Network


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

Hotspot


Dive into the research topics where Svetlana Gaidarova is active.

Publication


Featured researches published by Svetlana Gaidarova.


Leukemia | 2012

Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide

Antonia Lopez-Girona; Derek Mendy; Takumi Ito; Karen Miller; Anita Gandhi; Jian Kang; Satoki Karasawa; Gilles Carmel; Pilgrim Jackson; Mahan Abbasian; Afshin Mahmoudi; Brian E. Cathers; Emily Rychak; Svetlana Gaidarova; R Chen; Peter H. Schafer; Hiroshi Handa; Tom Daniel; Jilly F. Evans; Rajesh Chopra

Thalidomide and the immunomodulatory drug, lenalidomide, are therapeutically active in hematological malignancies. The ubiquitously expressed E3 ligase protein cereblon (CRBN) has been identified as the primary teratogenic target of thalidomide. Our studies demonstrate that thalidomide, lenalidomide and another immunomodulatory drug, pomalidomide, bound endogenous CRBN and recombinant CRBN–DNA damage binding protein-1 (DDB1) complexes. CRBN mediated antiproliferative activities of lenalidomide and pomalidomide in myeloma cells, as well as lenalidomide- and pomalidomide-induced cytokine production in T cells. Lenalidomide and pomalidomide inhibited autoubiquitination of CRBN in HEK293T cells expressing thalidomide-binding competent wild-type CRBN, but not thalidomide-binding defective CRBNYW/AA. Overexpression of CRBN wild-type protein, but not CRBNYW/AA mutant protein, in KMS12 myeloma cells, amplified pomalidomide-mediated reductions in c-myc and IRF4 expression and increases in p21WAF-1 expression. Long-term selection for lenalidomide resistance in H929 myeloma cell lines was accompanied by a reduction in CRBN, while in DF15R myeloma cells resistant to both pomalidomide and lenalidomide, CRBN protein was undetectable. Our biophysical, biochemical and gene silencing studies show that CRBN is a proximate, therapeutically important molecular target of lenalidomide and pomalidomide.


British Journal of Haematology | 2011

Lenalidomide downregulates the cell survival factor, interferon regulatory factor‐4, providing a potential mechanistic link for predicting response

Antonia Lopez-Girona; Daniel Heintel; Ling-Hua Zhang; Derek Mendy; Svetlana Gaidarova; Helen Brady; J. B. Bartlett; Peter H. Schafer; Martin Schreder; Arnold Bolomsky; Bernadette Hilgarth; Niklas Zojer; Heinz Gisslinger; Heinz Ludwig; Tom Daniel; Ulrich Jäger; Rajesh Chopra

Overexpression of the transcription factor interferon regulatory factor‐4 (IRF4), which is common in multiple myeloma (MM), is associated with poor prognosis. Patients with higher IRF4 expression have significantly poorer overall survival than those with low IRF4 expression. Lenalidomide is an IMiD® immunomodulatory compound that has both tumouricidal and immunomodulatory activity in MM. This study showed that lenalidomide downregulated IRF4 levels in MM cell lines and bone marrow samples within 8 h of drug exposure. This was associated with a decrease in MYC levels, as well as an initial G1 cell cycle arrest, decreased cell proliferation, and cell death by day 5 of treatment. In eight MM cell lines, high IRF4 levels correlated with increased lenalidomide sensitivity. The clinical significance of this observation was investigated in 154 patients with MM. Among MM patients with high levels of IRF4 expression, treatment with lenalidomide led to a significantly longer overall survival than other therapies in a retrospective analysis. These data confirm the central role of IRF4 in MM pathogenesis; indicate that this is an important mechanism by which lenalidomide exerts its antitumour effects; and may provide a mechanistic biomarker to predict response to lenalidomide.


British Journal of Haematology | 2014

Measuring cereblon as a biomarker of response or resistance to lenalidomide and pomalidomide requires use of standardized reagents and understanding of gene complexity.

Anita Gandhi; Derek Mendy; Michelle Waldman; Gengxin Chen; Emily Rychak; Karen Miller; Svetlana Gaidarova; Yan Ren; Maria Wang; Michael Breider; Gilles Carmel; Afshin Mahmoudi; Pilgrim Jackson; Mahan Abbasian; Brian E. Cathers; Peter H. Schafer; Tom Daniel; Antonia Lopez-Girona; Anjan Thakurta; Rajesh Chopra

Cereblon, a member of the cullin 4 ring ligase complex (CRL4), is the molecular target of the immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide and is required for the antiproliferative activity of these agents in multiple myeloma (MM) and immunomodulatory activity in T cells. Cereblons central role as a target of lenalidomide and pomalidomide suggests potential utility as a predictive biomarker of response or resistance to IMiD therapy. Our studies characterized a cereblon monoclonal antibody CRBN65, with high sensitivity and specificity in Western analysis and immunohistochemistry that is superior to commercially available antibodies. We identified multiple cereblon splice variants in both MM cell lines and primary cells, highlighting challenges with conventional gene expression assays given this gene complexity. Using CRBN65 antibody and TaqMan quantitative reverse transcription polymerase chain reaction assays, we showed lack of correlation between cereblon protein and mRNA levels. Furthermore, lack of correlation between cereblon expression in MM cell lines and sensitivity to lenalidomide was shown. In cell lines made resistant to lenalidomide and pomalidomide, cereblon protein is greatly reduced. These studies show limitations to the current approaches of cereblon measurement that rely on commercial reagents and assays. Standardized reagents and validated assays are needed to accurately assess the role of cereblon as a predictive biomarker.


Nature | 2016

A novel cereblon modulator recruits GSPT1 to the CRL4(CRBN) ubiquitin ligase.

Mary Matyskiela; Gang Lu; Takumi Ito; Barbra Pagarigan; Chin-Chun Lu; Karen Miller; Wei Fang; Nai-Yu Wang; Derek Nguyen; J. E. Houston; Gilles Carmel; Tam Tran; Mariko Riley; Lyn’Al Nosaka; Gabriel C. Lander; Svetlana Gaidarova; Shuichan Xu; Alexander L. Ruchelman; Hiroshi Handa; James Carmichael; Thomas O. Daniel; Brian E. Cathers; Antonia Lopez-Girona; Philip Chamberlain

Immunomodulatory drugs bind to cereblon (CRBN) to confer differentiated substrate specificity on the CRL4CRBN E3 ubiquitin ligase. Here we report the identification of a new cereblon modulator, CC-885, with potent anti-tumour activity. The anti-tumour activity of CC-885 is mediated through the cereblon-dependent ubiquitination and degradation of the translation termination factor GSPT1. Patient-derived acute myeloid leukaemia tumour cells exhibit high sensitivity to CC-885, indicating the clinical potential of this mechanism. Crystallographic studies of the CRBN–DDB1–CC-885–GSPT1 complex reveal that GSPT1 binds to cereblon through a surface turn containing a glycine residue at a key position, interacting with both CC-885 and a ‘hotspot’ on the cereblon surface. Although GSPT1 possesses no obvious structural, sequence or functional homology to previously known cereblon substrates, mutational analysis and modelling indicate that the cereblon substrate Ikaros uses a similar structural feature to bind cereblon, suggesting a common motif for substrate recruitment. These findings define a structural degron underlying cereblon ‘neosubstrate’ selectivity, and identify an anti-tumour target rendered druggable by cereblon modulation.


Blood | 2014

Lenalidomide inhibits the proliferation of CLL cells via a cereblon/ p21 WAF1/Cip1 -dependent mechanism independent of functional p53

Jessie-F. Fecteau; Laura G. Corral; Emanuela M. Ghia; Svetlana Gaidarova; Diahnn Futalan; Ila Sri Bharati; Brian E. Cathers; Maria Schwaederle; Bing Cui; Antonia Lopez-Girona; Davorka Messmer; Thomas J. Kipps

Lenalidomide has demonstrated clinical activity in patients with chronic lymphocytic leukemia (CLL), even though it is not cytotoxic for primary CLL cells in vitro. We examined the direct effect of lenalidomide on CLL-cell proliferation induced by CD154-expressing accessory cells in media containing interleukin-4 and -10. Treatment with lenalidomide significantly inhibited CLL-cell proliferation, an effect that was associated with the p53-independent upregulation of the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1) (p21). Silencing p21 with small interfering RNA impaired the capacity of lenalidomide to inhibit CLL-cell proliferation. Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. We isolated CLL cells from the blood of patients before and after short-term treatment with low-dose lenalidomide (5 mg per day) and found the leukemia cells were also induced to express p21 in vivo. These results indicate that lenalidomide can directly inhibit proliferation of CLL cells in a cereblon/p21-dependent but p53-independent manner, at concentrations achievable in vivo, potentially contributing to the capacity of this drug to inhibit disease-progression in patients with CLL.


Cancer Research | 2017

GSK3 inhibition drives maturation of NK cells and enhances their antitumor activity

Frank Cichocki; Bahram Valamehr; Ryan Bjordahl; Bin Zhang; Betsy Rezner; Paul Rogers; Svetlana Gaidarova; Stacey K Moreno; Katie Tuininga; Phillip Dougherty; Valarie McCullar; Peter Howard; Dhifaf Sarhan; Emily Taras; Heinrich Schlums; Stewart E Abbot; Daniel Shoemaker; Yenan T. Bryceson; Bruce R. Blazar; Scott Wolchko; Sarah Cooley; Jeffrey S. Miller

Maturation of human natural killer (NK) cells as defined by accumulation of cell-surface expression of CD57 is associated with increased cytotoxic character and TNF and IFNγ production upon target-cell recognition. Notably, multiple studies point to a unique role for CD57+ NK cells in cancer immunosurveillance, yet there is scant information about how they mature. In this study, we show that pharmacologic inhibition of GSK3 kinase in peripheral blood NK cells expanded ex vivo with IL15 greatly enhances CD57 upregulation and late-stage maturation. GSK3 inhibition elevated the expression of several transcription factors associated with late-stage NK-cell maturation including T-BET, ZEB2, and BLIMP-1 without affecting viability or proliferation. When exposed to human cancer cells, NK cell expanded ex vivo in the presence of a GSK3 inhibitor exhibited significantly higher production of TNF and IFNγ, elevated natural cytotoxicity, and increased antibody-dependent cellular cytotoxicity. In an established mouse xenograft model of ovarian cancer, adoptive transfer of NK cells conditioned in the same way also displayed more robust and durable tumor control. Our findings show how GSK3 kinase inhibition can greatly enhance the mature character of NK cells most desired for effective cancer immunotherapy. Cancer Res; 77(20); 5664-75. ©2017 AACR.


Cancer Research | 2018

Abstract 3576: FT500, an off-the-shelf NK cell cancer immunotherapy derived from a master pluripotent cell line, enhances T-cell activation and recruitment to overcome checkpoint blockade resistance

Ryan Bjordahl; Sajid Mahmood; Svetlana Gaidarova; Ramzey Abujarour; Raedun Clarke; Laurel Stokely; Paul Rogers; Moyar Ge; Megan Robinson; Betsy Rezner; Tom Tong Lee; Bahram Valamehr

The development of PD1/PDL1 targeting checkpoint inhibitors (CI) has transformed the oncology landscape, providing long term remissions in multiple indications. However, many tumor subtypes are resistant to checkpoint blockade therapy, and relapse remains a significant concern. Novel therapeutic approaches with the ability to overcome CI resistance are needed, and there is significant opportunity for therapies capable of additively or synergistically enhancing T-cell activation and recruitment when combined with CI. Adoptive transfer of NK cells from healthy donors has the potential to recruit T cells to the tumor microenvironment and augment T-cell activation at the tumor site. NK cells have both direct anti-tumor activity and the capacity to secrete inflammatory cytokines and chemokines upon activation, enabling the cells to play a unique and critical role in regulating anti-tumor T cell activity. We sought to determine whether FT500, an off-the-shelf NK cell product derived from a clonal master pluripotent cell line, could synergize with CI to relieve local immunosuppression and enhance T-cell activation and recruitment to the tumor site. FT500 is universally negative for cell surface PD1, and expression of PDL1 on tumor lines had no discernable effect on FT500 cytotoxicity. Similarly, addition of PDL1 blocking antibody had no effect on FT500 cytotoxicity or degranulation, suggesting that FT500 is inherently resistant to PDL1-PD1 mediated inhibition. Additionally, activation of FT500 induced the secretion of soluble factors capable of enhancing T-cell activation, as evidenced by increased upregulation of CD69. We hypothesized that FT500 might also enhance CI by promoting recruitment of T cells to the tumor site. Using conventional in vitro transwell migration assays, we found that FT500 produced soluble factors that promoted the migration of activated T cells. Additional profiling confirmed FT500 production of a range of chemokines, including CCL3, CCL4, CXCL10 and CCL22. Furthermore, using an in vivo recruitment model, FT500 was able to recruit T cells out of the circulation and into the peritoneal cavity. Similarly, utilizing a three-dimensional tumor spheroid model in vitro, infiltration of T cells into tumor spheroids was significantly enhanced when combined with FT500, suggesting that FT500 can enhance tumor infiltration of T cells. Our data suggest that FT500 is a potent producer of chemokines and can facilitate the recruitment of T cells to the tumor site. In addition to its direct cytotoxic potential, FT500 is also able to enhance T-cell activation, suggesting an ability to synergize with CI to reduce tumor burden. Together, our data provide evidence supporting the combination of FT500, an off-the-shelf NK cell cancer immunotherapy, with CI to overcome checkpoint blockade resistance. Citation Format: Ryan Bjordahl, Sajid Mahmood, Svetlana Gaidarova, Ramzey Abujarour, Raedun Clarke, Laurel Stokely, Paul Rogers, Moyar Ge, Megan Robinson, Betsy Rezner, Tom Tong Lee, Bahram Valamehr. FT500, an off-the-shelf NK cell cancer immunotherapy derived from a master pluripotent cell line, enhances T-cell activation and recruitment to overcome checkpoint blockade resistance [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3576.


Cancer Research | 2017

Abstract 609: Overcoming host histocompatibility barrier to create a renewable source of off-the-shelf effector lymphocytes for adoptive immunotherapy

Raedun Clarke; Matthieu Bauer; Ryan Bjordahl; Jeffrey Sasaki; Brian Groff; Svetlana Gaidarova; Tom Tong Lee; Weijie Lan; Michelle Burrascano; Ramzey Abujarour; Greg Bonello; Megan Robinson; Stewart Abbot; Scott Wolchko; Daniel Shoemaker; Bob Valamehr

Using epitope scanning of 272 short, synthetic peptides representing the amino acid sequence of the CB-11 peptide of type II collagen, we have shown that five strains of rat, immunized with type II collagen, produce antibodies to a region 37-45 amino acids from the amino end of CB-11 peptide. Antibodies to this region always gave the highest binding values suggesting that it is an immunodominant region. Wistar rats immunized with a synthetic peptide representing this region, coupled to keyhole limpet haemocyanin, produced antibodies to this peptide which could still be detected at 1:4000 to 1:8000 dilution but none developed clinical arthritis. All sera also showed binding of antibodies to denatured bovine type II collagen but not to native type II collagen, keyhole limpet haemocyanin or to bovine serum albumin by ELISA. Sera from peptide-immunized rats were examined for antibody binding to the 272 short peptides of the CB-11 peptide and to the synthetic peptides representing shortened forms of the immunodominant region and forms of it with substituted amino acids. These results showed that the antibodies in the peptide-immunized rats were not identical to those produced to that peptide by rats immunized with type II collagen but may represent subpopulations of them. These findings suggest caution in interpreting the role of antibodies to individual peptides in arthritis induction without knowledge of their fine specificity.


Cancer Research | 2017

Abstract 3755: Renewable and genetically engineered natural killer cells for off-the-shelf adoptive cellular immunotherapy

Ryan Bjordahl; Frank Cichocki; Raedun Clarke; Svetlana Gaidarova; Brian Groff; Paul Rogers; Stacey K. Moreno; Ramzey Abujarour; Greg Bonello; Thomas K. Lee; Weijie Lan; Matthieu Bauer; Dave Robbins; Betsy Rezner; Sarah Cooley; Bruce Walcheck; Stewart Abbot; Bruce R. Blazar; Scott Wolchko; Daniel Shoemaker; Jeffrey S. Miller; Bahram Valamehr

The unique attributes of a combinatorial tumor recognition system, diminished off-tumor cytotoxicity, and multifaceted effector function make natural killer (NK) cells a prime candidate for a universal approach to cancer immunotherapy. In addition, NK cells are the principal mediator of antibody-directed cellular cytotoxicity (ADCC). However, NK cell function is often impaired in the setting of cancer, reducing the effectiveness of the endogenous immune system and the therapeutic efficacy of monoclonal antibodies. To address the need for advanced and combinatorial cancer therapies, we developed a unique and effective strategy to create a renewable source of engineered “off-the-shelf” NK cells with augmented function, including enhanced ADCC and persistence. Key challenges associated with genetic editing, limited expansion, persistence and variability of peripheral blood (PB)-derived NK cells were overcome by utilizing our induced pluripotent stem cell (iPSC) technology as the unlimited starting material for the reproducible and consistent derivation of engineered NK cells. Through targeted transgene integration, we produced a clonal iPSC master cell line to continuously produce NK cells engineered to uniformly express a high affinity, non-cleavable version of CD16 (hnCD16-NK). In directed differentiation, the hnCD16-NK cells displayed homogeneous expression of CD16 (>95%) and a mature CD56+ NK cell phenotype, as exhibited by expression of KIR, NCRs, DNAM-1, and NKG2D. In contrast to endogenous CD16 expression, the engineered hnCD16 molecule was shown to be cleavage resistant upon NK cell activation (>95% CD16+ hnCD16-NK vs. Citation Format: Ryan Bjordahl, Frank Cichocki, Raedun Clarke, Svetlana Gaidarova, Brian Groff, Paul Rogers, Stacey Moreno, Ramzey Abujarour, Greg Bonello, Tom Lee, Weijie Lan, Matthieu Bauer, Dave Robbins, Betsy Rezner, Sarah Cooley, Bruce Walcheck, Stewart Abbot, Bruce Blazar, Scott Wolchko, Daniel Shoemaker, Jeffrey S. Miller, Bahram Valamehr. Renewable and genetically engineered natural killer cells for off-the-shelf adoptive cellular immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3755. doi:10.1158/1538-7445.AM2017-3755


Cancer Research | 2017

Abstract SY37-02: Ligand-directed degradation of GSPT1 by a novel cereblon modulator drives potent antitumor effects

Mary Matyskiela; Gang Lu; Takumi Ito; Barbra Pagarigan; Chin-Chu Lu; Karen K. Miller; Wei Fang; Nai-Yu Wang; Derek Nguyen; J. E. Houston; Gilles Carmel; Tam Tran; Mariko Riley; Lyn'Al Nosaka; Gabriel C. Lander; Svetlana Gaidarova; Shuichan Xu; Alexander L. Ruchelman; Hiroshi Handa; James Carmichale; Thomas O. Daniel; Brian E. Cathers; Antonia Lopez-Girona; P. P. Chamberlain

The protein cereblon is part of the CRL4-CRBN E3 ubiquitin ligase complex, and has been shown to be the molecular target for the drugs lenalidomide and pomalidomide. These drugs bind to the surface of cereblon, triggering the recruitment of substrate proteins to the ligase complex where they can be ubiquitinated and subsequently degraded by the proteosome. By this mechanism, lenalidomide and pomalidomide cause the degradation of the zinc finger transcription factors Ikaros and Aiolos, which mediate the antimyeloma activity of these compounds. Here we describe the discovery of CC-885, a novel cereblon modulator with potent and broad-spectrum antiproliferative activity against a panel of tumor cell lines. Acute myeloid leukemia (AML) cell lines and patient-derived AML cells show particular sensitivity to CC-885 treatment. CC-885 achieves this activity by causing the degradation of G1 to S phase transition 1 (GSPT1), a translation termination factor required for the release of nascent peptides from the ribosome. A crystal structure of cereblon in complex with the ligase adapter protein DDB1, as well as CC-885 and GSPT1, reveals that GSPT1 interacts with both CC-885 and the surface of cereblon. The principal molecular feature on GSPT1 that binds to cereblon is a beta-hairpin incorporating a glycine residue that docks against CC-885. Surprisingly, we found evidence that a similar molecular feature mediates Ikaros recruitment, even though there is no common structural fold or sequence homology other than the key glycine residue. We thereby define the common molecular feature, or degron, shared by the known cereblon neomorphic substrates. We further describe a novel therapeutic target, GSPT1, with promise in cancer such as AML. These results further show that cereblon-mediated protein degradation can be directed against new proteins and that this mechanism enables the targeting of multiple protein classes that may be considered undruggable with conventional approaches. Citation Format: Mary Matyskiela, Gang Lu, Takumi Ito, Barbra Pagarigan, Chin-Chu Lu, Karen Miller, Wei Fang, Nai-Yu Wang, Derek Nguyen, Jack Houston, Gilles Carmel, Tam Tran, Mariko Riley, Lyn9Al Nosaka, Gabriel Lander, Svetlana Gaidarova, Shuichan Xu, Alexander Ruchelman, Hiroshi Handa, James Carmichale, Thomas O. Daniel, Brian E. Cathers, Antonia Lopez-Girona, Philip Chamberlain. Ligand-directed degradation of GSPT1 by a novel cereblon modulator drives potent antitumor effects [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr SY37-02. doi:10.1158/1538-7445.AM2017-SY37-02

Collaboration


Dive into the Svetlana Gaidarova's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Raedun Clarke

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge