Rebecca Previs
Positions:
Adjunct Assistant Professor in the Department of Obstetrics and Gynecology
Obstetrics and Gynecology, Gynecologic Oncology
School of Medicine
Member of the Duke Cancer Institute
Duke Cancer Institute
School of Medicine
Education:
M.D. 2009
University of Virginia School of Medicine
Residency, Obstetrics And Gynecology
Duke University School of Medicine
Fellow, Gynecologic Oncology
University of Texas MD Anderson Cancer Center
Grants:
Janet Burros Ovarian Cancer Repository
Administered By
Obstetrics and Gynecology, Gynecologic Oncology
Awarded By
Cancer Data Initiative
Role
Co Investigator
Start Date
End Date
Evaluation of homologous recombination deficiency in uterine serous cancer
Administered By
Obstetrics and Gynecology, Gynecologic Oncology
Awarded By
Myriad Genetics, Inc.
Role
Principal Investigator
Start Date
End Date
A Role of Multilevel Healthcare Access Dimensions in Ovarian Cancer Disparities
Administered By
Population Health Sciences
Awarded By
National Institutes of Health
Role
Co Investigator
Start Date
End Date
Use of an oncolytic adenovirus to promote anti-tumor T-cell responses in ovarian cancer
Administered By
Obstetrics and Gynecology
Awarded By
Foundation for Women's Cancer
Role
Mentor
Start Date
End Date
Adrenergic signaling inhibition to enhance the immunogenicity of the ovarian tumor microenvironment prior to PD-1 checkpoint therapy
Administered By
Obstetrics and Gynecology, Gynecologic Oncology
Awarded By
Ponce Health Sciences University
Role
Principal Investigator
Start Date
End Date
Publications:
Multicenter Phase II Trial of the WEE1 Inhibitor Adavosertib in Refractory Solid Tumors Harboring CCNE1 Amplification.
PURPOSE: Preclinical cancer models harboring CCNE1 amplification were more sensitive to adavosertib treatment, a WEE1 kinase inhibitor, than models without amplification. Thus, we conducted this phase II study to assess the antitumor activity of adavosertib in patients with CCNE1-amplified, advanced refractory solid tumors. PATIENTS AND METHODS: Patients aged ≥ 18 years with measurable disease and refractory solid tumors harboring CCNE1 amplification, an Eastern Cooperative Oncology Group performance status of 0-1, and adequate organ function were studied. Patients received 300 mg of adavosertib once daily on days 1 through 5 and 8 through 12 of a 21-day cycle. The trial followed Bayesian optimal phase II design. The primary end point was objective response rate (ORR). RESULTS: Thirty patients were enrolled. The median follow-up duration was 9.9 months. Eight patients had partial responses (PRs), and three had stable disease (SD) ≥ 6 months, with an ORR of 27% (95% CI, 12 to 46), a SD ≥ 6 months/PR rate of 37% (95% CI, 20 to 56), a median progression-free survival duration of 4.1 months (95% CI, 1.8 to 6.4), and a median overall survival duration of 9.9 months (95% CI, 4.8 to 15). Fourteen patients with epithelial ovarian cancer showed an ORR of 36% (95% CI, 13 to 65) and SD ≥ 6 months/PR of 57% (95% CI, 29 to 82), a median progression-free survival duration of 6.3 months (95% CI, 2.4 to 10.2), and a median overall survival duration of 14.9 months (95% CI, 8.9 to 20.9). Common treatment-related toxicities were GI, hematologic toxicities, and fatigue. CONCLUSION: Adavosertib monotherapy demonstrates a manageable toxicity profile and promising clinical activity in refractory solid tumors harboring CCNE1 amplification, especially in epithelial ovarian cancer. Further study of adavosertib, alone or in combination with other therapeutic agents, in CCNE1-amplified epithelial ovarian cancer is warranted.
Authors
Fu, S; Yao, S; Yuan, Y; Previs, RA; Elias, AD; Carvajal, RD; George, TJ; Yuan, Y; Yu, L; Westin, SN; Xing, Y; Dumbrava, EE; Karp, DD; Piha-Paul, SA; Tsimberidou, AM; Ahnert, JR; Takebe, N; Lu, K; Keyomarsi, K; Meric-Bernstam, F
MLA Citation
Fu, Siqing, et al. “Multicenter Phase II Trial of the WEE1 Inhibitor Adavosertib in Refractory Solid Tumors Harboring CCNE1 Amplification.” J Clin Oncol, Dec. 2022, p. JCO2200830. Pubmed, doi:10.1200/JCO.22.00830.
URI
https://scholars.duke.edu/individual/pub1558883
PMID
36469840
Source
pubmed
Published In
Journal of Clinical Oncology
Published Date
Start Page
JCO2200830
DOI
10.1200/JCO.22.00830
Stress hormones are associated with inflammatory cytokines and attenuation of T-cell function in the ascites from patients with high grade serous ovarian cancer.
Mounting evidence suggests that chronic stress and subsequent distress can promote ovarian cancer progression. These altered psychological states have been linked to sustained release of stress hormones, activation of the β-adrenergic receptors in ovarian cancer cells, and induction of pro-tumoral signaling pathways. In addition, data suggest that chronic stress promotes an inflammatory landscape highlighted by increased infiltration of tumor-associated macrophages into the ovarian tumor microenvironment (TME). In ovarian cancer, ascites is a unique TME comprised of tumor, and immune cells, which secrete pro-tumoral cytokines and chemokines that modulate tumor-associated immunity. However, our knowledge about how stress hormones impact the ascites TME remains limited. We hypothesized that the ascites harbors measurable levels of stress hormones, and accumulation of these in the ascites generates a pro-tumorigenic, inflammatory, and immunosuppressive TME. We evaluated ascites samples from 49 patients with high grade serous ovarian cancer (HGSOC) and quantified cortisol and stress hormones metabolites, metanephrine (MN), and normetanephrine (NMN) in all samples. We also measured 38 individual cytokines in the ascites, including several pro-inflammatory cytokines, such as IL-6, which were positively correlated to MN or NMN levels of those samples. Conversely, we found cortisol levels were negatively correlated to several pro-inflammatory cytokines. As T-cells are integral to the TME and our analyses identified cytokines in the ascites known to modulate T-cell function, we characterized ascites-derived T-cells and assessed the impact of stress hormones on the T-cell phenotype. Our data show an altered CD4+/CD8+ T-cell ratio and a heterogeneous expression of exhaustion markers in T-cells from the ascites, while ascites-derived CD8+ T-cells exposed to epinephrine had decreased co-expression CD38 and Granzyme B. To extend these findings to animal models, we subjected ovarian cancer-bearing mice to daily restraint stress, which resulted in increased tumor growth in two models. Congruent with our human analyses, we detected corticosterone, MN, and NMN in the ascites from tumor-bearing mice, and these stress hormones correlated with several inflammatory cytokines. Moreover, daily restraint stress leads to increased CD4+PD-1+/CD8+PD-1+ T-cell ratio in the ovarian tumor microenvironment. Overall, these data highlight a role of stress hormones in the ascites TME as a driver of tumor-associated inflammation, T-cell suppression, and disease progression.
Authors
Aquino-Acevedo, AN; Knochenhauer, H; Castillo-Ocampo, Y; Ortiz-León, M; Rivera-López, YA; Morales-López, C; Cruz-Robles, ME; Hernández-Cordero, ER; Russell, S; Whitaker, R; Bonilla-Claudio, M; Chen, D-T; Dutil, J; Gaillard, SL; Yi, JS; Previs, RA; Armaiz-Pena, GN
MLA Citation
Aquino-Acevedo, Alexandra N., et al. “Stress hormones are associated with inflammatory cytokines and attenuation of T-cell function in the ascites from patients with high grade serous ovarian cancer.” Brain Behav Immun Health, vol. 26, Dec. 2022, p. 100558. Pubmed, doi:10.1016/j.bbih.2022.100558.
URI
https://scholars.duke.edu/individual/pub1557610
PMID
36439058
Source
pubmed
Published In
Brain, Behavior, & Immunity Health
Volume
26
Published Date
Start Page
100558
DOI
10.1016/j.bbih.2022.100558
Diversity and transparency in gynecologic oncology clinical trials.
PURPOSE: Clinical trials advance the standard of care for patients. Patients enrolled in trials should represent the population who would benefit from the intervention in clinical practice. The aim of this study was to assess whether clinical trials enrolling patients with gynecologic cancers report racial and ethnic participant composition and to examine the level of diversity in clinical trials. METHODS: Using ClinicalTrials.gov, we identified clinical trials enrolling patients with ovarian, uterine/endometrial, cervical, vaginal, and vulvar cancers from 1988 to 2019. Race and ethnicity data were extracted from participant demographics. Descriptive statistics on race, ethnicity, cancer type, location, study status, and sponsor type were calculated. Among trials which reported race and/or ethnicity, sub-analyses were performed on composition of race and ethnicity by funding source, location, and completed study status. RESULTS: A total of 1,882 trials met inclusion criteria; only 179 trials (9.5%) reported race information. Of these, the racial distribution of enrollees was 66.9% White, 8.6% Asian, 8.5% Black/African American, 0.4% Indian/Alaskan Native, 0.1% Native Hawaiian/Pacific Islander, 1.0% more than one race, and 14.5% unknown. Only 100 (5.3%) trials reported ethnicity. Except for trials enrolling patients with cervical cancer which enrolled 65.2% White and 62.1% Non-Hispanic/Latino/a patients, enrollees in trials for other gynecologic cancers were over 80% White and 88% Non-Hispanic/Latino/a. Industry funded trials enrolled higher proportions of White (68.4%) participants than non-industry funded trials (57.5%). Domestic trials report race (11.5%) and ethnicity (7.6%) at higher rates than international trials (6.9% and 2.3%, respectively). Reporting of race (1.7% vs. 13.9%) and ethnicity (1.7% vs. 11.1%) has increased over time for patients enrolled in 2000 vs. 2018. CONCLUSION: Less than 10% of trials enrolling patients with gynecologic malignancies report racial/ethnic participant composition on ClinicalTrials.gov. Accurate reporting of participant race/ethnicity is imperative to ensuring minority representation in clinical trials.
Authors
Montes de Oca, MK; Howell, EP; Spinosa, D; Knochenhauer, H; Peipert, BJ; Severson, E; Ramkissoon, S; Akinyemiju, TF; Previs, RA
MLA Citation
Montes de Oca, Mary Katherine, et al. “Diversity and transparency in gynecologic oncology clinical trials.” Cancer Causes Control, 2022. Pubmed, doi:10.1007/s10552-022-01646-y.
URI
https://scholars.duke.edu/individual/pub1496521
PMID
36284031
Source
pubmed
Published In
Cancer Causes Control
Published Date
DOI
10.1007/s10552-022-01646-y
Using genetically encoded fluorescent biosensors to interrogate ovarian cancer metabolism.
BACKGROUND: Epithelial ovarian cancer (OC) is the most lethal gynecological malignancy and patients present with significant metastatic burden, particularly to the adipose-rich microenvironment of the omentum. Recent evidence has highlighted the importance of metabolic adaptations in enabling this metastasis, leading to significant interest in evolving the arsenal of tools used to study OC metabolism. In this study, we demonstrate the capability of genetically encoded fluorescent biosensors to study OC, with a focus on 3D organoid models that better recapitulate in vivo tumor microenvironments. MATERIALS AND METHODS: Plasmids encoding the metabolic biosensors HyPer, iNap, Peredox, and Perceval were transfected into 15 ovarian cancer cell lines to assay oxidative stress, NADPH/NADP+, NADH/NAD+, and ATP/ADP, respectively. Fluorescence readings were used to assay dynamic metabolic responses to omental conditioned media (OCM) and 100 μM carboplatin treatment. SKOV3 cells expressing HyPer were imaged as 2D monolayers, 3D organoids, and as in vivo metastases via an intravital omental window. We further established organoids from ascites collected from Stage III/IV OC patients with carboplatin-resistant or carboplatin-sensitive tumors (n = 8 total). These patient-derived organoids (PDOs) were engineered to express HyPer, and metabolic readings of oxidative stress were performed during treatment with 100 μM carboplatin. RESULTS: Exposure to OCM or carboplatin induced heterogenous metabolic changes in 15 OC cell lines, as measured using metabolic sensors. Oxidative stress of in vivo omental metastases, measured via intravital imaging of metastasizing SKOV3-HyPer cells, was more closely recapitulated by SKOV3-HyPer organoids than by 2D monolayers. Finally, carboplatin treatment of HyPer-expressing PDOs induced higher oxidative stress in organoids derived from carboplatin-resistant patients than from those derived from carboplatin-sensitive patients. CONCLUSIONS: Our study showed that biosensors provide a useful method of studying dynamic metabolic changes in preclinical models of OC, including 3D organoids and intravital imaging. As 3D models of OC continue to evolve, the repertoire of biosensors will likely serve as valuable tools to probe the metabolic changes of clinical importance in OC.
Authors
MLA Citation
Bose, Shree, et al. “Using genetically encoded fluorescent biosensors to interrogate ovarian cancer metabolism.” J Ovarian Res, vol. 15, no. 1, Oct. 2022, p. 114. Pubmed, doi:10.1186/s13048-022-01046-5.
URI
https://scholars.duke.edu/individual/pub1554127
PMID
36266675
Source
pubmed
Published In
Journal of Ovarian Research
Volume
15
Published Date
Start Page
114
DOI
10.1186/s13048-022-01046-5
Endometrial cancer
The prior dichotomous classification of endometrial cancer into high and low risk subtypes based primarily on histology has been shown to be inadequate in predicting outcome and treatment response. More nuanced molecular categorization of these cancers provides improved targeted therapeutic options and a better understanding of the different disease processes.
Authors
Spinosa, D; Montes de Oca, MK; Watson, CH; Berchuck, A; Previs, RA
MLA Citation
Spinosa, D., et al. “Endometrial cancer.” Genomic and Precision Medicine: Oncology, Third Edition, 2022, pp. 191–203. Scopus, doi:10.1016/B978-0-12-800684-9.00014-9.
URI
https://scholars.duke.edu/individual/pub1550798
Source
scopus
Published Date
Start Page
191
End Page
203
DOI
10.1016/B978-0-12-800684-9.00014-9

Adjunct Assistant Professor in the Department of Obstetrics and Gynecology
Contact:
Division of Gynecologic Oncolo, DUMC Box 3079, Durham, NC 27710