Network


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

Hotspot


Dive into the research topics where Khristofer Garcia is active.

Publication


Featured researches published by Khristofer Garcia.


Cancer Research | 2010

Evaluation of the Proteasome Inhibitor MLN9708 in Preclinical Models of Human Cancer

Erik Kupperman; Edmund Lee; Yueying Cao; Bret Bannerman; Michael C. Fitzgerald; Allison Berger; Jie Yu; Yu Yang; Paul Hales; Frank J. Bruzzese; Jane Liu; Jonathan L. Blank; Khristofer Garcia; Christopher Tsu; Larry Dick; Paul Fleming; Li Yu; Mark Manfredi; Mark Rolfe; Joe Bolen

The proteasome was validated as an oncology target following the clinical success of VELCADE (bortezomib) for injection for the treatment of multiple myeloma and recurring mantle cell lymphoma. Consequently, several groups are pursuing the development of additional small-molecule proteasome inhibitors for both hematologic and solid tumor indications. Here, we describe MLN9708, a selective, orally bioavailable, second-generation proteasome inhibitor that is in phase I clinical development. MLN9708 has a shorter proteasome dissociation half-life and improved pharmacokinetics, pharmacodynamics, and antitumor activity compared with bortezomib. MLN9708 has a larger blood volume distribution at steady state, and analysis of 20S proteasome inhibition and markers of the unfolded protein response confirmed that MLN9708 has greater pharmacodynamic effects in tissues than bortezomib. MLN9708 showed activity in both solid tumor and hematologic preclinical xenograft models, and we found a correlation between greater pharmacodynamic responses and improved antitumor activity. Moreover, antitumor activity was shown via multiple dosing routes, including oral gavage. Taken together, these data support the clinical development of MLN9708 for both hematologic and solid tumor indications.


Biochemical Journal | 2010

Characterization of a new series of non-covalent proteasome inhibitors with exquisite potency and selectivity for the 20S β5-subunit

Christopher Blackburn; Kenneth M. Gigstad; Paul Hales; Khristofer Garcia; Marc D. Jones; Frank J. Bruzzese; Cynthia Barrett; Jane X. Liu; Teresa A. Soucy; Darshan S. Sappal; Nancy J. Bump; Edward J. Olhava; Paul Fleming; Lawrence R. Dick; Christopher Tsu; Jonathan L. Blank

The mammalian 26S proteasome is a 2500 kDa multi-catalytic complex involved in intracellular protein degradation. We describe the synthesis and properties of a novel series of non-covalent di-peptide inhibitors of the proteasome used on a capped tri-peptide that was first identified by high-throughput screening of a library of approx. 350000 compounds for inhibitors of the ubiquitin–proteasome system in cells. We show that these compounds are entirely selective for the β5 (chymotrypsin-like) site over the β1 (caspase-like) and β2 (trypsin-like) sites of the 20S core particle of the proteasome, and over a panel of less closely related proteases. Compound optimization, guided by X-ray crystallography of the liganded 20S core particle, confirmed their non-covalent binding mode and provided a structural basis for their enhanced in vitro and cellular potencies. We demonstrate that such compounds show low nanomolar IC50 values for the human 20S β5 site in vitro, and that pharmacological inhibition of this site in cells is sufficient to potently inhibit the degradation of a tetra-ubiquitin–luciferase reporter, activation of NFκB (nuclear factor κB) in response to TNF-α (tumour necrosis factor-α) and the proliferation of cancer cells. Finally, we identified capped di-peptides that show differential selectivity for the β5 site of the constitutively expressed proteasome and immunoproteasome in vitro and in B-cell lymphomas. Collectively, these studies describe the synthesis, activity and binding mode of a new series of non-covalent proteasome inhibitors with unprecedented potency and selectivity for the β5 site, and which can discriminate between the constitutive proteasome and immunoproteasome in vitro and in cells.The mammalian 26S proteasome is a 2500 kDa multi-catalytic complex involved in intracellular protein degradation. We describe the synthesis and properties of a novel series of non-covalent di-peptide inhibitors of the proteasome based [corrected] on a capped tri-peptide that was first identified by high-throughput screening of a library of approx. 350000 compounds for inhibitors of the ubiquitin-proteasome system in cells. We show that these compounds are entirely selective for the beta5 (chymotrypsin-like) site over the beta1 (caspase-like) and beta2 (trypsin-like) sites of the 20S core particle of the proteasome, and over a panel of less closely related proteases. Compound optimization, guided by X-ray crystallography of the liganded 20S core particle, confirmed their non-covalent binding mode and provided a structural basis for their enhanced in vitro and cellular potencies. We demonstrate that such compounds show low nanomolar IC50 values for the human 20S beta5 site in vitro, and that pharmacological inhibition of this site in cells is sufficient to potently inhibit the degradation of a tetra-ubiquitin-luciferase reporter, activation of NFkappaB (nuclear factor kappaB) in response to TNF-alpha (tumour necrosis factor-alpha) and the proliferation of cancer cells. Finally, we identified capped di-peptides that show differential selectivity for the beta5 site of the constitutively expressed proteasome and immunoproteasome in vitro and in B-cell lymphomas. Collectively, these studies describe the synthesis, activity and binding mode of a new series of non-covalent proteasome inhibitors with unprecedented potency and selectivity for the beta5 site, and which can discriminate between the constitutive proteasome and immunoproteasome in vitro and in cells.


Cancer Research | 2010

Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib

Siquan Chen; Jonathan L. Blank; Theodore Peters; Xiaozhen J. Liu; David M. Rappoli; Michael D. Pickard; Saurabh Menon; Jie Yu; Denise L. Driscoll; Trupti Lingaraj; Anne L. Burkhardt; Wei Chen; Khristofer Garcia; Darshan S. Sappal; Jesse Gray; Paul Hales; Patrick J. LeRoy; John Ringeling; Claudia Rabino; James J. Spelman; Jay P. Morgenstern

Multiple pathways have been proposed to explain how proteasome inhibition induces cell death, but mechanisms remain unclear. To approach this issue, we performed a genome-wide siRNA screen to evaluate the genetic determinants that confer sensitivity to bortezomib (Velcade (R); PS-341). This screen identified 100 genes whose knockdown affected lethality to bortezomib and to a structurally diverse set of other proteasome inhibitors. A comparison of three cell lines revealed that 39 of 100 genes were commonly linked to cell death. We causally linked bortezomib-induced cell death to the accumulation of ASF1B, Myc, ODC1, Noxa, BNIP3, Gadd45alpha, p-SMC1A, SREBF1, and p53. Our results suggest that proteasome inhibition promotes cell death primarily by dysregulating Myc and polyamines, interfering with protein translation, and disrupting essential DNA damage repair pathways, leading to programmed cell death.


Cancer Research | 2013

Novel DNA Damage Checkpoints Mediating Cell Death Induced by the NEDD8-Activating Enzyme Inhibitor MLN4924

Jonathan L. Blank; Xiaozhen J. Liu; Katherine Cosmopoulos; David C. Bouck; Khristofer Garcia; Hugues Bernard; Olga Tayber; Greg Hather; Ray Liu; Usha Narayanan; Michael Milhollen

MLN4924 is an investigational small-molecule inhibitor of the NEDD8-activating enzyme (NAE) in phase I clinical trials. NAE inhibition prevents the ubiquitination and proteasomal degradation of substrates for cullin-RING ubiquitin E3 ligases that support cancer pathophysiology, but the genetic determinants conferring sensitivity to NAE inhibition are unknown. To address this gap in knowledge, we conducted a genome-wide siRNA screen to identify genes and pathways that affect the lethality of MLN4924 in melanoma cells. Of the 154 genes identified, approximately one-half interfered with components of the cell cycle, apoptotic machinery, ubiquitin system, and DNA damage response pathways. In particular, genes involved in DNA replication, p53, BRCA1/BRCA2, transcription-coupled repair, and base excision repair seemed to be important for MLN4924 lethality. In contrast, genes within the G(2)-M checkpoint affected sensitivity to MLN4924 in colon cancer cells. Cell-cycle analysis in melanoma cells by flow cytometry following RNAi-mediated silencing showed that MLN4924 prevented the transition of cells from S-G(2) phase after induction of rereplication stress. Our analysis suggested an important role for the p21-dependent intra-S-phase checkpoint and extensive rereplication, whereas the ATR-dependent intra-S-phase checkpoint seemed to play a less dominant role. Unexpectedly, induction of the p21-dependent intra-S-phase checkpoint seemed to be independent of both Cdt1 stabilization and ATR signaling. Collectively, these data enhance our understanding of the mechanisms by which inhibition of NEDD8-dependent ubiquitination causes cell death, informing clinical development of MLN4924.


Molecular & Cellular Proteomics | 2011

Quantitative Proteomic Analysis of Cellular Protein Modulation upon Inhibition of the NEDD8-Activating Enzyme by MLN4924

Hua Liao; Xiaozhen J. Liu; Jonathan L. Blank; David C. Bouck; Hugues Bernard; Khristofer Garcia

Cullin-RING ubiquitin ligases (CRLs) are responsible for the ubiquitination of many cellular proteins, thereby targeting them for proteasomal degradation. In most cases the substrates of the CRLs have not been identified, although many of those that are known have cancer relevance. MLN4924, an investigational small molecule that is a potent and selective inhibitor of the Nedd8-activating enzyme (NAE), is currently being explored in Phase I clinical trials. Inhibition of Nedd8-activating enzyme by MLN4924 prevents the conjugation of cullin proteins with NEDD8, resulting in inactivation of the entire family of CRLs. We have performed stable isotope labeling with amino acids in cell culture analysis of A375 melanoma cells treated with MLN4924 to identify new CRL substrates, confidently identifying and quantitating 5122–6012 proteins per time point. Proteins such as MLX, EID1, KLF5, ORC6L, MAGEA6, MORF4L2, MRFAP1, MORF4L1, and TAX1BP1 are rapidly stabilized by MLN4924, suggesting that they are novel CRL substrates. Proteins up-regulated at later times were also identified and siRNA against their corresponding genes were used to evaluate their influence on MLN4924-induced cell death. Thirty-eight proteins were identified as being particularly important for the cytotoxicity of MLN4924. Strikingly, these proteins had roles in cell cycle, DNA damage repair, and ubiquitin transfer. Therefore, the combination of RNAi with stable isotope labeling with amino acids in cell culture provides a paradigm for understanding the mechanism of action of novel agents affecting the ubiquitin proteasome system and a path to identifying mechanistic biomarkers.


Bioorganic & Medicinal Chemistry Letters | 2010

Optimization of a series of dipeptides with a P3 threonine residue as non-covalent inhibitors of the chymotrypsin-like activity of the human 20S proteasome.

Christopher Blackburn; Cynthia Barrett; Jonathan L. Blank; Frank J. Bruzzese; Nancy J. Bump; Lawrence R. Dick; Paul Fleming; Khristofer Garcia; Paul Hales; Zhigen Hu; Matthew Jones; Jane X. Liu; Darshan S. Sappal; Michael D. Sintchak; Christopher Tsu; Kenneth M. Gigstad

Starting from a tripeptide screening hit, a series of dipeptide inhibitors of the proteasome with Thr as the P3 residue has been optimized with the aid of crystal structures in complex with the β-5/6 active site of y20S. Derivative 25, (β5 IC(50)=7.4 nM) inhibits only the chymotryptic activity of the proteasome, shows cellular activity against targets in the UPS, and inhibits proliferation.


Journal of Medicinal Chemistry | 2011

Design and Optimization of Potent and Orally Bioavailable Tetrahydronaphthalene Raf Inhibitors

Alexandra E. Gould; Ruth Adams; Sharmila Adhikari; Kathleen Aertgeerts; Roushan Afroze; Christopher Blackburn; Emily F. Calderwood; Ryan Chau; Jouhara Chouitar; Matthew O. Duffey; Dylan England; Cheryl A. Farrer; Nancy Forsyth; Khristofer Garcia; Jeffery Gaulin; Paul D. Greenspan; Ribo Guo; Sean Harrison; Shih-Chung Huang; Natalia Iartchouk; Dave Janowick; Mi-Sook Kim; Bheemashankar Kulkarni; Steven P. Langston; Jane X. Liu; Li-Ting Ma; Saurabh Menon; Hirotake Mizutani; Erin Paske; Christelle C. Renou

Inhibition of mutant B-Raf signaling, through either direct inhibition of the enzyme or inhibition of MEK, the direct substrate of Raf, has been demonstrated preclinically to inhibit tumor growth. Very recently, treatment of B-Raf mutant melanoma patients with a selective B-Raf inhibitor has resulted in promising preliminary evidence of antitumor activity. This article describes the design and optimization of tetrahydronaphthalene-derived compounds as potent inhibitors of the Raf pathway in vitro and in vivo. These compounds possess good pharmacokinetic properties in rodents and inhibit B-Raf mutant tumor growth in mouse xenograft models.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery and optimization of pyrazoline compounds as B-Raf inhibitors.

Matthew O. Duffey; Ruth Adams; Christopher Blackburn; Ryan W. Chau; Susan Chen; Katherine M. Galvin; Khristofer Garcia; Alexandra E. Gould; Paul D. Greenspan; Sean Harrison; Shih-Chung Huang; Mi-Sook Kim; Bheemashankar Kulkarni; Steven P. Langston; Jane X. Liu; Li-Ting Ma; Saurabh Menon; Masayuki Nagayoshi; R. Scott Rowland; Tricia J. Vos; Tianlin Xu; Johnny J. Yang; Shaoxia Yu; Qin Zhang

The discovery of novel pyrazoline derivatives as B-Raf (V600E) inhibitors is described in this report. Chemical modification of the pyrazoline scaffold led to the development of SAR and identified potent and selective inhibitors of B-Raf (V600E). Determination of the pharmacokinetic properties of selected inhibitors is also reported.


MedChemComm | 2012

Optimization of a series of dipeptides with a P3 β-neopentyl asparagine residue as non-covalent inhibitors of the chymotrypsin-like activity of human 20S proteasome

Christopher Blackburn; Cynthia Barrett; Jonathan L. Blank; Frank J. Bruzzese; Nancy J. Bump; Lawrence R. Dick; Paul Fleming; Khristofer Garcia; Paul Hales; Matthew Jones; Jane X. Liu; Masayuki Nagayoshi; Darshan S. Sappal; Michael D. Sintchak; Christopher Tsu; Cindy Q. Xia; Xiansi Zhou; Kenneth M. Gigstad

Inhibition of the proteasome by covalent inhibitors is a clinically proven anti-cancer therapy. We report here that dipeptides with a P3 neopentyl Asn residue are potent, reversible, non-covalent inhibitors selective for the chymotryptic activity of the 20S proteasome in vitro and in cells. The X-ray structure of compound 20 in complex with yeast 20S reveals the importance of hydrophobic bonding interactions of the neopentyl group within the S3 binding pocket of the 20S β5 sub-unit. Four compounds show comparable potencies to boronic acid inhibitors in a panel of assays.


Molecular Cancer Therapeutics | 2013

Abstract B92: Nedd8-activating enzyme inhibitor MLN4924 provides synergy in nonclinical models with mitomycin C through interactions with ATR, BRCA1/BRCA2 and chromatin dynamics pathways.

Khristofer Garcia; Jonathan L. Blank; David C. Bouck; Xiaozhen J. Liu; Greg Hather; Allison Berger; Katherine Cosmopoulos; Michael Thomas; Mike Kuranda; Michael D. Pickard; Ray Liu; Syamala Bandi; Peter G. Smith

MLN4924 is an investigational small molecule inhibitor of the Nedd8-activating enzyme (NAE) currently in Phase 1 clinical trials. MLN4924 induces DNA damage via rereplication in most cell lines. This distinct mechanism of DNA damage may affect its ability to combine with standards of care, including other DNA damaging agents. We studied the interaction of MLN4924 with other DNA damaging agents in a panel of 4 cell lines and found that mitomycin C, cisplatin, carboplatin, cytarabine, ultraviolet radiation, SN-38, and gemcitabine demonstrated synergy in combination with MLN4924 in at least 1 cell line. Further testing in xenograft-bearing mice demonstrated synergy of MLN4924 with mitomycin C and with carboplatin, and additivity with gemcitabine. Based in part on this data, MLN4924 is currently being evaluated in a Phase 1b trial ([NCT01862328][1]) with 3 combination arms: MLN4924 + carboplatin and paclitaxel, MLN4924 + gemcitabine, and MLN4924 + docetaxel. To evaluate the mechanism of synergy between MLN4924 and mitomycin C, in vitro experiments with RNAi were performed. Depletion of genes within the ATR and BRCA1/BRCA2 pathways, chromatin modification, and transcription-coupled repair reduced the synergy between mitomycin C and MLN4924. Our data suggest that mitomycin C causes stalled replication forks, which when combined with rereplication induced by MLN4924, results in frequent replication fork collisions, leading to cell death. This study provides a straightforward approach to understand the mechanism of synergy, which may be applied to additional combinations currently under clinical evaluation. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B92. Citation Format: Eric S. Lightcap, Khristofer Garcia, Jonathan L. Blank, David C. Bouck, Xiaozhen J. Liu, Greg Hather, Allison Berger, Katherine Cosmopoulos, Michael P. Thomas, Mike Kuranda, Michael D. Pickard, Ray Liu, Syamala Bandi, Peter G. Smith. Nedd8-activating enzyme inhibitor MLN4924 provides synergy in nonclinical models with mitomycin C through interactions with ATR, BRCA1/BRCA2 and chromatin dynamics pathways. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B92. [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01862328&atom=%2Fmolcanther%2F12%2F11_Supplement%2FB92.atom

Collaboration


Dive into the Khristofer Garcia's collaboration.

Top Co-Authors

Avatar

Jonathan L. Blank

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Paul Hales

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Christopher Tsu

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Frank J. Bruzzese

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Paul Fleming

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Saurabh Menon

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Darshan S. Sappal

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

David C. Bouck

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Jane X. Liu

Millennium Pharmaceuticals

View shared research outputs
Researchain Logo
Decentralizing Knowledge