Network


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

Hotspot


Dive into the research topics where Oriana Borquez-Ojeda is active.

Publication


Featured researches published by Oriana Borquez-Ojeda.


Science Translational Medicine | 2013

CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia.

Renier J. Brentjens; Marco L. Davila; Isabelle Riviere; Jae Park; Xiuyan Wang; Lindsay G. Cowell; Shirley Bartido; Jolanta Stefanski; Clare Taylor; Malgorzata Olszewska; Oriana Borquez-Ojeda; Jinrong Qu; Teresa Wasielewska; Qing He; Yvette Bernal; Ivelise Rijo; Cyrus V. Hedvat; Rachel Kobos; Kevin J. Curran; Peter G. Steinherz; Joseph G. Jurcic; Todd L. Rosenblat; P. Maslak; Mark G. Frattini; Michel Sadelain

Five adults with chemotherapy-refractory B-ALL were induced into molecular remissions after treatment with CD19 CAR-targeted T cells. CARving a Niche for Cancer Immunotherapy Acute lymphoblastic leukemia (ALL) is a cancer of the white blood cells that fend off infection. It’s most common in children but—as with many diseases that primarily affect children—has a much worse prognosis when it affects adults. Adults with relapsed disease have a very low chance of survival, and new therapies are desperately needed. Now, Brentjens et al. test T cells engineered to target CD19, which is expressed on both healthy B lymphocytes and B-ALL cells, in five chemotherapy-refractory adult B-ALL patients. Here, the authors treat patients with the patients’ own T cells altered to express not only CD19 but also a fusion of the costimulatory molecule CD28 with CD3ζ chain—so-called “second-generation chimeric antigen receptor (CAR) T cells.” All patients treated with these cells achieved tumor eradication and complete remission. These CAR T cells were well tolerated, although there was substantial cytokine release in some patients that correlated to tumor burden. These patients were treated with steroid therapy. Long-term follow-up in four of these patients was not possible because the CAR T cell therapy allowed these patients to be eligible for subsequent hematopoietic stem cell transplant (HSCT), which resulted in restored hematopoiesis. The remaining patient experienced a relapse of CD19+ cells that coincided with the lack of persistence of the CAR T cells from circulation. These data suggest that subsequent transfusions should be considered for patients unable to undergo HSCT. Adults with relapsed B cell acute lymphoblastic leukemia (B-ALL) have a dismal prognosis. Only those patients able to achieve a second remission with no minimal residual disease (MRD) have a hope for long-term survival in the context of a subsequent allogeneic hematopoietic stem cell transplantation (allo-HSCT). We have treated five relapsed B-ALL subjects with autologous T cells expressing a CD19-specific CD28/CD3ζ second-generation dual-signaling chimeric antigen receptor (CAR) termed 19-28z. All patients with persistent morphological disease or MRD+ disease upon T cell infusion demonstrated rapid tumor eradication and achieved MRD− complete remissions as assessed by deep sequencing polymerase chain reaction. Therapy was well tolerated, although significant cytokine elevations, specifically observed in those patients with morphologic evidence of disease at the time of treatment, required lymphotoxic steroid therapy to ameliorate cytokine-mediated toxicities. Indeed, cytokine elevations directly correlated to tumor burden at the time of CAR-modified T cell infusions. Tumor cells from one patient with relapsed disease after CAR-modified T cell therapy, who was ineligible for additional allo-HSCT or T cell therapy, exhibited persistent expression of CD19 and sensitivity to autologous 19-28z T cell–mediated cytotoxicity, which suggests potential clinical benefit of additional CAR-modified T cell infusions. These results demonstrate the marked antitumor efficacy of 19-28z CAR-modified T cells in patients with relapsed/refractory B-ALL and the reliability of this therapy to induce profound molecular remissions, forming a highly effective bridge to potentially curative therapy with subsequent allo-HSCT.


Science Translational Medicine | 2014

Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia.

Marco L. Davila; Isabelle Riviere; Xiuyan Wang; Shirley Bartido; Jae Park; Kevin J. Curran; Stephen S. Chung; Jolanta Stefanski; Oriana Borquez-Ojeda; Malgorzata Olszewska; Jinrong Qu; Teresa Wasielewska; Qing He; Mitsu Fink; Himaly Shinglot; Maher Youssif; Mark Satter; Yongzeng Wang; James Hosey; Hilda Quintanilla; Elizabeth Halton; Yvette Bernal; Diana C. G. Bouhassira; Maria E. Arcila; Mithat Gonen; Gail J. Roboz; P. Maslak; Dan Douer; Mark G. Frattini; Sergio Giralt

CD19 CAR T cell therapy induces complete remissions in 88% of 16 adult patients with relapsed or refractory acute lymphoblastic leukemia. CARving Out a Niche for CAR T Cell Immunotherapy Relapsed or refractory B acute lymphoblastic leukemia (B-ALL) in adults has a poor prognosis, with an expected median survival of less than 6 months. An emerging therapy for adult B-ALL is through T cells that target tumor cells with chimeric antigen receptors (CARs). Davila et al. now report the results of a phase 1 clinical trial of CAR T cells in 16 relapsed or refractory adult patients. The CD19-targeting CAR T cell therapy resulted in an 88% complete response rate, which allowed most of the patients to transition to allogeneic hematopoietic stem cell transplantation—the current standard of care. Moreover, the authors carefully characterized cytokine release syndrome (CRS), which is a series of toxicities associated with CAR T cell therapy. They found that serum C-reactive protein (CRP) associated with the severity of CRS, which should allow for identification of the subset of patients who will likely require therapeutic intervention with corticosteroids or interleukin-6 receptor blockade to curb the CRS. This is especially important because treatment for CRS may limit the efficacy of the CAR T cell therapy. These data support the need for further multicenter trials for CAR T cell therapy. We report on 16 patients with relapsed or refractory B cell acute lymphoblastic leukemia (B-ALL) that we treated with autologous T cells expressing the 19-28z chimeric antigen receptor (CAR) specific to the CD19 antigen. The overall complete response rate was 88%, which allowed us to transition most of these patients to a standard-of-care allogeneic hematopoietic stem cell transplant (allo-SCT). This therapy was as effective in high-risk patients with Philadelphia chromosome–positive (Ph+) disease as in those with relapsed disease after previous allo-SCT. Through systematic analysis of clinical data and serum cytokine levels over the first 21 days after T cell infusion, we have defined diagnostic criteria for a severe cytokine release syndrome (sCRS), with the goal of better identifying the subset of patients who will likely require therapeutic intervention with corticosteroids or interleukin-6 receptor blockade to curb the sCRS. Additionally, we found that serum C-reactive protein, a readily available laboratory study, can serve as a reliable indicator for the severity of the CRS. Together, our data provide strong support for conducting a multicenter phase 2 study to further evaluate 19-28z CAR T cells in B-ALL and a road map for patient management at centers now contemplating the use of CAR T cell therapy.


Journal of Immunotherapy | 2009

Manufacturing Validation of Biologically Functional T Cells Targeted to Cd19 Antigen for Autologous Adoptive Cell Therapy

Daniel Hollyman; Jolanta Stefanski; Mark Przybylowski; Shirley Bartido; Oriana Borquez-Ojeda; Clare Taylor; Raymond Yeh; Vanessa L. Capacio; Malgorzata Olszewska; James Hosey; Michel Sadelain; Renier J. Brentjens; Isabelle Riviere

On the basis of promising preclinical data demonstrating the eradication of systemic B-cell malignancies by CD19-targeted T lymphocytes in vivo in severe combined immunodeficient-beige mouse models, we are launching phase I clinical trials in patients with chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia. We present here the validation of the bioprocess which we developed for the production and expansion of clinical grade autologous T cells derived from patients with CLL. We demonstrate that T cells genetically modified with a replication-defective gammaretroviral vector derived from the Moloney murine leukemia virus encoding a chimeric antigen receptor (CAR) targeted to CD19 (1928z) can be expanded with Dynabeads CD3/CD28. This bioprocess allows us to generate clinical doses of 1928z+ T cells in approximately 2 to 3 weeks in a large-scale semiclosed culture system using the Wave Bioreactor. These 1928z+ T cells remain biologically functional not only in vitro but also in severe combined immunodeficient-beige mice bearing disseminated tumors. The validation requirements in terms of T-cell expansion, T-cell transduction with the 1928z CAR, biologic activity, quality control testing, and release criteria were met for all 4 validation runs using apheresis products from patients with CLL. Additionally, after expansion of the T cells, the diversity of the skewed Vβ T-cell receptor repertoire was significantly restored. This validated process will be used in phase I clinical trials in patients with chemorefractory CLL and in patients with relapsed acute lymphoblastic leukemia. It can also be adapted for other clinical trials involving the expansion and transduction of patient or donor T cells using any CAR or T-cell receptor.


Molecular and Cellular Biology | 2004

Vitamin C Is a Kinase Inhibitor: Dehydroascorbic Acid Inhibits IκBα Kinase β

Juan M. Cárcamo; Alicia Pedraza; Oriana Borquez-Ojeda; Bing Zhang; Roberto Sanchez; David W. Golde

ABSTRACT Reactive oxygen species (ROS) are key intermediates in cellular signal transduction pathways whose function may be counterbalanced by antioxidants. Acting as an antioxidant, ascorbic acid (AA) donates two electrons and becomes oxidized to dehydroascorbic acid (DHA). We discovered that DHA directly inhibits IκBα kinase β (IKKβ) and IKKα enzymatic activity in vitro, whereas AA did not have this effect. When cells were loaded with AA and induced to generate DHA by oxidative stress in cells expressing a constitutive active IKKβ, NF-κB activation was inhibited. Our results identify a dual molecular action of vitamin C in signal transduction and provide a direct linkage between the redox state of vitamin C and NF-κB signaling events. AA quenches ROS intermediates involved in the activation of NF-κB and is oxidized to DHA, which directly inhibits IKKβ and IKKα enzymatic activity. These findings define a function for vitamin C in signal transduction other than as an antioxidant and mechanistically illuminate how vitamin C down-modulates NF-κB signaling.


Journal of Biological Chemistry | 1997

Increased Uptake and Accumulation of Vitamin C in Human Immunodeficiency Virus 1-infected Hematopoietic Cell Lines*

Coralia I. Rivas; Juan Carlos Vera; Victor H. Guaiquil; Fernando V. Velásquez; Oriana Borquez-Ojeda; Juan G. Cárcamo; Ilona I. Concha; David W. Golde

Vitamin C (ascorbic acid) is required for normal host defense and functions importantly in cellular redox systems. To define the interrelationship between human immunodeficiency virus (HIV) infection and vitamin C flux at the cellular level, we analyzed vitamin C uptake and its effects on virus production and cellular proliferation in HIV-infected and uninfected human lymphoid, myeloid, and mononuclear phagocyte cell lines. Chronic or acute infection of these cell lines by HIV-1 led to increased expression of glucose transporter 1, associated with increased transport and accumulation of vitamin C. Infected cells also showed increased transport of glucose analogs. Exposure to vitamin C had a complex effect on cell proliferation and viral production. Low concentrations of vitamin C increased or decreased cell proliferation depending on the cell line and either had no effect or caused increased viral production. Exposure to high concentrations of vitamin C preferentially decreased the proliferation and survival of the HIV-infected cells and caused decreased viral production. These findings indicate that HIV infection in lymphocytic, monocytic, and myeloid cell lines leads to increased expression of glucose transporter 1 and consequent increased cellular vitamin C uptake. High concentrations of vitamin C were preferentially toxic to HIV-infected host defense cell lines in vitro.


Proceedings of the National Academy of Sciences of the United States of America | 2003

The laminin receptor modulates granulocyte–macrophage colony-stimulating factor receptor complex formation and modulates its signaling

Jian Chen; Juan M. Cárcamo; Oriana Borquez-Ojeda; Hediye Erdjument-Bromage; Paul Tempst; David W. Golde

Basement membrane matrix proteins are known to up-regulate granulocyte–macrophage colony-stimulating factor (GM-CSF) signaling in neutrophils and mononuclear phagocytes, but the mechanisms involved are poorly understood. We used the intracellular portion of the α subunit of the GM-CSF receptor (αGMR) to search for interacting proteins and identified the 67-kDa laminin receptor (LR), a nonintegrin matrix protein receptor expressed in several types of host defense cells and certain tumors, as a binding partner. LR was found to interact with the β subunit of the GMR (βGMR) as well. Whereas GM-CSF functions by engaging the αGMR and βGMR into receptor complexes, LR inhibited GM-CSF-induced receptor complex formation. Laminin and fibronectin binding to LR was found to prevent the binding of βGMR to LR and relieved the LR inhibition of GMR. These findings provide a mechanistic basis for enhancing host defense cell responsiveness to GM-CSF at transendothelial migration sites while suppressing it in circulation.


Molecular Therapy | 2016

459. Evaluation of Miltenyi ExpAct and TransAct CD3/28 Beads for CAR-T Cell Manufacturing

Xiuyan Wang; Jinrong Qu; Jolanta Stefanski; Oriana Borquez-Ojeda; Anniesha Hack; Qing He; Teresa Wasielewska; Fang Du; Michel Sadelain; Isabelle Riviere

Adoptive transfer of chimeric antigen receptor (CAR) engineered T cells is a promising emerging strategy to treat cancer patients. Large-scale manufacturing of cGMP-grade CAR T cells using patient T cells selected and activated by CTS™ Dynabeads® CD3/CD28 (Dynabeads) followed by transduction with retroviral vectors is being used in the context of many clinical trials by our laboratory and others. Although we have established a robust protocol using Dynabeads, it is important to explore alternative sources to pre-empt supply chain limitations of this critical reagent. To this end, we evaluated T cell activation with either Miltenyi TransAct CD3/28 (TransAct) beads or Miltenyi ExpAct Treg (ExpAct) beads. In small-scale experiments, PBMCs were directly activated with TransAct or ExpAct beads and compared with our standard T cell selection and activation using Dynabeads. Overall, the transduction efficiency and expansion of T cells were comparable upon activation with all three reagents. The TransAct bead-stimulated cells exhibited comparable effector memory (EM)/central memory (CM) phenotype to that of the Dynabeads stimulated cells. In line with the EM/CM phenotype, CAR T cells stimulated with either TransAct or Dynabeads and tranduced with CD19-targeted CAR demonstrated robust and comparable antitumor activity in a systemic NSG/CD19+ NALM6 tumor mouse model. We further tested the efficacy of TransAct beads using positively or negatively selected T cells in a large-scale cGMP grade CAR-T cell manufacturing setting. Both the transduction efficiency and expansion of selected CD3+ cells activated with TransAct beads and Dynabeads were comparable. CD19-targeted CAR T cells activated by either TransAct or Dynabead were subjected to an in vivo stress test by using decreasing amount of CAR-T cells to treat systemic CD19+NALM6 tumors in NSG mice. In this experimental setting, T cells stimulated with TransAct beads demonstrated equivalent if not better anti-tumor activity than T cells stimulated with Dynabeads. In conclusion, our pre-clinical results suggest that TransAct beads support efficient transduction and expansion of CAR T cells. TransAct activated T cells exhibit antitumor activity equivalent to Dynabeads activated T cells in our NSG/CD19+NAML6 stress test. Therefore, Miltenyi TransAct beads can be used as an alternative to Dynabeads to stimulate T cells in clinical trials aiming at evaluating CAR T cell safety and antitumor activity.


Molecular Therapy | 2015

80. Depletion of High-Content CD14+ Cells from Apheresis Products is Critical for the Successful Transduction and Expansion of CAR T Cells During Large-Scale cGMP Manufacturing

Xiuyan Wang; Jinrong Qu; Jolanta Stefanski; Fang Du; Oriana Borquez-Ojeda; Annisha Hack; Isabelle Riviere

Adoptive transfer of chimeric antigen receptor (CAR) engineered T cells holds great promise as a novel strategy to treat patients with cancer. Apheresis products are a major source of starting material for large-scale T cell selection using CD3/28 magnetic beads. At the time of collection, the composition in various cell lineages is highly variable. We have successfully manufactured CAR T cell products starting from apheresis products containing as few as 0.5% to 3% of CD3+ cells. However, we have experienced manufacturing challenges with apheresis products containing a high fraction of CD14+ cells. We conducted a study with an apheresis product derived from a patient with acute lymphoblastic leukemia containing 70% CD14+ cells. 350 × 106 CD3+ cells were selected from the cryopreserved apheresis product using CD3/28 magnetic beads either with or without a prior 1.5 hr monocyte/granulocyte adherence step. This short adherence procedure decreased the CD14+ cell composition from 70% to 45%. A second overnight depletion by adherence in T-flasks and re-stimulation with CD3/28 magnetic beads on day 1 were also performed in the depleted group to ensure the proper activation of CD3+ cells. On day 3, 350×106 vs 20 x106 total cells were recovered from each group without and with the depletion step, respectively. Selected cells from both groups were transduced using the same dilution of vector stocks at a cell concentration of either 0.4×106/mL or 0.2×106/mL in tissue culture bags. On day 7, we observed a drastic difference in transduction efficiency between the group without depletion (4.5%) and the group in which the CD14+ cells were depleted (38%). Moreover, starting with 108 × 106 selected cells on day 3, a mere total of 12.3 × 106 (14.8% CD3+) were recovered on day 11 in the group without depletion. By contrast, cells from the depleted group expanded 350 fold from day 3 to day 10 (100% CD3+). Our results suggest that high CD14+ cell content poses a manufacturing challenge when the selection of the CD3+ cells in the apheresis product is performed with CD3/CD28 magnetic beads. The CD14+ cell threshold for successful manufacturing without depletion remains to be determined. However, monitoring the CD14 cell content and depleting these cells from starting apheresis products with high CD14 content prior to CD3/CD28 magnetic bead selection are critical to ensure success of the CAR T cell manufacturing process.


Cancer Research | 2014

Abstract CT102: Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia

Michel Sadelain; Renier J. Brentjens; Marco L. Davila; Isabelle Riviere; Xiuyan Wang; Shirley Bartido; Jae Park; Diana C. G. Bouhassira; Kevin J. Curran; Stephen S. Chung; Jolanta Stefanski; Oriana Borquez-Ojeda; Sergio Giralt

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Chimeric antigen receptors (CARs) are artificial receptors for antigen that redirect antigen specificity, activate T cells and further enhance T cell function through their costimulatory component. Three groups, including our own, have reported objective tumor responses when infusing autologous T cells genetically modified with CD19-targeted CARs into patients with chronic lymphocytic leukemia (CLL), other indolent non-Hodgkin lymphomas (NHL) and, most dramatically in patients with relapsed or refractory B cell acute lymphoblastic leukemia (B-ALL). Here we report on 16 patients with relapsed or refractory B-ALL that we treated with autologous T cells expressing the 19-28z CAR specific to the CD19 antigen. The overall complete response rate was 88%, as assessed by morphological criteria and IgH deep sequencing. This allowed us to transition a majority of these patients to a standard of care allogeneic hematopoietic stem cell transplant (allo-SCT). This therapy was as effective in high-risk patients with Ph+ disease as in those with relapsed disease following prior allo-SCT. Through systematic analysis of clinical data and serum cytokine levels over the first 21 days following T cell infusion, we have defined diagnostic criteria for a severe cytokine release syndrome (sCRS), with the goal of better identifying the subset of patients who will likely require therapeutic intervention with corticosteroids or interleukin-6 receptor blockade to curb the CRS. Additionally, we found that serum C-reactive protein (CRP), a readily available laboratory study, can serve as a reliable indicator for the severity of the CRS and a road map for patient management at centers now contemplating the use of CAR T cell therapy. Based on these remarkably robust clinical results and the toxicity management algorithm we present here, we will soon open a multi-center Phase II clinical trial to further evaluate the efficacy of 19-28z CAR T cells and prospectively validate our proposed CRS monitoring and intervention guidelines in patients who are treated with CAR therapy. Citation Format: Michel Sadelain, Renier Brentjens, Marco Davila, Isabelle Riviere, Xiuyan Wang, Shirley Bartido, Jae Park, Diana Bouhassira, Kevin Curran, Stephen Chung, Jolanta Stefanski, Oriana Borquez-Ojeda, Sergio Giralt. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. [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 CT102. doi:10.1158/1538-7445.AM2014-CT102


Biochemistry | 2002

Vitamin C Suppresses TNFα-Induced NFκB Activation by Inhibiting IκBα Phosphorylation†

Juan M. Cárcamo; Alicia Pedraza; Oriana Borquez-Ojeda; David W. Golde

Collaboration


Dive into the Oriana Borquez-Ojeda's collaboration.

Top Co-Authors

Avatar

Isabelle Riviere

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jolanta Stefanski

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michel Sadelain

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shirley Bartido

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xiuyan Wang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David W. Golde

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jinrong Qu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Juan M. Cárcamo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Renier J. Brentjens

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Teresa Wasielewska

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge