Network


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

Hotspot


Dive into the research topics where Parul Doshi is active.

Publication


Featured researches published by Parul Doshi.


Transfusion | 2015

Resolving the daratumumab interference with blood compatibility testing

Claudia I. Chapuy; Rachel T. Nicholson; Maria Aguad; Bjoern Chapuy; Jacob P. Laubach; Paul G. Richardson; Parul Doshi; Richard M. Kaufman

Daratumumab (DARA), a promising novel therapy for multiple myeloma, is an IgG1κ monoclonal antibody that recognizes CD38 on myeloma cells. During routine compatibility testing, we observed that the plasma of five of five DARA‐treated patients demonstrated a positive antibody screen and panreactivity on red blood cell (RBC) panel testing. We hypothesized that the observed panreactivity reflected DARA binding to CD38 on reagent RBCs, and we investigated methods to prevent this binding.


Leukemia | 2015

Upregulation of CD38 expression on multiple myeloma cells by all- trans retinoic acid improves the efficacy of daratumumab

Inger S. Nijhof; Richard W.J. Groen; H M Lokhorst; B. van Kessel; Andries C. Bloem; J.F. van Velzen; R de Jong-Korlaar; H. Yuan; Willy Noort; Saskia K. Klein; Anton Martens; Parul Doshi; Kate Sasser; T Mutis; N W C J van de Donk

Daratumumab is an anti-CD38 monoclonal antibody with lytic activity against multiple myeloma (MM) cells, including ADCC (antibody-dependent cellular cytotoxicity) and CDC (complement-dependent cytotoxicity). Owing to a marked heterogeneity of response to daratumumab therapy in MM, we investigated determinants of the sensitivity of MM cells toward daratumumab-mediated ADCC and CDC. In bone marrow samples from 144 MM patients, we observed no difference in daratumumab-mediated lysis between newly diagnosed or relapsed/refractory patients. However, we discovered, next to an expected effect of effector (natural killer cells/monocytes) to target (MM cells) ratio on ADCC, a significant association between CD38 expression and daratumumab-mediated ADCC (127 patients), as well as CDC (56 patients). Similarly, experiments with isogenic MM cell lines expressing different levels of CD38 revealed that the level of CD38 expression is an important determinant of daratumumab-mediated ADCC and CDC. Importantly, all-trans retinoic acid (ATRA) increased CD38 expression levels but also reduced expression of the complement-inhibitory proteins CD55 and CD59 in both cell lines and primary MM samples. This resulted in a significant enhancement of the activity of daratumumab in vitro and in a humanized MM mouse model as well. Our results provide the preclinical rationale for further evaluation of daratumumab combined with ATRA in MM patients.


Transfusion | 2015

When blood transfusion medicine becomes complicated due to interference by monoclonal antibody therapy

Marlies Oostendorp; Jeroen J. Lammerts van Bueren; Parul Doshi; Imran Khan; Tahamtan Ahmadi; Paul Parren; Wouter W. van Solinge; Karen M.K. de Vooght

Monoclonal antibodies (MoAbs) are increasingly integrated in the standard of care. The notion that therapeutic MoAbs can interfere with clinical laboratory tests is an emerging concern that requires immediate recognition and the development of appropriate solutions. Here, we describe that treatment of multiple myeloma patients with daratumumab, a novel anti‐CD38 MoAb, resulted in false‐positive indirect antiglobulin tests (IATs) for all patients for 2 to 6 months after infusion. This precluded the correct identification of irregular blood group antibodies for patients requiring blood transfusion.


Journal of Thoracic Oncology | 2014

Antitumor efficacy of the anti-interleukin-6 (IL-6) antibody siltuximab in mouse xenograft models of lung cancer

Lanxi Song; Matthew A. Smith; Parul Doshi; Kate Sasser; William J. Fulp; Soner Altiok; Eric B. Haura

Introduction: Interleukin-6 (IL-6) can activate downstream signaling pathways in lung cancer cells, such as the STAT3 pathway, and is reported to be produced by tumor cells with activating EGFR mutations. We examined IL-6/STAT3 in lung cancer tumor tissues and the effects of siltuximab, a neutralizing antibody to human IL-6, in mouse models of lung cancer. Methods: IL-6 and STAT3 activation levels were compared with tumor histology and presence of KRAS mutations in snap-frozen, non–small-cell lung cancer tumors. The effects of siltuximab alone or in combination with erlotinib were examined in mouse xenograft models constructed using three cell line xenograft models and one primary explant mouse model. We examined the influence of cancer-associated fibroblasts (CAFs) on tumor growth and siltuximab effects. Results: IL-6 levels were higher in tumors of squamous cell versus adenocarcinoma histology and were not associated with presence of KRAS mutations. Tyrosine phosphorylation status of STAT3 did not correlate with tumor IL-6 levels. Serine phosphorylation of STAT3 was correlated with KRAS mutation status. Both tumor and stromal cells contributed to total IL-6 within tumors. Siltuximab had minimal effect as a single agent in xenografts with tumor cells alone; however, in models coadministered with CAFs, siltuximab had more potent effects on tumor inhibition. We observed no effects of combined erlotinib and siltuximab. Conclusions: IL-6 is elevated in subsets of human NSCLCs, especially with squamous cell histology. Tumors supported by stromal production of IL-6 seem to be the most vulnerable to tumor growth inhibition by siltuximab.


Molecular Oncology | 2014

Enhancement of paclitaxel and carboplatin therapies by CCL2 blockade in ovarian cancers

Francois Moisan; Edgar B. Francisco; Anamaria Brozovic; George E. Duran; Yan C. Wang; Shalini Chaturvedi; Shobha Seetharam; Linda A. Snyder; Parul Doshi; Branimir I. Sikic

Ovarian cancer is associated with a leukocyte infiltrate and high levels of chemokines such as CCL2. We tested the hypothesis that CCL2 inhibition can enhance chemotherapy with carboplatin and paclitaxel. Elevated CCL2 expression was found in three non‐MDR paclitaxel resistant ovarian cancer lines ES‐2/TP, MES‐OV/TP and OVCAR‐3/TP, compared to parental cells. Mice xenografted with these cells were treated with the anti‐human CCL2 antibody CNTO 888 and the anti‐mouse MCP‐1 antibody C1142, with and without paclitaxel or carboplatin. Our results show an additive effect of CCL2 blockade on the efficacy of paclitaxel and carboplatin. This therapeutic effect was largely due to inhibition of mouse stromal CCL2. We show that inhibition of CCL2 can enhance paclitaxel and carboplatin therapy of ovarian cancer.


Transfusion | 2016

International validation of a dithiothreitol (DTT)-based method to resolve the daratumumab interference with blood compatibility testing

Claudia I. Chapuy; Maria Aguad; Rachel T. Nicholson; James P. AuBuchon; Claudia S. Cohn; Meghan Delaney; Mark K. Fung; Meredith Unger; Parul Doshi; Michael F. Murphy; Larry J. Dumont; Richard M. Kaufman

Daratumumab (DARA) consistently interferes with routine blood bank serologic testing by directly binding to CD38 expressed on reagent red blood cells (RBCs). Treating RBCs with dithiothreitol (DTT) eliminates the DARA interference. We conducted an international, multicenter, blinded study aimed at validating the DTT method for use by blood bank laboratories worldwide.


Clinical Cancer Research | 2017

The Human CD38 Monoclonal Antibody Daratumumab Shows Antitumor Activity and Hampers Leukemia-Microenvironment Interactions in Chronic Lymphocytic Leukemia.

Alba Matas-Céspedes; Anna Vidal-Crespo; Vanina Rodriguez; Neus Villamor; Julio Delgado; Eva Giné; Heleia Roca-Ho; Pablo Menendez; Elias Campo; Armando López-Guillermo; Dolors Colomer; Gaël Roué; Adrian Wiestner; Paul Parren; Parul Doshi; Jeroen J. Lammerts van Bueren; Patricia Pérez-Galán

Purpose: To establish a proof-of-concept for the efficacy of the anti-CD38 antibody daratumumab in the poor prognosis CD38+ chronic lymphocytic leukemia (CLL) subtype. Experimental Design: The mechanism of action of daratumumab was assessed in CLL primary cells and cell lines using peripheral blood mononuclear cells to analyze antibody-dependent cell cytotoxicity (ADCC), murine and human macrophages to study antibody-dependent cell phagocytosis (ADCP), or human serum to analyze complement-dependent cytotoxicity (CDC). The effect of daratumumab on CLL cell migration and adhesion to extracellular matrix was characterized. Daratumumab activity was validated in two in vivo models. Results: Daratumumab demonstrated efficient lysis of patient-derived CLL cells and cell lines by ADCC in vitro and ADCP both in vitro and in vivo whereas exhibited negligible CDC in these cells. To demonstrate the therapeutic effect of daratumumab in CLL, we generated a disseminated CLL mouse model with the CD38+ MEC2 cell line and CLL patient–derived xenografts (CLL-PDX). Daratumumab significantly prolonged overall survival of MEC2 mice, completely eliminated cells from the infiltrated organs, and significantly reduced disease burden in the spleen of CLL-PDX. The effect of daratumumab on patient-derived CLL cell dissemination was demonstrated in vitro by its effect on CXCL12-induced migration and in vivo by interfering with CLL cell homing to spleen in NSG mice. Daratumumab also reduced adhesion of CLL cells to VCAM-1, accompanied by downregulation of the matrix metalloproteinase MMP9. Conclusions: These unique and substantial effects of daratumumab on CLL viability and dissemination support the investigation of its use in a clinical setting of CLL. Clin Cancer Res; 23(6); 1493–505. ©2016 AACR.


OncoImmunology | 2017

Mammary tumor-derived CCL2 enhances pro-metastatic systemic inflammation through upregulation of IL1β in tumor-associated macrophages

Kelly Kersten; Seth B. Coffelt; Marlous Hoogstraat; Niels J.M. Verstegen; Kim Vrijland; Metamia Ciampricotti; Chris W. Doornebal; Cheei-Sing Hau; Max D. Wellenstein; Camilla Salvagno; Parul Doshi; Esther H. Lips; Lodewyk F. A. Wessels; Karin E. de Visser

ABSTRACT Patients with primary solid malignancies frequently exhibit signs of systemic inflammation. Notably, elevated levels of neutrophils and their associated soluble mediators are regularly observed in cancer patients, and correlate with reduced survival and increased metastasis formation. Recently, we demonstrated a mechanistic link between mammary tumor-induced IL17-producing γδ T cells, systemic expansion of immunosuppressive neutrophils and metastasis formation in a genetically engineered mouse model for invasive breast cancer. How tumors orchestrate this systemic inflammatory cascade to facilitate dissemination remains unclear. Here we show that activation of this cascade relies on CCL2-mediated induction of IL1β in tumor-associated macrophages. In line with these findings, expression of CCL2 positively correlates with IL1Β and macrophage markers in human breast tumors. We demonstrate that blockade of CCL2 in mammary tumor-bearing mice results in reduced IL17 production by γδ T cells, decreased neutrophil expansion and enhanced CD8+ T cell activity. These results highlight a new role for CCL2 in facilitating the breast cancer-induced pro-metastatic systemic inflammatory γδ T cell – IL17 – neutrophil axis.


Haematologica | 2016

Sepantronium Bromide (YM155) improves daratumumab-mediated cellular lysis of multiple myeloma cells by abrogation of bone marrow stromal cell-induced resistance.

Sanne J. de Haart; Lisa Holthof; Willy Noort; Monique C. Minnema; Maarten Emmelot; Tineke Aarts-Riemens; Parul Doshi; Kate Sasser; Huipin Yuan; Joost D. de Bruijn; Anton Martens; Niels W.C.J. van de Donk; Henk M. Lokhorst; Richard W.J. Groen; Tuna Mutis

Targeted immunotherapy with the human anti-CD38 antibody daratumumab has recently emerged as a promising strategy for the treatment of multidrug resistant patients with multiple myeloma (MM). In experimental in vitro and in vivo settings, and in recently completed clinical trials, daratumumab


Clinical Lymphoma, Myeloma & Leukemia | 2018

Blood Transfusion Management and Transfusion-Related Outcomes in Daratumumab-Treated Patients With Relapsed or Refractory Multiple Myeloma

Ajai Chari; Suzanne Arinsburg; Sundar Jagannath; Toshihisa Satta; Ivey Treadwell; Donna Catamero; Gillian Morgan; Huaibao Feng; Clarissa Uhlar; Imran Khan; Parul Doshi; Saad Z Usmani

Introduction Daratumumab, a human CD38 monoclonal antibody approved for multiple myeloma (MM) treatment, binds red blood cells (RBCs), resulting in panagglutination in compatibility tests. Published mitigation methods avoid additional testing, ensuring timely release of blood products. Blood transfusion management and transfusion‐related outcomes of daratumumab‐treated patients in the SIRIUS study are reported, with emphasis on 2 clinical sites. Patients and Methods Patients had MM treated with ≥ 3 prior lines of therapy, including a proteasome inhibitor and an immunomodulatory drug, or were refractory to a proteasome inhibitor and an immunomodulatory drug. RBC typing and alloantibody screening were performed in gel cards. Antibody identification using RBC panels was performed on patients with positive antibody screens. Hematology panels and serum chemistry were analyzed ≤ 2 days before each daratumumab infusion and the first daratumumab dose within each treatment cycle, respectively. Pre‐ and posttransfusion hemoglobin values were analyzed retrospectively. Results At clinical cutoff, patients received 236 transfusions; 47 (37.9%) of 124 patients received 147 packed RBC transfusions, and 17 (13.7%) received 89 platelet transfusions. No hemolysis was reported, and 1 platelet transfusion reaction was observed. At Mount Sinai, no transfusion adverse events were observed, no new unexpected RBC alloantibodies were identified, and transfusions increased hemoglobin values (median, 1.2 g/dL). At Levine Cancer Institute, 6 of 7 patients responded to transfusions, with a median hemoglobin change of 1.7 g/dL. Conclusion In SIRIUS, no RBC transfusion reactions, including hemolysis, were observed. Observations from Mount Sinai and Levine Cancer Institute confirm that transfusions may be administered safely to daratumumab‐treated patients. Graphical Abstract Figure. No Caption available. Micro‐Abstract Daratumumab binds CD38 on red blood cells (RBCs), resulting in panagglutination in blood compatibility tests. RBC transfusions and transfusion‐related adverse events (AEs) from the phase 2 SIRIUS clinical trial were analyzed and, an in‐depth analysis from 2 clinical study sites were performed. RBC transfusion–related AEs, including hemolysis, were not observed among daratumumab‐treated patients, confirming that transfusions may be safely administered.

Collaboration


Dive into the Parul Doshi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ajai Chari

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Aguad

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge