Network


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

Hotspot


Dive into the research topics where Jasgit C. Sachdev is active.

Publication


Featured researches published by Jasgit C. Sachdev.


Cancer Research | 2016

Abstract S5-07: Preliminary efficacy and safety of pembrolizumab (MK-3475) in patients with PD-L1–positive, estrogen receptor-positive (ER+)/HER2-negative advanced breast cancer enrolled in KEYNOTE-028

Hope S. Rugo; J-P Delord; S-A Im; Patrick A. Ott; Sarina Anne Piha-Paul; Philippe L. Bedard; Jasgit C. Sachdev; C. Le Tourneau; E. van Brummelen; A. Varga; Sanatan Saraf; Dina Pietrangelo; Vassiliki Karantza; Antoinette R. Tan

Background: The programmed cell death 1 (PD-1) pathway is used by tumors to evade immune surveillance. Pembrolizumab is a humanized anti–PD-1 monoclonal antibody that blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2. Pembrolizumab has shown robust antitumor activity against several advanced malignancies, including triple-negative breast cancer. We assessed the safety and efficacy of pembrolizumab in patients with PD-L1–positive, ER+/HER2-negative advanced breast cancer. Methods: KEYNOTE-028 (ClinicalTrials.gov, NCT02054806) is an ongoing multicohort, open-label phase 1b study evaluating the safety and efficacy of pembrolizumab in patients with PD-L1–positive advanced solid tumors. Key eligibility criteria for this cohort include ER+ and HER2-negative tumor status defined by institutional standards, locally advanced or metastatic disease, ECOG performance status of 0 or 1, failure of or inability to receive standard therapy, and PD-L1 expression in stroma or in ≥1% of tumor cells as assessed at a central laboratory using a prototype immunohistochemistry assay with the 22C3 antibody (Merck). Pembrolizumab was administered at a dose of 10 mg/kg every 2 weeks for up to 24 months or until confirmed progression or intolerable toxicity. Response is based on RECIST v1.1 as assessed by investigator review every 8 weeks for the first 6 months and every 12 weeks thereafter. Primary efficacy end point is overall response rate (ORR). Results: Of the 248 patients with ER+/HER2-negative breast cancer who had evaluable tumor samples screened for PD-L1 expression, 48 (19%) had PD-L1–positive tumors. Of these, 25 patients were enrolled. Median age was 53 years (range, 36-79), and 44% of patients had an ECOG performance status of 1. Patients were heavily pretreated, with 76% having received ≥3 prior lines of therapy for advanced disease, including 48.0% who received ≥5 prior lines. Analyses of ORR, duration of response, and adverse events are ongoing and will be completed by September 4, 2015. Conclusion: Data from this KEYNOTE-028 cohort will provide information on the antitumor activity and safety of pembrolizumab in patients with heavily pretreated, PD-L1–positive, ER+/HER2-negative advanced breast cancer. Citation Format: Rugo HS, Delord J-P, Im S-A, Ott PA, Piha-Paul SA, Bedard PL, Sachdev J, Le Tourneau C, van Brummelen E, Varga A, Saraf S, Pietrangelo D, Karantza V, Tan A. Preliminary efficacy and safety of pembrolizumab (MK-3475) in patients with PD-L1–positive, estrogen receptor-positive (ER+)/HER2-negative advanced breast cancer enrolled in KEYNOTE-028. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr S5-07.


Clinical Cancer Research | 2016

Phase I study of PSMA-targeted docetaxel-containing nanoparticle BIND-014 in patients with advanced solid tumors

Daniel D. Von Hoff; Monica M. Mita; Ramesh K. Ramanathan; Glen J. Weiss; Alain C. Mita; Patricia LoRusso; Howard A. Burris; Lowell L. Hart; Susan C. Low; Donald Parsons; Stephen E. Zale; Jason Summa; Hagop Youssoufian; Jasgit C. Sachdev

Purpose: First-in-human phase I trial to determine the safety, pharmacokinetics, and antitumor activity of BIND-014, a novel, tumor prostate-specific membrane antigen (PSMA)–targeted nanoparticle, containing docetaxel. Experimental Design: Patients with advanced solid tumors received BIND-014 every three weeks (n = 28) or weekly (n = 27), with dose levels ranging from 3.5 to 75 mg/m2 and 15 to 45 mg/m2, respectively. Results: BIND-014 was generally well tolerated, with no unexpected toxicities. The most common drug-related toxicities (>20% of patients) on either schedule included neutropenia, fatigue, anemia, alopecia, and diarrhea. BIND-014 demonstrated a dose-linear pharmacokinetic profile, distinct from docetaxel, with prolonged persistence of docetaxel-encapsulated circulating nanoparticles. Of the 52 patients evaluable for response, one had a complete response (cervical cancer on the every three week schedule) and five had partial responses (ampullary adenocarcinoma, non–small cell lung, and prostate cancers on the every-three-week schedule, and breast and gastroesophageal cancers on the weekly schedule). Responses were noted in both PSMA-detectable and -undetectable tumors. Conclusions: BIND-014 was generally well tolerated, with predictable and manageable toxicity and a unique pharmacokinetic profile compared with conventional docetaxel. Clinical activity was noted in multiple tumor types. The recommended phase II dose of BIND-014 is 60 mg/m2 every three weeks or 40 mg/m2 weekly. Clin Cancer Res; 22(13); 3157–63. ©2016 AACR.


Cancer Discovery | 2017

Phase I, dose-escalation, two-part trial of the PARP inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers

Johann S. de Bono; Ramesh K. Ramanathan; Lida Mina; Rashmi Chugh; John A. Glaspy; Saeed Rafii; Stan B. Kaye; Jasgit C. Sachdev; John V. Heymach; David C. Smith; Joshua W. Henshaw; Ashleigh Herriott; Miranda J. Patterson; Nicola J. Curtin; Lauren Averett Byers; Zev A. Wainberg

Talazoparib inhibits PARP catalytic activity, trapping PARP1 on damaged DNA and causing cell death in BRCA1/2-mutated cells. We evaluated talazoparib therapy in this two-part, phase I, first-in-human trial. Antitumor activity, MTD, pharmacokinetics, and pharmacodynamics of once-daily talazoparib were determined in an open-label, multicenter, dose-escalation study (NCT01286987). The MTD was 1.0 mg/day, with an elimination half-life of 50 hours. Treatment-related adverse events included fatigue (26/71 patients; 37%) and anemia (25/71 patients; 35%). Grade 3 to 4 adverse events included anemia (17/71 patients; 24%) and thrombocytopenia (13/71 patients; 18%). Sustained PARP inhibition was observed at doses ≥0.60 mg/day. At 1.0 mg/day, confirmed responses were observed in 7 of 14 (50%) and 5 of 12 (42%) patients with BRCA mutation-associated breast and ovarian cancers, respectively, and in patients with pancreatic and small cell lung cancer. Talazoparib demonstrated single-agent antitumor activity and was well tolerated in patients at the recommended dose of 1.0 mg/day.Significance: In this clinical trial, we show that talazoparib has single-agent antitumor activity and a tolerable safety profile. At its recommended phase II dose of 1.0 mg/day, confirmed responses were observed in patients with BRCA mutation-associated breast and ovarian cancers and in patients with pancreatic and small cell lung cancer. Cancer Discov; 7(6); 620-9. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 539.


British Journal of Clinical Pharmacology | 2016

AKT in cancer: new molecular insights and advances in drug development

Prabhjot Singh Mundi; Jasgit C. Sachdev; Carolyn K. McCourt; Kevin Kalinsky

The phosphatidylinositol‐3 kinase (PI3K)–AKT pathway is one of the most commonly dysregulated pathways in all of cancer, with somatic mutations, copy number alterations, aberrant epigenetic regulation and increased expression in a number of cancers. The carefully maintained homeostatic balance of cell division and growth on one hand, and programmed cell death on the other, is universally disturbed in tumorigenesis, and downstream effectors of the PI3K–AKT pathway play an important role in this disturbance. With a wide array of downstream effectors involved in cell survival and proliferation, the well‐characterized direct interactions of AKT make it a highly attractive yet elusive target for cancer therapy. Here, we review the salient features of this pathway, evidence of its role in promoting tumorigenesis and recent progress in the development of therapeutic agents that target AKT.


Clinical Breast Cancer | 2008

Evolution of Bevacizumab-Based Therapy in the Management of Breast Cancer

Jasgit C. Sachdev; Mohammad Jahanzeb

Combination therapy comprising bevacizumab with paclitaxel recently received accelerated approval from the US Food and Drug Administration (FDA) for use in the first-line treatment of patients with metastatic breast cancer. Bevacizumab is a monoclonal antibody that targets vascular endothelial growth factor (VEGF), providing direct inhibition of angiogenesis. The evolution of bevacizumab use in the first-line treatment of patients with breast cancer has been characterized by a logical progression of phase II and III trials that have demonstrated that angiogenesis plays an important role throughout all stages of breast cancer growth and progression. In the phase III clinical trial E2100, which provided the basis for FDA approval, the use of bevacizumab (10 mg/kg on days 1 and 15) plus paclitaxel (90 mg/m2 days 1, 8, and 15 every 28 days) given until disease progression approximately doubled median progression-free survival (PFS; 11.8 months vs. 5.9 months; hazard ratio = 0.60; P < .001) compared with paclitaxel alone; by contrast, a statistically significant improvement in overall survival was not seen with the addition of bevacizumab, although a post hoc analysis demonstrated a significant increase in 1-year survival for the combination arm. The E2100 study, as well as the majority of clinical trial designs for bevacizumab, has used PFS as the primary efficacy endpoint, and, in this review, the development of PFS as a measure of clinical benefit is outlined. This review also discusses the importance of VEGF signaling in early phases of breast tumor progression, which has provided a rationale for the investigation of bevacizumab in early-stage settings.


Clinical Breast Cancer | 2012

Blockade of the HER Family of Receptors in the Treatment of HER2-Positive Metastatic Breast Cancer

Jasgit C. Sachdev; Mohammad Jahanzeb

Breast cancer is the most common type of cancer among women and the second leading cause of cancer death in the United States. Metastatic breast cancer is considered incurable, and treatment is aimed at palliating symptoms, achieving remission, and prolonging survival. Treatment options for metastatic disease vary based on tumor surface markers and clinical factors in an individual patient and include cytotoxic chemotherapy, hormonal therapy, biological therapy, or some combination of these. An important molecular determinant of therapy is the human epidermal growth factor receptor 2 (HER2) positivity of the tumor, which affects response to HER2-targeted treatment. HER2 is a member of the human epidermal growth factor receptor family of receptor tyrosine kinases, also known as the HER family, which activates signaling that promotes tumorigenic cellular processes such as proliferation and evasion of apoptosis. Several targeted agents, including monoclonal antibodies and tyrosine kinase inhibitors that inhibit one or more HER family receptors have been developed that affect signaling through this pathway. Some of these, such as trastuzumab and lapatinib, have been approved for breast cancer treatment. Resistance to therapy is a challenge that limits the duration of benefit achieved with these agents. Therefore, combinations of HER family-targeted agents with other therapies such as cytotoxic agents, hormonal therapy, or inhibitors of other cellular pathways, are being developed to exploit synergy and overcome resistance mechanisms. Here we review the HER family-targeted agents currently approved or in development for HER2-positive metastatic breast cancer with a focus on strategies to overcome tumor resistance.


Cancer Research | 2015

Abstract PD5-2: Ph1b study of the PI3K inhibitor taselisib (GDC-0032) in combination with letrozole in patients with hormone receptor-positive advanced breast cancer

Cristina Saura; Jasgit C. Sachdev; Manish R. Patel; A. Cervantes; Dejan Juric; Jeffrey R. Infante; Donald A. Richards; Sandra Sanabria; Xuyang Lu; Joseph A. Ware; Timothy R. Wilson; Hema Parmar; Jerry Y. Hsu; Mafalda Oliveira; Daniel D. Von Hoff; José Baselga; Ian E. Krop

Background: Taselisib (GDC-0032) is a next-generation PI3K inhibitor with increased anti-tumor activity against PIK3CA mutant (MT) cancers. Taselisib is an orally bioavailable, potent, and selective inhibitor of Class I PI3K alpha, delta, and gamma isoforms, with 30-fold less inhibition of the PI3K beta isoform relative to the PI3K alpha isoform. Preclinical data show that taselisib has enhanced activity against PI3K alpha isoform (PIK3CA) MT breast cancer cell lines and enhanced antitumor activity when combined with letrozole. Clinical data with single-agent taselisib also showed increased tumor shrinkage in patients with PIK3CA MT breast cancer as compared to patients with PIK3CA wildtype (WT) breast cancer. Material and Methods: A Phase 1b dose escalation study was conducted with evaluation of taselisib doses ranging from 6-9 mg QD in combination with letrozole 2.5mg QD in a modified 3+3 design. A dose expansion cohort was conducted with taselisib 6 mg QD. Safety and tolerability of GDC-0032 was assessed, as well as pharmacokinetics (PK), pharmacodynamic (PD) assessment by FDG-PET, and anti-tumor activity by RECIST. Results: As of 31 January 2014, 28 patients were enrolled onto this study with the completion of dose escalation and the dose expansion cohort. No dose limiting toxicities (DLTs) were observed at either the 6 mg (n = 20) or 9 mg (n = 8) dose levels. Adverse events (AEs) assessed by the investigator as related to taselisib in ≥10% of patients (any grade) included diarrhea, nausea, stomatitis, fatigue, rash, decreased appetite, hyperglycemia, dysgeusia, mucosal inflammation, vomiting, muscle spasms, asthenia, dry mouth, dry skin, pruritus, and aspartate aminotransferase increased. Grade 3 and 4 adverse events assessed by the investigator as drug-related and occurring in greater than one patient included diarrhea (14%), hyperglycemia (7%), and mucosal inflammation (7%). No apparent PK interactions were observed between taselisib and letrozole. The median number of prior systemic therapies was six, and promising efficacy data has been observed in these heavily pretreated patients. Metabolic partial responses via FDG-PET (≥ 20% decrease in mean SUVmax) were observed in 11 out of 18 patients assessed (61%). Confirmed partial responses by RECIST have been observed at both the 6mg and 9mg taselisib dose levels. For patients with measurable disease at baseline, the overall response rate of 38% was observed in patients with PIK3CA MT breast cancer and 9% in patients with PIK3CA WT breast cancer. Updated data on safety, PD, efficacy, and biomarker correlates will be presented. Conclusions: The combination of taselisib and letrozole is a well-tolerated regimen with promising preliminary efficacy in PIK3CA MT breast cancer patients. This preliminary Ph1b clinical data is consistent with taselisib preclinical and single-agent clinical data showing increased anti-tumor activity for taselisib in PIK3CA MT breast cancer as compared to PIK3CA WT breast cancer. Taselisib is being further investigated in the neoadjuvant setting in combination with letrozole in the LORELEI study in patients with untreated hormone receptor-positive breast cancer. Citation Format: Cristina Saura, Jasgit Sachdev, Manish R Patel, Andres Cervantes, Dejan Juric, Jeffrey R Infante, Donald Richards, Sandra Sanabria, Xuyang Lu, Joseph Ware, Timothy R Wilson, Hema Parmar, Jerry Y Hsu, Mafalda Oliveira, Eric P Winer, Daniel D Von Hoff, Jose Baselga, Ian E Krop. Ph1b study of the PI3K inhibitor taselisib (GDC-0032) in combination with letrozole in patients with hormone receptor-positive advanced breast cancer [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr PD5-2.


Clinical Cancer Research | 2016

Safety, Pharmacokinetics and Pharmacodynamics of a Humanized anti-Semaphorin 4D Antibody, in a First-In-Human Study of Patients with Advanced Solid Tumors

Amita Patnaik; Glen J. Weiss; John E. Leonard; Drew W. Rasco; Jasgit C. Sachdev; Terrence L. Fisher; Laurie A. Winter; Christine Reilly; Robert B. Parker; Danielle Mutz; Lisa Blaydorn; Anthony W. Tolcher; Maurice Zauderer; Ramesh K. Ramanathan

Purpose: Study objectives included evaluating the safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and antitumor activity of VX15/2503 in advanced solid tumor patients. Experimental Design: Weekly i.v. doses were administered on a 28-day cycle. Safety, immunogenicity, PK, efficacy, T-cell membrane–associated SEMA4D (cSEMA4D) expression and saturation, soluble SEMA4D (sSEMA4D) serum levels, and serum biomarker levels were evaluated. Results: Forty-two patients were enrolled into seven sequential cohorts and an expansion cohort (20 mg/kg). VX15/2503 was well tolerated. Treatment-related adverse events were primarily grade 1 or 2 and included nausea (14.3%) and fatigue (11.9%); arthralgia, decreased appetite, infusion-related reaction, and pyrexia were each 7.3%. One pancreatic cancer patient (15 mg/kg) experienced a Grade 3 dose-limiting toxicity; elevated γ-glutamyl transferase. Complete cSEMA4D saturation was generally observed at serum antibody concentrations ≥0.3 μg/mL, resulting in decreased cSEMA4D expression. Soluble SEMA4D levels increased with dose and infusion number. Neutralizing anti-VX15/2503 antibodies led to treatment discontinuation for 1 patient. VX15/2503 Cmax and AUC generally increased with dose and dose number. One patient (20 mg/kg) experienced a partial response, 19 patients (45.2%) exhibited SD for ≥8 weeks, and 8 (19%) had SD for ≥16 weeks. Subjects with elevated B/T lymphocytes exhibited longer progression-free survival. Conclusions: VX15/2503 was well tolerated and produced expected PD effects. The correlation between immune cell levels at baseline and progression-free survival is consistent with an immune-mediated mechanism of action. Future investigations will be in combination with immunomodulatory agents. Clin Cancer Res; 22(4); 827–36. ©2015 AACR.


Clinical Cancer Research | 2017

Correlation between Ferumoxytol Uptake in Tumor Lesions by MRI and Response to Nanoliposomal Irinotecan in Patients with Advanced Solid Tumors: A Pilot Study

Ramesh K. Ramanathan; Ronald L. Korn; Natarajan Raghunand; Jasgit C. Sachdev; Ronald G. Newbold; Gayle S. Jameson; Gerald J. Fetterly; Joshua Prey; Stephan Klinz; Jaeyeon Kim; Jason E. Cain; Bart S. Hendriks; Daryl C. Drummond; Eliel Bayever; Jonathan Fitzgerald

Purpose: To determine whether deposition characteristics of ferumoxytol (FMX) iron nanoparticles in tumors, identified by quantitative MRI, may predict tumor lesion response to nanoliposomal irinotecan (nal-IRI). Experimental Design: Eligible patients with previously treated solid tumors had FMX-MRI scans before and following (1, 24, and 72 hours) FMX injection. After MRI acquisition, R2* signal was used to calculate FMX levels in plasma, reference tissue, and tumor lesions by comparison with a phantom-based standard curve. Patients then received nal-IRI (70 mg/m2 free base strength) biweekly until progression. Two percutaneous core biopsies were collected from selected tumor lesions 72 hours after FMX or nal-IRI. Results: Iron particle levels were quantified by FMX-MRI in plasma, reference tissues, and tumor lesions in 13 of 15 eligible patients. On the basis of a mechanistic pharmacokinetic model, tissue permeability to FMX correlated with early FMX-MRI signals at 1 and 24 hours, while FMX tissue binding contributed at 72 hours. Higher FMX levels (ranked relative to median value of multiple evaluable lesions from 9 patients) were significantly associated with reduction in lesion size by RECIST v1.1 at early time points (P < 0.001 at 1 hour and P < 0.003 at 24 hours FMX-MRI, one-way ANOVA). No association was observed with post-FMX levels at 72 hours. Irinotecan drug levels in lesions correlated with patients time on treatment (Spearman ρ = 0.7824; P = 0.0016). Conclusions: Correlation between FMX levels in tumor lesions and nal-IRI activity suggests that lesion permeability to FMX and subsequent tumor uptake may be a useful noninvasive and predictive biomarker for nal-IRI response in patients with solid tumors. Clin Cancer Res; 23(14); 3638–48. ©2017 AACR.


Cancer Research | 2014

Abstract CT327: Multicenter phase I study of MRX34, a first-in-class microRNA miR-34 mimic liposomal injection

Muhammad Shaalan Beg; Mitesh J. Borad; Jasgit C. Sachdev; David S. Hong; Susan Smith; Andres Bader; Jay Stoudemire; Sinil Kim; Andrew Brenner

Background: MRX34 is a liposome-formulated double-stranded mimic of tumor suppressor microRNA-34 (miR-34), which is lost or expressed at reduced levels in many solid and hematologic malignancies. miR-34 functions in the p53 tumor suppressor pathway and regulates the expression of more than 20 cancer-related genes including BCL2, E2F3, HDAC1, MET, MEK1, CDK4/6, PDGFR-ɑ, SIRT1, WNT1/3, NOTCH-1, β-catenin, CD44, Nanog and AXL. miR-34 also suppresses stem-like properties in cancer cells and sensitizes cancer cells to cytotoxic and targeted therapies. A multicenter phase I clinical trial of MRX34 is being conducted in patients with unresectable hepatocellular carcinoma or metastatic cancer with liver involvement. Methods: Eligible patients are enrolled in a standard 3 + 3 dose escalation study at MRX34 starting dose of 10 mg/m2, administered IV on days 1, 4, 8, 11, 15, and 18 of 28-day cycles. Dexamethasone premedication was added after the first 2 dose levels. The primary objective is to determine the RP2D, and secondary objectives include assessments of safety, PK and objective response rate (ORR). Response is evaluated by CT/MRI every 8 weeks. Once RP2D has been established, expansion cohort will be treated at the RP2D, with specific focus on determining biological response in tumor biopsy material. The latter will include assessing tumor uptake of miR-34 mimic and down-regulation of target gene expression as was observed in animal models of cancer. Results: Twenty-five patients received study drug, and enrollment is currently ongoing at the 5th dose level (70 mg/m2) and MTD has not been reached. There were 18 Caucasians, 15 males, with a median age of 62.5 years and a median of 4 prior treatments. Liposomal formulation-associated infusion reactions have been observed as characterized by the following treatment emergent adverse events (AEs) out of 20 patients: fever (13), chills (12), fatigue (12), thrombocytopenia (8), diarrhea (8), back/flank pain (6), nausea (6), and neutropenia (4). Most common Gr 3/4 AEs were neutropenia (4), thrombocytopenia (3), fatigue (2), AST/ALT elevation (4) and back/flank pain (2). Addition of pre-dose dexamethasone with higher dose levels ameliorated infusion reactions. One patient died on study from disease progression. One dose-limiting toxicity was observed at the 2nd dose level (20 mg/m2; Gr 3 acute kidney injury associated with nausea, vomiting, diarrhea and dehydration) and one at the 5th dose level (70 mg/m2; Gr 3 increased AST). Conclusions: MRX34 has a manageable safety profile in patients with unresectable primary liver cancer or metastatic cancer with liver involvement. Determination of the R2PD, assessment of clinical response, and analyses of molecular markers are ongoing. Citation Format: Muhammad Shaalan Beg, Mitesh Borad, Jasgit Sachdev, David S. Hong, Susan Smith, Andres Bader, Jay Stoudemire, Sinil Kim, Andrew Brenner. Multicenter phase I study of MRX34, a first-in-class microRNA miR-34 mimic liposomal injection. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr CT327. doi:10.1158/1538-7445.AM2014-CT327

Collaboration


Dive into the Jasgit C. Sachdev's collaboration.

Top Co-Authors

Avatar

Daniel D. Von Hoff

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Monica M. Mita

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lowell L. Hart

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Glen J. Weiss

Cancer Treatment Centers of America

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gayle S. Jameson

Translational Genomics Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge