Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katherine V. Ferry-Galow is active.

Publication


Featured researches published by Katherine V. Ferry-Galow.


Clinical Cancer Research | 2016

Phase I Safety, Pharmacokinetic, and Pharmacodynamic Study of the Poly(ADP-ribose) Polymerase (PARP) Inhibitor Veliparib (ABT-888) in Combination with Irinotecan in Patients with Advanced Solid Tumors

Patricia LoRusso; Jing Li; Angelika Burger; Lance K. Heilbrun; Edward A. Sausville; Scott A. Boerner; Daryn Smith; Mary Jo Pilat; Jie Zhang; Sara M. Tolaney; James M. Cleary; Alice P. Chen; Lawrence Rubinstein; Julie L. Boerner; Adam Bowditch; Dongpo Cai; Tracy Bell; Andrew Wolanski; Allison Marrero; Yiping Zhang; Jiuping Ji; Katherine V. Ferry-Galow; Robert J. Kinders; Geoffrey I. Shapiro

Purpose: PARP is essential for recognition and repair of DNA damage. In preclinical models, PARP inhibitors modulate topoisomerase I inhibitor–mediated DNA damage. This phase I study determined the MTD, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics (PD) of veliparib, an orally bioavailable PARP1/2 inhibitor, in combination with irinotecan. Experimental Design: Patients with advanced solid tumors were treated with 100 mg/m2 irinotecan on days 1 and 8 of a 21-day cycle. Twice-daily oral dosing of veliparib (10–50 mg) occurred on days 3 to 14 (cycle 1) and days −1 to 14 (subsequent cycles) followed by a 6-day rest. PK studies were conducted with both agents alone and in combination. Paired tumor biopsies were obtained after irinotecan alone and veliparib/irinotecan to evaluate PARP1/2 inhibition and explore DNA damage signals (nuclear γ-H2AX and pNBS1). Results: Thirty-five patients were treated. DLTs included fatigue, diarrhea, febrile neutropenia, and neutropenia. The MTD was 100 mg/m2 irinotecan (days 1 and 8) combined with veliparib 40 mg twice daily (days −1–14) on a 21-day cycle. Of 31 response-evaluable patients, there were six (19%) partial responses. Veliparib exhibited linear PK, and there were no apparent PK interactions between veliparib and irinotecan. At all dose levels, veliparib reduced tumor poly(ADP-ribose) (PAR) content in the presence of irinotecan. Several samples showed increases in γ-H2AX and pNBS1 after veliparib/irinotecan compared with irinotecan alone. Conclusions: Veliparib can be safely combined with irinotecan at doses that inhibit PARP catalytic activity. Preliminary antitumor activity justifies further evaluation of the combination. Clin Cancer Res; 22(13); 3227–37. ©2016 AACR.


Clinical Cancer Research | 2017

Efficacy of the PARP inhibitor veliparib with carboplatin or as a single agent in patients with germline BRCA1- or BRCA2-associated metastatic breast cancer: California Cancer Consortium trial NCT01149083

George Somlo; Paul Frankel; Banu Arun; Cynthia X. Ma; Agustin A. Garcia; Tessa Cigler; Leah Cream; Harold A. Harvey; Joseph A. Sparano; Rita Nanda; Helen K. Chew; Timothy J. Moynihan; Linda T. Vahdat; Matthew P. Goetz; Jan H. Beumer; Arti Hurria; Joanne E. Mortimer; Richard Piekarz; Sharon Sand; Josef Herzog; Lily Ravel Van Tongeren; Katherine V. Ferry-Galow; Alice P. Chen; Christopher Ruel; Edward M. Newman; David R. Gandara; Jeffrey N. Weitzel

Purpose: We aimed to establish the MTD of the poly (ADP-ribose) (PAR) polymerase inhibitor, veliparib, in combination with carboplatin in germline BRCA1- and BRCA2- (BRCA)-associated metastatic breast cancer (MBC), to assess the efficacy of single-agent veliparib, and of the combination treatment after progression, and to correlate PAR levels with clinical outcome. Experimental Design: Phase I patients received carboplatin (AUC of 5–6, every 21 days), with escalating doses (50-20 mg) of oral twice-daily (BID) veliparib. In a companion phase II trial, patients received single-agent veliparib (400 mg BID), and upon progression, received the combination at MTD. Peripheral blood mononuclear cell PAR and serum veliparib levels were assessed and correlated with outcome. Results: Twenty-seven phase I trial patients were evaluable. Dose-limiting toxicities were nausea, dehydration, and thrombocytopenia [MTD: veliparib 150 mg po BID and carboplatin (AUC of 5)]. Response rate (RR) was 56%; 3 patients remain in complete response (CR) beyond 3 years. Progression-free survival (PFS) and overall survival (OS) were 8.7 and 18.8 months. The PFS and OS were 5.2 and 14.5 months in the 44 patients in the phase II trial, with a 14% RR in BRCA1 (n = 22) and 36% in BRCA2 (n = 22). One of 30 patients responded to the combination therapy after progression on veliparib. Higher baseline PAR was associated with clinical benefit. Conclusions: Safety and efficacy are encouraging with veliparib alone and in combination with carboplatin in BRCA-associated MBC. Lasting CRs were observed when the combination was administered first in the phase I trial. Further investigation of PAR level association with clinical outcomes is warranted. Clin Cancer Res; 23(15); 4066–76. ©2017 AACR.


PLOS ONE | 2017

Antibody-independent capture of circulating tumor cells of non-epithelial origin with the ApoStream® system

Priya Balasubramanian; Robert J. Kinders; Shivaani Kummar; Vishal Gupta; David K. Hasegawa; Anoop Menachery; Scott M. Lawrence; Lihua Wang; Katherine V. Ferry-Galow; Darren W. Davis; Ralph E. Parchment; Joseph E. Tomaszewski; James H. Doroshow; Joseph Najbauer

Circulating tumor cells (CTCs) are increasingly employed for research and clinical monitoring of cancer, though most current methods do not permit the isolation of non-epithelial tumor cells. Furthermore, CTCs isolated with antibody-dependent methods are not suitable for downstream experimental uses, including in vitro culturing and implantation in vivo. In the present study, we describe the development, validation, and transfer across laboratories of a new antibody-independent device for the enrichment of CTCs from blood samples of patients with various cancer diagnoses. The ApoStream® device uses dielectrophoresis (DEP) field-flow assist to separate non-hematopoietic cells from the peripheral blood mononuclear fraction by exposing cells in a laminar flow stream to a critical alternating current frequency. The ApoStream® device was calibrated and validated in a formal cross-laboratory protocol using 3 different cancer cell lines spanning a range of distinct phenotypes (A549, MDA-MB-231, and ASPS-1). In spike-recovery experiments, cancer cell recovery efficiencies appeared independent of spiking level and averaged between 68% and 55%, depending on the cell line. No inter-run carryover was detected in control samples. Moreover, the clinical-readiness of the device in the context of non-epithelial cancers was evaluated with blood specimens from fifteen patients with metastatic sarcoma. The ApoStream® device successfully isolated CTCs from all patients with sarcomas examined, and the phenotypic heterogeneity of the enriched cells was demonstrated by fluorescence in situ hybridization or with multiplex immunophenotyping panels. Therefore, the ApoStream® technology expands the clinical utility of CTC evaluation to mesenchymal cancers.


Clinical Cancer Research | 2016

Phase I safety, pharmacokinetic and pharmacodynamic study of the poly (ADP-ribose) polymerase inhibitor veliparib with irinotecan in patients with advanced tumors

Patricia LoRusso; Jing Li; Angelika Burger; Lance K. Heilbrun; Edward A. Sausville; Scott Boerner; Daryn Smith; Mary Jo Pilat; Jie Zhang; Sara M. Tolaney; James M. Cleary; Alice Chen; Lawrence Rubinstein; Julie L. Boerner; Adam Bowditch; Dongpo Cai; Tracy Bell; Andrew Wolanski; Allison Marrero; Yiping Zhang; Jiuping Ji; Katherine V. Ferry-Galow; Robert J. Kinders; Ralph E. Parchment; Geoffrey I. Shapiro

Purpose: PARP is essential for recognition and repair of DNA damage. In preclinical models, PARP inhibitors modulate topoisomerase I inhibitor–mediated DNA damage. This phase I study determined the MTD, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics (PD) of veliparib, an orally bioavailable PARP1/2 inhibitor, in combination with irinotecan. Experimental Design: Patients with advanced solid tumors were treated with 100 mg/m2 irinotecan on days 1 and 8 of a 21-day cycle. Twice-daily oral dosing of veliparib (10–50 mg) occurred on days 3 to 14 (cycle 1) and days −1 to 14 (subsequent cycles) followed by a 6-day rest. PK studies were conducted with both agents alone and in combination. Paired tumor biopsies were obtained after irinotecan alone and veliparib/irinotecan to evaluate PARP1/2 inhibition and explore DNA damage signals (nuclear γ-H2AX and pNBS1). Results: Thirty-five patients were treated. DLTs included fatigue, diarrhea, febrile neutropenia, and neutropenia. The MTD was 100 mg/m2 irinotecan (days 1 and 8) combined with veliparib 40 mg twice daily (days −1–14) on a 21-day cycle. Of 31 response-evaluable patients, there were six (19%) partial responses. Veliparib exhibited linear PK, and there were no apparent PK interactions between veliparib and irinotecan. At all dose levels, veliparib reduced tumor poly(ADP-ribose) (PAR) content in the presence of irinotecan. Several samples showed increases in γ-H2AX and pNBS1 after veliparib/irinotecan compared with irinotecan alone. Conclusions: Veliparib can be safely combined with irinotecan at doses that inhibit PARP catalytic activity. Preliminary antitumor activity justifies further evaluation of the combination. Clin Cancer Res; 22(13); 3227–37. ©2016 AACR.


Archive | 2018

Integrating Biomarkers in Early-Phase Trials

Katherine V. Ferry-Galow; James H. Doroshow

Abstract The clinical success of a targeted cancer therapy is associated with clinical proof of mechanism (PoM): demonstration of the molecular mechanism of action (MoA) of an agent in patients. Fundamental to assessing MoA hypotheses in early-phase clinical trials is measurement of molecular pharmacodynamic (PD) biomarkers, which are altered in response to drug. Here, we describe how PD biomarkers can be used for clinical PoM, outlining considerations for biomarker and assay selection, development, and clinical readiness, including the importance of specimen quality and use of surrogate tissues. We also explore additional clinical applications for PD biomarkers: molecular response–based dosage regimen optimization, design of combination therapies, and development of companion diagnostic biomarkers. We emphasize the incorporation of PD assessment into early clinical trial design, providing a breakdown of PD biomarker data that can be realistically obtained at each trial phase and a framework for constructing clinical protocols to include PD biomarker studies.


Journal of Oncology Practice | 2018

What Can Be Done to Improve Research Biopsy Quality in Oncology Clinical Trials

Katherine V. Ferry-Galow; Vivekananda Datta; Hala Makhlouf; John J. Wright; Bradford J. Wood; Elliot Levy; Etta D. Pisano; Alda L. Tam; Susanna I. Lee; Umar Mahmood; Lawrence Rubinstein; James H. Doroshow; Alice P. Chen

PURPOSE: Research biopsy specimens collected in clinical trials often present requirements beyond those of tumor biopsy specimens collected for diagnostic purposes. Research biopsies underpin hypothesis-driven drug development, pharmacodynamic assessment of molecularly targeted anticancer agents, and, increasingly, genomic assessment for precision medicine; insufficient biopsy specimen quality or quantity therefore compromises the scientific value of a study and the resources devoted to it, as well as each patients contribution to and potential benefit from a clinical trial. METHODS: To improve research biopsy specimen quality, we consulted with other translational oncology teams and reviewed current best practices. RESULTS: Among the recommendations were improving communication between oncologists and interventional radiologists, providing feedback on specimen sufficiency, increasing academic recognition and financial support for the time investment required by radiologists to collect and preserve research biopsy specimens, and improving real-time assessment of tissue quality. CONCLUSION: Implementing these recommendations at the National Cancer Institutes Developmental Therapeutics Clinic has demonstrably improved the quality of biopsy specimens collected; more widespread dissemination of these recommendations beyond large clinical cancer centers is possible and will be of value to the community in improving clinical research and, ultimately, patient care.


Cancer Research | 2018

Abstract 4537: Cryogenic biopsy device for preservation of labile biomarkers

Erez Nevo; Melinda G. Hollingshead; Katherine V. Ferry-Galow; Jeevan Prasaad Govindharajulu; Shoshan Nevo; Kay D. Gray; Ralph E. Parchment; James H. Doroshow; Apurva K. Srivastava

Background: Preanalytical factors such as the handling of biospecimens between resection and analysis can have a significant impact on assay results which may adversely affect patient care. Global phosphoprotein analysis indicates that a 5 minute delay in time-to-stabilization (ischemia time) can impact 20% of phospho-serine and 50% of phospho-tyrosine residues in kinases. Consequently, inadequate tumor biopsy processing is a significant impediment to the use of phosphorylated biomarkers in oncology. We aim to develop new biopsy devices and methodologies to stabilize highly dynamic biomarkers and minimize the influence of preanalytical factors. Methods: A cryobiopsy device (CBD) was developed by integrating a 19G cryoablation needle with a 16G cutting cannula of a clinical biopsy device. The device enables in-situ freezing of the tissue sample before it is cut and removed from the tumor. We measured the stabilization of a highly labile biomarker, phosphorylated-MET (pMET), to compare between percutaneous biopsy samples acquired by the CBD (zero ischemic time) and by a clinical grade 18G biopsy device (Temno) (10-20 second ischemic time). The animal protocol was approved by the FNLCR9s IACUC. Athymic nude mice (nu/nu NCr) were implanted with human gastric carcinoma cell line SNU-5. Tumors were biopsied 3 weeks after implantation (tumor size range 150-250mm3). The needle was passed through the skin into the tumor, a tissue sample was collected, and the biopsy needle was retracted. The tissue sample of the Temno device was rapidly frozen in a cryovial that was precooled in liquid nitrogen [1]. The already frozen tissue sample of the CBD was rapidly transferred to a second precooled cryovial. The vials were sealed and returned to liquid nitrogen. Frozen specimens were stored at −80°C until processing. Each tumor was biopsied once and the animal was euthanized. The methodology to determine the pMET/MET levels in the biopsy samples is detailed in [2]. Results: The pMET/MET ratio was not significantly different between samples acquired by the CBD and by the Temno device (mean±st dev): pY1234/35-MET/MET ratio: CBD 0.75±0.11; Temno 0.71±0.10 pY1356-MET/MET ratio: CBD 0.56±0.08; Temno 0.50±0.06 pY1235-MET/MET ratio: CBD 0.90±0.15; Temno 0.83±0.14 Conclusions: Past studies showed that tissue levels of highly labile biomarkers like pMET can change significantly in 1-2 minutes from the time of tissue harvesting [2]. The current study demonstrates that rapid freezing of the tissue within less than 20 seconds from tissue harvesting maintains the in-vivo levels of pMET similarly to tissue freezing before tissue harvesting. This result suggests that devices that rapidly freeze tissue samples after removal from the body, which are more suitable for clinical use, may be sufficient for preservation and accurate quantitation of highly labile biomarkers. 1. PMID: 18980982 2. PMID: 27001313 Supported by NCI Contracts HHSN261200800001E and HHSN261201200050C Citation Format: Erez Nevo, Melinda G. Hollingshead, Katherine Ferry-Galow, Jeevan P. Govindharajulu, Shoshan Nevo, Kay D. Gray, Ralph E. Parchment, James H. Doroshow, Apurva K. Srivastava. Cryogenic biopsy device for preservation of labile biomarkers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4537.


Cancer Research | 2017

Abstract 4678: Pilot trial of talazoparib (BMN 673), an oral PARP inhibitor, in patients with advanced solid tumors carrying deleterious BRCA mutations

Robert S. Meehan; Alice P. Chen; Geraldine O’Sullivan Coyne; Shivaani Kummar; Jiuping Ji; Rasa Vilimas; Lamin Juwara; Robert J. Kinders; Katherine V. Ferry-Galow; Deborah Wilsker; Yiping Zhang; Angie B. Dull; Tony Navas; Lihua Wang; Ralph E. Parchment; James H. Doroshow

Inhibition of poly (ADP-ribose) polymerase (PARP) sensitizes tumor cells to DNA damage that would normally be repaired through the base excision repair pathway. PARP inhibitors are active clinically against BRCA-deficient ovarian cancers. The PARP inhibitor talazoparib produces cytotoxicity in human cancer cell lines and animal models of tumors that harbor mutations that compromise DNA repair pathways. In this study, single agent talazoparib (1000 µg/day) was administered to patients with deleterious BRCA1 or BRCA2 mutations and advanced solid tumors in 28 day cycles. The primary objective of the trial was to examine pharmacodynamic (PD) effects of talazoparib; the secondary objective was to determine response rate in patients whose tumors carry BRCA mutations. Mandatory paired tumor biopsies were obtained pre-treatment and 3-6 hrs post-treatment on cycle 1 day 8. Optional biopsies were collected at the time of progression. One core from each time point was analyzed for PARP inhibition by a validated ELISA assay while the other core was used for IFA analysis of γH2AX. A total of 9 patients (pts) were enrolled and treated before this trial was closed due to lack of drug availability: [prostate (3), ovarian (2), breast (2), uterine sarcoma (1), pancreatic (1)]. Median age was 63 (range: 33-73 yrs); male-to-female ratio was 4:5; and the median number of prior treatments was 6 (range: 1-12). All 9 pts were evaluable for PD endpoints. One pt progressed during the first cycle of treatment; 8 pts were evaluable for clinical response. Mean time on study for evaluable pts was 8 cycles (range: 2-18); 5 of 8 (62%) pts experienced a documented partial responses [ovarian (2), prostate (2), breast (1)] lasting between 4 and 12 cycles (median: 6 cycles). Two pts had stable disease for 4 to 6 cycles, and one progressed after 2 cycles. The agent was well tolerated; the most frequent adverse events were hematologic including grade (gr) 4 anemia (1) and thrombocytopenia (1), and gr 3 anemia (2), neutropenia (1), lymphopenia (1). Decreases in PAR levels (>75%) were observed in all cycle 1 day 8 biopsy pairs, documenting a primary PD effect. Increased γH2AX expression was observed for 4/6 pts in post-dose biopsies; pre-treatment γH2AX levels, measured as %Nuclear Associated Protein (NAP), increased from a mean ± SD of 1.33 ± 1.08 to a post-treatment %NAP mean of 5.60 ± 0.78; (p=0.018), supporting a role for drug-enhanced DNA double strand breaks in the mechanism of action of talazoparib for BRCA mutant tumors. In summary, talazoparib demonstrated significant clinical activity as a single agent in patients with BRCA-deficient tumors and produced substantial reductions in tumor PAR levels in matched pre and post-treatment tumor biopsies. Citation Format: Robert S. Meehan, Alice P. Chen, Geraldine O’Sullivan Coyne, Shivaani Kummar, Jiuping Ji, Rasa Vilimas, Lamin Juwara, Robert J. Kinders, Katherine Ferry-Galow, Deborah Wilsker, Yiping Zhang, Angie B. Dull, Tony Navas, Lihua Wang, Ralph E. Parchment, James H. Doroshow. Pilot trial of talazoparib (BMN 673), an oral PARP inhibitor, in patients with advanced solid tumors carrying deleterious BRCA mutations [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 4678. doi:10.1158/1538-7445.AM2017-4678


Seminars in Oncology | 2016

The root causes of pharmacodynamic assay failure

Katherine V. Ferry-Galow; Hala R. Makhlouf; Deborah Wilsker; Scott M. Lawrence; Thomas D. Pfister; Allison M. Marrero; Kristina M. Bigelow; William Yutzy; Jiuping J. Ji; Donna Butcher; Brad A. Gouker; Shivaani Kummar; Alice P. Chen; Robert J. Kinders; James H. Doroshow

Robust pharmacodynamic assay results are valuable for informing go/no-go decisions about continued development of new anti-cancer agents and for identifying combinations of targeted agents, but often pharmacodynamic results are too incomplete or variable to fulfill this role. Our experience suggests that variable reagent and specimen quality are two major contributors to this problem. Minimizing all potential sources of variability in procedures for specimen collection, processing, and assay measurements is essential for meaningful comparison of pharmacodynamic biomarkers across sample time points. This is especially true in the evaluation of pre- and post-dose tumor biopsies, which suffer from high levels of tumor insufficiency due to variations in biopsy collection techniques and significant specimen heterogeneity within and across patients. Developing methods to assess heterogeneous biopsies is necessary in order to evaluate a majority of tumor biopsies collected for pharmacodynamic biomarker studies. Improved collection devices and standardization of methods are being sought in order to improve the tumor content and quality of tumor biopsies. In terms of reagent variability, we have found that stringent initial reagent qualification and quality control of R&D-grade reagents is critical to minimize lot-to-lot variability and prevent assay failures, especially for clinical pharmacodynamic questions, which often demand assay performance that meets or exceeds clinical diagnostic assay standards. Rigorous reagent specifications and use of appropriate assay quality control methodologies help to ensure consistency between assay runs, laboratories and trials to provide much needed pharmacodynamic insights into the activity of investigational agents.


Clinical Cancer Research | 2016

Phase I safety, pharmacokinetic and pharmacodynamic study of the poly (ADP-ribose) polymerase inhibitor veliparib with irinotecan in patients with advanced solid tumors

Patricia LoRusso; Jing Li; Angelika Burger; Lance K. Heilbrun; Edward A. Sausville; Scott Boerner; Daryn Smith; Mary Jo Pilat; Jie Zhang; Sara M. Tolaney; James M. Cleary; Alice Chen; Lawrence Rubinstein; Julie L. Boerner; Adam Bowditch; Dongpo Cai; Tracy Bell; Andrew Wolanski; Allison Marrero; Yiping Zhang; Jiuping Ji; Katherine V. Ferry-Galow; Robert J. Kinders; Ralph E. Parchment; Geoffrey I. Shapiro

Purpose: PARP is essential for recognition and repair of DNA damage. In preclinical models, PARP inhibitors modulate topoisomerase I inhibitor–mediated DNA damage. This phase I study determined the MTD, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics (PD) of veliparib, an orally bioavailable PARP1/2 inhibitor, in combination with irinotecan. Experimental Design: Patients with advanced solid tumors were treated with 100 mg/m2 irinotecan on days 1 and 8 of a 21-day cycle. Twice-daily oral dosing of veliparib (10–50 mg) occurred on days 3 to 14 (cycle 1) and days −1 to 14 (subsequent cycles) followed by a 6-day rest. PK studies were conducted with both agents alone and in combination. Paired tumor biopsies were obtained after irinotecan alone and veliparib/irinotecan to evaluate PARP1/2 inhibition and explore DNA damage signals (nuclear γ-H2AX and pNBS1). Results: Thirty-five patients were treated. DLTs included fatigue, diarrhea, febrile neutropenia, and neutropenia. The MTD was 100 mg/m2 irinotecan (days 1 and 8) combined with veliparib 40 mg twice daily (days −1–14) on a 21-day cycle. Of 31 response-evaluable patients, there were six (19%) partial responses. Veliparib exhibited linear PK, and there were no apparent PK interactions between veliparib and irinotecan. At all dose levels, veliparib reduced tumor poly(ADP-ribose) (PAR) content in the presence of irinotecan. Several samples showed increases in γ-H2AX and pNBS1 after veliparib/irinotecan compared with irinotecan alone. Conclusions: Veliparib can be safely combined with irinotecan at doses that inhibit PARP catalytic activity. Preliminary antitumor activity justifies further evaluation of the combination. Clin Cancer Res; 22(13); 3227–37. ©2016 AACR.

Collaboration


Dive into the Katherine V. Ferry-Galow's collaboration.

Top Co-Authors

Avatar

James H. Doroshow

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alice P. Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ralph E. Parchment

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yiping Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hala Makhlouf

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lawrence Rubinstein

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge