Network


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

Hotspot


Dive into the research topics where Andrea Arruda is active.

Publication


Featured researches published by Andrea Arruda.


Nature | 2016

A 17-gene stemness score for rapid determination of risk in acute leukaemia.

Stanley W.K. Ng; Amanda Mitchell; James A. Kennedy; Weihsu C. Chen; Jessica McLeod; Narmin Ibrahimova; Andrea Arruda; Andreea C. Popescu; Vikas Gupta; Aaron D. Schimmer; Andre C. Schuh; Karen Yee; Lars Bullinger; Tobias Herold; Dennis Görlich; Thomas Büchner; Wolfgang Hiddemann; Wolfgang E. Berdel; Bernhard Wörmann; Meyling Cheok; Claude Preudhomme; Hervé Dombret; Klaus H. Metzeler; Christian Buske; Bob Löwenberg; Peter W. Zandstra; Mark D. Minden; John E. Dick; Jean C.Y. Wang

Refractoriness to induction chemotherapy and relapse after achievement of remission are the main obstacles to cure in acute myeloid leukaemia (AML). After standard induction chemotherapy, patients are assigned to different post-remission strategies on the basis of cytogenetic and molecular abnormalities that broadly define adverse, intermediate and favourable risk categories. However, some patients do not respond to induction therapy and another subset will eventually relapse despite the lack of adverse risk factors. There is an urgent need for better biomarkers to identify these high-risk patients before starting induction chemotherapy, to enable testing of alternative induction strategies in clinical trials. The high rate of relapse in AML has been attributed to the persistence of leukaemia stem cells (LSCs), which possess a number of stem cell properties, including quiescence, that are linked to therapy resistance. Here, to develop predictive and/or prognostic biomarkers related to stemness, we generated a list of genes that are differentially expressed between 138 LSC+ and 89 LSC− cell fractions from 78 AML patients validated by xenotransplantation. To extract the core transcriptional components of stemness relevant to clinical outcomes, we performed sparse regression analysis of LSC gene expression against survival in a large training cohort, generating a 17-gene LSC score (LSC17). The LSC17 score was highly prognostic in five independent cohorts comprising patients of diverse AML subtypes (n = 908) and contributed greatly to accurate prediction of initial therapy resistance. Patients with high LSC17 scores had poor outcomes with current treatments including allogeneic stem cell transplantation. The LSC17 score provides clinicians with a rapid and powerful tool to identify AML patients who do not benefit from standard therapy and who should be enrolled in trials evaluating novel upfront or post-remission strategies.


Blood Advances | 2017

Impact of genomic alterations on outcomes in myelofibrosis patients undergoing JAK1/2 inhibitor therapy

Jay Y. Spiegel; Caroline Jane McNamara; James A. Kennedy; Tony Panzarella; Andrea Arruda; Tracy Stockley; Mahadeo A. Sukhai; Mariam Thomas; Justyna Bartoszko; Jenny M. Ho; Nancy Siddiq; Dawn Maze; Aaron D. Schimmer; Andre C. Schuh; Hassan Sibai; Karen Yee; Jamie Claudio; Rebecca Devlin; Mark D. Minden; Suzanne Kamel-Reid; Vikas Gupta

In myelofibrosis (MF), driver mutations in JAK2, MPL, or CALR impact survival and progression to blast phase, with the greatest risk conferred by triple-negative status. Subclonal mutations, including mutations in high-molecular risk (HMR) genes, such as ASXL1, EZH2, IDH1/2, and SRSF2 have also been associated with inferior prognosis. However, data evaluating the impact of next-generation sequencing in MF patients treated with JAK1/2 inhibitors are lacking. Using a 54-gene myeloid panel, we performed targeted sequencing on 100 MF patients treated with ruxolitinib (n = 77) or momelotinib (n = 23) and correlated mutational profiles with treatment outcomes. Ninety-nine patients had at least 1 mutation identified, 46 (46%) had 2 mutations, and 34 (34%) patients had ≥3 mutations. Seventy-nine patients carried a mutation in JAK2V617F, 14 patients had mutations in CALR, 6 patients had an MPL mutation, and 2 patients were triple negative. No mutation was significantly associated with spleen or anemia response. A high Dynamic International Prognostic Scoring System score and pretreatment transfusion dependence were associated with a shorter time to treatment failure (TTF), and this association retained significance on multivariable analysis. Patients with ASXL1 (hazard ratio [HR], 1.86; P = .03) and EZH2 mutations (HR, 2.94; P = .009) and an HMR profile (HR, 2.06; P = .01) had shorter TTF. On multivariate analysis, ASXL1 or EZH2 mutations were independently associated with shorter TTF and overall survival. These findings help identify patients unlikely to have a durable response with current JAK1/2 inhibitors and provide a framework for future studies.


Molecular therapy. Methods & clinical development | 2016

Preclinical validation: LV/IL-12 transduction of patient leukemia cells for immunotherapy of AML

Ju Huang; Yuanfeng Liu; Bryan Au; Dwayne L. Barber; Andrea Arruda; Axel Schambach; Michael Rothe; Mark D. Minden; Christopher J. Paige; Jeffrey A. Medin

Interleukin-12 (IL-12) is a potent cytokine that may be harnessed to treat cancer. To date, nearly 100 IL-12-based clinical trials have been initiated worldwide. Yet systemic administration of IL-12 is toxic. Different strategies are being developed to reduce such toxicities by restricting IL-12 distribution. Our previous studies employed lentivector-mediated expression of murine IL-12 in tumor cells and demonstrated effective protection in both mouse leukemia and solid tumor challenge models. In this study, we carried out preclinical validation studies using a novel lentivector to engineer expression of human IL-12 in acute myeloid leukemia blast cells isolated from 21 patients. Acute myeloid leukemia cells were transduced with a bicistronic lentivector that encodes the human IL-12 cDNA as a fusion, as well as a LNGFR (ΔLNGFR)/mutant thymidylate kinase cassette as a marking and cell-fate control element. A range of 20–70% functional transduction efficiencies was achieved. Transduced acute myeloid leukemia cells produced bioactive IL-12 protein and displayed dose-dependent sensitivity to the prodrug 3′-azido-3′-deoxythymidine. In vitro immortalization assays using transduced mouse hematopoietic stem cells demonstrated minimal genotoxic risk from our IL-12 vector. Scale-up transduction and cell processing was subsequently validated in a GMP facility to support our (now approved) Clinical Trial Application (CTA).


Clinical Cancer Research | 2018

Allogeneic Human Double Negative T Cells as a Novel Immunotherapy for Acute Myeloid Leukemia and Its Underlying Mechanisms

Jong Bok Lee; Mark D. Minden; Weihsu C. Chen; Elena Streck; Branson Chen; Hyeonjeong Kang; Andrea Arruda; Dalam Ly; Sandy D. Der; Sohyeong Kang; Paulina Achita; Cheryl D'Souza; Yueyang Li; Richard Childs; John E. Dick; Li Zhang

Purpose: To explore the potential of ex vivo expanded healthy donor–derived allogeneic CD4 and CD8 double-negative cells (DNT) as a novel cellular immunotherapy for leukemia patients. Experimental Design: Clinical-grade DNTs from peripheral blood of healthy donors were expanded and their antileukemic activity and safety were examined using flow cytometry–based in vitro killing assays and xenograft models against AML patient blasts and healthy donor–derived hematopoietic cells. Mechanism of action was investigated using antibody-mediated blocking assays and recombinant protein treatment assays. Results: Expanded DNTs from healthy donors target a majority (36/46) of primary AML cells, including 9 chemotherapy-resistant patient samples in vitro, and significantly reduce the leukemia load in patient-derived xenograft models in a DNT donor–unrestricted manner. Importantly, allogeneic DNTs do not attack normal hematopoietic cells or affect hematopoietic stem/progenitor cell engraftment and differentiation, or cause xenogeneic GVHD in recipients. Mechanistically, DNTs express high levels of NKG2D and DNAM-1 that bind to cognate ligands preferentially expressed on AML cells. Upon recognition of AML cells, DNTs rapidly release IFNγ, which further increases NKG2D and DNAM-1 ligands’ expression on AML cells. IFNγ pretreatment enhances the susceptibility of AML cells to DNT-mediated cytotoxicity, including primary AML samples that are otherwise resistant to DNTs, and the effect of IFNγ treatment is abrogated by NKG2D and DNAM-1–blocking antibodies. Conclusions: This study supports healthy donor–derived allogeneic DNTs as a therapy to treat patients with chemotherapy-resistant AML and also reveals interrelated roles of NKG2D, DNAM-1, and IFNγ in selective targeting of AML by DNTs. Clin Cancer Res; 24(2); 370–82. ©2017 AACR.


Molecular Therapy | 2015

413. Pre-Clinical Preparation and Validation of Tumor Cell-Based IL-12 Immunotherapy for Acute Myeloid Leukemia

Bryan Au; Yuanfeng Liu; Ju Huang; Megan Nelles; Andrea Arruda; Michael Rothe; Gabi Paul; Axel Schambach; Dwayne L. Barber; Mark D. Minden; Christopher J. Paige; Jeffrey A. Medin

Interleukin(IL)-12 is a potent pro-inflammatory cytokine that stimulates a variety of effector cells involved in anti-tumor immunity. Systemic administration of IL-12 has associated toxicities, however. Various strategies are being developed to reduce such toxicities by restricting IL-12 distribution. Options here include generating fusions with tumor-targeting molecules and directing gene delivery to specific cells. First – we used lentivirus vectors (LVs) to engineer expression of murine IL-12 in tumor cells ex vivo and subsequently infused such modified cells into recipient mice. This strategy restricts IL-12 to the local tumor microenvironment thereby promoting immune activation in the context of tumor-associated antigens (TAAs). Using mouse models of both leukemia and solid tumors, we found that this cell-based approach generated effective anti-tumor protection when as little as 1% of the tumor burden expressed IL-12 as long as threshold expression levels on a per cell basis were reached. Second – our groups showed that anti-tumor mechanisms here involve CD4+ killer T cells, dendritic cells, and direct cell-cell contact with effectors. Clinical translation of this cell-based IL-12 therapy is in progress in Toronto for AML using a novel LV to modify patients’ own blast cells. Patient AML cells collected to date (n = 21) were stratified based on in vivo growth kinetics and transduced with a near-GMP-grade bicistronic LV that encodes the human IL-12 cDNA as a p40-p70 fusion, as well as a mutant thymidylate kinase (tmpK) fused to the ectodomain of LNGFR (trLNGFR) as a suicide (cell-fate control) cassette. The trLNGFR/tmpK element allows selection and also selective ablation of transduced cells by administration of AZT. Furthermore, it also allows tracking of transduced cells and quantification of transduction frequencies/transgene expression levels. With our current protocol, functional transduction efficiencies of primary patient AML blasts ranged from 20% to 70% (n=17). Transduced AML cells displayed a strong correlation between vector copy number and trLNGFR/tmpK + IL-12 levels and dose-dependent sensitivity to AZT. In vitro immortalization (IVIM) assays determined that the near-GMP LV/IL-12 vector displayed minimal genotoxic risk in transduced lin- cells; insertion site analyses carried out on expanded clones displayed poly-to oligo-clonality patterns. Pre-clinical data on toxicity and scale-up considerations are being accumulated in preparation for a Clinical Trial Application to Health Canada targeting AML. This LV/IL-12 immunotherapy platform targeting tumor cells themselves thus holds potential to be effective against a wide variety of cancers.


Blood Advances | 2018

The mutational landscape of accelerated- and blast-phase myeloproliferative neoplasms impacts patient outcomes

Caroline Jane McNamara; Tony Panzarella; James A. Kennedy; Andrea Arruda; Jaime O. Claudio; Georgina Daher-Reyes; Jenny M. Ho; Nancy Siddiq; Rebecca Devlin; Hubert Tsui; Jie Su; Tracy L. Stockley; Mahadeo A. Sukhai; Nisha Kanwar; Steven M. Chan; Dawn Maze; Aaron D. Schimmer; Andre C. Schuh; Hassan Sibai; Auro Viswabandya; Karen Yee; Mark D. Minden; Suzanne Kamel-Reid; Vikas Gupta

There is a paucity of data regarding the impact of mutations on outcomes in accelerated-phase (AP) and blast-phase (BP) myeloproliferative neoplasms (MPNs). Moreover, it is unknown whether mutational status affects survival, as seen in chronic-phase MPNs. Therefore, we performed a retrospective analysis of all patients treated at our institution with AP/BP MPNs (N = 122; AP = 14; BP = 108) to comprehensively describe the mutational profile and correlate with clinical outcomes. Targeted sequencing with a 54-gene panel was performed. Forty-four patients were treated with intensive therapy, 27 with nonintensive therapy, and 51 with best supportive care (BSC). The most common mutation was JAK2V617F, occurring in 55% of subjects; CALR was found in 13% of patients and MPL in 6%. Thirty-two (26%) patients were triple negative. Other frequently mutated genes were ASXL1 (30%), TET2 (25%), SRSF2 (22%), RUNX1 (20%), and TP53 (17%). Mutations in 1, 2, 3, and ≥4 genes were seen in 15%, 13%, 25%, and 46% of patients, respectively. There was no difference in survival between patients treated with intensive vs nonintensive therapy, and the benefit of intensive therapy was limited to patients who were able to undergo transplantation. TP53 was the only individual mutation to correlate with shorter overall survival (hazard ratio, 1.89; P = .03). In the multivariate analysis, mutated TP53, ≥4 mutations, low albumin, increased peripheral blood blasts, ≥3 cytogenetic abnormalities, and BSC were associated with shorter survival. In conclusion, mutational data enhance the understanding of patients with AP/BP MPN who are likely to benefit from current therapeutic options.


Blood | 2018

Distinct patterns of clonal evolution in patients with concurrent myelo- and lymphoproliferative neoplasms

James A. Kennedy; Jessie J. F. Medeiros; Stephanie M. Dobson; Andrea Arruda; Mahadeo A. Sukhai; Tracy L. Stockley; Anne Tierens; Mark D. Minden; Suzanne Kamel-Reid; John E. Dick; Vikas Gupta

TO THE EDITOR: BCR-ABL -negative myeloproliferative neoplasms (MPNs) are a group of clonal stem cell disorders characterized by the overproduction of mature myeloid cells. However, MPN patients also have a 2.8- to 3.4-fold increased risk of developing a lymphoproliferative disorder (LPD) compared


Blood | 2013

Leukemic Engraftment In NOD. SCID Mice Is Correlated With Clinical Parameters and Predicts Outcome In Human AML

Amanda Mitchell; Weihsu Claire Chen; Jessica McLeod; Andreea C. Popescu; Andrea Arruda; Mark D. Minden; John E. Dick; Jean C.Y. Wang


HemaSphere | 2018

Clinical Utility of Next-generation Sequencing in the Management of Myeloproliferative Neoplasms: A Single-Center Experience

Waleed Alduaij; Caroline Jane McNamara; Andre C. Schuh; Andrea Arruda; Mahadeo A. Sukhai; Nisha Kanwar; Mariam Thomas; Jay Y. Spiegel; James A. Kennedy; Tracy Stockley; Hubert Tsui; Rebecca Devlin; Hassan Sibai; Dawn Maze; Aaron D. Schimmer; Karen Yee; Steven M. Chan; Suzanne Kamel-Reid; Vikas Gupta


Blood | 2016

Impact of Genomic Alterations on Outcomes in Myelofibrosis Patients Undergoing JAK1/2 Inhibitor Therapy

Jay Y. Spiegel; Caroline Jane McNamara; Andrea Arruda; Tony Panzarella; James A. Kennedy; Tracy L. Stockley; Mahadeo A. Sukhai; Mariam Thomas; Justyna Bartoszko; Jenny M. Ho; Nancy Siddiq; Aaron D. Schimmer; Andre C. Schuh; Hassan Sibai; Karen Yee; Jaime O. Claudio; Rebecca Devlin; Mark D. Minden; Suzanne Kamel-Reid; Vikas Gupta

Collaboration


Dive into the Andrea Arruda's collaboration.

Top Co-Authors

Avatar

Mark D. Minden

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vikas Gupta

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Andre C. Schuh

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

John E. Dick

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Karen Yee

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Mahadeo A. Sukhai

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Suzanne Kamel-Reid

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Aaron D. Schimmer

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Caroline Jane McNamara

Princess Margaret Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge