Michelle Kuhne
Bristol-Myers Squibb
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Michelle Kuhne.
Clinical Cancer Research | 2013
Michelle Kuhne; Tanya Mulvey; Blake Belanger; Sharline Chen; Chin Pan; Colin Chong; Fei Cao; Wafa Niekro; Tom Kempe; Karla A. Henning; Lewis J. Cohen; Alan J. Korman; Pina M. Cardarelli
Purpose: CXCR4 has been identified as a prognostic marker for acute myeloid leukemia (AML) and other malignancies. We describe the development and characterization of a fully human antibody to CXCR4 and its application for therapy of AML, non–Hodgkin lymphoma (NHL), chronic lymphoid leukemia (CLL), and multiple myeloma. Experimental Design: Human transgenic mice were immunized with CXCR4-expressing cells, and antibodies reactive with CXCR4 were analyzed for apoptosis induction and ability to interfere with CXCL12-induced migration and calcium flux. In vivo efficacy was determined in multiple AML, NHL, and multiple myeloma xenograft tumors in severe combined immunodeficient mice. Results: BMS-936564/MDX-1338 is a fully human IgG4 monoclonal antibody that specifically recognizes human CXCR4. In vitro studies show that MDX-1338 binds to CXCR4-expressing cells with low nanomolar affinity, blocks CXCL12 binding to CXCR4-expressing cells, and inhibits CXCL12-induced migration and calcium flux with low nanomolar EC50 values. When given as monotherapy, MDX-1338 exhibits antitumor activity in established tumors including AML, NHL, and multiple myeloma xenograft models. In addition, we show that MDX-1338 induced apoptosis on a panel of cell lines and propose that antibody-induced apoptosis is one of the mechanisms of tumor growth inhibition. Conclusions: BMS-936564/MDX-1338 is a potent CXCR4 antagonist which is efficacious as monotherapy in tumor-bearing mice and is currently in phase I for the treatment of relapsed/refractory AML, NHL, CLL, and multiple myeloma. Clin Cancer Res; 19(2); 357–66. ©2012 AACR.
Blood | 2014
Aldo M. Roccaro; Antonio Sacco; Cristina Jiménez; Patricia Maiso; Michele Moschetta; Yuji Mishima; Yosra Aljawai; Ilyas Sahin; Michelle Kuhne; Pina M. Cardarelli; Lewis J. Cohen; Jesús F. San Miguel; Ramón García-Sanz; Irene M. Ghobrial
The C-X-C chemokine receptor type 4 (CXCR4) plays a crucial role in modulating cell trafficking in hematopoietic stem cells and clonal B cells. We screened 418 patients with B-cell lymphoproliferative disorders and described the presence of the C1013G/CXCR4 warts, hypogammaglobulinemia, infections, and myelokathexis-associated mutation in 28.2% (37/131) of patients with lymphoplasmacytic lymphoma (Waldenström macroglobulinemia [WM]), being either absent or present in only 7% of other B-cell lymphomas. In vivo functional characterization demonstrates its activating role in WM cells, as demonstrated by significant tumor proliferation and dissemination to extramedullary organs, leading to disease progression and decreased survival. The use of a monoclonal antibody anti-CXCR4 led to significant tumor reduction in a C1013G/CXCR4 WM model, whereas drug resistance was observed in mutated WM cells exposed to Brutons tyrosine kinase, mammalian target of rapamycin, and phosphatidylinositol 3-kinase inhibitors, but not proteasome inhibitors. These findings demonstrate that C1013G/CXCR4 is an activating mutation in WM and support its role as a critical regulator of WM molecular pathogenesis and as an important therapeutic target.
Cell Reports | 2015
Aldo M. Roccaro; Yuji Mishima; Antonio Sacco; Michele Moschetta; Yu-Tzu Tai; Jiantao Shi; Yong Zhang; Michaela R. Reagan; Daisy Huynh; Yawara Kawano; Ilyas Sahin; Marco Chiarini; Salomon Manier; Michele Cea; Yosra Aljawai; Siobhan Glavey; Elizabeth A. Morgan; Chin Pan; Franziska Michor; Pina M. Cardarelli; Michelle Kuhne; Irene M. Ghobrial
Extra-medullary disease (EMD) in multiple myeloma (MM) is associated with poor prognosis and resistance to chemotherapy. However, molecular alterations that lead to EMD have not been well defined. We developed bone marrow (BM)- and EMD-prone MM syngeneic cell lines; identified that epithelial-to-mesenchymal transition (EMT) transcriptional patterns were significantly enriched in both clones compared to parental cells, together with higher levels of CXCR4 protein; and demonstrated that CXCR4 enhanced the acquisition of an EMT-like phenotype in MM cells with a phenotypic conversion for invasion, leading to higher bone metastasis and EMD dissemination in vivo. In contrast, CXCR4 silencing led to inhibited tumor growth and reduced survival. Ulocuplumab, a monoclonal anti-CXCR4 antibody, inhibited MM cell dissemination, supported by suppression of the CXCR4-driven EMT-like phenotype. These results suggest that targeting CXCR4 may act as a regulator of EMD through EMT-like transcriptional modulation, thus representing a potential therapeutic strategy to prevent MM disease progression.
Clinical Cancer Research | 2009
Pina M. Cardarelli; Maria-Cristina Moldovan-Loomis; Ben Preston; Amelia Black; David Passmore; Tseng-Hui Chen; Sharline Chen; Jie Liu; Michelle Kuhne; Mohan Srinivasan; Albert Assad; Alison Witte; Robert F. Graziano; David John King
Purpose: This study was undertaken to evaluate the effects of MDX-1401, a nonfucosylated fully human monoclonal antibody that binds to human CD30, and to determine whether it exhibits greater in vitro and in vivo activity than its parental antibody. Experimental Design: Assays measuring antibody binding to CD30-expressing cells and FcγRIIIa (CD16) transfectants as well as antibody-dependent cellular cytotoxicity (ADCC) were conducted. Antitumor activity was determined using a Karpas-299 systemic model. Results: The binding of MDX-1401 to CD30 antigen was identical to fucose-containing parental anti-CD30 antibody (MDX-060). In contrast, MDX-1401 showed increased binding affinity to FcγRIIIa-transfected cells resulting in increased effector function. MDX-1401 greatly improved ADCC activity as evidenced by a decrease in half-maximal effective concentration (EC50) and an increase in maximum cell lysis when compared with MDX-060. Increased ADCC activity was observed among a panel of cell lines, including one with very low CD30 antigen expression in which parental antibody failed to induce any detectable ADCC. MDX-1401 activity with all FcγRIIIa polymorphic variants, including less active Phe/Phe158 and Phe/Val158 effector cells, was shown. Furthermore, MDX-1401 was efficacious in inhibiting tumor growth in CD30+ lymphoma xenografts. Conclusions: The low doses of antibody required for ADCC activity irrespective of donor genotype, the ability to mediate ADCC in target cells expressing low levels of CD30, and increased in vivo efficacy support the development of MDX-1401 for treatment of malignant lymphoma.
Oncotarget | 2016
Manoj Kumar Kashyap; Deepak Kumar; Harrison Jones; Carlos I. Amaya-Chanaga; Michael Y. Choi; Johanna Melo-Cardenas; Amine Ale-Ali; Michelle Kuhne; Peter Sabbatini; Lewis J. Cohen; Suresh Shelat; Laura Z. Rassenti; Thomas J. Kipps; Pina M. Cardarelli; Januario E. Castro
The CXCR4 receptor (Chemokine C-X-C motif receptor 4) is highly expressed in different hematological malignancies including chronic lymphocytic leukemia (CLL). The CXCR4 ligand (CXCL12) stimulates CXCR4 promoting cell survival and proliferation, and may contribute to the tropism of leukemia cells towards lymphoid tissues. Therefore, strategies targeting CXCR4 may constitute an effective therapeutic approach for CLL. To address that question, we studied the effect of Ulocuplumab (BMS-936564), a fully human IgG4 anti-CXCR4 antibody, using a stroma – CLL cells co-culture model. We found that Ulocuplumab (BMS-936564) inhibited CXCL12 mediated CXCR4 activation-migration of CLL cells at nanomolar concentrations. This effect was comparable to AMD3100 (Plerixafor - Mozobil), a small molecule CXCR4 inhibitor. However, Ulocuplumab (BMS-936564) but not AMD3100 induced apoptosis in CLL at nanomolar concentrations in the presence or absence of stromal cell support. This pro-apoptotic effect was independent of CLL high-risk prognostic markers, was associated with production of reactive oxygen species and did not require caspase activation. Overall, these findings are evidence that Ulocuplumab (BMS-936564) has biological activity in CLL, highlight the relevance of the CXCR4-CXCL12 pathway as a therapeutic target in CLL, and provide biological rationale for ongoing clinical trials in CLL and other hematological malignancies.
Archive | 2012
Michelle Kuhne; Chin Pan; Josephine M. Cardarelli
Gastroenterology | 2008
Alison Witte; Michelle Kuhne; Benjamin T. Preston; Steven R. Wallace; Sharline Chen; Geetha Vasudevan; Michael Yellin; Kiron M. Das; Pina M. Cardarelli
Blood | 2013
Carlos I. Amaya-Chanaga; Harrison Jones; Eiman A. AlMahasnah; Michael Y. Choi; Michelle Kuhne; Lewis J. Cohen; Peter Sabbatini; Thomas J. Kipps; Pina M. Cardarelli; Januario E. Castro
Blood | 2011
Michelle Kuhne; Tanya Mulvey; Sharline Chen; Chin Pan; Colin Chong; Wafa Niekro; Tom Kempe; Karla A. Henning; Lewis J. Cohen; Alan J. Korman; Pina M. Cardarelli
Blood | 2012
Manoj Kumar Kashyap; Deepak Kumar; Harrison Jones Jones; Michael Y. Choi; Johanna Melo-Cardenas; Michelle Kuhne; Peter Sabbatini; Lewis J. Cohen; Suresh Shelat; Pina M. Cardarelli; Thomas J. Kipps; Januario E. Castro