Network


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

Hotspot


Dive into the research topics where Keyvan Rezai is active.

Publication


Featured researches published by Keyvan Rezai.


The Lancet Haematology | 2016

Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study

Sandy Amorim; Anastasios Stathis; Mary Gleeson; Sunil Iyengar; Valeria Magarotto; Xavier Leleu; Franck Morschhauser; Lionel Karlin; Florence Broussais; Keyvan Rezai; Patrice Herait; Carmen Kahatt; François Lokiec; Gilles Salles; Thierry Facon; Antonio Palumbo; David Cunningham; Emanuele Zucca; Catherine Thieblemont

BACKGROUND The first-in-class small molecule inhibitor OTX015 (MK-8628) specifically binds to bromodomain motifs BRD2, BRD3, and BRD4 of bromodomain and extraterminal (BET) proteins, inhibiting them from binding to acetylated histones, which occurs preferentially at super-enhancer regions that control oncogene expression. OTX015 is active in haematological preclinical entities including leukaemia, lymphoma, and myeloma. We aimed to establish the recommended dose of OTX015 in patients with haematological malignancies. We report the results from a cohort of patients with lymphoma or multiple myeloma (non-leukaemia cohort). METHODS In this dose-escalation, open-label, phase 1 study, we recruited patients from seven university hospital centres (in France [four], Switzerland [one], UK [one], and Italy [one]). Adult patients with non-leukaemia haematological malignancies who had disease progression on standard therapies were eligible to participate. Patients were treated with oral OTX015 once a day continuously over five doses (10 mg, 20 mg, 40 mg, 80 mg, and 120 mg), using a conventional 3 + 3 design, with allowance for evaluation of alternative administration schedules. The primary endpoint was dose-limiting toxicity (DLT) in the first treatment cycle (21 days). Secondary objectives were to evaluate safety, pharmacokinetics, and preliminary clinical activity of OTX015. The study is ongoing and is registered with ClinicalTrials.gov, number NCT01713582. FINDINGS Between Feb 4, 2013, and Sept 5, 2014, 45 patients (33 with lymphoma and 12 with myeloma), with a median age of 66 years (IQR 55-72) and a median of four lines of prior therapy (IQR 3-5), were enrolled and treated. No DLTs were observed in the doses up to and including 80 mg once a day (first three patients). We then explored a schedule of 40 mg twice a day (21 of 21 days). DLTs were reported in five of six patients receiving OTX015 at this dose and schedule (all five patients had grade 4 thrombocytopenia). We explored various schedules at 120 mg once a day but none was tolerable, with DLTs of thrombocytopenia, gastrointestinal events (diarrhoea, vomiting, dysgeusia, mucositis), fatigue, and hyponatraemia in 11 of 18 evaluable patients. At this point, the Safety Monitoring Committee decided to establish the feasibility of 80 mg once a day on a continuous basis, and four additional patients were enrolled at this dose. DLTs (grade 4 thrombocytopenia) was noted in two of the patients. In light of these DLTs and other toxicities noted at 120 mg, the dose of 80 mg once a day was selected, although on a schedule of 14 days on, 7 days off. Common toxic effects reported in the study were thrombocytopenia (43 [96%] patients), anaemia (41 [91%]), neutropenia (23 [51%]), diarrhoea (21 [47%]), fatigue (12 [27%]), and nausea (11 [24%]). Grade 3-4 adverse events were infrequent other than thrombocytopenia (26 [58%]). OTX015 plasma peak concentrations and areas under the concentration versus time curve increased proportionally with dose. Trough concentrations increased less than proportionally at lower doses, but reached or exceeded the in-vitro active range at 40 mg twice a day and 120 mg once a day. Three patients with diffuse large B-cell lymphoma achieved durable objective responses (two complete responses at 120 mg once a day, and one partial response at 80 mg once a day), and six additional patients (two with diffuse large B-cell lymphoma, four with indolent lymphomas) had evidence of clinical activity, albeit not meeting objective response criteria. INTERPRETATION The once-daily recommended dose for oral, single agent oral OTX015 in patients with lymphoma is 80 mg on a 14 days on, 7 days off schedule, for phase 2 studies. OTX015 is under evaluation in expansion cohorts using this intermittent administration (14 days every 3 weeks) to allow for recovery from toxic effects. FUNDING Oncoethix GmbH (a wholly owned subsidiary of Merck Sharp & Dohme Corp).


The Lancet Haematology | 2016

Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study

Céline Berthon; Emmanuel Raffoux; Xavier Thomas; Norbert Vey; Carlos Gomez-Roca; Karen Yee; David Taussig; Keyvan Rezai; Christophe Roumier; Patrice Herait; Carmen Kahatt; Bruno Quesnel; M. Michallet; Christian Recher; François Lokiec; Claude Preudhomme; Hervé Dombret

BACKGROUND Bromodomain and extraterminal (BET) proteins are chromatin readers that preferentially affect the transcription of genes with super-enhancers, including oncogenes. BET proteins bind acetylated histone tails via their bromodomain, bringing the elongation complex to the promoter region. OTX015 (MK-8628) specifically binds to BRD2, BRD3, and BRD4, preventing BET proteins from binding to the chromatin, thus inhibiting gene transcription. OTX015 inhibits proliferation in many haematological malignancy cell lines and patient cells, in vitro and in vivo. We aimed to establish the recommended dose of OTX015 in patients with haematological malignancies. We report the results of patients with acute leukaemia (leukaemia cohort). METHODS In this dose-escalation, phase 1 study we recruited patients from seven university hospital centres (in France [five], UK [one], and Canada [one]). Adults with acute leukaemia who had failed or had a contraindication to standard therapies were eligible to participate. OTX015 was given orally at increasing doses from 10 mg/day to 160 mg/day (14 of 21 days), using a conventional 3 + 3 design. In this open-label trial, OTX015 was initially administered once a day, with allowance for exploration of other schedules. The primary endpoint was dose-limiting toxicity (DLT), assessed during the first treatment cycle (21 days). The study is ongoing and is registered with ClinicalTrials.gov, NCT01713582. FINDINGS Between Jan 18, 2013, and Sept 9, 2014, 41 patients, 36 with acute myeloid leukaemia, a median age of 70 years (IQR 60-75) and two lines of previous therapy, were recruited and treated across six dose levels of OTX015. No DLT was recorded until 160 mg/day, when one patient had grade 3 diarrhoea and another had grade 3 fatigue. However, concomitant grade 1-2 non-DLT toxic effects (ie, gastrointestinal, fatigue, or cutaneous) from 120 mg doses hampered patient compliance and 80 mg once a day was judged the recommended dose with a 14 days on, 7 days off schedule. Common toxic effects for all OTX015 doses were fatigue (including grade 3 in three patients) and bilirubin concentration increases (including grade 3-4 in two patients). OTX015 plasma exposure increased proportionally up to 120 mg/day with trough concentrations in the in-vitro active range from 80 mg/day (274 nmol/L). Three patients (receiving 40 mg/day, 80 mg/day, and 160 mg/day) achieved complete remission or complete remission with incomplete recovery of platelets lasting 2-5 months, and two additional patients had partial blast clearance. No predictive biomarkers for response have been identified so far. INTERPRETATION The once-daily recommended dose for oral, single agent oral OTX015 use in patients with acute leukaemia for further phase 2 studies is 80 mg on a 14 days on, 7 days off schedule. FUNDING Oncoethix GmbH, a wholly owned subsidiary of Merck Sharp & Dohme Corp.


Cancer | 2012

Safety, pharmacokinetics, and activity of EZN-2208, a novel conjugate of polyethylene glycol and SN38, in patients with advanced malignancies

Razelle Kurzrock; Sanjay Goel; Jennifer J. Wheler; David S. Hong; Siqing Fu; Keyvan Rezai; Sonia K. Morgan-Linnell; Saik Urien; Sridhar Mani; Imran Chaudhary; Mohammed Ghalib; Cm Aby Buchbinder Md; François Lokiec; Mary F. Mulcahy

EZN‐2208 is a water‐soluble, polyethylene glycol drug conjugate of SN38, which is the active moiety of irinotecan. In this study, the authors evaluated the tolerability, pharmacokinetics (PK), and activity of EZN‐2208 in adult patients with advanced solid tumors.


International Journal of Cancer | 2016

OTX015 (MK-8628), a novel BET inhibitor, displays in vitro and in vivo antitumor effects alone and in combination with conventional therapies in glioblastoma models.

Caroline Berenguer-Daizé; Lucile Astorgues-Xerri; Elodie Odore; Mylène Cayol; Esteban Cvitkovic; Kay Noel; Mohamed Bekradda; Sarah MacKenzie; Keyvan Rezai; François Lokiec; Maria Eugenia Riveiro; L'Houcine Ouafik

Bromodomain and extraterminal (BET) bromodomain (BRD) proteins are epigenetic readers that bind to acetylated lysine residues on chromatin, acting as co‐activators or co‐repressors of gene expression. BRD2 and BRD4, members of the BET family, are significantly increased in glioblastoma multiforme (GBM), the most common primary adult brain cancer. OTX015 (MK‐8628), a novel BRD2/3/4 inhibitor, is under evaluation in dose‐finding studies in solid tumors, including GBM. We investigated the pharmacologic characteristics of OTX015 as a single agent and combined with targeted therapy or conventional chemotherapies in glioblastoma cell lines. OTX015 displayed higher antiproliferative effects compared to its analog JQ1, with GI50 values of approximately 0.2 µM. In addition, C‐MYC and CDKN1A mRNA levels increased transiently after 4 h‐exposure to OTX015, while BRD2, SESN3, HEXIM‐1, HIST2H2BE, and HIST1H2BK were rapidly upregulated and sustained after 24 h. Studies in three additional GBM cell lines supported the antiproliferative effects of OTX015. In U87MG cells, OTX015 showed synergistic to additive activity when administered concomitant to or before SN38, temozolomide or everolimus. Single agent oral OTX015 significantly increased survival in mice bearing orthotopic or heterotopic U87MG xenografts. OTX015 combined simultaneously with temozolomide improved mice survival over either single agent. The passage of OTX015 across the blood–brain barrier was demonstrated with OTX015 tumor levels 7 to 15‐fold higher than in normal tissues, along with preferential binding of OTX015 to tumor tissue. The significant antitumor effects seen with OTX015 in GBM xenograft models highlight its therapeutic potential in GBM patients, alone or combined with conventional chemotherapies.


Cancer Research | 2014

Abstract CT231: BET-bromodomain inhibitor OTX015 shows clinically meaningful activity at nontoxic doses: interim results of an ongoing phase I trial in hematologic malignancies

Patrice Herait; Céline Berthon; Catherine Thieblemont; Emmanuel Raffoux; Valeria Magarotto; Anastasios Stathis; Xavier Thomas; Xavier Leleu; Carlos Gomez-Roca; Elodie Odore; Christophe Roumier; Fabrice Bourdel; Bruno Quesnel; Emanuele Zucca; Mauricette Michallet; Christian Recher; Esteban Cvitkovic; Keyvan Rezai; Claude Preudhomme; Thierry Facon; Antonio Palumbo; Hervé Dombret

Aim: The bromodomain and extraterminal (BET) subfamily of human bromodomain (BRD) proteins associates with acetylated chromatin and plays a key role in the epigenetic control of transcriptional activation, notably of genes with super-enhancers, such as the MYC oncogene. OTX015, a potent small molecule inhibitor of BRD2/3/4 (Noel et al, EORTC-NCI-AACR 2013), inhibits proliferation of a wide range of hematologic malignancies (HMs) in vitro (Bonetti et al, EORTC-NCI-AACR 2012; Boi et al, EORTC-NCI-AACR 2013; Braun et al, ASH 2013). This phase I study, designed to determine the recommended dose and pharmacokinetics of oral OTX015 as a single agent, is the first reported clinical study evaluating the effects of BRD inhibition in patients (pts) with HMs. Methods: Two independent cohorts of pts having failed all standard therapies were given ascending oral doses of OTX015 in a conventional 3+3 design, with acute leukemias (AL) treated 14 days on/7 days off and other HMs (OHM) treated continuously in 21-day cycles. OTX015 was given once daily (QD), then twice daily (BID). Results: From Jan to Dec 2013, 16 pts with AL (14 AML, 2 ALL) and 17 with OHM (6 DLBCL, 5 other lymphomas, 6 multiple myelomas) were enrolled over 4 dose levels, 10, 20, 40 and 80 mg QD. Exposure increased dose-proportionally. Plasma trough concentrations at 80 mg QD and 12h concentrations at 40 mg QD were ≥ IC50 values in vitro (250 nM), justifying the shift to a BID schedule. The 40 mg BID cohort is ongoing. Pts have a median age of 70 years (range 32-83) and median of 2 (1-8) prior therapies; 10 of 16 AL pts had AML secondary to pre-existing conditions or chemotherapy. No dose limiting toxicity was observed up to 80 mg QD/40 mg BID. Adverse events (AEs) were mainly grade (G) 1-2 hematologic and gastrointestinal events and diabetes aggravation. G 3-4 AEs were reversible thrombocytopenia in 3 pts with OHM (40 and 80 mg), and neutropenia, diarrhea, and elevated transaminases in 1 pt each. No cumulative toxicity was observed. Nine pts received >3 (range 4-7) cycles without or with minor interruptions. Among 28 pts evaluable for response, 6 had clinically meaningful activity, with 4 of 6 treated at 80 mg. Four pts with refractory/relapsed secondary or post-treatment AML achieved significant peripheral and bone marrow blast decrease or clearance, including 1 complete remission (CR) and 1 CR with incomplete recovery. Among OHM, 1 DLBCL had a partial response (PR) and 1 lymphoplasmacytic lymphoma had metabolic PR on cycle 2 PET-scan. Treatment of 5 of 6 responding pts is ongoing. Responses occurred in pts with various clinical, cytogenetic and molecular profiles. Conclusion: OTX015 is the first BRD inhibitor demonstrating clinical activity. Maximum tolerated dose was not reached at 80 mg QD or 40 mg BID; dose escalation is ongoing with the BID schedule and further schedule optimization. Updated results will be presented. Citation Format: Patrice E. Herait, Celine Berthon, Catherine Thieblemont, Emmanuel Raffoux, Valeria Magarotto, Anastasios Stathis, Xavier Thomas, Xavier Leleu, Carlos Gomez-Roca, Elodie Odore, Christophe Roumier, Fabrice Bourdel, Bruno Quesnel, Emanuele Zucca, Mauricette Michallet, Christian Recher, Esteban Cvitkovic, Keyvan Rezai, Claude Preudhomme, Thierry Facon, Antonio Palumbo, Herve Dombret. BET-bromodomain inhibitor OTX015 shows clinically meaningful activity at nontoxic doses: interim results of an ongoing phase I trial in hematologic malignancies. [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 CT231. doi:10.1158/1538-7445.AM2014-CT231


The Journal of Infectious Diseases | 2014

Extracellular Adenosine Triphosphate Affects the Response of Human Macrophages Infected With Mycobacterium tuberculosis

Nicolas Dubois-Colas; Laetitia Petit-Jentreau; Luis B. Barreiro; Sylvère Durand; Guillaume Soubigou; Cécile Lecointe; Jihène Klibi; Keyvan Rezai; François Lokiec; Jean-Yves Coppée; Brigitte Gicquel; Ludovic Tailleux

Granulomas are the hallmark of Mycobacterium tuberculosis infection. As the host fails to control the bacteria, the center of the granuloma exhibits necrosis resulting from the dying of infected macrophages. The release of the intracellular pool of nucleotides into the surrounding medium may modulate the response of newly infected macrophages, although this has never been investigated. Here, we show that extracellular adenosine triphosphate (ATP) indirectly modulates the expression of 272 genes in human macrophages infected with M. tuberculosis and that it induces their alternative activation. ATP is rapidly hydrolyzed by the ecto-ATPase CD39 into adenosine monophosphate (AMP), and it is AMP that regulates the macrophage response through the adenosine A2A receptor. Our findings reveal a previously unrecognized role for the purinergic pathway in the host response to M. tuberculosis. Dampening inflammation through signaling via the adenosine A2A receptor may limit tissue damage but may also favor bacterial immune escape.


Drug Metabolism and Disposition | 2006

Pharmacokinetics, Metabolism, and Routes of Excretion of Intravenous Irofulven in Patients with Advanced Solid Tumors

Angelo Paci; Keyvan Rezai; Alain Deroussent; Dominique De Valeriola; Micheline Re; Sophie Weill; Esteban Cvitkovic; Carmen Kahatt; Anwar Shah; Stephen J. Waters; Gary Weems; Gilles Vassal; François Lokiec

Irofulven is currently in Phase 2 clinical trials against a wide variety of solid tumors and has demonstrated activity in ovarian, prostate, gastrointestinal, and non–small cell lung cancer. The objectives of this study were to determine its pharmacokinetics and route of excretion and to characterize its metabolites in human plasma and urine samples after a 30-min i.v. infusion at a dose of 0.55 mg/kg in patients with advanced solid tumors. Three patients were administered i.v. 100 μCi of [14C]irofulven over a 30-min infusion on day 1 of cycle 1. Serial blood and plasma samples were drawn at 0 (before irofulven infusion) and up to 144 h after the start of infusion. Urine and fecal samples were collected for up to 144 h after the start of infusion. The mean urinary and fecal excretion of radioactivity up to 144 h were 71.2 and 2.9%, respectively, indicating renal excretion was the major route of elimination of [14C]irofulven. The Cmax, AUC0-∞, and terminal half-life values for total radioactivity were 1130 ng-Eq/ml, 24,400 ng-Eq · h/ml, and 116.5 h, respectively, and the corresponding values for irofulven were 82.7 ng/ml, 65.5 ng · h/ml, and 0.3 h, respectively, suggesting that the total radioactivity in human plasma was a result of the metabolites. Twelve metabolites of irofulven were detected in human urine and plasma by electrospray ionization/tandem mass spectrometry. Among these metabolites, the cyclopropane ring-opened metabolite (M2) of irofulven was found, and seven others were proposed as glucuronide and glutathione conjugates.


Oncotarget | 2017

The bromodomain inhibitor OTX015 (MK-8628) exerts anti-tumor activity in triple-negative breast cancer models as single agent and in combination with everolimus

Ramiro Vázquez; Maria Eugenia Riveiro; Lucile Astorgues-Xerri; Elodie Odore; Keyvan Rezai; Eugenio Erba; Nicolò Panini; Andrea Rinaldi; Ivo Kwee; Luca Beltrame; Mohamed Bekradda; Esteban Cvitkovic; Francesco Bertoni; Roberta Frapolli; Maurizio D'Incalci

Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous subgroup of breast tumors clinically defined by the lack of estrogen, progesterone and HER2 receptors, limiting the use of the targeted therapies employed in other breast malignancies. Recent evidence indicates that c-MYC is a key driver of TNBC. The BET-bromodomain inhibitor OTX015 (MK-8628) has potent antiproliferative activity accompanied by c-MYC down-regulation in several tumor types, and has demonstrated synergism with the mTOR inhibitor everolimus in different models. The aim of this study was to evaluate the anti-tumor activity of OTX015 as single agent and in combination with everolimus in TNBC models. OTX015 was assayed in three human TNBC-derived cell lines, HCC1937, MDA-MB-231 and MDA-MB-468, all showing antiproliferative activity after 72 h (GI50 = 75–650 nM). This was accompanied by cell cycle arrest and decreased expression of cancer stem cells markers. However, c-MYC protein and mRNA levels were only down-regulated in MDA-MB-468 cells. Gene set enrichment analysis showed up-regulation of genes involved in epigenetic control of transcription, chromatin and the cell cycle, and down-regulation of stemness-related genes. In vitro, combination with everolimus was additive in HCC1937 and MDA-MB-231 cells, but antagonistic in MDA-MB-468 cells. In MDA-MB-231 murine xenografts, tumor mass was significantly (p < 0.05) reduced by OTX015 with respect to vehicle-treated animals (best T/C = 40.7%). Although everolimus alone was not active, the combination was more effective than OTX015 alone (best T/C = 20.7%). This work supports current clinical trials with OTX015 in TNBC (NCT02259114).


Radiotherapy and Oncology | 2011

A phase I trial combining oral cisplatin (CP Ethypharm) with radiotherapy in patients with locally advanced head and neck squamous cell carcinoma

Yungan Tao; Keyvan Rezai; Etienne Brain; Atoussa Etessami; Antoine Lusinchi; Stéphane Temam; Saïk Urien; Marie-Louise Vo Van; Françoise Vauzelle-Kervroedan; François Lokiec; Nicolas Daly-Schveitzer; Jean Bourhis

PURPOSE To determine the maximum tolerated dose (MTD) of oral cisplatin (CP Ethypharm®) in combination with radiotherapy in head and neck squamous cell carcinoma (HNSCC) and the recommended dose for phase II trials. PATIENTS AND METHODS Phase I, multicenter, open-labelled, non-comparative and dose escalating trial. CP Ethypharm® was administered on five consecutive days every other week for 7 weeks (4 treatment cycles) in combination with radiotherapy. Eighteen patients with locally advanced HNSCC were allocated to four cisplatin dose levels: 10 mg/m(2)/day: 4 patients; 15 mg/m(2)/day: 4, 20 mg/m(2)/day: 5 and 25 mg/m(2)/day: 5. The inclusion of patients was dictated by occurrence of dose limiting toxicities (DLTs) at each dosing level. RESULTS The most frequently experienced AEs were gastrointestinal (GI) disorders. Five DLTs were observed, including three at 25 mg/m(2) level (two grade 2 renal toxicities, one grade 3 GI and renal toxicities), one at 20 mg/m(2) level (grade 3 GI disorders), one at 10 mg/m(2) level (grade 4 mucositis). PK analysis showed no significant difference of C(max) values between day 1 and day 5 of treatment at each dose level (total & ultrafilterable platinum). CONCLUSION Due to 3 DLTs experienced at 25 mg/m(2)/day, MTD was reached and the recommended dose for phase II studies was determined as 20 mg/m(2)/day.


Oncotarget | 2016

OTX015 (MK-8628), a novel BET inhibitor, exhibits antitumor activity in non-small cell and small cell lung cancer models harboring different oncogenic mutations

Maria Eugenia Riveiro; Lucile Astorgues-Xerri; Ramiro Vázquez; Roberta Frapolli; Ivo Kwee; Andrea Rinaldi; Elodie Odore; Keyvan Rezai; Mohamed Bekradda; Giorgio Inghirami; Maurizio D’Incalci; Kay Noel; Esteban Cvitkovic; Eric Raymond; Francesco Bertoni

Inhibitors targeting epigenetic control points of oncogenes offer a potential mean of blocking tumor progression in small cell and non-small cell lung carcinomas (SCLC, NSCLC). OTX015 (MK-8628) is a BET inhibitor selectively blocking BRD2/3/4. OTX015 was evaluated in a panel of NSCLC or SCLC models harboring different oncogenic mutations. Cell proliferation inhibition and cell cycle arrest were seen in sensitive NSCLC cells. MYC and MYCN were downregulated at both the mRNA and protein levels. In addition, OTX015-treatment significantly downregulated various stemness cell markers, including NANOG, Musashi-1, CD113 and EpCAM in H3122-tumors in vivo. Conversely, in SCLC models, weak antitumor activity was observed with OTX015, both in vitro and in vivo. No predictive biomarkers of OTX015 activity were identified in a large panel of candidate genes known to be affected by BET inhibition. In NSCLC models, OTX015 was equally active in both EML4-ALK positive and negative cell lines, whereas in SCLC models the presence of functional RB1 protein, which controls cell progression at G1, may be related to the final biological outcome of OTX015. Gene expression profiling in NSCLC and SCLC cell lines showed that OTX015 affects important genes and pathways with a very high overlapping between both sensitive and resistant cell lines. These data support the rationale for the OTX015 Phase Ib (NCT02259114) in solid tumors, where NSCLC patients with rearranged ALK gene or KRAS-positive mutations are currently being treated.

Collaboration


Dive into the Keyvan Rezai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anastasios Stathis

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander Zukiwski

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge