Network


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

Hotspot


Dive into the research topics where Justin M. Watts is active.

Publication


Featured researches published by Justin M. Watts.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic malignancies

Stephen E. Kurtz; Christopher A. Eide; Andy Kaempf; Vishesh Khanna; Samantha L. Savage; Angela Rofelty; Isabel English; Hibery Ho; Ravi Pandya; William J. Bolosky; Hoifung Poon; Michael W. Deininger; Robert H. Collins; Ronan Swords; Justin M. Watts; Daniel A. Pollyea; Bruno C. Medeiros; Elie Traer; Cristina E. Tognon; Motomi Mori; Brian J. Druker; Jeffrey W. Tyner

Significance Mononuclear cells obtained from freshly isolated patient samples with various hematologic malignancies were evaluated for sensitivities to combinations of drugs that target specific cell-signaling pathways. The diagnostic, genetic/cytogenetic, and cellular features of the patient samples were correlated with effective drug combinations. For myeloid-derived tumors, such as acute myeloid leukemia, several combinations of targeted agents that include a kinase inhibitor and venetoclax, a selective inhibitor of BCL2, are effective. Translating the genetic and epigenetic heterogeneity underlying human cancers into therapeutic strategies is an ongoing challenge. Large-scale sequencing efforts have uncovered a spectrum of mutations in many hematologic malignancies, including acute myeloid leukemia (AML), suggesting that combinations of agents will be required to treat these diseases effectively. Combinatorial approaches will also be critical for combating the emergence of genetically heterogeneous subclones, rescue signals in the microenvironment, and tumor-intrinsic feedback pathways that all contribute to disease relapse. To identify novel and effective drug combinations, we performed ex vivo sensitivity profiling of 122 primary patient samples from a variety of hematologic malignancies against a panel of 48 drug combinations. The combinations were designed as drug pairs that target nonoverlapping biological pathways and comprise drugs from different classes, preferably with Food and Drug Administration approval. A combination ratio (CR) was derived for each drug pair, and CRs were evaluated with respect to diagnostic categories as well as against genetic, cytogenetic, and cellular phenotypes of specimens from the two largest disease categories: AML and chronic lymphocytic leukemia (CLL). Nearly all tested combinations involving a BCL2 inhibitor showed additional benefit in patients with myeloid malignancies, whereas select combinations involving PI3K, CSF1R, or bromodomain inhibitors showed preferential benefit in lymphoid malignancies. Expanded analyses of patients with AML and CLL revealed specific patterns of ex vivo drug combination efficacy that were associated with select genetic, cytogenetic, and phenotypic disease subsets, warranting further evaluation. These findings highlight the heuristic value of an integrated functional genomic approach to the identification of novel treatment strategies for hematologic malignancies.


Leukemia Research | 2016

KB004, a first in class monoclonal antibody targeting the receptor tyrosine kinase EphA3, in patients with advanced hematologic malignancies: Results from a phase 1 study.

Ronan Swords; Peter L. Greenberg; Andrew Wei; Simon Durrant; Anjali S. Advani; Mark Hertzberg; Ian D. Lewis; Gabriel Rivera; Dita Gratzinger; Alice C. Fan; Dean W. Felsher; Jorge Cortes; Justin M. Watts; Geoff T. Yarranton; Jackie M. Walling; Jeffrey E. Lancet

EphA3 is an Ephrin receptor tyrosine kinase that is overexpressed in most hematologic malignancies. We performed a first-in-human multicenter phase I study of the anti-EphA3 monoclonal antibody KB004 in refractory hematologic malignancies in order to determine safety and tolerability, along with the secondary objectives of pharmacokinetics (PK) and pharmacodynamics (PD) assessments, as well as preliminary assessment of efficacy. Patients were enrolled on a dose escalation phase (DEP) initially, followed by a cohort expansion phase (CEP). KB004 was administered by intravenous infusion on days 1, 8, and 15 of each 21-day cycle in escalating doses. A total of 50 patients (AML 39, MDS/MPN 3, MDS 4, DLBCL 1, MF 3) received KB004 in the DEP; an additional 14 patients were treated on the CEP (AML 8, MDS 6). The most common toxicities were transient grade 1 and grade 2 infusion reactions (IRs) in 79% of patients. IRs were dose limiting above 250mg. Sustained exposure exceeding the predicted effective concentration (1ug/mL) and covering the 7-day interval between doses was achieved above 190mg. Responses were observed in patients with AML, MF, MDS/MPN and MDS. In this study, KB004 was well tolerated and clinically active when given as a weekly infusion.


Blood Cancer Journal | 2017

Expanded safety analysis of pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, in patients with acute myeloid leukemia and myelodysplastic syndromes

Ronan Swords; Justin M. Watts; Harry P. Erba; Jessica K. Altman; Michael B. Maris; Faiz Anwer; Zhaowei Hua; H. Stein; Hélène M. Faessel; Farhad Sedarati; Bruce J. Dezube; Frank Giles; Bruno C. Medeiros; Daniel J. DeAngelo

Expanded safety analysis of pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, in patients with acute myeloid leukemia and myelodysplastic syndromes


Leukemia Research | 2016

Telomere length and associations with somatic mutations and clinical outcomes in acute myeloid leukemia.

Justin M. Watts; Bogdan Dumitriu; Patrick Hilden; Ashwin Kishtagari; Franck Rapaport; Christina Chen; Jihae Ahn; Sean M. Devlin; Eytan M. Stein; Raajit Rampal; Ross L. Levine; Neal S. Young; Martin S. Tallman

We examined the genetic implications and clinical impact of telomere length (TL) in 67 patients with acute myeloid leukemia (AML). There was a trend toward improved survival at 6 months in patients with longer TL. We found that patients with activating mutations, such as FLT3-ITD, had shorter TL, while those with mutations in epigenetic modifying enzymes, particularly IDH1 and IDH2, had longer TL. These are intriguing findings that warrant further investigation in larger cohorts. Our data show the potential of TL as a predictive biomarker in AML and identify genetic subsets that may be particularly vulnerable to telomere-targeted therapies.


British Journal of Haematology | 2015

Melanoma and non‐melanoma skin cancers in hairy cell leukaemia: a Surveillance, Epidemiology and End Results population analysis and the 30‐year experience at Memorial Sloan Kettering Cancer Center

Justin M. Watts; Ashwin Kishtagari; Meier Hsu; Mario E. Lacouture; Michael A. Postow; Jae H. Park; Eytan M. Stein; Julie Teruya-Feldstein; Omar Abdel-Wahab; Sean M. Devlin; Martin S. Tallman

Few studies have examined melanoma and non‐melanoma skin cancer (NMSC) incidence rates after a diagnosis of hairy cell leukaemia (HCL). We assessed 267 HCL patients treated at Memorial Sloan Kettering Cancer Center (MSKCC) and Surveillance, Epidemiology and End Results (SEER) data for melanoma and NMSC incidence rates after HCL. Incidence data from MSKCC patients demonstrated a 10‐year combined melanoma and NMSC skin cancer rate of 11·3%, melanoma 4·4% and NMSC 6·9%. Molecular analysis of skin cancers from MSKCC patients revealed activating RAS mutations in 3/9 patients, including one patient with melanoma. Of 4750 SEER patients with HCL, 55 (1·2%) had a subsequent diagnosis of melanoma. Standardized incidence ratios (SIRs) did not show that melanoma was more common in HCL patients versus the general population (SIR 1·3, 95% CI 0·78–2·03). Analysis of SEER HCL patients diagnosed before and after 1990 (approximately before and after purine analogue therapy was introduced) showed no evidence of an increased incidence after 1990. A better understanding of any potential association between HCL and skin cancer is highly relevant given ongoing trials using BRAF inhibitors, such as vemurafenib, for relapsed HCL, as RAS‐mutant skin cancers could be paradoxically activated in these patients.


Journal of Clinical Investigation | 2018

Oncogenic TRK fusions are amenable to inhibition in hematologic malignancies

Justin Taylor; Dean Pavlick; Akihide Yoshimi; Christina Marcelus; Stephen S. Chung; Jaclyn F. Hechtman; Ryma Benayed; Emiliano Cocco; Benjamin H. Durham; Lillian Bitner; Daichi Inoue; Young Rock Chung; Kerry Mullaney; Justin M. Watts; Eli L. Diamond; Lee A. Albacker; Tariq I. Mughal; Kevin Ebata; Brian B. Tuch; Nora Ku; Maurizio Scaltriti; Mikhail Roshal; Maria E. Arcila; Siraj M. Ali; David M. Hyman; Jae H. Park; Omar Abdel-Wahab

Rearrangements involving the neurotrophic receptor kinase genes (NTRK1, NTRK2, and NTRK3; hereafter referred to as TRK) produce oncogenic fusions in a wide variety of cancers in adults and children. Although TRK fusions occur in fewer than 1% of all solid tumors, inhibition of TRK results in profound therapeutic responses, resulting in Breakthrough Therapy FDA approval of the TRK inhibitor larotrectinib for adult and pediatric patients with solid tumors, regardless of histology. In contrast to solid tumors, the frequency of TRK fusions and the clinical effects of targeting TRK in hematologic malignancies are unknown. Here, through an evaluation for TRK fusions across more than 7,000 patients with hematologic malignancies, we identified TRK fusions in acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), histiocytosis, multiple myeloma, and dendritic cell neoplasms. Although TRK fusions occurred in only 0.1% of patients (8 of 7,311 patients), they conferred responsiveness to TRK inhibition in vitro and in vivo in a patient-derived xenograft and a corresponding AML patient with ETV6-NTRK2 fusion. These data identify that despite their individual rarity, collectively, TRK fusions are present in a wide variety of hematologic malignancies and predict clinically significant therapeutic responses to TRK inhibition.


Blood Cancer Journal | 2016

Acute leukemia in adult Hispanic Americans: a large-population study

Ronan Swords; J Sznol; Roy Elias; Justin M. Watts; A Zelent; E Martin; Fernando Vargas; S Bethel-Ellison; E Kobetz

Acute leukemia (AL) is a diverse group of clonal hematopoietic disorders that are broadly categorized into two types: acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL).1, 2, 3 AML is further grouped into acute promyelocytic leukemia (APL, a highly curable disease with a unique and pathognomonic genetic lesion) and non-APL AML. ALL is also further lineage classified into B-cell ALL or T-cell ALL. Unlike older classification systems that defined AL according to how leukemia cells looked and stained under the light microscope,1 the current iteration of the World Health Organization (WHO) classification of hematopoietic neoplasms incorporates cytogenetic and molecular data to provide prognostic and therapeutic information of value to the treating clinician.2, 3 When applied to large-population databases, the WHO framework also provides useful insights into the distribution of AL among ethnic groups, informing on the causative factors of AL, which are at this time poorly understood. For example, in the United States, whites have higher rates of AML compared with other groups.4 B-ALL is uncommon in blacks and Asians, whereas Hispanics have the highest incidence rates (IR) of this leukemia.5 AML appears less common in Hispanics when compared with whites, however, APL appears comparatively more common in Hispanics.6 The varied distribution of AL among these ethnic groups suggests that host susceptibility factors are critical determinants of disease in one group, but not in another. The extent to which the environment interacts with these factors is unknown. In Florida, Hispanics comprise 23.6% of the population, with up to 65% of this group residing in South Florida. About 51% of Hispanics in Florida are native born, 49% are foreign born. In 2015, it is estimated that 3930 new cases of AL will be diagnosed.7 Given the known interaction between ethnicity and AL incidence, we sought for the first time, to better understand the epidemiological patterns of AL distribution throughout Florida. Utilizing the Florida Cancer Data System (FCDS), we analyzed the patterns of B-cell ALL, T-cell ALL, non-APL AML and APL AML among Hispanics and non Hispanic Whites.


Modern Pathology | 2018

Myeloid neoplasms with features intermediate between primary myelofibrosis and chronic myelomonocytic leukemia

Jennifer R. Chapman; Julia T. Geyer; Mahsa Khanlari; Adrienne Moul; Carmen E. Casas; Scot T. Connor; Yao Shan Fan; Justin M. Watts; Ronan Swords; Francisco Vega; Attilio Orazi

Monocytosis can develop during disease course in primary myelofibrosis simulating that seen in chronic myelomonocytic leukemia, and should not lead to disease reclassification. In contrast, at presentation, rare cases have clinical, morphologic, and molecular genetic features truly intermediate between primary myelofibrosis and chronic myelomonocytic leukemia. The taxonomy and natural history of these diseases are unclear. We identified cases which either: (1) fulfilled the 2008 World Health Organization criteria for primary myelofibrosis but had absolute monocytosis and, when available, chronic myelomonocytic leukemia-related mutations (ASXL1, SRSF2, TET2) or (2) fulfilled criteria of chronic myelomonocytic leukemia but had megakaryocytic proliferation and atypia, marrow fibrosis, and myeloproliferative-type driver mutations (JAK2, MPL, CALR). Patients with established primary myelofibrosis who developed monocytosis and those with chronic myelomonocytic leukemia with marrow fibrosis were excluded. By combining the pathology databases of two large institutions, six eligible cases were identified. Patients were predominantly male and elderly with monocytosis at diagnosis (average 17.5%/2.3 × 103/μl), organomegaly, primary myelofibrosis-like atypical megakaryocytes admixed with a variable number of chronic myelomonocytic leukemia-like hypolobated forms, variable myelodysplasia, marrow fibrosis and osteosclerosis. All had a normal karyotype and no myelodysplasia-associated cytogenetic abnormalities. Five of the patients in whom a more extensive molecular characterization was performed showed co-mutations involving JAK2 or MPL and ASXL1, SRSF2, TET2, NRAS, and/or KRAS. Disease progression has occurred in all and two have died. Rare patients present with features that overlap between primary myelofibrosis and chronic myelomonocytic leukemia and are thus difficult to classify based on current World Health Organization criteria. Biologically, these cases likely represent primary myelofibrosis with monocytosis, dysplasia, and secondary (non-driver) mutations at presentation. Alternatively, they may represent a true gray zone of neoplasms. Their clinical behavior appears aggressive and innovative therapeutic approaches may be beneficial in this particular subset.


Therapeutic advances in hematology | 2017

Biological and clinical implications of telomere dysfunction in myeloid malignancies

Ashwin Kishtagari; Justin M. Watts

Telomeres at the ends of linear chromosomes protect the genome. Telomeres shorten with each round of cell division, placing a finite limit on cell growth. Telomere attrition is associated with cell senescence and apoptosis. Telomerase, a specialized ribonucleoprotein complex, maintains telomeres homeostasis through repeat addition of telomere sequences to the 3′ telomeric overhang. Telomere biology is closely related to cancer and normal aging. Upregulation of telomerase or activation of the alternative pathway of telomere lengthening is a hallmark of cancer cells, making telomerase an attractive target for cancer therapeutics. In this review, we will discuss telomere biology and the prognostic implications of telomere length in acute myeloid leukemia, and review exciting new investigational approaches using telomerase inhibitors in acute myeloid leukemia and other myeloid malignancies.


International Journal of Molecular Sciences | 2017

A Case of AML Characterized by a Novel t(4;15)(q31;q22) Translocation That Confers a Growth-Stimulatory Response to Retinoid-Based Therapy

Justin M. Watts; Aymee Perez; Lutecia Pereira; Yao Shan Fan; Geoffrey Brown; Francisco Vega; Kevin Petrie; Ronan Swords; Arthur Zelent

Here we report the case of a 30-year-old woman with relapsed acute myeloid leukemia (AML) who was treated with all-trans retinoic acid (ATRA) as part of investigational therapy (NCT02273102). The patient died from rapid disease progression following eight days of continuous treatment with ATRA. Karyotype analysis and RNA-Seq revealed the presence of a novel t(4;15)(q31;q22) reciprocal translocation involving the TMEM154 and RASGRF1 genes. Analysis of primary cells from the patient revealed the expression of TMEM154-RASGRF1 mRNA and the resulting fusion protein, but no expression of the reciprocal RASGRF1-TMEM154 fusion. Consistent with the response of the patient to ATRA therapy, we observed a rapid proliferation of t(4;15) primary cells following ATRA treatment ex vivo. Preliminary characterization of the retinoid response of t(4;15) AML revealed that in stark contrast to non-t(4;15) AML, these cells proliferate in response to specific agonists of RARα and RARγ. Furthermore, we observed an increase in the levels of nuclear RARγ upon ATRA treatment. In summary, the identification of the novel t(4;15)(q31;q22) reciprocal translocation opens new avenues in the study of retinoid resistance and provides potential for a new biomarker for therapy of AML.

Collaboration


Dive into the Justin M. Watts'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ashwin Kishtagari

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge