Network


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

Hotspot


Dive into the research topics where Janet Ayello is active.

Publication


Featured researches published by Janet Ayello.


Biology of Blood and Marrow Transplantation | 2010

An Age-Dependent Pharmacokinetic Study of Intravenous and Oral Mycophenolate Mofetil in Combination with Tacrolimus for GVHD Prophylaxis in Pediatric Allogeneic Stem Cell Transplantation Recipients

Monica Bhatia; Olga Militano; Zhezhen Jin; Michal Figurski; Leslie M. Shaw; Virginia Moore; Erin Morris; Bradford Tallamy; Carmella van deVen; Janet Ayello; L.A. Baxter-Lowe; Prakash Satwani; Diane George; James Garvin; Mitchell S. Cairo

Acute graft-versus-host disease (aGVHD) still remains a major limiting factor following allogeneic stem cell transplantation (AlloSCT) in pediatric recipients. Mycophenolate mofetil (MMF), an uncompetitive selective inhibitor of inosine monophosphate dehydrogenase, is a new immunosuppressant agent without major mucosal, hepatic, or renal toxicity compared to other prophylactic aGVHD immunosuppressant drugs. Although there has been an extensive pharmacokinetic (PK) experience with MMF administration following solid organ transplantation in children, there is a paucity of PK data following its use in pediatric AlloSCT recipients. We investigated the safety and PK of MMF as GVHD prophylaxis following intravenous (i.v.) and oral (p.o.) administration (900 mg/m(2) every 6 hours) in conjunction with tacrolimus, after myeloablative (MA) and nonmyeloablative (NMA) conditioning and AlloSCT in 3 distinct age groups of pediatric AlloSCT recipients (0-6 years, 6-12 years, and 12-16 years). Mycophenolic acid (MPA) in plasma samples was measured either by high-performance liquid chromatography (HPLC) or liquid chromatography/mass spectrometry (LC/MS/MS) as we have previously described. Plasma samples were obtained at baseline and at 0.5, 1, 2, 3, 4, and 6 hours after i.v. dosing on days +1, +7, +14, and at 2 time points between day +45 and +100 after p.o. administration post AlloSCT. MPA PK analysis included AUC (0-6 hours), C(max), T(max), C(ss), V(ss), C trough (C(0)), CL, and T((1/2).) Thirty-eight patients, with a median age of 8 years (0.33-16 years), 20/18 M:F ratio, 21/17 malignant/nonmalignant disease, 17/21 MA: NMA conditioning, 16 of 22 related/unrelated allografts. Median time to myeloid and platelet engraftment was 18 and 31 days, respectively. Mean donor chimerism on day +60 and +100 was 83% and 90%, respectively. Probability of developing aGVHD grade II-IV and extensive chronic GVHD (cGVHD) was 54% and 34%, respectively. There was significant intra- and interpatient MMF PK variability. There was a significant increase in i.v. MPA area under the curve (AUC)(0-6 hour) and C(max) (P < .0003) and a significant decrease in CL(ss) (P < .002) and V(ss) (P < .001) on day +14 versus day +7. Children <12 years of age had a significant increase in i.v. MPA T(max) (P = .01), V(ss) (P = .028), and CL(ss) (P < .001) compared to the older age group. There was a trend in increased i.v. MPA CL(ss) following MA versus NMA conditioning (P < .054); i.v. and p.o. MMF administration (900 mg/m(2) every 6 hours) in combination with tacrolimus was well tolerated in pediatric AlloSCT recipients. There was a significant increase in MPA exposure on day +14 versus day +7, suggesting improved enterohepatic recirculation at day +14 post-AlloSCT. Children <12 years of age appear to have a significantly different MPA PK profile compared to older children and adolescents and may require more frequent dosing.


