Network


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

Hotspot


Dive into the research topics where Daniel J. Maslyar is active.

Publication


Featured researches published by Daniel J. Maslyar.


Molecular Cancer Therapeutics | 2016

Phase I Study of DMOT4039A, an Antibody-Drug Conjugate Targeting Mesothelin, in Patients with Unresectable Pancreatic or Platinum-Resistant Ovarian Cancer

Colin D. Weekes; Laetitia E. Lamberts; Mitesh J. Borad; Johannes Voortman; Robert R. McWilliams; Jennifer R. Diamond; Elisabeth G.E. de Vries; Henk M.W. Verheul; Christopher Hanyoung Lieu; George P. Kim; Yulei Wang; Suzie J. Scales; Divya Samineni; Flavia Brunstein; YounJeong Choi; Daniel J. Maslyar; Gerardo Colon-Otero

DMOT4039A, a humanized anti-mesothelin mAb conjugated to the antimitotic agent monomethyl auristatin E (MMAE), was given to patients with pancreatic and ovarian cancer every 3 weeks (0.2–2.8 mg/kg; q3w) or weekly (0.8–1.2 mg/kg). A 3+3 design was used for dose escalation followed by expansion at the recommended phase II dose (RP2D) to evaluate safety and pharmacokinetics. Antitumor response was evaluated per RECIST 1.1 and serum CA19-9 or CA125 declines. Tumor mesothelin expression was determined by IHC. Seventy-one patients (40 pancreatic cancer; 31 ovarian cancer) were treated with DMOT4039A. For the q3w schedule (n = 54), the MTD and RP2D was 2.4 mg/kg, with dose-limiting toxicities of grade 3 hyperglycemia and grade 3 hypophosphatemia at 2.8 mg/kg. For the weekly schedule (n = 17), the maximum assessed dose was 1.2 mg/kg, with further dose escalations deferred because of toxicities limiting scheduled retreatment in later cycles, and therefore the RP2D level for the weekly regimen was determined to be 1 mg/kg. Across both schedules, the most common toxicities were gastrointestinal and constitutional. Treatment-related serious adverse events occurred in 6 patients; 4 patients continued treatment following dose reductions. Drug exposure as measured by antibody-conjugated MMAE and total antibody was generally dose proportional over all dose levels on both schedules. A total of 6 patients had confirmed partial responses (4 ovarian; 2 pancreatic) with DMOT4039A at 2.4 to 2.8 mg/kg i.v. q3w. DMOT4039A administered at doses up to 2.4 mg/kg q3w and 1.0 mg/kg weekly has a tolerable safety profile and antitumor activity in both pancreatic and ovarian cancer. Mol Cancer Ther; 15(3); 439–47. ©2016 AACR.


Clinical Cancer Research | 2016

ImmunoPET with Anti-Mesothelin Antibody in Patients with Pancreatic and Ovarian Cancer before Anti-Mesothelin Antibody-Drug Conjugate Treatment

Titia E Lamberts; Catharina Willemien Menke-van der Houven van Oordt; Eva J. ter Weele; Frederike Bensch; Michiel M. Smeenk; Johannes Voortman; Otto S. Hoekstra; Simon Williams; Bernard M. Fine; Daniel J. Maslyar; Johan R. de Jong; Jourik A. Gietema; Carolina P. Schröder; Alfons H. H. Bongaerts; Marjolijn N. Lub-de Hooge; Henk M.W. Verheul; Sandra Sanabria Bohorquez; Andor W. J. M. Glaudemans; Elisabeth G.E. de Vries

