Explore This Lab

Overview

The Rueda lab has been involved in basic, translational and clinical research focused on women’s reproductive health. The lab specializes in reproductive and cancer biology, with an emphasis on the investigation of novel signaling pathways that contribute to the pathology of benign gynecologic diseases, malignant transformation, and the contribution of cancer stem cells to the pathology of ovarian and uterine tumors. A major effort in the Rueda laboratory has been to identify and functionally characterize inherent and acquired mechanisms that contribute to metastasis, resistance, and recurrence of gynecological cancers to better inform the development and testing of novel or repurposed drugs that can target both cancer stem and non-stem tumor cells. 

The Rueda Lab and its collaborators were responsible for the establishment of the VCRB/MGH GYN Tissue Repository.  The repository was established to develop a collection of clinically applicable tissue and correlate biologic samples (ex. blood, ascites) collected from women diagnosed with gynecologic conditions and or gynecologic cancers along with their annotated medical information. From the inception one of our objectives was to establish an infrastructure whereby all gynecologic samples were collected under a single carefully maintained IRB approved repository protocol. Moreover, the presence of an established well-maintained repository that has appropriate management would be the best way to ensure a quality product, equitable distribution, and oversight. At present the repository has collected over 3000 samples. The VCRB/MGH Repository is led by Dr. Bo Rueda who serves as the Executive Director. Other leaders include members of the MGH Gynecologic Oncology Research Consortium (Drs. Eric Eisenhauer, David Spriggs and Cesar Castro).

This rich resource serves to support our investigators whose research is focused on developing or improving early diagnostics, defining molecular pathways contributing to the genesis of the tumors, developing pre-clinical models for testing of novel anti-cancer therapies and or combination drug strategies including primary derived organoids and xenografts, and discerning mechanisms contributing to inherent and acquired treatment resistance.

The overall success of our banking efforts is the byproduct of collegial interactions between our gyn surgeons, medical oncologists, pathology staff, gyn oncology fellows, post-doctoral fellows, clinical research coordinators, clinical support staff and research technologists. Importantly, we recognize the unselfish generosity of the patients themselves for providing informed consent which allows us to collect the discarded samples and the annotated clinical information which we hope will benefit others suffering from similar diagnoses and conditions. Similarly, we appreciate that this resource was initiated with the support of the Nile Albright Research Foundation (A.K.A Advanced Medical Research Foundation) and the Vincent Memorial Hospital Foundation.

The benefits of this effort are evidenced by the increase in clinically meaningful publications in high quality peer reviewed publications, increased extramural funding from federal funding agencies and foundations. Also of importance is that this resource serves to promote active collaborations with pharmaceutical companies to test novel anti-cancer agents while recognizing the potential for opportunities to move forward into phase I trials should they be successful.

Mass General Gynecologic Oncology Research Consortium

The Mass General Gynecologic Oncology Research Consortium (MGH Gyn ORC) was formalized to unite basic, translational and clinician scientists to engage in transformative research that will enhance our understanding of the molecular and cellular factors that contribute to the genesis, progression and resistance of gynecologic cancers enabling improved diagnosis, treatment, and prevention.

The objectives/goals of the Gyn ORC are:
  • Develop and support an open interactive research infrastructure to promote the integration of knowledge, technology, resources, and personnel needed to combat gynecologic malignancies at MGH.
  • Support sample and annotated data collection from our patient cohort to develop better diagnostics, understand how treatment alters the tumor landscape, and to appreciate mechanisms contributing to inherent and acquired resistance to inform clinical care.
  • Develop applicable primary derived organoids and xenografts from our patients gyn malignancies for testing of novel drugs and or combination drug treatment strategies.
  • Continue to develop interactive educational strategies for our clinicians in training, postdoctoral fellows, and graduate students.
  • Promote diversity, equity, and inclusion with regards to appreciating how malignancies impact everyone. Recognizing how some individuals representing different communities, cultures, and or race may be differentially affected and how we might better serve those in need.
  • Support and integrate Clinical and Outcomes based research to improve patient’s outcome and quality of life.
The MGH Gyn ORC leadership includes Drs. Eric Eisenhauer, Cesar Castro and Bo Rueda. Dr. Eisenhauer represents the Division of Gynecologic Oncology and Dr. Rueda represents, the Vincent Center for Reproductive Biology in the Department of Obstetrics and Gynecology. Dr. Castro represents the gynecological cancer focused efforts of the Mass General Cancer Center and the Gyn Medical Oncologists in the Department of Medicine.

View all consortium members

Sara Bouberhan, MD, Gynecologic Medical Oncology

Dr. Bouberhan’s research is focused on identifying blood-based biomarkers in ovarian cancer. She is currently working on a project to detect patient-specific p53 mutations in plasma using droplet-digital PCR. This project relies on an ongoing collaboration with the Gyn Tissue Repository. Banked specimens from patients’ initial surgical resection are accessed to identify tumor-specific DNA sequences. These sequences are then used to develop customized DNA sequencing primers and detection probes which are applied to blood samples. Blood collected through the tissue banking protocol at the time of patients’ initial surgery is used as a substrate to identify circulating tumor DNA. Dr. Bouberhan also collaborates with the tissue repository team to prospectively collect blood and ascites specimens to support this and future projects. 

Amy Bregar, MD, Gynecologic Oncology

Amy Bregar, MD, is an Assistant Professor in Obstetrics, Gynecology and Reproductive Biology at Harvard Medical School. She is the director of the gynecologic oncology sub-internship for medical students and is lead faculty for the resident gynecologic oncology rotation at Massachusetts General Hospital.

Dr. Bregar’s research interest includes clinical trial development, medical education, and quality improvement. She is a member of multiple professional organizations, including the New Investigators Committee and the Older Adults Working Group of the national NRG (formally Gynecologic Oncology Group or GOG). Dr. Bregar is a principal investigator of clinical trials evaluating novel therapies in the treatment of ovarian cancer

Alexander Melamed, MD, MPH

Alexander Melamed, MD, MPH, focuses his research on the evaluation of the effectiveness of health care service, with an emphasis on the role of surgery in improving outcomes among women with gynecologic cancer. To overcome the challenge of unmeasured confounding when attempting to estimate causal association in observational data, Dr. Melamed employs sophisticated analytic methods including acyclic graphs, propensity score methods, and quasi- experimental study designs. His studies have been published in the New England Journal of Medicine, JAMA, BMJ, and high impact cancer journals. Dr. Melamed is currently on the management committee of the Laparoscopic Cytoreduction After Neoadjuvant Chemotherapy (LANCE) trial, an international integrated phase III/pilot randomized trial designed to assess the efficacy of minimally invasive surgery for interval cytoreduction of advanced ovarian cancer.

David Pépin, PhD

Dr. Pépin’s laboratory studies female reproductive development, and particularly how the TGF-b superfamily of hormones regulates ovarian and uterine formation and subsequent functions. His group focuses on Anti-Mullerian Hormone, aka AMH, and the mechanisms by which it controls ovarian folliculogenesis. Discoveries regarding AMH and other related hormones are being translated to the clinic, with the development of a new class of contraceptives, infertility treatments, drugs protecting the fertility of children and women receiving chemotherapies, and even treatments for ovarian cancer. Importantly, his team is developing multiple treatment strategies, from biologics, to gene therapies, and high-throughput drug screens to address unmet needs in women’s health.

Andrea Russo, MD, Gynecologic Radiation Oncology

As Director of the Gynecologic Radiation Oncology Program at MGH, Andrea Russo, MD, dedicates her time and skills to the care of patients with gynecologic cancers. Dr. Russo is also an Assistant Professor at Harvard Medical School. She is actively engaged in clinical and translational research to improve patient outcomes.

With Dr. Russo's expertise in the latest radiation technologies including brachytherapy, IMRT/VMAT, and proton therapy, she is committed to providing the highest quality of evidence-based and compassionate care.

Steven Skates, PhD

Dr. Skates has been dedicated to studying early detection of ovarian cancer including biomarker discovery and validation. He spearheaded development of multiple longitudinal early detection algorithms which were implemented in five prospective early detection clinical trials. More specifically, Dr. Skates risk of ovarian cancer algorithm has been implemented in a definitive trial of 200,000 postmenopausal women with half screened annually for up to 11 years and with ovarian cancer mortality as the “gold standard” endpoint. Dr. Skates has served as a co-investigator for multiple proposal s led MGH, BWH and DFCI based investigators on identification and validation of biomarker(s) for ovarian cancer detection. He is recruited by Gyn ORC members to develop statistical models for proposed research applications.

David Spriggs, MD, Gynecologic Medical Oncology

Dr. Spriggs’ research is focused on cell membrane glyco-proteins that promote the patho-biology of ovarian cancer. The serum marker, CA125, has been used to manage ovarian cancer since the 1980’s but its function(s) have yet to be completely identified. CA125 is the cleaved portion of the tethered mucin MUC16. The Spriggs Lab have provided insights into the function of MUC16.

It is now apparent that MUC16 regulates growth, invasion and metastatic disease disease through the structure of sugars (glycosylation) on the surface of normal and cancer cells. This regulation requires interaction with specialized sugar binding proteins, Galectins, which are key components of the tumor microenvironment. We are actively developing antibodies against MUC16 and Galectin 3 for diagnosis, imaging and treatments. Our work has shown that antibodies which inhibit these cell – cell interactions can slow tumor growth and inhibit metastasis.

Cheng Wang, PhD

Dr. Wang’s group focuses on the identification of key regulatory proteins in the Hippo-Yap signaling pathway that regulate the normal physiology of the cervix and ovary. More importantly his team is defining how aberrant Hippo-Yap signaling can promote malignant transformation, invasive and metastatic properties.

Recently, Dr. Wang has shown that increased Yap signaling can promote an immunotolerant environment. Delineating the mechanisms by which Yap promotes immunotolerance will allow for the development of novel treatment strategies to overcome this tumor resistant micro-environment. To accomplish these goals Dr. Cheng’s lab develops novel transgenic mouse models to determine the functional contributions of the members of the Hippo-Yap signaling pathway in cervical and ovarian cancer.

Oladapo Yeku, MD, PhD, FACP, Gynecologic Medical Oncology

Dr. Yeku’s research efforts are focused on delineating the functional contributions of the tumor microenvironment. Specifically, he and his team are designing and testing immunologic approaches for the management of ovarian cancer. His research utilizes syngeneic immune competent mouse models of metastatic ovarian cancer with subsequent validation in appropriate genetically engineered and xenograft models. This approach allows for analysis of the effects of this combinatorial therapy in recruiting and harnessing the hosts’ effector immune cells against cancer cells. Furthermore, this approach allows critical evaluation of the immunosuppressive cytokine, cellular and tumor microenvironment in response to therapy.

The MGH Gyn ORC maintains the VCRB/ MGH Gynecologic Tissue Repository which actively collects tissue and correlates biologics from informed patients who have provided written consent for the collection of excess material collected as part of a scheduled gynecologic procedure. 
Learn more about the Tissue Repository
We primarily focus on cases that have the greatest potential to yield tumor tissue/ascites and blood samples along with their corresponding clinical data.
 
We aim to generate a cohort of representative patient-derived xenograft and patient-derived organoid models from gynecologic tumor cells collected pre-and post-treatment.
 
When feasible we genomically, molecularly, and functionally characterize our gynecologic cancer samples.

Current Projects

Delineating the diagnostic and therapeutic potential of the tumor-associated carbohydrate (TACA), sialyl-Tn in ovarian cancer. Working in collaboration with our pharma collaborators we identified and characterized mouse anti-STn specific antibodies for cancer therapeutic applications in binding assays, and in vitro, and in vivo efficacy assays. Our collaborative efforts provided evidence to support the hypothesis that an STn positive fraction recovered from OvCa cells displays cancer stem-like properties. This work was followed our teems demonstrating the efficacy of a humanized anti-STn-ADCs in in vitro and in vivo models of ovarian cancer. We recently completed additional in vivo studies illustrating the effectiveness of the anti-STn with and without cytotoxics. We now have evidence to suggest that a custom STn ELISA we developed may serve to help identify patients that an anti-STn strategy may benefit.  Working with Dr. Steve Skates we are assessing whether the STn based ELISA could augment CA125 as a diagnostic and or for following treatment response. In addition, we have preliminary data that supports the concept that STn is immunosuppressive and are working to validate this finding.

Determining whether tumor cell plasticity can account for the enrichment of cancer stem cell populations post treatment. Numerous reports support the concept of a subpopulation of cancer cells that have or can obtain the capacity to gain stem like properties and are often referred to as caner stem cells (CSCs). The inherent or acquired mechanisms by which these stem-like properties might be (re)activated remains incompletely understood. Appropriately, it is believed that the tumor microenvironment contributes to this change in phenotype via crosstalk, including that cancer associated fibroblasts can influence tumor cells to acquire more stem-like properties. Similarly, it has been proposed the cytokines and/or chemokines secreted directly from bulk tumor cells might influence the stem sources.  More recently, however, it was proposed that extracellular vesicles (EVs) may be responsible for sharing the necessary cellular machinery (ex. miRNAs, lncRNAs, demethylases) to influence these tumor cells to gain stem-like properties.  This led us to hypothesize that EVs extruded from ovarian CSC in response to treatment might contain factors needed to convert neighboring tumor cells to become more stem-like. We are currently focusing our efforts on defining the potential mechanisms by which this could occur and with an emphasis on the direct transfer or through upregulation of methyltransferases which can promote stem like properties. 

Investigating mechanisms contributing to resistance to current HER2 treatment strategies in endometrial cancer.  A significant percentage of high grade endometrioid and uterine serous cancers cancer display HER2 amplifications. Utilizing in vitro and in vivo approaches, we continue to gain multi-dimensional insights into how cells may attain resistance to HER2 directed therapy. We believe this work will improve how we might apply the anti HER2 agents underdevelopment towards high grade endometrial cancer.  For this we have generated or accumulated multiple HER2 resistant endometrial cancer models for which we are testing combination strategies with the intent to overcome inherent and acquired resistance to ensure a more durable response.

Provide pre-clinical rationale for identifying unique endometrial tumor molecular characteristics that will influence clinical decisions.  Endometrial cancer mortality has increased over the past three decades despite medical advances in our understanding of the genomic profile and the advent of immunotherapy. Therapies for recurrent disease still have limited durability highlighting the need for novel therapeutic strategies that exploit the molecular underpinnings of an individual tumor. While encouraging anti-tumor activity has been observed utilizing immune checkpoint blockade in recurrent endometrial cancer, over 50% of women do not respond despite harboring the molecular signatures associated with response. Improved response to immune checkpoint blockade has been observed in ovarian tumors when poly-ADP-ribose polymerase (PARP) inhibitors have been used simultaneously suggesting synergistic activity. The degree of homologous repair deficiency (HRD) has only been characterized on a limited basis in endometrial cancers, but much pre-clinical data has demonstrated that genomic events, such as PIK3CA mutation and loss of PTEN create genomic instability mimicking HRD. We hypothesize that a subset of endometrial tumors harbors a significant degree of HRD that associates with the expression of immune checkpoint proteins. We are currently analyzing data derived from testing high grade endometrioid and serous endometrial carcinomas using the FoundationOne platform to detect tumor gain of function mutations, mutational burden, loss of heterozygosity, checkpoint protein RNA dosage and PD-1 expression. Associations amongst the tumor molecular characteristics will be correlated with clinical outcomes. We plan to functionally test our findings by treating correlate primary derived organoids and assessing their response. It is anticipated that these data will provide some preclinical rationale for clinical trials investigating the role of dual immune checkpoint and PARP inhibition in women with recurrent endometrial cancer.

Delineating the functional contribution of Galectin 3 to the pathology of ovarian endometrial cancers. Over the past two years we have expanded our interests in glycobiology to better understand how Galectin-3 functionally contributes to the pathology of high grade ovarian and uterine cancer. Our research efforts are focused on how it promotes an immunosuppressive environment, promotes stem like properties and influences metastasis. In doing so we have teamed up with Drs. David Spriggs and Oladapo Yeku to focus on testing small molecule inhibitors in combination with a novel anti-Gal-3 antibody-based strategy to neutralize Gal-3’s protumor properties in uterine and ovarian carcinoma.

Working with our Pharma colleagues/collaborators: We have multiple collaborative and SRA related projects whereby we are actively working with Pharma to develop and validate diagnostics/biomarkers in blood and/or tumor samples, identify new targets contributing to treatment resistant recurrent disease, testing the efficacy of novel anti-cancer agents in our pre-clinical cancer models, and developing novel drugs for treatment of gynecologic cancers.

Who We Are

Bo Rueda, PhD

Director, Vincent Center for Reproductive Biology, Massachusetts General Hospital
Associate Professor, Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School

Affiliated Faculty, Harvard Stem Cell Institute

Affiliated Faculty, Massachusetts General Hospital Cancer Center

Meet our team

Paula DiBenedettoPaula DiBenedetto
Research Technician I

Paula DiBenedetto is originally from Northbrook, Illinois and obtained her bachelor's degree in Biochemistry from the University of Michigan. She now works as a Research Technician in the Rueda Lab, where she helps with ongoing research projects focused on identifying targetable pathways contributing to drug resistant gynecologic cancers. In addition, Paula plays a pivotal role in the Center’s effort to expand its Gyn sample banking efforts and pre-clinical model development. Paula hopes to learn from the unique intersection of this clinical, translation and research experience in Dr. Rueda's lab before attending medical school. When she is not in the lab, Paula spends her time reading, running, and exploring Boston with friends.


Ursula Winter, PhD
Postdoctoral Fellow

Ursula Winter, PhD, is a postdoctoral fellow of Im Hyungsoon Lab at the Center for Systems Biology within the Radiology Department at Massachusetts General Hospital. With a robust background in genetics and a doctorate in pharmaco-oncology, Dr. Winter's expertise lies in cell biology, particularly in identifying crucial biomarkers, refining treatment strategies, and monitoring drug responses, with a primary focus on advancing patient care in breast and ovarian cancer. She actively engages in preclinical development and validation of medical devices, pioneering the integration of extracellular vesicle characterization as a non-invasive alternative to traditional liquid biopsies.

Additionally, Dr. Winters spearheads research into nanostructures aimed at enhancing signal detection and revolutionizing diagnostic capabilities. "Through collaborative efforts with interdisciplinary teams, like Dr. Rueda’s Lab, we seamlessly bridge scientific inquiry with clinical implementation, fostering innovative solutions to address the needs of oncology patients", she shares.


Maryam Azimi, PhDMaryam Azimi, PhD
Postdoctoral Fellow

Maryam Azimi, PhD, received her PhD in Medical Immunology from Tehran University of Medical Science, Tehran, Iran. Her PhD was focused on the immunomodulatory effects of Regulatory T cells derived Exosomes (EVs) on Helper T cells (CD4+ T cells) in immunologic disorders. One main objective of her study was to find diagnostic and therapeutic biomarkers, especially MicroRNAs encapsulated in EVs.

After her graduation, Dr. Azimi continued her research field as a researcher at the Immunology Research Center at the Iran University of Medical Sciences. In 2020, she resumed her work as an Assistant Professor. During this time, she worked on the immunomodulatory effects of mesenchymal stem cell-derived exosomes in suppressing immune responses in tumors. Due to her interest in obtaining international experience, she joined Dr. Bo Rueda's Laboratory at Massachusetts General Hospital, Harvard Medical School, as a postdoctoral research fellow in August 2022.

Dr. Azimi's current research focuses on understanding the role of retrotransposons, specifically Long Interspersed Element-1 (LINE-1), in DNA damage and chromosomal instability leading to tumorigenesis in gynecological tissue. She is addressing various questions about the underlying roles of LINE-1 by establishing primary Fallopian tube epithelial cells in both 2D and 3D culture models. Outside of the lab, she likes photography, reading books, and working out.


Eugene KimEugene Kim
Research Technician II

Eugene Kim grew up in Edison, NJ before receiving his bachelor’s degree in mathematics at Amherst College. He currently works as a research technician in the Rueda Lab and supports the tissue banking efforts as well as other post-docs/fellows with their projects. Eugene hopes to attend medical school in the future and attributes a lot of his motivation to his experiences at this job. In his free time, he enjoys running, working out, and spending time with his friends.


Venkatesh (Venky) Pooladanda, MS (Pharm.), PhDVenkatesh (Venky) Pooladanda, MS (Pharm.), PhD
Postdoctoral Fellow

Venkatesh (Venky) Pooladanda, MS (Pharm.), PhD, received his doctoral degree in Pharmacology and Toxicology from National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India where he studied the effect of acute lung inflammation on tumor metastasis. He also has training in immunology, nano medicine, and infectious diseases. Dr. Pooladanda has the knowledge of both basic and translational research. He is passionate about academic research in immuno-oncology and is looking to translate the laboratory outcomes to clinical benefits. He loves taking on new adventures in science and explore new signaling pathways and molecular therapies. Apart from the science, he enjoys meditation and hiking.


Lissah JohnsonLissah Johnson
Student

Lissah Johnson is a PhD candidate in the Biological Sciences in Public Health program at Harvard T.H. Chan School of Public Health and works jointly in the laboratories of Dr. Bo Rueda and Dr. Kristopher Sarosiek. In the Rueda Lab, her project focuses on understanding how ovulation-related exposures can trigger pro-cancerous changes in fallopian tube epithelial cells — where high grade serous ovarian cancer originates. Prior to starting graduate school, she received a BS in Chemistry with Honors from the University of Redlands in Southern California and then worked as a chemist in an environmental health laboratory at the California Department of Public Health. Outside of the laboratory, she enjoys dancing salsa and bachata, spending time outdoors running or hiking, and exploring the art, culture, history and cuisine of new places.


Publications

Publications

Al-Alem, L.F.; Baker, A.T.; Pandya, U.M.; Eisenhauer, E.L.; Rueda, B.R. Understanding and Targeting Apoptotic Pathways in Ovarian Cancer. Cancers. 2019 Oct 24; 11(11):1631. doi:10.3390/cancers11111631

Bellio, C.; DiGloria, C.; Spriggs, D.R.; Foster, R.; Growdon, W.B.; Rueda, B.R. The Metabolic Inhibitor CPI-613 Negates Treatment Enrichment of Ovarian Cancer Stem CellsCancers. 2019 Oct 29; 11(11):1678. doi:10.3390/cancers11111678

Bellio C, DiGloria C, Foster R, James K, Konstantinopoulos PA, Growdon WB, Rueda BR. PARP inhibition induces enrichment of DNA repair proficient CD133 and CD117 positive ovarian cancer stem cells. Mol Cancer Res. 2018 Nov 6. pii: molcanres.0594.2018. doi: 10.1158/1541-7786.MCR-18-0594. [Epub ahead of print] PMID:30401718.

Chiu YH, Karmon AE, Gaskins AJ, Arvizu M, Williams PL, Souter I, Rueda BR, Hauser R, Chavarro JE; Serum omega-3 fatty acids and treatment outcomes among women undergoing assisted reproduction. EARTH Study Team.Hum Reprod. 2018 Jan 1;33(1):156-165. doi: 10.1093/humrep/dex335.PMID:29136189

Cardozo ER, Foster R, Karmon AE, Lee AE, Gatune LW, Rueda BR, Styer AK. MicroRNA 21a-5p overexpression impacts mediators of extracellular matrix formation in uterine leiomyoma. Reprod Biol Endocrinol. 2018 May 11;16(1):46. doi: 10.1186/s12958-018-0364-8. PMID:29747655

Starbuck K, Al-Alem L, Eavarone DA, Hernandez SF, Bellio C, Prendergast JM, Stein J, Dransfield DT, Zarrella B, Growdon WB, Behrens J, Foster R, Rueda BR. Treatment of ovarian cancer by targeting the tumor stem cell-associated carbohydrate antigen, Sialyl-Thomsen-nouveau. Oncotarget. 2018 May 1;9(33):23289-23305. doi: 10.18632/oncotarget.25289. eCollection 2018 May 1.PMID:29796189

Eavarone DA, Al-Alem L, Lugovskoy A, Prendergast JM, Nazer RI, Stein JN, Dransfield DT, Behrens J, Rueda BR. Humanized anti-Sialyl-Tn antibodies for the treatment of ovarian carcinoma. PLoS One. 2018 Jul 27;13(7):e0201314. doi: 10.1371/journal.pone.0201314. eCollection 2018.PMID:30052649

Bregar A, Deshpande A, Grange C, Zi T, Stall J, Hirsch H, Reeves J, Sathyanarayanan S, Growdon WB, Rueda BR. Characterization of immune regulatory molecules B7-H4 and PD-L1 in low and high grade endometrial tumors. Gynecologic oncology. 2017; 145(3):446-452. PubMed [journal] PMID:28347512

Prendergast JM, Galvao da Silva AP, Eavarone DA, Ghaderi D, Zhang M, Brady D, Wicks J, DeSander J, Behrens J, Rueda BR. Novel anti-Sialyl-Tn monoclonal antibodies and antibody-drug conjugates demonstrate tumor specificity and anti-tumor activity. mAbs. 2017; 9(4):615-627. PubMed [journal] PMID: 28281872PMCID: PMC5419082

Bajwa P, Nielsen S, Lombard JM, Rassam L, Nahar P, Rueda BR, Wilkinson JE, Miller RA, Tanwar PS. Overactive mTOR signaling leads to endometrial hyperplasia in aged women and mice. Oncotarget. 2017; 8(5):7265-7275. PubMed [journal] PMID: 27980219 PMCID: PMC5352319

Garrett LA, Growdon WB, Rueda BR, Foster R. Influence of a novel histone deacetylase inhibitor panobinostat (LBH589) on the growth of ovarian cancer. Journal of ovarian research. 2016; 9(1):58. PubMed [journal] PMID: 27633667 PMCID: PMC5025559

Guan Y, Guo L, Zukerberg L, Rueda BR, Styer AK. MicroRNA-15b regulates reversion-inducing cysteine-rich protein with Kazal motifs (RECK) expression in human uterine leiomyoma. Reproductive biology and endocrinology : RB&E. 2016; 14(1):45. PubMed [journal] PMID: 27530410 PMCID: PMC4988044

Styer AK, Rueda BR. The Epidemiology and Genetics of Uterine Leiomyoma. Best practice & research. Clinical obstetrics & gynaecology. 2016; 34:3-12. PubMed [journal] PMID: 26725703

Hernandez SF, Chisholm S, Borger D, Foster R, Rueda BR, Growdon WB. Ridaforolimus improves the anti-tumor activity of dual HER2 blockade in uterine serous carcinoma in vivo models with HER2 gene amplification and PIK3CA mutation. Gynecol Oncol. 2016 Apr 1. pii: S0090-8258(16)30083- X. doi: 10.1016/j.ygyno.2016.03.027. 141(3):570-579. PubMed [journal] PMID: 27017985

Roussel-Gervais A, Couture C, Langlais D, Takayasu S, Balsalobre A, Rueda BR, Zukerberg LR, Figarella-Branger D, Brue T, Drouin J. The Cables1 Gene in Glucocorticoid Regulation of Pituitary Corticotrope Growth and Cushing Disease. The Journal of clinical endocrinology and metabolism. 2016; 101(2):513-22. PubMed [journal] PMID: 26695862

Clark NC, Friel AM, Pru CA, Zhang L, Shioda T, Rueda BR, Peluso JJ, Pru JK. Progesterone receptor membrane component 1 promotes survival of human breast cancer cells and the growth of xenograft tumors. Cancer biology & therapy. 2016; 17(3):262-71. PubMed [journal] PMID: 26785864 PMCID: PMC4847980

Hernandez SF, Vahidi NA, Park S, Weitzel RP, Tisdale J, Rueda BR, Wolff EF. Characterization of extracellular DDX4- or Ddx4-positive ovarian cells. Nature medicine. 2015; 21(10):1114-6. PubMed [journal] PMID: 26444630

Complete list of published work