Experimental Hematology | 2002

Ex vivo expansion, maturation, and activation of umbilical cord blood–derived T lymphocytes with IL-2, IL-12, anti-CD3, and IL-7: Potential for adoptive cellular immunotherapy post–umbilical cord blood transplantation

Kathleen L Robinson; Janet Ayello; Rose Hughes; Carmella van de Ven; Linda Issitt; Joanne Kurtzberg; Mitchell S. Cairo

OBJECTIVES We investigated whether umbilical cord blood (UCB) T cells could be ex vivo expanded and activated in short-term culture for potential utilization as adoptive cellular immunotherapy post-umbilical cord blood transplantation (UCBT). METHODS Fresh UCB mononuclear cells (MNCs) were isolated by Ficoll density centrifugation. Cryopreserved UCB mononuclear cells were thawed and washed with 2.5% human serum albumin and 5% dextrose in isotonic saline. The nonadherent MNC fraction were then plated in a serum-free cocktail of IL-2, IL-12, and anti-CD3 with and without IL-7 for 48 hours. Proliferation, cytotoxicity, TH1 (IFN-gamma), CD25, and CD45RO assays were performed. RESULTS Proliferation studies demonstrated a significant increase in the proliferative ability of the UCB MNCs incubated in anti-CD3, IL-2, IL-12, and IL-7 (fresh--p < 0.005, and thawed--p < 0.001). The combination of all four agonists significantly induced expression of CD45 RO (fresh--p < 0.05, and thawed--p < 0.001) in both the CD4(+) and CD8(+) T cells expressing CD25 (fresh UCB--p < 0.01 [CD4] and p < 0.005 [CD8], respectively; thawed UCB--p < 0.001 [CD4] and p < 0.001 [CD8]). Intracellular cytokine profiles also revealed a significant increase in the production of IFN-gamma (TH1 cells) (fresh UCB--p < 0.005, and thawed UCB--p < 0.001). The combination also significantly increased the killing of K562-labeled target cells (fresh--p < 0.0001, and thawed--0.731 +/- 0.03 vs 0.16 +/- 0.01) (p < 0.001). CONCLUSIONS These data suggest that the ex vivo combination of IL-2, IL-12, anti-CD3, and IL-7 significantly enhances the proliferation, activation, maturation, and cytotoxic potential of UCB T cells of both fresh and thawed UCB MNC. Further studies, however, are required to determine whether these ex vivo--expanded MNC could also potentially exacerbate acute or chronic graft-vs-host disease and/or other toxicities if utilized post-UCBT.


Journal of Clinical Oncology | 2011

Safety, Pharmacokinetics, and Immunomodulatory Effects of Lenalidomide in Children and Adolescents With Relapsed/Refractory Solid Tumors or Myelodysplastic Syndrome: A Children's Oncology Group Phase I Consortium Report

Stacey L. Berg; Mitchell S. Cairo; Heidi V. Russell; Janet Ayello; Ashish M. Ingle; Henry Lau; Nianhang Chen; Peter C. Adamson; Susan M. Blaney

PURPOSE To determine the maximum-tolerated or recommended phase II dose, dose-limiting toxicities (DLTs), pharmacokinetics (PK), and immunomodulatory effects of lenalidomide in children with recurrent or refractory solid tumors or myelodysplastic syndrome (MDS). PATIENTS AND METHODS Cohorts of children with solid tumors received lenalidomide once daily for 21 days, every 28 days at dose levels of 15 to 70 mg/m(2)/dose. Children with MDS received a fixed dose of 5 mg/m(2)/dose. Specimens for PK and immune modulation were obtained in the first cycle. RESULTS Forty-nine patients (46 solid tumor, three MDS), median age 16 years (range, 1 to 21 years), were enrolled, and 42 were fully assessable for toxicity. One patient had a cerebrovascular ischemic event of uncertain relationship to lenalidomide. DLTs included hypercalcemia at 15 mg/m(2); hypophosphatemia/hypokalemia, neutropenia, and somnolence at 40 mg/m(2); and urticaria at 55 mg/m(2). At the highest dose level evaluated (70 mg/m(2)), zero of six patients had DLT. A maximum-tolerated dose was not reached. No objective responses were observed. PK studies (n = 29) showed that clearance is faster in children younger than 12 years of age. Immunomodulatory studies (n = 26) showed a significant increase in serum interleukin (IL) -2, IL-15, granulocyte-macrophage colony-stimulating factor, natural killer (NK) cells, NK cytotoxicity, and lymphokine activated killer (LAK) cytoxicity, and a significant decrease in CD4(+)/CD25(+) regulatory T cells. CONCLUSION Lenalidomide is well-tolerated at doses up to 70 mg/m(2)/d for 21 days in children with solid tumors. Drug clearance in children younger than 12 years is faster than in adolescents and young adults. Lenalidomide significantly upregulates cellular immunity, including NK and LAK activity.


Cancer immunology research | 2015

Targeting CD20+ Aggressive B-cell Non–Hodgkin Lymphoma by Anti-CD20 CAR mRNA-Modified Expanded Natural Killer Cells In Vitro and in NSG Mice

Yaya Chu; Jessica Hochberg; Ashlin Yahr; Janet Ayello; Carmella van de Ven; Matthew J. Barth; Myron S. Czuczman; Mitchell S. Cairo

Chu and colleagues show that K562-mbIL15-41BBL–expanded peripheral blood NK cells, modified with mRNA nucleofection of an anti-CD20 CAR, significantly enhanced cytotoxicity against CD20+B-cell non-Hodgkin lymphoma, extended survival time, and reduced tumor size in xenografted NSG mice. The prognosis is very dismal for patients with relapsed CD20+ B-cell non-Hodgkin lymphoma (B-NHL). Facilitating the development of alternative novel therapeutic strategies is required to improve outcomes in patients with recurrent/refractory CD20+ B-NHL. In this study, we investigated functional activities of anti-CD20 CAR-modified, expanded peripheral blood NK cells (exPBNK) following mRNA nucleofection against CD20+ B-NHL in vitro and in vivo. CAR+ exPBNK had significantly enhanced in vitro cytotoxicity, compared with CAR− exPBNK against CD20+ Ramos (P < 0.05), Daudi, Raji, and two rituximab-resistant cell lines, Raji-2R and Raji-4RH (P < 0.001). As expected, there was no significant difference against CD20− RS4;11 and Jurkat cells. CD107a degranulation and intracellular IFNγ production were also enhanced in CAR+ exPBNK in response to CD20+ B-NHL–specific stimulation. In Raji-Luc and Raji-2R-Luc xenografted NOD/SCID/γ-chain−/− (NSG) mice, the luciferase signals measured in the CAR+ exPBNK-treated group were significantly reduced, compared with the signals measured in the untreated mice and in mice treated with the CAR− exPBNK. Furthermore, the CAR exPBNK-treated mice had significantly extended survival time (P < 0.001) and reduced tumor size, compared with those of the untreated and the CAR− exPBNK-treated mice (P < 0.05). These preclinical data suggest that ex vivo–exPBNK modified with anti-CD20 CAR may have therapeutic potential for treating patients with poor-risk CD20+ hematologic malignancies. Cancer Immunol Res; 3(4); 333–44. ©2014 AACR.


British Journal of Haematology | 2015

Obinutuzumab (GA101) compared to rituximab significantly enhances cell death and antibody-dependent cytotoxicity and improves overall survival against CD20(+) rituximab-sensitive/-resistant Burkitt lymphoma (BL) and precursor B-acute lymphoblastic leukaemia (pre-B-ALL): potential targeted therapy in patients with poor risk CD20(+) BL and pre-B-ALL.

Awasthi A; Janet Ayello; van de Ven C; Elmacken M; Sabulski A; Matthew J. Barth; Myron S. Czuczman; Islam H; Christian Klein; Cairo Ms

Obinutuzumab is a novel glycoengineered Type‐II CD20 monoclonal antibody. CD20 is expressed in approximately 100% of children and adolescents with Burkitt lymphoma (BL) and 40% with precursor B‐cell acute lymphoblastic leukaemia (pre‐B‐ALL). We evaluated the anti‐tumour activity of obinutuzumab versus rituximab against rituximab‐resistant (Raji 4RH) and ‐sensitive (Raji) BL and pre‐B‐ALL (U698‐M) cells in vitro and in human BL or Pre‐B‐ALL xenografted mice. We demonstrated that obinutuzumab compared to rituximab significantly enhanced cell death against Raji 35·6 ± 3·1% vs. 25·1 ± 2·0%, (P = 0·001), Raji4RH 19·7 ± 2·2% vs. 7·9 ± 1·5% (P = 0·001) and U‐698‐M 47·3 ± 4·9% vs. 23·2 ± 0·5% (P = 0·001), respectively. Obinutuzumab versus rituximab also induced a significant increase in antibody‐dependent cellular cytotoxicity (ADCC) with K562‐IL15‐41BBL expanded NK cells against Raji 73·8 ± 8·1% vs. 56·81 ± 4·6% (P = 0·001), Raji‐4RH 40·0 ± 1·6% vs. 0·5 ± 1·1% (P = 0·001) and U‐698‐M 70·0 ± 1·6% vs. 45·5 ± 0·1% (P = 0·001), respectively. Overall survival in tumour xenografted mice receiving 30 mg/kg of obinutuzumab was significantly increased when compared to those receiving 30 mg/kg of rituximab in BL; Raji (P = 0·05), Raji4RH (P = 0·02) and U698‐M (P = 0·03), respectively. These preclinical data suggest obinutuzumab is significantly superior to rituximab in inducing cell death, ADCC and against rituximab‐sensitive/‐resistant BL and pre‐B‐ALL xenografted mice. Taken together, these preclinical results provide evidence to suggest that future investigation of obinutuzumab is warranted in patients with relapsed/refractory CD20+ BL and/or pre‐B‐ALL.


Experimental Hematology | 2009

Characterization of natural killer and natural killer-like T cells derived from ex vivo expanded and activated cord blood mononuclear cells: implications for adoptive cellular immunotherapy.

Janet Ayello; Carmella van de Ven; Evan Cairo; Jessica Hochberg; Laxmi Baxi; Prakash Satwani; Mitchell S. Cairo

OBJECTIVE Cord blood (CB) is limited by the absence of available donor effector cells for post-unrelated CB transplantation adoptive cellular immunotherapy. We reported the ability to ex vivo expand (EvE) CB mononuclear cells (MNC) after short-term incubation with anti-CD3, interleukin (IL)-2, IL-7, and IL-12 (antibody/cytokine [AB/CY]) into subpopulations of CD3(-)/56(+) natural killer (NK) cells with enhanced in vitro and in vivo tumor cytotoxicity. MATERIALS AND METHODS We compared 2- vs 7-day EvE of rethawed CB MNCs in AB/CY and activation of NK and NK-like T (NKT) cell (CD3(+)/56(+)) subsets expressing specific NK-cell receptors along with IL-15, IL-18, and interferon-gamma production. RESULTS Nonadherent total cell number were significantly increased at day 7 (p<0.001) along with NK-cell number (20-fold) and an enrichment in NKT-like subsets (36-fold). There was no change in the NK(dim) subset; yet the NKT(bright) and NKT KIR3DL1(dim) subsets were significantly increased (p<0.05). NK cells expressing the inhibitory natural cytoxicity receptor CD94/NKG2A were decreased (p<0.001), while those expressing activating natural cytoxicity receptor CD94/NKG2D receptor and activating NK and NKT KIR2DS4 subsets were significantly increased (p<0.001). IL-18 and interferon-gamma protein production was also significantly increased (p<0.001 and p<0.05, respectively). Lysosomal-associated membrane protein-1 and granzyme B expression were increased (p<0.001 and p>0.01, respectively), which correlated with the significant increase in NK, LAK, and tumor cytotoxicity of the EvE cells. CONCLUSION This study demonstrates that previously cryopreserved and rethawed CB MNCs can be EvE up to 7 days to yield viable and activated NK and NKT-like subsets that appear to be cytolytic based on the cell repertoire and could be utilized in the future as adoptive cellular immunotherapy post-unrelated CB transplantation.


Clinical Cancer Research | 2007

A Phase I Study of 90Yttrium-Ibritumomab-Tiuxetan in Children and Adolescents with Relapsed/Refractory CD20-Positive Non–Hodgkin's Lymphoma: A Children's Oncology Group Study

Erin M. Cooney-Qualter; Mark Krailo; Anne L. Angiolillo; Rashid A. Fawwaz; Gregory A. Wiseman; Lauren Harrison; Virginia Kohl; Peter C. Adamson; Janet Ayello; Carmella Vandeven; Sherrie L. Perkins; Mitchell S. Cairo

Purpose: The prognosis for children with recurrent CD20+ non–Hodgkins lymphoma is dismal. A radiolabeled anti-CD20 antibody, 90yttrium-ibritumomab-tiuxetan (90Y-IT), is Food and Drug Administration approved for adults with recurrent indolent CD20+ B cell–non–Hodgkins lymphoma. There is no data on the safety and feasibility of 90Y-IT in refractory childhood CD20+ lymphoma. Experimental Design: Children and adolescents with refractory/relapsed CD20+ lymphoma were eligible for this phase I radioimmunotherapy study. Patients (n = 5) received rituximab (250 mg/m2 i.v.) on days 0 and 7 and indium-111 ibritumomab-tiuxetan (5 mCi i.v.) on day 0. Dosimetry studies were done on days 0, 1, 3, and 6. Immediately after rituximab on day 7, patients received 90Y-IT if dosimetry studies showed <2000 cGy exposure to all solid organs and <300 cGy to marrow, as well as 0.4 mCi/kg in patients with good marrow reserve (n = 3) and 0.1 mCi/kg in patients with poor marrow reserve (after bone marrow transplant; n = 2). Results: No patients experienced nonhematologic or hematologic dose-limiting toxicity. Human antimurine antibody/human antichimeric antibody incidence was 0%. One patient experienced grade II infusion–related chills associated with rituximab. The following are the means of organ radiation exposure (cGy): kidneys 341 (112-515), liver 345 (83-798), lungs 309 (155-519), marrow 46 (20-78), spleen 565 (161-816), and total body 42 (14-68). Conclusions: Based on these findings, an expanded investigator-initiated limited institutional phase II study has been designed to further evaluate the safety, tolerability, and response rate with 90Y-IT dose stratification based on marrow reserve.


British Journal of Haematology | 2012

T cell depletion utilizing CD34+ stem cell selection and CD3+ addback from unrelated adult donors in paediatric allogeneic stem cell transplantation recipients

Mark B. Geyer; Angela Ricci; Judith S. Jacobson; Robbie G. Majzner; Deirdre Duffy; Carmella van de Ven; Janet Ayello; Monica Bhatia; James Garvin; Diane George; Prakash Satwani; Lauren Harrison; Erin Morris; Mildred Semidei-Pomales; Joseph E. Schwartz; Bachir Alobeid; Lee Ann Baxter-Lowe; Mitchell S. Cairo

CD34‐selected haploidentical and unrelated donor allogeneic stem cell transplantation (AlloSCT) in paediatric recipients is associated with sustained engraftment and low risk of acute graft‐versus‐host disease (aGVHD), but limited by delayed immune reconstitution and increased risk of viral and fungal infection. The optimal dose of donor T cells to prevent graft failure and minimize risk of early opportunistic infection and post‐transplant lymphoproliferative disorder (PTLD), while avoiding severe aGVHD, remains unknown. We prospectively studied CD34‐selected 8–10/10 human leucocyte antigen (HLA)‐matched unrelated donor (MUD) peripheral blood stem cell transplantation (PBSCT) in a cohort of 19 paediatric AlloSCT recipients with malignant (n = 13) or non‐malignant (n = 6) diseases. T cells were added back to achieve total dose 1·0–2·5 × 105 CD3+/kg. GVHD pharmacoprophylaxis consisted only of tacrolimus. All patients engrafted neutrophils. Probabilities of grade II–IV aGVHD, limited chronic GVHD (cGVHD), and extensive cGVHD were 15·8%, 23·3%, and 0%, respectively. One patient developed PTLD. One‐year infection‐related mortality was 5·6%. T cell immune reconstitution was delayed. One‐year overall survival was 82·3%. Five patients with malignant disease ultimately died from progressive disease. CD34‐selected MUD PBSCT using a defined dose of T cell add‐back resulted in high rates of engraftment and low risk of grade II–IV aGVHD, early transplantation‐related mortality, and extensive cGVHD.


Cytotherapy | 2011

Interleukin (IL)-15 in combination with IL-2, fms-like tyrosine kinase-3 ligand and anti-CD3 significantly enhances umbilical cord blood natural killer (NK) cell and NK-cell subset expansion and NK function

Prakash Satwani; Carmella van de Ven; Janet Ayello; Dustin Cairo; Lynn L. Simpson; Laxmi V. Baxi; Mitchell S. Cairo

BACKGROUND AIMS Interleukin (IL)-15 and fms-like tyrosine kinase-3 (FLT-3) are crucial factors for the development of human and murine natural killer (NK) cells. Previously, we have demonstrated significant ex vivo expansion and activation of unrelated cord blood (UCB) NK cells with an antibody/cytokine cocktail consisting of anti-CD3 + IL-2 + IL-12 + IL-7 and anti-CD3 + IL-2 + IL-12 + IL-18. METHODS In the current experiments, we investigated the effects of short-term culture with anti-CD3 + IL-2 + FLT-3 + IL-15 on cord blood (CB) NK cell and NK-cell subset expansion and function. CB mononuclear cells were cultured for 48 h in AIM-V media or AIM-V + IL-2 (5 ng/mL) + anti-CD3 (50 ng/mL) + FLT-3 (50 ng/mL) ± escalating doses of IL-15 (1, 10 or 100 ng/mL). Flow cytometric analysis was performed using various fluorescent-conjugated monoclonal antibodies. In vitro cytotoxicity was determined with a standard europium assay against K562 and Daudi cells. RESULTS There was a 4.8-fold significant increase in NK-cell population (CD3(-)/16(+)/56(+); P < 0.03), 21-fold significant increase in CD3(-)/56(+)/158a(+) (KIR2DL1/S1; P < 0.002), 46-fold significant increase in CD3(-)/56(+)/158b(+) (KIR2DL1/S2; P < 0.002) and 11.5-fold significant increase in CD3(-)/56(+)/NKB1(+) (KIR3DL1; P < 0.01). We also noted a significant increase in both NK and lymphokine-activated killer (LAK) cytotoxicity with IL-2 + anti-CD3 + FLT-3 + IL-15 (100 ng/mL) compared with IL-2 + anti-CD3 + FLT-3 and media alone against K562 (P < 0.01) and Daudi (P < 0.001), respectively. CONCLUSIONS We have demonstrated a significant increase in UCB NK cells and NK cells expressing a variety of killer immunoglobulin-like receptor (KIR) receptors after short-term culture with anti-CD3, IL-2, FLT-3 and IL-15. Furthermore, there was a significant increase in in vitro NK/LAK cell cytotoxicity.


Clinical Cancer Research | 2005

A Phase I Clinical, Pharmacologic, and Biologic Study of Thrombopoietin and Granulocyte Colony-Stimulating Factor in Children Receiving Ifosfamide, Carboplatin, and Etoposide Chemotherapy for Recurrent or Refractory Solid Tumors: A Children's Oncology Group Experience

Anne L. Angiolillo; Virginia Davenport; Mary Ann Bonilla; Carmella van de Ven; Janet Ayello; Olga Militano; Langdon L. Miller; Mark Krailo; Gregory H. Reaman; Mitchell S. Cairo

Purpose: Ifosfamide, carboplatin, and etoposide (ICE) are associated with grade III/IV dose-limiting thrombocytopenia. The Childrens Oncology Group conducted a phase I dose escalation, pharmacokinetic, and biological study of recombinant human thrombopoietin (rhTPO) after ICE in children with recurrent/refractory solid tumors (CCG-09717) to assess the toxicity and maximum tolerated dose of rhTPO administered at 1.2, 2.4, or 3.6 μg/kg per dose. Experimental Design: Children received ifosfamide 1,800 mg/m2 on days 0 to 4, carboplatin 400 mg/m2 on days 0 to 1, and etoposide 100 mg/m2 on days 0 to 4. rhTPO was administered i.v. on days +4, +6, +8, +10, and +12 at 1.2, 2.4, or 3.6 μg/kg per dose. Results: rhTPO was well tolerated and maximum tolerated dose was not reached. Median time to platelet recovery ≥100,000/μL of rhTPO at 1.2, 2.4, and 3.6 μg/kg/d was 24 days (22-24d), 25 days (23-29d), and 22 days (16-37d), respectively. Patients required a median of 2 days of platelet transfusions (0-7 days). Mean (± SD) rhTPO maximum serum concentrations were 63.3 ± 9.7 and 89.3 ± 15.7 ng/mL and terminal half-lives were 47 ± 13 and 64 ± 42 hours after 2.4 and 3.6 μg/kg/d, respectively. There was a significant increase in colony-forming unit megakaryocyte upon WBC count recovery. Conclusions: rhTPO was well tolerated. Time to hematologic recovery and median number of platelet transfusions seem to be improved compared with historical controls receiving ICE + granulocyte colony-stimulating factor (CCG-0894).

Collaboration


Dive into the Janet Ayello's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Prakash Satwani

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erin Morris

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Megan S. Lim

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Changhong Yin

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Barth

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Yaya Chu

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Sanghoon Lee

New York Medical College

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge