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:
Validation of the Labcorp Plasma Focus Test to Facilitate Precision Oncology Through Cell-Free DNA Genomic Profiling of Solid Tumors.
Genomic profiling is critical for precision oncology to guide treatment decisions. Liquid biopsy testing is a complementary approach to tissue testing, particularly when tissue is not readily available. The Labcorp Plasma Focus test is a circulating cell-free DNA genomic profiling test that identifies actionable variants in solid cancers, including non-small-cell lung, colorectal, melanoma, breast, esophageal, gastroesophageal junction, and gastric cancers. This study highlights the analytical validation of the test, including accuracy compared with orthogonal methods, as well as sensitivity, specificity, precision, reproducibility, and repeatability. Concordance with orthogonal methods showed percent positive agreement of 98.7%, 89.3%, and 96.2% for single nucleotide variants (SNVs), insertion/deletions (indels), and copy number amplifications (CNAs), respectively, and 100.0% for translocations and microsatellite instability (MSI). Analytical sensitivity revealed a median limit of detection of 0.7% and 0.6% for SNVs and indels, 1.4-fold for CNAs, 0.5% variant allele frequency for translocations, and 0.6% for MSI. Specificity was >99% for SNVs/indels and 100% for CNAs, translocations, and MSI. Average positive agreement from precision, reproducibility, and repeatability experiments was 97.5% and 88.9% for SNVs/indels and CNAs, and 100% for translocations and MSI. Taken together, these data show that the Labcorp Plasma Focus test is a highly accurate, sensitive, and specific approach for cell-free DNA genomic profiling to supplement tissue testing and inform treatment decisions.
Authors
Verner, EL; Jackson, JB; Severson, E; Valkenburg, KC; Greer, AE; Riley, DR; Sausen, M; Maddox, C; McGregor, PM; Karandikar, A; Hastings, SB; Previs, RA; Reddy, VP; Jensen, TJ; Ramkissoon, SH
MLA Citation
Verner, Ellen L., et al. “Validation of the Labcorp Plasma Focus Test to Facilitate Precision Oncology Through Cell-Free DNA Genomic Profiling of Solid Tumors.” J Mol Diagn, vol. 25, no. 7, July 2023, pp. 477–89. Pubmed, doi:10.1016/j.jmoldx.2023.03.008.
URI
https://scholars.duke.edu/individual/pub1586211
PMID
37068734
Source
pubmed
Published In
J Mol Diagn
Volume
25
Published Date
Start Page
477
End Page
489
DOI
10.1016/j.jmoldx.2023.03.008
Targeting CaMKK2 inhibits actin cytoskeletal assembly to suppress cancer metastasis.
Triple-negative breast cancers (TNBCs) tend to become invasive and metastatic at early stages in their development. Despite some treatment successes in early stage localized TNBC, the rate of distant recurrence remains high, and long-term survival outcomes remain poor. In a search for new therapeutic targets for this disease, we observed that elevated expression of the serine/threonine kinase calcium/calmodulin (CaM)-dependent protein kinase kinase 2 (CaMKK2) is highly correlated with tumor invasiveness. In validation studies, genetic disruption of CaMKK2 expression or inhibition of its activity with small molecule inhibitors disrupted spontaneous metastatic outgrowth from primary tumors in murine xenograft models of TNBC. High-grade serous ovarian cancer (HGSOC), a high-risk, poor prognosis ovarian cancer subtype, shares many features with TNBC, and CaMKK2 inhibition effectively blocked metastatic progression in a validated xenograft model of this disease. Mechanistically, CaMKK2 increased the expression of the phosphodiesterase PDE1A which hydrolyzed cyclic guanosine monophosphate (cGMP) to decrease the cGMP-dependent activity of protein kinase G1 (PKG1). Inhibition of PKG1 resulted in decreased phosphorylation of vasodilator stimulated phosphoprotein (VASP), which in its hypophosphorylated state binds to and regulates F-actin assembly to facilitate cell movement. Together, these findings establish a targetable CaMKK2-PDE1A-PKG1-VASP signaling pathway that controls cancer cell motility and metastasis by impacting the actin cytoskeleton. Further, it identifies CaMKK2 as a potential therapeutic target that can be exploited to restrict tumor invasiveness in patients diagnosed with early-stage TNBC or localized HGSOC.
Authors
Mukherjee, D; Previs, RA; Haines, C; Al Abo, M; Juras, PK; Strickland, KC; Chakraborty, B; Artham, S; Whitaker, RS; Hebert, K; Fontenot, J; Patierno, SR; Freedman, JA; Lau, FH; Burow, ME; Chang, C-Y; McDonnell, DP
MLA Citation
Mukherjee, Debarati, et al. “Targeting CaMKK2 inhibits actin cytoskeletal assembly to suppress cancer metastasis.” Cancer Res, June 2023. Pubmed, doi:10.1158/0008-5472.CAN-22-1622.
URI
https://scholars.duke.edu/individual/pub1583916
PMID
37335130
Source
pubmed
Published In
Cancer Res
Published Date
DOI
10.1158/0008-5472.CAN-22-1622
Association of Genomic Instability Score, Tumor Mutational Burden, and Tumor-Infiltrating Lymphocytes as Biomarkers in Uterine Serous Carcinoma.
Background: Uterine serous carcinomas represent 10% of uterine carcinomas but account for nearly 40% of deaths from the disease. Improved molecular characterization of these tumors is instrumental in guiding targeted treatment and improving outcomes. This study assessed the genomic instability score (GIS), tumor mutational burden (TMB), and tumor-infiltrating lymphocytes (TILs) in patients with USC. Methods: A retrospective cohort study evaluated patients with USC following staging surgery. The GIS and TMB were determined from archived specimens. We evaluated the tumoral expression of CD3, CD4, CD8, FOXP3, and CD68 using immunohistochemistry. T-tests were used to assess associations of TILs with the GIS. Results: We evaluated 53 patients with USC. The median GIS was 31 (range: 0−52) and a higher GIS was not associated with progression-free (PFS) or overall survival (OS). The median TMB was 1.35 mt/Mb; patients with TMB > 1.35 mt/Mb had improved PFS and OS (p = 0.005; p = 0.002, respectively). Tumors with increased CD3+ and CD4+ immune cells had a higher mean GIS (p = 0.013, p = 0.002). Conclusions: TMB > 1.35 mt/Mb was associated with improved survival in USC patients, whereas the GIS was not. Lower TMB thresholds may provide prognostic value for less immunogenic tumors such as USC. In this limited cohort, we observed that increased TIL populations were correlated with a higher GIS.
Authors
Bloom, EA; Peters, PN; Whitaker, R; Russell, S; Albright, B; Cummings, S; Timms, KM; Slavin, T; Probst, B; Strickland, KC; Previs, RA
MLA Citation
Bloom, Elizabeth A., et al. “Association of Genomic Instability Score, Tumor Mutational Burden, and Tumor-Infiltrating Lymphocytes as Biomarkers in Uterine Serous Carcinoma.” Cancers (Basel), vol. 15, no. 2, Jan. 2023. Pubmed, doi:10.3390/cancers15020528.
URI
https://scholars.duke.edu/individual/pub1563293
PMID
36672477
Source
pubmed
Published In
Cancers
Volume
15
Published Date
DOI
10.3390/cancers15020528
Telemedicine and gynecologic oncology: caring for patients remotely during a global pandemic.
BACKGROUND: Telemedicine uses technology to deliver medical care remotely and has been shown to provide similar patient satisfaction and care outcomes compared with in-person visits. OBJECTIVE: This study aimed to assess the gynecologic oncology patient telehealth experience. STUDY DESIGN: All patients receiving telehealth care between March 23, 2020, to May 14, 2020, from a single institution's gynecologic oncology division were offered postvisit surveys to assess satisfaction. Basic demographic and clinical data were collected and analyzed with descriptive statistics. Patient zip code data were correlated with Community Need Index scores and visualized using heat maps. RESULTS: Of 286 telehealth visits, 112 postvisit surveys (39.2%) were collected. Survey responses demonstrated high patient satisfaction with responders agreeing that privacy was respected (97.3%), diagnosis and treatment options were adequately explained (92%), they could easily ask questions (97.3%), and they established a good rapport with their provider (96.4%). Additional benefits included reduced travel (92.9%), time (83.0%), cost (67.9%), and family interruption (57.1%). Among 11 patients receiving treatment on a clinical trial, 10 (90.9%) were able to continue on trial without disruption. Most responders (87.5%) preferred future visits to occur via telehealth or a mixture of telehealth and in-person visits. No difference in satisfaction was found among patients residing in zip codes associated with higher Community Need Index scores or increased distance from the institution. CONCLUSION: The use of telemedicine in providing gynecologic oncology care was associated with high patient satisfaction and had the benefits of reduced time, cost, travel, and interruption to family time.
Authors
MLA Citation
Wong, Janice, et al. “Telemedicine and gynecologic oncology: caring for patients remotely during a global pandemic.” Ajog Glob Rep, vol. 2, no. 4, Nov. 2022, p. 100124. Pubmed, doi:10.1016/j.xagr.2022.100124.
URI
https://scholars.duke.edu/individual/pub1558036
PMID
36451896
Source
pubmed
Published In
Ajog Global Reports
Volume
2
Published Date
Start Page
100124
DOI
10.1016/j.xagr.2022.100124
RNA Sequencing Identifies Novel NRG1 Fusions in Solid Tumors that Lack Co-Occurring Oncogenic Drivers.
NRG1 gene fusions are rare, therapeutically relevant, oncogenic drivers that occur across solid tumor types. To understand the landscape of NRG1 gene fusions, 4397 solid tumor formalin-fixed, paraffin-embedded samples consecutively tested by comprehensive genomic and immune profiling during standard care were analyzed. Nineteen NRG1 fusions were found in 17 unique patients, across multiple tumor types, including non-small-cell lung (n = 7), breast (n = 2), colorectal (n = 3), esophageal (n = 2), ovarian (n = 1), pancreatic (n = 1), and unknown primary (n = 1) carcinomas, with a cumulative incidence of 0.38%. Fusions were identified with breakpoints across four NRG1 introns spanning 1.4 megabases, with a mixture of known (n = 8) and previously unreported (n = 11) fusion partners. Co-occurring driver alterations in tumors with NRG1 fusions were uncommon, except colorectal carcinoma, where concurrent alterations in APC, BRAF, and ERBB2 were present in a subset of cases. The overall lack of co-occurring drivers highlights the importance of identifying NRG1 gene fusions, as these patients are unlikely to harbor other targetable alterations. In addition, RNA sequencing is important to identify NRG1 gene fusions given the variety of fusion partners and large genomic areas where breakpoints can occur.
Authors
Severson, E; Achyut, BR; Nesline, M; Pabla, S; Previs, RA; Kannan, G; Chenn, A; Zhang, S; Klein, R; Conroy, J; Sausen, M; Sathyan, P; Saini, KS; Ghosh, A; Jensen, TJ; Reddy, P; Ramkissoon, SH
MLA Citation
Severson, Eric, et al. “RNA Sequencing Identifies Novel NRG1 Fusions in Solid Tumors that Lack Co-Occurring Oncogenic Drivers.” J Mol Diagn, vol. 25, no. 7, July 2023, pp. 454–66. Pubmed, doi:10.1016/j.jmoldx.2023.03.011.
URI
https://scholars.duke.edu/individual/pub1575133
PMID
37164276
Source
pubmed
Published In
J Mol Diagn
Volume
25
Published Date
Start Page
454
End Page
466
DOI
10.1016/j.jmoldx.2023.03.011

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