Purpose: Mesothelin (MSLN) is frequently overexpressed in pancreatic and ovarian cancers, making it a potential drug target. We performed an 89Zr-PET imaging study with MMOT0530A, a MSLN antibody, in conjunction with a phase I study with the antibody–drug conjugate DMOT4039A, containing MMOT0530A bound to MMAE. The aim was to study antibody tumor uptake, whole-body distribution, and relation between uptake, response to treatment, and MSLN expression. Experimental Design: Before DMOT4039A treatment, patients received 37 MBq 89Zr-MMOT0530A followed by PET/CT imaging 2, 4, and 7 days postinjection. Tracer uptake was expressed as standardized uptake value (SUV). MSLN expression was determined with immunohistochemistry (IHC) on archival tumor tissue. Results: Eleven patients were included, 7 with pancreatic and 4 with ovarian cancer. IHC MSLN expression varied from absent to strong. Suitable tracer antibody dose was 10 mg MMOT0530A and optimal imaging time was 4 and 7 days postinjection. Tumor tracer uptake occurred in 37 lesions with mean SUVmax of 13.1 (±7.5) on PET 4 days postinjection, with 11.5 (±7.5) in (N = 17) pancreatic and 14.5 (±8.7) in (N = 20) ovarian cancer lesions. Within patients, a mean 2.4-fold (±1.10) difference in uptake between tumor lesions existed. Uptake in blood, liver, kidneys, spleen, and intestine reflected normal antibody distribution. Tracer tumor uptake was correlated to IHC. Best response to DMOT4039A was partial response in one patient. Conclusions: With 89Zr-MMOT0530A-PET, pancreatic and ovarian cancer lesions as well as antibody biodistribution could be visualized. This technique can potentially guide individualized antibody-based treatment. Clin Cancer Res; 22(7); 1642–52. ©2015 AACR.


Cancer Research | 2013

Abstract LB-290: Targeting MUC16 with the antibody-drug conjugate (ADC) DMUC5754A in patients with platinum-resistant ovarian cancer: A phase I study of safety and pharmacokinetics.

Joyce Liu; Kathleen N. Moore; Michael J. Birrer; Suzanne Berlin; Ursula A. Matulonis; Jeffrey R. Infante; Jian Xi; Robert Kahn; Yulei Wang; Katie Wood; Daniel Coleman; Daniel J. Maslyar; Eric Humke; Howard A. Burris

Background: MUC16 is a large transmembrane protein overexpressed by the majority of ovarian cancers (OC), compared with normal tissues. The role of MUC16 in the pathogenesis of ovarian cancer is unknown; however, MUC16 may facilitate the binding of ovarian tumor cells to mesothelial cells lining the peritoneal cavity, and may inhibit natural killer cell-mediated anti-tumor cytotoxic responses. Conjugation of a highly potent cytotoxic drug to a MUC16-specific monoclonal antibody represents a novel approach to treatment of MUC16-expressing tumors. DMUC5754A is an ADC containing the humanized IgG1 anti-MUC16 monoclonal antibody linked to the potent anti-mitotic agent MMAE and demonstrates anti-tumor activity in MUC16-expressing tumor xenograft models. Methods: This phase I study evaluated safety, pharmacokinetics (PK), and pharmacodynamic (PD) activity of DMUC5754A (0.3-3.2 mg/kg) given every 3 weeks (q3w) to patients with advanced recurrent platinum-resistant OC. A standard 3+3 design was used to determine the maximum-tolerated dose, followed by cohort expansion. Tumor tissue was assessed for expression of MUC16 and other relevant markers. Clinical activity was evaluated per RECIST. Results: Forty-four patients (22 escalation, 22 expansion at 2.4 mg/kg), median age 62 (44-79), ECOG PS 0-1, received a median of 4 doses (range 1-20) of DMUC5754A. Two DLTs, 1 Grade 4 neutropenia and 1 Grade 4 uric acid increase, occurred at the maximally administered dose of 3.2 mg/kg. Grade ≥ 3 related adverse events (AE) occurring in ≥ 5% of patients included fatigue (4 at 2.4 mg/kg; 9% total) and neutropenia (1 at 3.2 mg/kg, 3 at 2.4 mg/kg; 9% total). The most common related AEs over all dose levels were fatigue (57%), nausea (34%), vomiting (27%), decreased appetite (25%), peripheral neuropathy (25%), and diarrhea (23%). Serious drug-related AEs (SAE) were small intestine obstruction (2 patients), hypocalcemia (1 patient), and neutropenia (1 patient). Total antibody and conjugated MMAE were not impacted by circulating CA125 and displayed dose-dependent PK with clearance decreasing as dose increased. Accumulation of total antibody, conjugated MMAE and free MMAE was not observed due to their short half-lives (≤5 days). Tumor MUC16 expression was evaluable in 42 patients, showing 20% IHC 0, 16% IHC 2+, and 64% IHC 3+. All confirmed responses (1 CR and 4 PRs) occurred in patients treated at 2.4 mg/kg and whose tumors were MUC16 IHC 2+ or 3+. Six additional patients had minor responses (3 at 2.4 mg/kg). Sixteen of the twenty-nine patients at 2.4 mg/kg were on study ≥ 105 days. Human epididymis protein 4 was a potential surrogate marker for serologic response measures in the presence of anti-MUC16/CA125-binding therapy. Conclusions: DMUC5754A at 2.4 mg/kg q3w has an encouraging safety profile and evidence of anti-tumor activity in MUC16-expressing OC. Further studies are planned. Citation Format: Joyce Liu, Kathleen Moore, Michael Birrer, Suzanne Berlin, Ursula Matulonis, Jeffrey Infante, Jian Xi, Robert Kahn, Yulei Wang, Katie Wood, Daniel Coleman, Daniel Maslyar, Eric Humke, Howard Burris. Targeting MUC16 with the antibody-drug conjugate (ADC) DMUC5754A in patients with platinum-resistant ovarian cancer: A phase I study of safety and pharmacokinetics. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr LB-290. doi:10.1158/1538-7445.AM2013-LB-290


Annals of Oncology | 2014

1598PTRANSLATIONAL PKPD OF DNIB0600A, AN ANTI-NAPI2B-VC-MMAE ADC IN OVARIAN AND NSCLC CANCERS

Kedan Lin; S. Sukumaran; J. Xu; Crystal Zhang; YounJeong Choi; Shang-Fan Yu; Paul Polakis; Daniel J. Maslyar

ABSTRACT Aim: Antibody-drug conjugates (ADCs) represent an expanding class of therapeutic molecules in preclinical and clinical development for oncologic indications. Understanding the relationship between pre-clinical to clinical studies with strategic application of pharmacokinetic/pharmacodynamics modeling may allow for optimized dosing strategies for ADCs. NaPi2b is a multi-transmembrane, sodium-dependent phosphate transporter that is expressed in human lung, ovarian, and thyroid cancers. DNIB0600A, which consists of an anti-NaPi2b monoclonal antibody conjugated to the cytotoxic drug MMAE through a cleavable VC linker, is currently in Phase I/II clinical trials. The purpose of this study is to develop a PKPD model based on preclinical data and to utilize the preclinical exposure response relationships to predict clinical outcomes. Methods: DNIB0600A PK was evaluated in normal SCID mice. Dose ranging in-vivo efficacy studies were performed in NaPi2b-expressing xenograft mouse models of ovarian and lung cancers. A semi-mechanistic PKPD model was developed to describe exposure-efficacy relationships in both types of the tumor models. Human PK and PD data were collected from ongoing Phase I/II trials. Results: DNIB0600A demonstrated differential anti-tumor activities in the models, with the ovarian model being more responsive when compared with lung tumor model to NaPi2b ADC treatment. Human efficacious doses for treating ovarian cancer and NSCLC were predicted based on the pre-clinical PKPD relationships. Observed efficacy data from preliminary analysis of Phase I/II trials were in general agreements with model predictions. Conclusions: We built an integrated PKPD model to predict clinical outcome. This approach can be extended to other vc-MMAE based ADCs, and can help in preclinical model validation and ADC optimization. Disclosure: All authors have declared no conflicts of interest.


Annals of Oncology | 2018

Anti-NaPi2b antibody–drug conjugate lifastuzumab vedotin (DNIB0600A) compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer in a randomized, open-label, phase II study

Susana Banerjee; Amit M. Oza; Michael J. Birrer; Erika Paige Hamilton; J Hasan; Alexandra Leary; Kathleen N. Moore; B Mackowiak-Matejczyk; J Pikiel; Isabelle Ray-Coquard; Peter C Trask; Kedan Lin; E Schuth; Anjali Vaze; YounJeong Choi; Jim Marsters; Daniel J. Maslyar; Lemahieu; Yulei Wang; Eric Humke; Joyce Liu

Background Lifastuzumab vedotin (LIFA) is a humanized anti-NaPi2b monoclonal antibody conjugated to a potent antimitotic agent, monomethyl auristatin E, which inhibits cell division by blocking the polymerization of tubulin. This study is the first to compare an antibody-drug conjugate (ADC) to standard-of-care in ovarian cancer (OC) patients. Patients and methods Platinum-resistant OC patients were randomized to receive LIFA [2.4 mg/kg, intravenously, every 3 weeks (Q3W)] or pegylated liposomal doxorubicin (PLD) (40 mg/m2, intravenously, Q4W). NaPi2b expression and serum CA-125 and HE4 levels were assessed. The primary end point was progression-free survival (PFS) in intent-to-treat (ITT) and NaPi2b-high patients. Results Ninety-five patients were randomized (47 LIFA; 48 PLD). The stratified PFS hazard ratio was 0.78 [95% confidence interval (95% CI), 0.46-1.31; P = 0.34] with a median PFS of 5.3 versus 3.1 months (LIFA versus PLD arm, respectively) in the ITT population, and 0.71 (95% CI, 0.40-1.26; P = 0.24) with a median PFS of 5.3 months versus 3.4 months (LIFA versus PLD arm, respectively) in NaPi2b-high patients. The objective response rate was 34% (95% CI, 22% to 49%, LIFA) versus 15% (95% CI, 7% to 28%, PLD) in the ITT population (P = 0.03), and 36% (95% CI, 22% to 52%, LIFA) versus 14% (95% CI, 6% to 27%, PLD) in NaPi2b-high patients (P = 0.02). Toxicities included grade ≥3 adverse events (AEs) (46% LIFA; 51% PLD), serious AEs (30% both arms), and AEs leading to discontinuation of drug (9% LIFA; 8% PLD). Five (11%) LIFA versus 2 (4%) PLD patients had grade ≥2 neuropathy. Conclusion LIFA Q3W was well tolerated and improved objective response rate with a modest, nonstatistically significant improvement of PFS compared with PLD in platinum-resistant OC. While the response rate for the monomethyl auristatin E-containing ADC was promising, response durations were relatively short, thereby highlighting the importance of evaluating both response rates and duration of response when evaluating ADCs in OC. Clinical trials.gov NCT01991210.


Cancer Research | 2015

Abstract 4310: Predictive biomarkers of tumor sensitivity to STEAP1 antibody-drug conjugate (ADC) in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC)

Daniel C. Danila; Howard I. Scher; Edith Szafer-Glusman; Amrita Herkal; Rebecca Suttmann; Martin Fleisher; Nicole A. Schreiber; Kristen Rebecca Curtis; Houston Gilbert; Daniel J. Maslyar; Bernard M. Fine; Ron Firestein; Michael Mamounas; Mark R. Lackner; Omar Kabbarah

ADCs hold promise for enhancing the therapeutic index of cytotoxics. STEAP1 is overexpressed in mCRPC and is the target of an ADC currently under clinical development. To identify pts most likely to benefit from the ADC, we explored STEAP1 expression as a predictive biomarker in tumor tissue using a CLIA-certified IHC assay, and on circulating tumor cells (CTCs) in blood using the CellSearch and EPIC platforms. Sixty pts with progressive mCRPC received doses ranging from 0.3 to 2.8 mg/kg once every three weeks. Response was defined as a ≥50% decline in PSA from baseline, and time on study for >6 months was consistent with continued clinical benefit. At doses of ≥2 mg/kg, the response rate (RR) was 10/45 (22%, 95% CI 9.9-34.1) and was highest in the STEAP1 IHC 3+ group, as was the fraction of patients who remained on study for >6 months. At ≥2 mg/kg, 20/45 pts (44%, 95% CI 29.5-58.5) had unfavorable CTC counts of ≥5/7.5mL at baseline and were considered evaluable. After treatment, conversions from unfavorable to favorable CTC counts of An immuno-fluorescence-based assay developed to measure STEAP1 expression levels on CTCs showed readily detectable signal in 18/42 samples (43%, 95% CI 28-58), with a range from 1-56% of STEAP1-positive CTCs per case. After treatment, a decrease in the fraction of patients with STEAP1-positive and an increase in the STEAP1-negative CTCs was observed compared to baseline (see Table). We are prospectively selecting pts with STEAP1 IHC 2+/3+ tumors and assessing STEAP1 levels on CTCs in an ongoing Ph1 expansion study with the goal of developing a companion diagnostic to enrich for mCRPC pts most likely to benefit from treatment with the STEAP1 ADC. Citation Format: Daniel C. Danila, Howard I. Scher, Edith Szafer-Glusman, Amrita Herkal, Rebecca Suttmann, Martin Fleisher, Nicole A. Schreiber, Kristen Curtis, Houston Gilbert, Daniel Maslyar, Bernard Fine, Ron Firestein, Michael Mamounas, Mark R. Lackner, Omar Kabbarah. Predictive biomarkers of tumor sensitivity to STEAP1 antibody-drug conjugate (ADC) in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC). [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4310. doi:10.1158/1538-7445.AM2015-4310


Annals of Oncology | 2014

1627PPET-IMAGING WITH 89ZR-LABELED ANTI-MESOTHELIN (MSLN) ANTIBODY IN PATIENTS WITH PANCREATIC CANCER (PC) OR OVARIAN CANCER (OC)

Laetitia E. Lamberts; C W Menke-van der Houven van Oordt; Frederike Bensch; J Voortman; Otto S. Hoekstra; Daniel J. Maslyar; Simon Williams; Bernard M. Fine; Alphons H. H. Bongaerts; Jourik A. Gietema; Carolien P. Schröder; E J Ter Weele; M. N. Lub-de Hooge; Henk M.W. Verheul; S Sanabria; A. W. J. M. Glaudemans; de Elisabeth G. E. Vries

Aim: The tumor antigen MSLN is frequently overexpressed in PC and OC. A 89Zr-PET study (NCT01832116) with MMOT0530A, an anti-MSLN antibody, was initiated in conjunction with a phase 1 study of the antibody-drug conjugate DMOT4039A (containing MMOT0530A linked to the anti-mitotic agent MMAE, NCT01469793). This imaging study aims to investigate antibody tumor uptake, whole body distribution and organ pharmacokinetics and to explore the relation between uptake and MSLN expression and response to DMOT40392A treatment in patients with unresectable PC or platinum-resistant OC. Methods: Before receiving DMOT4039A, patients were injected with 37 MBq 89Zr-MMOT0530A +/- additional unlabeled MMOT0530A, followed by PET/CT imaging 2, 4 and 7 days post injection (pi). Tracer uptake was quantified with standardized uptake value (SUV) and expressed as mean (±SD). MSLN expression was determined in archival tumor tissue with an exploratory immunohistochemical (IHC) assay. Results: 7 PC and 4 OC patients were included. MSLN expression varied from 0 to 3+. The optimal antibody protein dose resulting in sufficient circulating tracer was 10 mg MMOT0530A and the optimal imaging time was 4 or 7 days pi. Tumor tracer uptake was observed in 37 quantifiable tumor lesions (all patients) with mean SUV of 10.7 (±6.3) on PET 4 days pi. The mean SUV per patient (1-8 lesions/patient) was 10.9 (±5.7), with 9.2 (±4.5) in PC and 11.9 (±7.4) in OC lesions on PET 4 days pi. Within patients, a mean 2.4-fold (±1.10) difference in tumor uptake between lesions was found. Two measurable lesions on diagnostic CT (according to RECIST 1.1) were not visible on PET. Uptake in blood, liver, kidneys, spleen and intestine reflected normal antibody distribution with mean SUV at day 4 pi of 5.6, 7.8, 6.1, 4.1 and 3.2, respectively, while low uptake was observed in muscle, lung, brain and bone (0.6, 1.0, 0.2 and 0.7, respectively). Tracer tumor uptake was lower in the 2 patients with IHC scores 0 and 1. Best response on DMOT4039A was stable disease in ten patients. An association between iPET tumor uptake and clinical response could not be determined. Conclusions: 89Zr-MMOT0530A-PET shows antibody uptake in primary and metastatic PC and OC tumor lesions. This technique can potentially guide antibody-based therapy development.


Cancer Research | 2013

Abstract 2659: Imaging human pancreatic tumor xenografts with 89Zr-labeled anti-mesothelin antibody.

Eva J. ter Weele; Marjolijn N. Lub-de Hooge; Daniel J. Maslyar; Anton G.T. Terwisscha van Scheltinga; Jos G. W. Kosterink; Elisabeth G.E. de Vries; Simon Williams

Background: Mesothelin (MSLN) is a tumor differentiation antigen that is highly expressed by cells of many epithelial tumors, with limited expression in normal human tissues. Our understanding of therapeutic antibodies targeting MSLN might benefit from immunoPET imaging of antibody uptake. We developed and preclinically validated an 89Zr labeled anti-MSLN antibody (“89Zr-AMA”) for this noninvasive imaging of tumor and normal organ uptake. Methods: 89Zr was attached to an anti-MSLN humanized IgG1 monoclonal antibody derivatized with the bifunctional chelator reagent N-succinyldesferrioxamine-B-tetrafluorphenol. The 89Zr-AMA was characterized in terms of conjugation ratio, aggregation, radiochemical purity, stability, and immunoreactivity. Two human MSLN-expressing pancreatic tumor cell lines, HPAC and CAPAN-2, were used for xenograft studies in mice. Tumor uptake and organ distribution of 89Zr-AMA were studied in the HPAC line at three protein doses (10, 25 and 100 μg) labeled with 1 MBq 89Zr and results were compared with nonspecific 111In-IgG. After dose-finding, CAPAN-2 and HPAC tumor xenograft-bearing mice were scanned with μPET at 1, 3, and 6 days after tracer injection of the optimal AMA dose labeled with 5 MBq 89Zr, followed by ex vivo biodistribution at day 6. Tracer uptake was quantified and expressed as mean standardized uptake values (SUVmean). Results: 89Zr-AMA formed with high specific activity (> 500 MBq/mg), high yield (> 90% without further purification), and high purity (> 95% determined by SE-HPLC analysis). In vitro validation of 89Zr-AMA showed a fully preserved immunoreactivity with a long (> 1 week) stability in 0.9% NaCl. Biodistribution analyses of the dose-finding groups revealed a dose-dependent 89Zr-AMA tumor uptake, with the highest fractional tumor uptake in the 10 μg dose group, 14.2 %ID/g on day 6. Tumor uptake of the non-specific control antibody, 111In-IgG, was lower than that of the 89Zr-AMA (P Conclusion: 89Zr-AMA tumor uptake is antigen-specific in MSLN-expressing tumors. This tracer can be translated to the clinic for serial non-invasive PET imaging. Citation Format: Eva J. ter Weele, Marjolijn N. Lub-de Hooge, Daniel Maslyar, Anton G.T. Terwisscha van Scheltinga, Jos G.W. Kosterink, Elisabeth G.E. de Vries, Simon P. Williams. Imaging human pancreatic tumor xenografts with 89Zr-labeled anti-mesothelin antibody. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2659. doi:10.1158/1538-7445.AM2013-2659


Annals of Oncology | 2016

PTEN loss as a predictive biomarker for the Akt inhibitor ipatasertib combined with abiraterone acetate in patients with metastatic castration-resistant prostate cancer (mCRPC)

J. S. De Bono; U. De Giorgi; Christophe Massard; Sergio Bracarda; D. Nava Rodrigues; Ivo Kocak; A. Font; J. Arranz Arija; Kent C. Shih; George Daniel Radavoi; Wei Yu; Wai Y. Chan; Steven Gendreau; Liying Zhang; Ruth Riisnaes; M.J. Wongchenko; Daniel J. Maslyar; Viorel Jinga


Annals of Oncology | 2016

Phase I study of safety and pharmacokinetics of the anti-MUC16 antibody–drug conjugate DMUC5754A in patients with platinum-resistant ovarian cancer or unresectable pancreatic cancer

Joyce Liu; Kathleen N. Moore; Michael J. Birrer; Suzanne Berlin; Ursula A. Matulonis; Jeffrey R. Infante; Brian M. Wolpin; K. A. Poon; R. Firestein; J. Xu; Robert Kahn; Yulei Wang; Katie Wood; Walter C. Darbonne; Mark R. Lackner; S. K. Kelley; X. Lu; YounJeong Choi; Daniel J. Maslyar; Eric Humke; Howard A. Burris

Collaboration


Dive into the Daniel J. Maslyar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge