ,Tumor Cells, Cultured ,Genetics ,medicine ,Animals ,Humans ,Mesenchymal stem cell ,Gene signature ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Primary tumor ,3. Good health ,Histone Deacetylase Inhibitors ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Female ,Mammosphere ,Biomarkers ,Chemoresistance ,Research Article - Abstract
Background Triple-negative breast cancer (TNBC) represents an aggressive subtype with limited therapeutic options. Experimental preclinical models that recapitulate their tumors of origin can accelerate target identification, thereby potentially improving therapeutic efficacy. Patient-derived xenografts (PDXs), due to their genomic and transcriptomic fidelity to the tumors from which they are derived, are poised to improve the preclinical testing of drug-target combinations in translational models. Despite the previous development of breast and TNBC PDX models, those derived from patients with demonstrated health-disparities are lacking. Methods We use an aggressive TNBC PDX model propagated in SCID/Beige mice that was established from an African-American woman, TU-BcX-2 K1, and assess its metastatic potential and drug sensitivities under distinct in vitro conditions. Cellular derivatives of the primary tumor or the PDX were grown in 2D culture conditions or grown in mammospheres 3D culture. Flow cytometry and fluorescence staining was used to quantify cancer stem cell-like populations. qRT-PCR was used to describe the mesenchymal gene signature of the tumor. The sensitivity of TU-BcX-2 K1-derived cells to anti-neoplastic oncology drugs was compared in adherent cells and mammospheres. Drug response was evaluated using a live/dead staining kit and crystal violet staining. Results TU-BcX-2 K1 has a low propensity for metastasis, reflects a mesenchymal state, and contains a large burden of cancer stem cells. We show that TU-BcX-2 K1 cells have differential responses to cytotoxic and targeted therapies in 2D compared to 3D culture conditions insofar as several drug classes conferred sensitivity in 2D but not in 3D culture, or cells grown as mammospheres. Conclusions Here we introduce a new TNBC PDX model and demonstrate the differences in evaluating drug sensitivity in adherent cells compared to mammosphere, or suspension, culture. Electronic supplementary material The online version of this article (10.1186/s12885-019-5401-2) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
16. Obesity-Altered Adipose Stem Cells Promote ER+ Breast Cancer Metastasis through Estrogen Independent Pathways
- Author
-
Adam Beighley, Paulina Giacomelli, Matthew E. Burow, Rachel M. Wise, Guangdi Wang, Ben A. O’Donnnell, Bruce A. Bunnell, Jacob D. Lampenfeld, Rachel A. Sabol, Brianne N. Sullivan, Melyssa R. Bratton, Margarite D. Matossian, Bridgette M. Collins-Burow, and Mark Harrison
- Subjects
obesity ,Estrogen receptor ,Adipose tissue ,Metastasis ,lcsh:Chemistry ,Mice ,0302 clinical medicine ,Cell Movement ,Adipocytes ,Aromatase ,Neoplasm Metastasis ,lcsh:QH301-705.5 ,Spectroscopy ,Cells, Cultured ,0303 health sciences ,biology ,Chemistry ,Stem Cells ,digestive, oral, and skin physiology ,General Medicine ,Flow Cytometry ,3. Good health ,Computer Science Applications ,Gene Expression Regulation, Neoplastic ,Receptors, Estrogen ,030220 oncology & carcinogenesis ,Female ,Stem cell ,hormones, hormone substitutes, and hormone antagonists ,estrogen receptor ,Signal Transduction ,medicine.drug_class ,Ovariectomy ,Breast Neoplasms ,Catalysis ,Article ,Cell Line ,Inorganic Chemistry ,03 medical and health sciences ,breast cancer ,medicine ,metastasis ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,Tumor microenvironment ,Organic Chemistry ,Estrogens ,medicine.disease ,adipose stem cells ,lcsh:Biology (General) ,lcsh:QD1-999 ,Estrogen ,Cancer research ,biology.protein ,Estrogen receptor alpha - Abstract
Adipose stem cells (ASCs) play an essential role in tumor microenvironments. These cells are altered by obesity (obASCs) and previous studies have shown that obASCs secrete higher levels of leptin. Increased leptin, which upregulates estrogen receptor alpha (ER&alpha, ) and aromatase, enhances estrogen bioavailability and signaling in estrogen receptor positive (ER+) breast cancer (BC) tumor growth and metastasis. In this study, we evaluate the effect of obASCs on ER+BC outside of the ER&alpha, signaling axis using breast cancer models with constitutively active ER&alpha, resulting from clinically relevant mutations (Y537S and D538G). We found that while obASCs promote tumor growth and proliferation, it occurs mostly through abrogated estrogen signaling when BC has constitutive ER activity. However, obASCs have a similar promotion of metastasis irrespective of ER status, demonstrating that obASC promotion of metastasis may not be completely estrogen dependent. We found that obASCs upregulate two genes in both ER wild type (WT) and ER mutant (MUT) BC: SERPINE1 and ABCB1. This study demonstrates that obASCs promote metastasis in ER WT and MUT xenografts and an ER MUT patient derived xenograft (PDX) model. However, obASCs promote tumor growth only in ER WT xenografts.
- Published
- 2019
17. Correction: Evaluation of deacetylase inhibition in metaplastic breast carcinoma using multiple derivations of preclinical models of a new patient-derived tumor
- Author
-
Matthew E. Burow, Tiffany C. Chang, Bridgette M. Collins-Burow, Hope E. Burks, Rachel A. Sabol, Lucio Miele, Elizabeth A. Martin, Bruce A. Bunnell, Margarite D. Matossian, Sukhmani Gill, Henri Wathieu, Steven G. Elliott, Adam I. Riker, Deniz A. Ucar, Luis Del Valle, and Jovanny Zabaleta
- Subjects
0301 basic medicine ,Multidisciplinary ,business.industry ,Science ,Metaplastic Breast Carcinoma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0226464.].
- Published
- 2021
- Full Text
- View/download PDF
18. Family history and pathogenic/likely pathogenic germline variants in prostate cancer patients
- Author
-
Jodi Lyn Layton, Elisa Ledet, Pedro C. Barata, Brian E. Lewis, A. Oliver Sartor, Rachel A. Sabol, Marcus Marie Moses, and Ellen Jaeger
- Subjects
Cancer Research ,Prostate cancer ,Oncology ,medicine.diagnostic_test ,business.industry ,Medicine ,Family history ,business ,medicine.disease ,Bioinformatics ,Likely pathogenic ,Germline ,Genetic testing - Abstract
143 Background: Recent literature highlights the importance of germline genetic testing in prostate cancer (PCa) patients. Surprisingly, a literature review indicates that family history records are incomplete in published studies. Methods: Prospective and complete family history data were gathered from 496 men in a single institution with a personal history of PCa who underwent germline genetic testing using a panel of at least 79 genes (Invitae testing) from 2016-2020. Comprehensive FH were obtained in all PCa patients in this database and analysis of prevalent FH was assessed at the time of sample collection. Age, race, metastastes at any time, and Gleason score were also ascertained. MUTYH heterozygotes were not considered pathogenic. Results: Pathogenic/likely pathogenic variants (PV/LPVs) were not associated with age at diagnosis, race, or presence of metastasis. Men with Gleason scores 8-10 at time of diagnosis were more likely to have PV/LPV ( P= 0.004). One or more first degree relatives (FDR) with any cancer with was not predictive for germline PV/LPVs for men with PCa ( P= 0.96). Analysis of patients with one or more FDR with breast, prostate, ovarian, or pancreatic cancer revealed that only FDR with breast cancer ( P = 0.028) or ovarian cancer ( P = 0.015) was predictive for PV/LPVs. Though one or more FDR with prostate cancer did not predict a PV/LPV in the overall panel, further analysis indicate that a history of a FDR with PCa was predictive for PV/LPV in a DNA damage repair (DDR) gene ( P= 0.044). Conclusions: In men with a personal history of PCa, germline PV/LPVs were associated with a FDR with breast or ovarian cancer. A FDR with PCa was predictive for PV/LPV in DDR genes. These data emphasize the contribution of FH to germline genetic testing results in a cohort with complete ascertainment of cancer in first degree relatives.
- Published
- 2021
- Full Text
- View/download PDF
19. Abstract 3866: Investigating tumor infiltrating immune cells signature in obese triple negative breast cancer
- Author
-
Keli Xu, Fokhrul Hossain, Matthew Dean, Giulia Monticone, Jovanny Zabaleta, Lucio Miele, Luis Del Valle, Maria Dulfary Sanchez-Pino, Samarpan Majumder, Deniz A. Ucar, Rachel A. Sabol, Bruce A. Bunnell, and Dorota Wyczechowska
- Subjects
Cancer Research ,Immune system ,Oncology ,business.industry ,Cancer research ,Medicine ,business ,Signature (topology) ,Triple-negative breast cancer - Abstract
Triple negative breast cancer (TNBC) is a heterogeneous group of clinically aggressive breast cancers and TNBC patients are pathologically negative for estrogen receptor (ER−), progesterone receptor (PR−), and human epidermal growth factor receptor 2 (HER2−) amplification, which impedes the use of targeted therapies used in other breast cancer subtypes. Obesity is a chronic state of inflammation and is associated with increased secretion of pro-inflammatory cytokines, increased infiltration of immune cells and the development of a microenvironment that supports tumor growth. Several epidemiological studies support the association of obesity with TNBC; however, the underlining molecular mechanisms remain unknown. We developed an immunocompetent obese FVB (female) mouse model by feeding ‘Western diet' or control diet for four months and then injecting with syngeneic C0321 (mouse TNBC) cells to investigate the immune signatures and to study the role of obesity related factors in TNBC progression. We found significant body weight increases in ‘Western diet' fed mice group as expected. The percentage of CD8+ and CD4+ T cells, and macrophages in spleen, liver and peripheral blood in obese mice did not change significantly compared to lean mice. However, the percentages of immunosuppressive Myeloid Derived Suppressor cells (MDSCs), particularly Monocytic-MDSCs were elevated in liver and visceral fat tissue in obese mice. Importantly, tumors in ‘Western Diet' fed mice achieved significantly larger volumes. We found no noticeable difference in the percentage of CD8+ and CD4+ T cells, MDSC or macrophages in the spleen of tumor bearing obese mice. Interestingly, there was a decreasing trend in tumor infiltrating CD8+ and CD4+ T cells in tumor bearing obese mice. Further, tumor infiltrating myeloid cells, MDSCs and macrophages were significantly increased in obese mice, suggesting the formation of an immunosuppressive environment. More importantly, tumor infiltrating MDSCs from obese mice were more immunosuppressive than those from lean mice. In summary, our results reveal alterations in the number and function of tumor infiltrating immune cells in obese mice. RNA-Sequencing of lean vs obese tumors revealed significant changes in pathways related to Hypertrophic and Dilated Cardiomyopathy, Adrenergic signaling in Cardiomyocytes, as well as signaling pathways of Insulin, cGMP-PKG, Glucagon, Calcium and Adipocytokine and others. Our data warrants further investigation on the immunosuppressive tumor microenvironment in obese TNBC patients. Citation Format: Fokhrul Hossain, Deniz A Ucar, Maria Sanchez-Pino, Matthew Dean, Samarpan Majumder, Dorota Wyczechowska, Giulia Monticone, Rachel Sabol, Keli Xu, Luis D Valle, Jovanny Zabaleta, Bruce Bunnell, Lucio Miele. Investigating tumor infiltrating immune cells signature in obese triple negative breast cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3866.
- Published
- 2020
- Full Text
- View/download PDF
20. Abstract C110: Applications of patient-derived triple-negative breast cancer xenografts that represent understudied patients in Louisiana in targeted therapeutic research
- Author
-
Bridgette M. Collins-Burow, Hope E. Burks, Elizabeth C. Martin, Henri Wathieu, Bruce A. Bunnell, Alex Alfortish, Fokhrul Hossain, Van T. Hoang, Steven Elliott, Jovanny Zabaleta, Adam Riker, Margarite D. Matossian, Rachel A. Sabol, Krzysztof Moroz, Nicholas C. Pashos, Matthew E. Burow, Steven D. Jones, Lucio Miele, Maryl K. Wright, Tiffany R. Chang, Deniz A. Ucar, and Arnold H. Zea
- Subjects
Oncology ,High rate ,medicine.medical_specialty ,Epidemiology ,business.industry ,Cancer ,medicine.disease ,Tumor Cell Biology ,Metastasis ,Breast cancer ,Stroma ,Internal medicine ,Medicine ,In patient ,business ,Triple-negative breast cancer - Abstract
Triple-negative breast cancers (TNBCs) constitute approximately 12% of all breast cancer cases and are approximately twice as prevalent in African-American populations. Louisiana has a high proportion of African-American residents (32.3% in 2017), and New Orleans has among the highest incidences of TNBC in the country. Louisiana patients also have a high incidence of co-morbidities that affect breast cancer biology and outcomes, including type 2 diabetes and obesity. TNBCs have an aggressive clinical presentation due to high rates of metastasis, recurrence and chemoresistance. There are currently no clinically approved targeted therapies for TNBC; cytotoxic chemotherapy is the first-line treatment for TNBC, and recurrent, chemoresistant cancers are usually fatal. TNBCs are molecularly heterogeneous, consisting of at least four molecular subgroups, and immunologically heterogeneous. Both molecular and immunologic properties are associated with clinical outcomes and are seriously understudied in patients under-represented in biomedical research. Patient-derived xenografts (PDXs), as well as patient-derived organoids (PDO), are currently the best model for translational oncology therapeutic research because they accurately recapitulate the complex architecture and heterogenous genetic and molecular composition of solid cancers. To date, the majority of TNBC research has been based on Caucasian patients, although incidence rates of TNBC are higher in African-American cohorts. Our collaborative team aims to overcome this obstacle by establishing and characterizing TNBC PDX models that represent this understudied cohort. We currently have ten TNBC PDX models representing different patient ethnicities, responsiveness to chemotherapies, as well as different TNBC molecular subtypes and metastatic behavior. We dissect and evaluate the various individual components (tumor cell biology, stroma, immune, extracellular matrix) of TNBC tumors. We utilize these models in vivo, ex vivo and in vitro to examine how unique kinases and targeted inhibitors affect the distinct tumor characteristics. In addition to in vivo treatment studies, we generated cell lines and PDOs and we utilize novel techniques such as tissue decellularization to examine extracellular matrix components. We also analyze mechanistically relevant transcript (qRT-PCR) and protein (Western blot, immunohistochemistry) expression patterns that are unique to each PDX model to evaluate the effects of targeted therapies. We work with surrounding laboratories in the greater New Orleans area (Tulane, LSU, Xavier) that are also focused on therapeutic discovery of TNBC in a collaborative effort to provide translational models for their projects. Our aim is to leverage novel PDX models from understudied patients with a range of clinical and molecular presentations to guide the selection of therapeutically targetable pathways and therapeutic agents in specific molecular subtypes of TNBC. Citation Format: Margarite D. Matossian, Steven Elliott, Hope E. Burks, Maryl Wright, Rachel A. Sabol, Van T. Hoang, Deniz A. Ucar, Alex Alfortish, Jovanny Zabaleta, Fokhrul Hossain, Tiffany Chang, Henri Wathieu, Nicholas Pashos, Bruce Bunnell, Krzysztof Moroz, Arnold Zea, Adam Riker, Steven D. Jones, Elizabeth C. Martin, Lucio Miele, Bridgette M. Collins-Burow, Matthew E. Burow. Applications of patient-derived triple-negative breast cancer xenografts that represent understudied patients in Louisiana in targeted therapeutic research [abstract]. In: Proceedings of the Eleventh AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2018 Nov 2-5; New Orleans, LA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2020;29(6 Suppl):Abstract nr C110.
- Published
- 2020
- Full Text
- View/download PDF
21. Obesity-Altered Adipose Stem Cells Promote Radiation Resistance of Estrogen Receptor Positive Breast Cancer through Paracrine Signaling
- Author
-
Benjamin T. Freeman, Alifiani B. Hartono, Maxwell B. Sandler, Rachel A. Sabol, Rachel M. Wise, Vidal A. Villela, Lucio Miele, Alexandra Denys, Fokhrul Hossain, Bruce A. Bunnell, and Mark Harrison
- Subjects
Leptin ,obesity ,Estrogen receptor ,Adipose tissue ,Apoptosis ,lcsh:Chemistry ,Mice ,Tumor Microenvironment ,Medicine ,RNA, Small Interfering ,skin and connective tissue diseases ,lcsh:QH301-705.5 ,Spectroscopy ,Radiation ,Receptors, Notch ,General Medicine ,Computer Science Applications ,Adipose Tissue ,Receptors, Estrogen ,Gene Knockdown Techniques ,S Phase Cell Cycle Checkpoints ,MCF-7 Cells ,Female ,Stem cell ,Signal Transduction ,Stromal cell ,Cell Survival ,Notch signaling pathway ,Breast Neoplasms ,Article ,Catalysis ,Inorganic Chemistry ,breast cancer ,Breast cancer ,Paracrine Communication ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,radiotherapy ,Cell Proliferation ,radiation resistance ,Tumor microenvironment ,Interleukin-6 ,business.industry ,Organic Chemistry ,Mesenchymal Stem Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,Coculture Techniques ,Oxidative Stress ,adipose stem cells ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer research ,business ,DNA Damage - Abstract
Obesity is associated with poorer responses to chemo- and radiation therapy for breast cancer, which leads to higher mortality rates for obese women who develop breast cancer. Adipose stem cells (ASCs) are an integral stromal component of the tumor microenvironment (TME). In this study, the effects of obesity-altered ASCs (obASCs) on estrogen receptor positive breast cancer cell&rsquo, s (ER+BCCs) response to radiotherapy (RT) were evaluated. We determined that BCCs had a decreased apoptotic index and increased surviving fraction following RT when co-cultured with obASCs compared to lnASCs or non-co-cultured cells. Further, obASCs reduced oxidative stress and induced IL-6 expression in co-cultured BCCs after radiation. obASCs produce increased levels of leptin relative to ASCs from normal-weight individuals (lnASCs). obASCs upregulate the expression of IL-6 compared to non-co-cultured BCCs, but BCCs co-cultured with leptin knockdown obASCs did not upregulate IL-6. The impact of shLeptin obASCs on radiation resistance of ER+BCCs demonstrate a decreased radioprotective ability compared to shControl obASCs. Key NOTCH signaling players were enhanced in ER+BBCs following co-culture with shCtrl obASCs but not shLep obASCs. This work demonstrates that obesity-altered ASCs, via enhanced secretion of leptin, promote IL-6 and NOTCH signaling pathways in ER+BCCs leading to radiation resistance.
- Published
- 2020
- Full Text
- View/download PDF
22. Abstract P6-03-17: Effect of histone deacetylase inhibitors on patient-derived neoadjuvant chemotherapy resistant triple negative breast cancer xenografts that represent understudied patients
- Author
-
Rachel A. Sabol, Bruce A. Bunnell, Steven Elliott, Krzysztof Moroz, Alex Alfortish, Elizabeth C. Martin, Steven D. Jones, Maryl K. Wright, Gabrielle O. Windsor, Arnold H. Zea, Bridgette M. Collins-Burow, Lucio Miele, Jovanny Zabaleta, Van T. Hoang, Adam Riker, Margarite D. Matossian, Matthew E. Burow, Deniz A. Ucar, Madlin Alzoubi, Thomas J. Yan, Hope E. Burks, Tiffany R. Chang, Fokhrul Hossain, and Henri Wathieu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,Cancer ,Drug resistance ,medicine.disease ,medicine.disease_cause ,Metastasis ,CDH1 ,Breast cancer ,Internal medicine ,medicine ,biology.protein ,Histone deacetylase ,Carcinogenesis ,business ,Triple-negative breast cancer - Abstract
Triple negative breast cancers (TNBCs) are a clinically and biologically aggressive breast cancer (BC) subtype; TNBC tumors have higher rates of metastasis, relapse and acquired/inherent drug resistance. Incidence and mortality rates of TNBC are stratified based on patient ethnicity - patients with African ancestry have higher mortality rates and diagnoses of invasive cancers compared to patients representing other ethnicities. Louisiana has a high proportion of African-American residents (32.7% in 2018), and New Orleans has among the highest incidences of TNBC in the country. Many of our patients present with TNBC tumors that are partially or completely resistant to neoadjuvant chemotherapies. There are currently no clinically approved targeted therapies for TNBC. Current therapeutic discovery focused TNBC research does not aptly address the knowledge gap regarding ethnic disparity in TNBC incidence/mortality rates and TNBC biology. To date, most TNBC-related research and knowledge has been acquired from Caucasian patients, although patients with African and Hispanic ancestries represent the majority of TNBC cases. Patient-derived xenografts (PDXs) are extensively used in BC research, as they mimic complex microanatomy, oncoarchitecture, and cell-cell/cell-stroma interactions of tumors. Here, we demonstrated the unique composition of PDX tumors is not dramatically affected by serial transplantation in mice, based on molecular phenotypes (examined using qRT-PCR and RNA sequencing) and the oncoarchitecture of the extracellular matrix (based on cryogenic scanning electron microscopy). Using these models in basic research facilitates translation of laboratory findings to the clinical setting, and dramatically enhanced drug discovery research. We have established over twelve TNBC PDX models, 90% of which represent patients of African ancestry, and most of which are resistant to neoadjuvant regimens. We focus on dissecting and evaluating kinase inhibitor/targeted drug response to various individual components (tumor cell biology, stroma, immune, extracellular matrix) of chemotherapy resistant TNBC tumors. Histone deacetylase inhibitors (DACi) are a promising therapeutic agent in TNBC systems; they have been shown to suppress tumorigenesis and metastasis in TNBC through suppression of the mesenchymal phenotype in cell line-based studies. In this study we utilized various TNBC PDX models (TU-BcX-2K1, -2O0, 4IC, -4M4, -4QAN, -4QX) to assess these findings in more translational systems. Interestingly, we showed that DACi effect on tumorigenesis and metastasis varied depending on specific TNBC PDXs utilized. These data implicate specific genes/signaling pathways exist in individual patient tumors that can predict tumor responsiveness to DACi. Preliminary data using the NCI oncology drug set implicated the MEK1/2 pathway contributed to sensitization of TNBC cells. Furthermore, we found a disconnect in gene expressions that were previously shown to be affected by DACi therapy (CDH1, VIM, ZEB1, ZEB2) in various derivations of PDX models (cells, PDX-Os, ex vivo, in vivo). These findings demonstrate that testing various derivations of PDX models is crucial to parsing out specific mechanisms of targeted therapies. Our methods presented here to assess targeted drug response and drug resistance using PDX models can be applied to any area of cancer research and is not limited to breast cancer. Citation Format: Margarite Matossian, Steven Elliott, Maryl Wright, Tiffany Chang, Madlin Alzoubi, Henri Wathieu, Rachel Sabol, Alex Alfortish, Hope Burks, Van Hoang, Deniz Ucar, Gabrielle Windsor, Thomas Yan, Jovanny Zabaleta, Fokhrul Hossain, Bruce Bunnell, Krzysztof Moroz, Arnold Zea, Adam Riker, Steven Jones, Elizabeth Martin, Lucio Miele, Bridgette Collins-Burow, Matthew Burow. Effect of histone deacetylase inhibitors on patient-derived neoadjuvant chemotherapy resistant triple negative breast cancer xenografts that represent understudied patients [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P6-03-17.
- Published
- 2020
- Full Text
- View/download PDF
23. Therapeutic Potential of Adipose Stem Cells
- Author
-
Rachel A, Sabol, Annie C, Bowles, Alexandra, Côté, Rachel, Wise, Nicholas, Pashos, and Bruce A, Bunnell
- Subjects
Adipose Tissue ,Stem Cells ,Adipocytes ,Humans ,Cell Differentiation ,Mesenchymal Stem Cells - Abstract
Adipose stem cells (ASCs) have gained attention in the fields of stem cells regenerative medicine due to their multifaceted therapeutic capabilities. Promising preclinical evidence of ASCs has supported the substantial interest in the use of these cells as therapy for human disease. ASCs are an adult stem cell resident in adipose tissue with the potential to differentiation along mesenchymal lineages. They also are known to be recruited to sites of inflammation where they exhibit strong immunomodulatory capabilities to promote wound healing and regeneration. ASCs can be isolated from adipose tissue at a relatively high yield compared to their mesenchymal cell counterparts: bone marrow-derived mesenchymal stem cells (BM-MSCs). Like BM-MSCs, ASCs are easily culture expanded and have a reduced immunogenicity or are perhaps immune privileged, making them attractive options for cellular therapy. Additionally, the heterogeneous cellular product obtained after digestion of adipose tissue, called the stromal vascular fraction (SVF), contains ASCs and several populations of stromal and immune cells. Both the SVF and culture expanded ASCs have the potential to be therapeutic in various diseases. This review will focus on the preclinical and clinical evidence of SVF and ASCs, which make them potential candidates for therapy in regenerative medicine and inflammatory disease processes.
- Published
- 2018
24. Therapeutic Potential of Adipose Stem Cells
- Author
-
Nicholas C. Pashos, Rachel M. Wise, Annie C. Bowles, Bruce A. Bunnell, Alexandra Côté, and Rachel A. Sabol
- Subjects
0301 basic medicine ,business.industry ,animal diseases ,medicine.medical_treatment ,Mesenchymal stem cell ,Adipose tissue ,chemical and pharmacologic phenomena ,Stem-cell therapy ,Stromal vascular fraction ,Regenerative medicine ,Cell therapy ,03 medical and health sciences ,030104 developmental biology ,medicine ,Cancer research ,Stem cell ,business ,Adult stem cell - Abstract
Adipose stem cells (ASCs) have gained attention in the fields of stem cells regenerative medicine due to their multifaceted therapeutic capabilities. Promising preclinical evidence of ASCs has supported the substantial interest in the use of these cells as therapy for human disease. ASCs are an adult stem cell resident in adipose tissue with the potential to differentiation along mesenchymal lineages. They also are known to be recruited to sites of inflammation where they exhibit strong immunomodulatory capabilities to promote wound healing and regeneration. ASCs can be isolated from adipose tissue at a relatively high yield compared to their mesenchymal cell counterparts: bone marrow-derived mesenchymal stem cells (BM-MSCs). Like BM-MSCs, ASCs are easily culture expanded and have a reduced immunogenicity or are perhaps immune privileged, making them attractive options for cellular therapy. Additionally, the heterogeneous cellular product obtained after digestion of adipose tissue, called the stromal vascular fraction (SVF), contains ASCs and several populations of stromal and immune cells. Both the SVF and culture expanded ASCs have the potential to be therapeutic in various diseases. This review will focus on the preclinical and clinical evidence of SVF and ASCs, which make them potential candidates for therapy in regenerative medicine and inflammatory disease processes.
- Published
- 2018
- Full Text
- View/download PDF
25. Discussion
- Author
-
Rachel A. Sabol and Bruce A. Bunnell
- Subjects
Gene knockdown ,Breast cancer ,business.industry ,Cancer research ,CRISPR ,Medicine ,Adipose tissue ,Surgery ,Stem cell ,business ,medicine.disease - Published
- 2019
- Full Text
- View/download PDF
26. A novel patient-derived xenograft model for claudin-low triple-negative breast cancer
- Author
-
Steven Elliott, Krzysztof Moroz, Nicholas C. Pashos, Kristin S. Miller, Benjamen O’Donnell, Elizabeth C. Martin, Van T. Hoang, Steven D. Jones, Lucio Miele, Bridgette M. Collins-Burow, Bahia M. Wahba, Matthew E. Burow, Lyndsay V. Rhodes, Margarite D. Matossian, Fokhrul Hossain, Hope E. Burks, Augusto C. Ochoa, Arnold H. Zea, Amir A. Al-Khami, Adam I. Riker, Rachel A. Sabol, Annie C. Bowles, and Bruce A. Bunnell
- Subjects
0301 basic medicine ,Cancer Research ,Triple Negative Breast Neoplasms ,Article ,Extracellular matrix ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Breast cancer ,Western blot ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Triple-negative breast cancer ,Cell Proliferation ,medicine.diagnostic_test ,business.industry ,Mesenchymal stem cell ,medicine.disease ,Claudin-Low ,Phenotype ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Claudins ,Cancer research ,Immunohistochemistry ,Female ,Neoplasm Recurrence, Local ,business - Abstract
BACKGROUND: Triple negative breast cancer (TNBC) subtypes are clinically aggressive and are treated with targeted therapeutics commonly used in other BC subtypes. The claudin-low (CL) molecular subtype of TNBC has high rates of metastases, chemoresistance and recurrence. There exists an urgent need to identify novel therapeutic targets in TNBC; however, existing models utilized in target discovery research have limitations. Patient-derived xenograft (PDX) models have emerged as superior models for target discovery experiments because they recapitulate features of patient tumors that are limited by xenograft methods. METHODS: We utilize immunohistochemistry, qRT-PCR and Western Blot to visualize tumor architecture, cellular composition, genomic and protein expressions of a new TNBC PDX model (TU-BcX-2O0). We utilize tissue decellularization techniques to examine extracellular matrix composition of TU-BcX-2O0. RESULTS: Our laboratory successfully established a TNBC PDX tumor, TU-BCX-2O0, which represents a CL-TNBC subtype and maintains this phenotype throughout subsequent passaging. We dissected TU-BCx-2O0 to examine aspects of this complex tumor that can be targeted by developing therapeutics, including specific cell populations within the tumor, the extracellular matrix, and cancer stem-like cell populations. CONCLUSIONS: Here we characterize a claudin-low TNBC patient-derived xenograft model that can be utilized for therapeutic research studies.
- Published
- 2017
27. 2070 High-intensity focused ultrasound (HIFU) can be used synergistically with tamoxifen to overcome resistance in preclinical and patient derived xenograft models
- Author
-
Rachel A. Sabol, Bruce A. Bunnell, Hakm Y. Murad, Damir B. Khismatullin, and Matthew E. Burow
- Subjects
business.industry ,medicine.medical_treatment ,Cancer research ,Medicine ,General Medicine ,Basic/Translational Science/Team Science ,business ,Tamoxifen ,High-intensity focused ultrasound ,Tumor xenograft ,medicine.drug - Abstract
OBJECTIVES/SPECIFIC AIMS: The goal of this study is to evaluate a potential strategy to overcome tamoxifen (tam) resistance by using tam in combination with high-intensity focused ultrasound (HIFU). Tam is the most commonly used anti-cancer therapeutic agent in estrogen receptor positive (ER+) breast cancer (BC) which accounts for ~70% of BC cases. Tam treatment decreases a woman’s risk of recurrence by 50%; however, BC that is initially responsive to tam often develops resistance. METHODS/STUDY POPULATION: HIFU deposits acoustic energy locally to a cancerous region, which induces strong vibrations of molecules inside and outside of the cells. The resulting absorption causes rapid heating and mechanical disruption. This clinically relevant, noninvasive, and nonionizing physical force modality, has been shown to synergistically enhance chemical anticancer therapies. RESULTS/ANTICIPATED RESULTS: In this study we found that treatment of MCF7 cells with HIFU and tam has additive antiproliferative effects and mediates increased cell death. Additionally, we used tam resistant (TR) MCF7 cells that had been exposed to low-dose tam over time until they acquired resistance. When MCF7 TR are treated with tam there is no change in viability; however, treatment with HIFU in combination with tam decreased viability of both MCF7 and MCF7 TR to 19% and the viability of the cell lines was indistinguishable. We next evaluated the effect on MCF7 Y537S mutant ESR1, where ER is mutated to be constitutively active. Treatment of MCF7 Y537S had no significant decrease in viability of combination therapy compared with viability after HIFU alone. Analysis of ERalpha gene expression showed that HIFU treatment increased ERalpha expression in MCF7 TR cells, thus resensitizing these cells to tam and allowing these therapies to work synergistically. Our team developed a system to evaluate the potential of this combination of therapies in a patient-derived xenografts (PDX) model. PDX have emerged as a novel translational tool for cancer research with the potential to more accurately recapitulate the molecular and behavioral aspects of cancer. The WHIM20 PDX is a tamoxifen resistant tumor where the patient developed the Y537S mutation in ESR1. Ex vivo experiments on PDX tumor pieces demonstrated that combination therapy of HIFU and tam work synergistically to increase cell death of these tumors. Further, cryogenic-scanning electron microscopy was utilized to directly demonstrate the physical disruption to both cellular and tumor microenvironment post exposure to combination treatment. DISCUSSION/SIGNIFICANCE OF IMPACT: These studies present a novel translational strategy to overcome tamoxifen resistance in ER+BC.
- Published
- 2018
28. Abstract A01: Application of patient-derived models from understudied patient populations to discover therapeutically targetable pathways in triple-negative breast cancer systems
- Author
-
Van T. Hoang, Krzysztof Moroz, Elizabeth C. Martin, Nicholas C. Pashos, Alex Alfortish, Benjamen O’Donnell, Hope E. Burks, Steven D. Jones, Kristin S. Miller, Lucio Miele, Adam Riker, Ayse D. Ucar Bilyeu, Bruce A. Bunnell, Annie C. Bowles, Fokhrul Hossain, Margarite D. Matossian, Rachel A. Sabol, Bridgette M. Collins-Burow, Tiffany R. Chang, Arnold H. Zea, and Matthew E. Burow
- Subjects
Cancer Research ,Kinase ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,Tumor Cell Biology ,Metastasis ,Targeted therapy ,Breast cancer ,Oncology ,medicine ,Cancer research ,Epigenetics ,business ,Molecular Biology ,Triple-negative breast cancer - Abstract
Triple-negative breast cancers (TNBCs) constitute approximately 12% of all breast cancers, and are approximately twice more prevalent in African-American populations. Louisiana has a high proportion of African-American residents (32.5% in 2015), and among the highest incidences of TNBC in the country. Louisiana patients also have a high incidence of comorbidities that affect breast cancer biology and outcomes, including type 2 diabetes and obesity. TNBCs have an aggressive clinical presentation due to high rates of metastasis, recurrence, and chemoresistance, and targeted therapy remains elusive. Discovery of novel therapeutic targets, including a subset of previously uncharacterized kinases in TNBC could provide important insights into future targeted therapies. However, current models utilized in target discovery research are limited by the inability to accurately recapitulate the complex architecture and heterogenous genetic and molecular composition of breast cancer. Furthermore, immortalized cell lines have been selected in a two-dimensional environment and may have lost important epigenetic features of original tumors they derived from. Recently, our laboratory has successfully established four TNBC patient-derived xenograft (PDX) models representing different patient ethnicities, responsiveness to chemotherapies, and different TNBC molecular subtypes and metastatic behavior. Our primary objective was to dissect and evaluate the various individual components (tumor cell biology, stroma, immune, extracellular matrix) that drive complex interactions within TNBC tumors. We utilize these models in vivo, ex vivo, and in vitro to examine how unique kinases and small molecule inhibitors affect the distinct tumor characteristics. In addition to in vivo treatment studies we generated cell lines and mammospheres (TU-BcX-2K1, TU-BcX-2O0, TU-BcX-49S, TU-BcX-4IC), and we utilize novel techniques such as tissue decellularization to examine extracellular matrix components. We also analyze mechanistically relevant transcript (qRT-PCR) and protein (Western blot, immunohistochemistry) expression patterns that are unique to each PDX model to evaluate the effect of small-molecule inhibitors on these transcripts and proteins. Our aim is to leverage novel patient-derived models from understudied patients with a range of clinical presentations to guide the selection of therapeutically targetable pathways and molecules in specific molecular subtypes of TNBC. Citation Format: Margarite Matossian, Hope Burks, Tiffany Chang, Annie Bowles, Rachel Sabol, Van Hoang, Bruce Bunnell, Alex Alfortish, Krzysztof Moroz, Arnold Zea, Adam Riker, Steven Jones, Ayse D. Ucar Bilyeu, Fokhrul Hossain, Kristin Miller, Elizabeth Martin, Benjamen O’Donnell, Nicholas Pashos, Lucio Miele, Matthew Burow, Bridgette Collins-Burow. Application of patient-derived models from understudied patient populations to discover therapeutically targetable pathways in triple-negative breast cancer systems [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A01.
- Published
- 2018
- Full Text
- View/download PDF
29. 2057 L1 expression analysis in adipose-derived stem cells
- Author
-
Prescott L. Deininger, Maria E. Morales, Tiffany Kaul, Rachel A. Sabol, and Bruce A. Bunnell
- Subjects
Expression analysis ,Adipose tissue ,hemic and immune systems ,General Medicine ,Basic/Translational Science/Team Science ,Biology ,Cell biology - Abstract
OBJECTIVES/SPECIFIC AIMS: Long interspersed element-1s (L1s) are autonomous, mobile elements that are able to copy and insert themselves throughout the genome with their own reverse transcriptase and endonuclease. These elements make up 17% of the human genome with over 500,000 copies, though the vast majority of L1s are defective with only a few dozen potentially responsible for L1 activity. Full-length L1s have the potential to contribute to mutagenesis through random insertion and increased genetic instability. Here we set out to study L1 expression at the specific loci level in bone marrow-derived stem cells (bmSCs) and adipose-derived stem cells (ASCs) and compare the levels of expression from ASCs from donor patients who are young and lean, obese, and old. Our hypothesis is that L1-related damage may contribute to mutation and inflammation that alters the function of these stem cells throughout the life of an individual. METHODS/STUDY POPULATION: ASCs and bmSCs were isolated from patient donors. The following samples were collected: ASCs from 3 young (under the age of 59) and lean (BMI30, and bmSCs from 4 young and lean patients. Cytoplasmic RNA from the cell populations was isolated and sequenced by RNA-Seq from the cell populations. Using our recently developed bioinformatics pipeline, we set out to quantify L1 expression and identify the few culprit L1s at specific loci that are actively transcribing to RNA in the ASC and bmSC samples. RESULTS/ANTICIPATED RESULTS: Here we provide proof of concept with the application of this novel method in characterizing full-length expressed L1s at the specific loci level in ASCs and bmSCs. We identified L1 loci that are commonly expressed in these cell types and observed an increase in L1 expression in the obese and old ASC cells compared with the young, lean ASCs and bmSCs. DISCUSSION/SIGNIFICANCE OF IMPACT: ASCs hold the promise of broad application in the biomedical field including regenerative treatment. There are reports that ASCs cultivated from older and obese donors are less effective in regenerative treatments. By demonstrating an increased expression of the mutagenic L1 element in ASCs from obese and old donors, this study provides further evidence suggesting the preferable use of ASCs from young and lean donors for regenerative therapies. These studies will also help us to understand the potential contribution of L1 expression to loss of stem cell function during the aging process.
- Published
- 2018
- Full Text
- View/download PDF
30. Obesity-altered adipose stem cells promote breast cancer progression and metastasis
- Author
-
Amy L. Strong, Rachel A. Sabol, Bruce A. Bunnell, and Matthew E. Burow
- Subjects
Cancer Research ,Transplantation ,business.industry ,Immunology ,Adipose tissue ,Cell Biology ,medicine.disease ,Obesity ,Metastasis ,Breast cancer ,Oncology ,medicine ,Cancer research ,Immunology and Allergy ,Stem cell ,business ,Genetics (clinical) - Published
- 2018
- Full Text
- View/download PDF
31. Abstract P5-05-05: Patient-derived triple negative breast cancer xenografts as a translational model to screen for novel kinase pathways
- Author
-
Sean Bong Lee, Van T. Hoang, David H. Drewry, Margarite D. Matossian, Rachel A. Sabol, Carrow I. Wells, Arnold H. Zea, Hope E. Burks, Krzysztof Moroz, Matthew E. Burow, Steven Elliott, Steven D. Jones, William J. Zuercher, Bruce A. Bunnell, Alex Alfortish, Alifiani B. Hartono, Annie C. Bowles, and Bridgette M. Collins-Burow
- Subjects
Cancer Research ,Oncology ,Kinase ,business.industry ,Cancer research ,Medicine ,business ,Triple-negative breast cancer - Abstract
Overall, triple negative breast cancers (TNBCs) constitute 12% of all breast cancers, and is approximately twice more prevalent in African-American populations. Louisiana has a high proportion of African-American residents (32.5% in 2015), and thus hosts a higher population of TNBC patients. TNBCs have an aggressive phenotype that is elusive to the targeted therapeutics used to treat other breast cancer subtypes. Certain kinase families have been extensively studied as regulators of epithelial-mesenchymal transition (EMT), a process involved in the initiation of cancer metastasis. Discovery of novel kinase targets within the subset of uncharacterized kinases could provide important insight into future targeted therapies. However, current models utilized in target discovery research are limited by the inability to accurately recapitulate the complex stromal architecture and heterogenous genetic and molecular composition of breast cancer. Furthermore, immortalized cell lines are limited to a 2D environment and over time acquire mutations that may not reflect the primary tumor. Recently, our laboratory has successfully established two TNBC patient-derived xenograft (PDX) models derived from African-American patients, and generated cell lines (TU-BCx-2K1, TU-BCx-2O0) and mammospheres. One of these models, 2O0, presents tumor architecture, cellular composition, genomic (qRT-PCR) and protein (western blot) expressions that are concordant with a claudin-low subtype, which has higher rates of metastasis and recurrence. Furthermore, we show that both TNBC models metastasize to the lungs, and exhibit molecular characteristics consistent with mesenchymal phenotypes. We utilized these translational PDX models to screen a library of small molecule inhibitors that represent a variety of kinase pathways to identify novel therapeutic targets and/or pathways that are specific to TNBC subtypes. We found in a preliminary cell morphology screen using three TNBC cell lines (MDA-MB-231, BT549, MDA-MB-157), two small molecule inhibitors that increased epithelial marker (CDH1) gene expression, suppressed mesenchymal (VIM, c-FOS, SNAI1, ZEB1) expression and/or suppressed cellular motility in transwell migration assays. We observed after ex vivo treatments with our PDX tumors the two compounds increase the epithelial marker CDH1 expression, and suppress mesenchymal markers (VIM, MMP2, c-FOS, SNAI1, ZEB1) expressions. We confirm these findings in the TU-BCx-2K1 cell line. Kinase array data revealed candidate kinases responsible for the observed EMT changes in the two compounds of interest (NEK5, NEK9, NEK1 potentially affect cell motility; SRC-family kinases, TAOK2, STK10 potentially affect EMT gene changes); we plan to utilize the PDX cell lines to characterize these kinases in EMT. We aim to ultimately discover novel therapeutic targets specific to different TNBC molecular subtypes. Citation Format: Matossian M, Burks H, Bowles A, Sabol R, Hoang V, Elliott S, Bunnell B, Zuercher W, Drewry D, Wells C, Alfortish A, Lee S, Hartono A, Jones S, Moroz K, Zea A, Burow M, Collins-Burow B. Patient-derived triple negative breast cancer xenografts as a translational model to screen for novel kinase pathways [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P5-05-05.
- Published
- 2018
- Full Text
- View/download PDF
32. Abstract 1117: Triple negative breast cancer patient-derived xenografts as a translational model for discovery of novel therapeutic targets
- Author
-
Van T. Hoang, Annie C. Bowles, Krzysztof Moroz, Bridgette M. Collins-Burow, Matthew E. Burow, Rachel A. Sabol, Bruce A. Bunnell, Hope E. Burks, Margarite D. Matossian, William J. Zuercher, David H. Drewry, and Carrow I. Wells
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,Kinase ,Population ,Cancer ,Biology ,Cell morphology ,medicine.disease ,Primary tumor ,Breast cancer ,Internal medicine ,SNAI1 ,medicine ,education ,Triple-negative breast cancer - Abstract
Triple negative breast cancers (TNBCs) constitute 12% of all breast cancers, and is approximately twice more prevalent in African-American populations. Louisiana has a high proportion of African-American residents (32.5% in 2015), and thus hosts a higher population of TNBC patients. TNBCs have an aggressive phenotype that is elusive to the targeted therapeutics used to treat other breast cancer subtypes. The claudin-low molecular subtype has higher rates of metastases and recurrence. Certain kinase families have been extensively studied as regulators of epithelial-mesenchymal transition (EMT), a process involved in the initiation of cancer metastasis. Discovery of novel kinase targets within the subset of uncharacterized kinases could provide important insight into future targeted therapies. However, current models utilized in target discovery research are limited by the inability to accurately recapitulate the complex stromal architecture and heterogenous genetic and molecular composition of breast cancer. Furthermore, immortalized cell lines are limited to a 2D environment and over time acquire mutations that may not reflect the primary tumor. Recently, our laboratory has successfully established two TNBC patient-derived xenograft (PDX) models derived from African-American patients, and generated cell lines (TU-BCx-2K1, TU-BCx-2O0) and mammospheres. One of these models, 2O0, presents tumor architecture, cellular composition, genomic (qRT-PCR) and protein (western blot) expressions that are concordant with a claudin-low subtype. Furthermore, we show that both TNBC models metastasize to the lungs, and exhibit molecular characteristics consistent with mesenchymal phenotypes. We utilized these translational PDX models to screen a library of small molecule inhibitors that represent a variety of kinase pathways to identify novel therapeutic targets and/or pathways that are specific to TNBC subtypes. We found in a preliminary cell morphology screen using three TNBC cell lines (MDA-MB-231, BT549, MDA-MB-157), two small molecule inhibitors that increased epithelial marker (CDH1) gene expression, suppressed mesenchymal (VIM, c-FOS, SNAI1, ZEB1) expression and/or suppressed cellular motility in transwell migration assays. We observed after ex vivo treatments with our PDX tumors the two compounds increase the epithelial marker CDH1 expression, and suppress mesenchymal markers (VIM, MMP2) expressions. We confirm these findings in the TU-BCx-2K1 cell line. Kinase array data revealed candidate kinases responsible for the observed EMT changes in the two compounds of interest (NEK5, NEK9, NEK1 potentially affect cell motility; SRC-family kinases, TAOK2, STK10 potentially affect EMT gene changes); we plan to utilize the PDX cell lines to characterize these kinases in EMT. We aim to ultimately discover novel therapeutic targets specific to different TNBC molecular subtypes. Citation Format: Margarite D. Matossian, Hope E. Burks, Annie C. Bowles, Rachel A. Sabol, Van T. Hoang, Bruce Bunnell, William J. Zuercher, David H. Drewry, Carrow Wells, Krzysztof Moroz, Matthew E. Burow, Bridgette Collins-Burow. Triple negative breast cancer patient-derived xenografts as a translational model for discovery of novel therapeutic targets [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1117. doi:10.1158/1538-7445.AM2017-1117
- Published
- 2017
- Full Text
- View/download PDF
33. Correction: Evaluation of deacetylase inhibition in metaplastic breast carcinoma using multiple derivations of preclinical models of a new patient-derived tumor.
- Author
-
Tiffany C Chang, Margarite D Matossian, Steven Elliott, Hope E Burks, Rachel A Sabol, Deniz A Ucar, Henri Wathieu, Jovanny Zabaleta, Luis Del Valle, Sukhmani Gill, Elizabeth Martin, Adam I Riker, Lucio Miele, Bruce A Bunnell, Matthew E Burow, and Bridgette M Collins-Burow
- Subjects
Medicine ,Science - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0226464.].
- Published
- 2021
- Full Text
- View/download PDF
34. Evaluation of deacetylase inhibition in metaplastic breast carcinoma using multiple derivations of preclinical models of a new patient-derived tumor.
- Author
-
Tiffany C Chang, Margarite D Matossian, Steven Elliott, Hope E Burks, Rachel A Sabol, Deniz A Ucar, Henri Wathieu, Jovanny Zabaleta, Luis Del Valle, Sukhmani Gill, Elizabeth Martin, Adam I Riker, Lucio Miele, Bruce A Bunnell, Matthew E Burow, and Bridgette M Collins-Burow
- Subjects
Medicine ,Science - Abstract
Metaplastic breast carcinoma (MBC) is a clinically aggressive and rare subtype of breast cancer, with similar features to basal-like breast cancers. Due to rapid growth rates and characteristic heterogeneity, MBC is often unresponsive to standard chemotherapies; and novel targeted therapeutic discovery is urgently needed. Histone deacetylase inhibitors (DACi) suppress tumor growth and metastasis through regulation of the epithelial-to-mesenchymal transition axis in various cancers, including basal-like breast cancers. We utilized a new MBC patient-derived xenograft (PDX) to examine the effect of DACi therapy on MBC. Cell morphology, cell cycle-associated gene expressions, transwell migration, and metastasis were evaluated in patient-derived cells and tumors after treatment with romidepsin and panobinostat. Derivations of our PDX model, including cells, spheres, organoids, explants, and in vivo implanted tumors were treated. Finally, we tested the effects of combining DACi with approved chemotherapeutics on relative cell biomass. DACi significantly suppressed the total number of lung metastasis in vivo using our PDX model, suggesting a role for DACi in preventing circulating tumor cells from seeding distal tissue sites. These data were supported by our findings that DACi reduced cell migration, populations, and expression of mesenchymal-associated genes. While DACi treatment did affect cell cycle-regulating genes in vitro, tumor growth was not affected compared to controls. Importantly, gene expression results varied depending on the cellular or tumor system used, emphasizing the importance of using multiple derivations of cancer models in preclinical therapeutic discovery research. Furthermore, DACi sensitized and produced a synergistic effect with approved oncology therapeutics on inherently resistant MBC. This study introduced a role for DACi in suppressing the migratory and mesenchymal phenotype of MBC cells through regulation of the epithelial-mesenchymal transition axis and suppression of the CTC population. Preliminary evidence that DACi treatment in combination with MEK1/2 inhibitors exerts a synergistic effect on MBC cells was also demonstrated.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.