Network


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

Hotspot


Dive into the research topics where Kristi Elkins is active.

Publication


Featured researches published by Kristi Elkins.


Cancer Research | 2009

Antibody-Drug Conjugates for the Treatment of Non–Hodgkin's Lymphoma: Target and Linker-Drug Selection

Andrew Polson; Jill Calemine-Fenaux; Pamela Chan; Wesley Chang; Erin H. Christensen; Suzanna Clark; Frederic J. de Sauvage; Dan L. Eaton; Kristi Elkins; J. Michael Elliott; Gretchen Frantz; Reina N. Fuji; Alane Gray; Kristin Harden; Gladys Ingle; Noelyn M. Kljavin; Hartmut Koeppen; Christopher P. Nelson; Saileta Prabhu; Helga Raab; Sarajane Ross; Jean-Philippe Stephan; Suzie J. Scales; Susan D. Spencer; Richard Vandlen; Bernd Wranik; Shang-Fan Yu; Bing Zheng; Allen Ebens

Antibody-drug conjugates (ADC), potent cytotoxic drugs covalently linked to antibodies via chemical linkers, provide a means to increase the effectiveness of chemotherapy by targeting the drug to neoplastic cells while reducing side effects. Here, we systematically examine the potential targets and linker-drug combinations that could provide an optimal ADC for the treatment for non-Hodgkins lymphoma. We identified seven antigens (CD19, CD20, CD21, CD22, CD72, CD79b, and CD180) for potential treatment of non-Hodgkins lymphoma with ADCs. ADCs with cleavable linkers mediated in vivo efficacy via all these targets; ADCs with uncleavable linkers were only effective when targeted to CD22 and CD79b. In target-independent safety studies in rats, the uncleavable linker ADCs showed reduced toxicity, presumably due to the reduced release of free drug or other toxic metabolites into the circulation. Thus, our data suggest that ADCs with cleavable linkers work on a broad range of targets, and for specific targets, ADCs with uncleavable linkers provide a promising opportunity to improve the therapeutic window for ADCs in humans.


Blood | 2009

Therapeutic potential of an anti-CD79b antibody–drug conjugate, anti–CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma

David Dornan; Bennett F; Yung-Hsiang Chen; Mark S. Dennis; Dan L. Eaton; Kristi Elkins; Dorothy French; MaryAnn Go; Andrew Jack; Junutula; Hartmut Koeppen; Jeffrey Lau; Jacqueline McBride; Andy C. Rawstron; Xiaoyan Shi; Nancy Yu; Shang-Fan Yu; Peng Yue; Bing Zheng; Allen Ebens; Andrew G. Polson

Here we describe the generation of an antibody-drug conjugate (ADC) consisting of a humanized anti-CD79b antibody that is conjugated to monomethylauristatin E (MMAE) through engineered cysteines (THIOMABs) by a protease cleavable linker. By using flow cytometry, we detected the surface expression of CD79b in almost all non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia patients, suggesting that anti-CD79b-vcMMAE could be widely used in these malignancies. By using NHL cell lines to simulate a patient population we discovered that a minimal cell-surface expression level of CD79b was required for in vitro activity. Within the subpopulation of cell lines above this minimal threshold, we found that sensitivity to free MMAE, mutation of cancer genes, and cell doubling time were poorly correlated with in vitro activity; however, the expression level of BCL-XL was correlated with reduced sensitivity to anti-CD79b-vcMMAE. This observation was supported by in vivo data showing that a Bcl-2 family inhibitor, ABT-263, strikingly enhanced the activity of anti-CD79b-vcMMAE. Furthermore, anti-CD79b-vcMMAE was significantly more effective than a standard-of-care regimen, R-CHOP (ie, rituximab with a single intravenous injection of 30 mg/kg cyclophosphamide, 2.475 mg/kg doxorubicin, 0.375 mg/kg vincristine, and oral dosing of 0.15 mg/kg prednisone once a day for 5 days), in 3 xenograft models of NHL. Together, these data suggest that anti-CD79b-vcMMAE could be broadly efficacious for the treatment of NHL.


Molecular Cancer Therapeutics | 2013

DCDT2980S, an Anti-CD22-Monomethyl Auristatin E Antibody–Drug Conjugate, Is a Potential Treatment for Non-Hodgkin Lymphoma

Dongwei Li; Kirsten Achilles Poon; Shang-Fan Yu; Randall Dere; MaryAnn Go; Jeffrey Lau; Bing Zheng; Kristi Elkins; Dimitry M. Danilenko; Katherine R. Kozak; Pamela Chan; Josefa Chuh; Xiaoyan Shi; Denise Nazzal; Franklin Fuh; Jacqueline McBride; Vanitha Ramakrishnan; Ruth de Tute; Andy C. Rawstron; Andrew Jack; Rong Deng; Yu-Waye Chu; David Dornan; Marna Williams; William Ho; Allen Ebens; Saileta Prabhu; Andrew G. Polson

Antibody–drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via chemical linkers, allow specific targeting of drugs to neoplastic cells. We have used this technology to develop the ADC DCDT2980S that targets CD22, an antigen with expression limited to B cells and the vast majority of non-Hodgkin lymphomas (NHL). DCDT2980S consists of a humanized anti-CD22 monoclonal IgG1 antibody with a potent microtubule-disrupting agent, monomethyl auristatin E (MMAE), linked to the reduced cysteines of the antibody via a protease cleavable linker, maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl (MC-vc-PAB). We describe the efficacy, safety, and pharmacokinetics of DCDT2980S in animal models to assess its potential as a therapeutic for the treatment of B-cell malignancies. We did not find a strong correlation between in vitro or in vivo efficacy and CD22 surface expression, nor a correlation of sensitivity to free drug and in vitro potency. We show that DCDT2980S was capable of inducing complete tumor regression in xenograft mouse models of NHL and can be more effective than rituximab plus combination chemotherapy at drug exposures that were well tolerated in cynomolgus monkeys. These results suggest that DCDT2980S has an efficacy, safety, and pharmacokinetics profile that support potential treatment of NHL. Mol Cancer Ther; 12(7); 1255–65. ©2013 AACR.


Molecular Cancer Therapeutics | 2012

FcRL5 as a Target of Antibody–Drug Conjugates for the Treatment of Multiple Myeloma

Kristi Elkins; Bing Zheng; MaryAnn Go; Dionysos Slaga; Changchung Du; Suzie J. Scales; Shang-Fan Yu; Jacqueline McBride; Ruth de Tute; Andy C. Rawstron; Andrew Jack; Allen Ebens; Andrew Polson

Fc receptor-like 5 (FcRL5/FcRH5/IRTA2/CD307) is a surface protein expressed selectively on B cells and plasma cells. We found that FcRL5 was expressed at elevated levels on the surface of plasma cells from the bone marrow of patients diagnosed with multiple myeloma. This prevalence in multiple myeloma and narrow pattern of normal expression indicate that FcRL5 could be a target for antibody-based therapies for multiple myeloma, particularly antibody–drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via specialized chemical linkers, where limited expression on normal tissues is a key component to their safety. We found that FcRL5 is internalized upon antibody binding, indicating that ADCs to FcRL5 could be effective. Indeed, we found that FcRL5 ADCs were efficacious in vitro and in vivo but the unconjugated antibody was not. The two most effective consisted of our anti-FcRL5 antibody conjugated through cysteines to monomethylauristatin E (MMAE) by a maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl (MC-vcPAB) linker (anti-FcRL5-MC-vcPAB-MMAE) or conjugated via lysines to the maytansinoid DM4 through a disulfide linker (anti-FcRL5-SPDB-DM4). These two ADCs were highly effective in vivo in combination with bortezomib or lenalidomide, drugs in use for the treatment of multiple myeloma. These data show that the FcRL5 ADCs described herein show promise as an effective treatment for multiple myeloma. Mol Cancer Ther; 11(10); 2222–32. ©2012 AACR.


Clinical Cancer Research | 2013

Loss of NAPRT1 Expression by Tumor-specific Promoter Methylation Provides a Novel Predictive Biomarker for NAMPT Inhibitors

David S. Shames; Kristi Elkins; Kimberly Walter; Thomas Holcomb; Pan Du; Dane Mohl; Yang Xiao; Thinh Q. Pham; Peter M. Haverty; Bianca M. Liederer; Xiaorong Liang; Robert L. Yauch; Thomas O'Brien; Richard Bourgon; Hartmut Koeppen; Lisa D. Belmont

Purpose: We sought to identify predictive biomarkers for a novel nicotinamide phosphoribosyltransferase (NAMPT) inhibitor. Experimental Design: We use a NAMPT inhibitor, GNE-617, to evaluate nicotinic acid rescue status in a panel of more than 400 cancer cell lines. Using correlative analysis and RNA interference (RNAi), we identify a specific biomarker for nicotinic acid rescue status. We next determine the mechanism of regulation of expression of the biomarker. Finally, we develop immunohistochemical (IHC) and DNA methylation assays and evaluate cancer tissue for prevalence of the biomarker across indications. Results: Nicotinate phosphoribosyltransferase (NAPRT1) is necessary for nicotinic acid rescue and its expression is the major determinant of rescue status. We demonstrate that NAPRT1 promoter methylation accounts for NAPRT1 deficiency in cancer cells, and NAPRT1 methylation is predictive of rescue status in cancer cell lines. Bisulfite next-generation sequencing mapping of the NAPRT1 promoter identified tumor-specific sites of NAPRT1 DNA methylation and enabled the development of a quantitative methylation-specific PCR (QMSP) assay suitable for use on archival formalin-fixed paraffin-embedded tumor tissue. Conclusions: Tumor-specific promoter hypermethylation of NAPRT1 inactivates one of two NAD salvage pathways, resulting in synthetic lethality with the coadministration of a NAMPT inhibitor. NAPRT1 expression is lost due to promoter hypermethylation in most cancer types evaluated at frequencies ranging from 5% to 65%. NAPRT1-specific immunohistochemical or DNA methylation assays can be used on archival formalin paraffin-embedded cancer tissue to identify patients likely to benefit from coadministration of a Nampt inhibitor and nicotinic acid. Clin Cancer Res; 19(24); 6912–23. ©2013 AACR.


Science Translational Medicine | 2011

CD40 Pathway Activation Status Predicts Response to CD40 Therapy in Diffuse Large B Cell Lymphoma

Bart Burington; Peng Yue; Xiaoyan Shi; Ranjana H. Advani; Jeffrey Lau; Jenille Tan; Susanna Stinson; Jeremy Stinson; Thomas Januario; Sven de Vos; Stephen M. Ansell; Andres Forero-Torres; Grazyna Fedorowicz; Teddy T C Yang; Kristi Elkins; Changchun Du; Sankar Mohan; Nancy Yu; Zora Modrusan; Somasekar Seshagiri; Shang Fan Yu; Ajay Pandita; Hartmut Koeppen; Dorothy French; Andrew G. Polson; Rienk Offringa; Nancy Whiting; Allen Ebens; David Dornan

A 15-gene expression signature predicts whether a patient with diffuse large B cell lymphoma will respond to dacetuzumab, a therapeutic antibody. Matching Treatment to Tumor If physicians could predict the future, it would take the guess work out of designing the right treatment regimen for every patient’s cancer. The results presented by Burington et al. move us closer to clearing the crystal ball for diffuse large B cell lymphomas, a common type of non-Hodgkin’s lymphoma in which a cell surface receptor, CD40, presents a seemingly attractive target for therapy. Although stimulation of CD40 by ligand binding can cause apoptosis of B cells—a trait that one would predict to be desirable for a B cell lymphoma drug—it can also induce undesirable proliferation of some lymphoma cells. This murky paradox makes it unclear when to prescribe dacetuzumab, a CD40-targeted therapeutic monoclonal antibody. The authors have now identified a 15-gene expression signature that signals the biochemical status of a lymphoma, thus clarifying whether it can be subdued by anti-CD40 therapy. The authors collected an array of cell lines derived from non-Hodgkin’s lymphomas that show a range of sensitivity to anti-CD40 therapy. By assessing gene expression before and after CD40 stimulation and creating a score that reflected CD40 pathway activation, Burington et al. found that cell lines with higher baseline activation of the CD40 pathway tended to be unresponsive to anti-CD40 stimulation. The researchers then identified a group of 15 active genes whose expression in formalin-fixed tissue (as would be obtained from patients) correctly predicted susceptibility to anti-CD40 treatment 77% of the time, a result verified in another set of cells lines and by quantitative polymerase chain reaction (PCR). Next, in a real-world test of the utility of this 15-gene predictor, tumor tissue samples from 39 patients who had been treated with dacetuzumab were scored for CD40 pathway activation with the new gene signature. A large majority (88%) of the patients predicted by the gene signature to be resistant to therapy in fact did not respond to therapy, showing a median progression-free survival of 40 days; 67% of those predicted to respond to dacetuzumab did so, with median progression-free survival extended to 169 days. These results encourage further testing in a prospective clinical trial designed to examine the ability of the 15-gene signature to predict which lymphoma patients will benefit from dacetuzumab treatment. If this index proves useful, it can be added to the catalog of prognostic tools at the service of the oncologist as they match drug to patient—without the need of a crystal ball. The primary function of B cells, critical components of the adaptive immune response, is to produce antibodies against foreign antigens, as well as to perform isotype class switching, which changes the heavy chain of an antibody so that it can interact with different repertoires of effector cells. CD40 is a member of the tumor necrosis factor superfamily of cell surface receptors that transmits survival signals to B cells. In contrast, in B cell cancers, stimulation of CD40 signaling results in a heterogeneous response in which cells can sometimes undergo cell death in response to treatment, depending on the system studied. We found an association between sensitivity to CD40 stimulation and mutation of the tumor suppressor p53 in a panel of non-Hodgkin’s lymphoma cell lines. Consistent with p53’s tumor suppressor role, we found that higher levels of intrinsic DNA damage and increased proliferation rates, as well as higher levels of BCL6, a transcriptional repressor proto-oncogene, were associated with sensitivity to CD40 stimulation. In addition, CD40 treatment–resistant cell lines were sensitized to CD40 stimulation after the introduction of DNA-damaging agents. Using gene expression analysis, we also showed that resistant cell lines exhibited a preexisting activated CD40 pathway and that an mRNA expression signature comprising CD40 target genes predicted sensitivity and resistance to CD40-activating agents in cell lines and mouse xenograft models. Finally, the gene signature predicted tumor shrinkage and progression-free survival in patients with diffuse large B cell lymphoma treated with dacetuzumab, a monoclonal antibody with partial CD40 agonist activity. These data show that CD40 pathway activation status may be useful in predicting the antitumor activity of CD40-stimulating therapeutic drugs.


PLOS ONE | 2014

Structural Basis for Resistance to Diverse Classes of NAMPT Inhibitors.

Weiru Wang; Kristi Elkins; Angela Oh; Yen-Ching Ho; Jiansheng Wu; Hong Li; Yang Xiao; Mandy Kwong; Mary Coons; Bobby Brillantes; Eric Cheng; Lisa Crocker; Peter S. Dragovich; Deepak Sampath; Xiaozhang Zheng; Kenneth W. Bair; Thomas O'Brien; Lisa D. Belmont

Inhibiting NAD biosynthesis by blocking the function of nicotinamide phosphoribosyl transferase (NAMPT) is an attractive therapeutic strategy for targeting tumor metabolism. However, the development of drug resistance commonly limits the efficacy of cancer therapeutics. This study identifies mutations in NAMPT that confer resistance to a novel NAMPT inhibitor, GNE-618, in cell culture and in vivo, thus demonstrating that the cytotoxicity of GNE-618 is on target. We determine the crystal structures of six NAMPT mutants in the apo form and in complex with various inhibitors and use cellular, biochemical and structural data to elucidate two resistance mechanisms. One is the surprising finding of allosteric modulation by mutation of residue Ser165, resulting in unwinding of an α-helix that binds the NAMPT substrate 5-phosphoribosyl-1-pyrophosphate (PRPP). The other mechanism is orthosteric blocking of inhibitor binding by mutations of Gly217. Furthermore, by evaluating a panel of diverse small molecule inhibitors, we unravel inhibitor structure activity relationships on the mutant enzymes. These results provide valuable insights into the design of next generation NAMPT inhibitors that offer improved therapeutic potential by evading certain mechanisms of resistance.


Archive | 2008

Anti-CD79b antibodies and immunoconjugates and methods of use

Yvonne Chen; Mark S. Dennis; David Dornan; Kristi Elkins; Jagath R. Junutula; Andrew G. Polson; Bing Zheng


International Immunology | 2006

Expression pattern of the human FcRH/IRTA receptors in normal tissue and in B-chronic lymphocytic leukemia

Andrew G. Polson; Bing Zheng; Kristi Elkins; Wesley Chang; Changchun Du; Patrick Dowd; Lulu Yen; Christine Tan; Jo-Anne Hongo; Hartmut Koeppen; Allen Ebens


Archive | 2004

Compositions and methods for the treatment of tumor of hematopoietic origin

Craig Crowley; Frederic J. de Sauvage; Dan L. Eaton; Allen Ebens; Kristi Elkins; Jo-Anne Hongo; Jagath R. Junutula; Andrew G. Polson; Sarajane Ross; Victoria Smith; Richard Vandlen; Bing Zheng

Collaboration


Dive into the Kristi Elkins's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Polson

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge