Network


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

Hotspot


Dive into the research topics where Kevin Lin is active.

Publication


Featured researches published by Kevin Lin.


Cancer Discovery | 2013

Discovery of a Mutant-Selective Covalent Inhibitor of EGFR that Overcomes T790M-Mediated Resistance in NSCLC

Annette O Walter; Robert Tjin Tham Sjin; Henry J Haringsma; Kadoaki Ohashi; Jing Sun; Kwangho Lee; Aleksander Dubrovskiy; Matthew T. Labenski; Zhendong Zhu; Zhigang Wang; Michael Sheets; Thia St Martin; Russell Karp; Dan van Kalken; Prasoon Chaturvedi; Deqiang Niu; M. Nacht; Russell C. Petter; William F. Westlin; Kevin Lin; Sarah S. Jaw-Tsai; Mitch Raponi; Terry Van Dyke; Jeff Etter; Zoe Weaver; William Pao; Juswinder Singh; Andrew Simmons; Thomas Harding; Andrew E. Allen

UNLABELLED Patients with non-small cell lung cancer (NSCLC) with activating EGF receptor (EGFR) mutations initially respond to first-generation reversible EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by acquired resistance, frequently driven by the EGFR(T790M) mutation. CO-1686 is a novel, irreversible, and orally delivered kinase inhibitor that specifically targets the mutant forms of EGFR, including T790M, while exhibiting minimal activity toward the wild-type (WT) receptor. Oral administration of CO-1686 as single agent induces tumor regression in EGFR-mutated NSCLC tumor xenograft and transgenic models. Minimal activity of CO-1686 against the WT EGFR receptor was observed. In NSCLC cells with acquired resistance to CO-1686 in vitro, there was no evidence of additional mutations or amplification of the EGFR gene, but resistant cells exhibited signs of epithelial-mesenchymal transition and demonstrated increased sensitivity to AKT inhibitors. These results suggest that CO-1686 may offer a novel therapeutic option for patients with mutant EGFR NSCLC. SIGNIFICANCE We report the preclinical development of a novel covalent inhibitor, CO-1686, that irreversibly and selectively inhibits mutant EGFR, in particular the T790M drug-resistance mutation, in NSCLC models. CO-1686 is the fi rst drug of its class in clinical development for the treatment of T790M-positive NSCLC, potentially offering potent inhibition of mutant EGFR while avoiding the on-target toxicity observed with inhibition of the WT EGFR.


Immunity | 2008

The Rap GTPases Regulate B Cell Morphology, Immune-Synapse Formation, and Signaling by Particulate B Cell Receptor Ligands

Kevin Lin; Spencer A. Freeman; Saba Zabetian; Hayley K. Brugger; Michele Weber; Victor Lei; May Dang-Lawson; Kathy W.K. Tse; Rene Santamaria; Facundo D. Batista; Michael R. Gold

B lymphocytes spread and extend membrane processes when searching for antigens and form immune synapses upon contacting cells that display antigens on their surface. Although these dynamic morphological changes facilitate B cell activation, the signaling pathways underlying these processes are not fully understood. We found that activation of the Rap GTPases was essential for these changes in B cell morphology. Rap activation was important for B cell receptor (BCR)- and lymphocyte-function-associated antigen-1 (LFA-1)-induced spreading, for BCR-induced immune-synapse formation, and for particulate BCR ligands to induce localized F-actin assembly and membrane-process extension. Rap activation and F-actin assembly were also required for optimal BCR signaling in response to particulate antigens but not soluble antigens. Thus by controlling B cell morphology and cytoskeletal organization, Rap might play a key role in the activation of B cells by particulate and cell-associated antigens.


Cancer Discovery | 2017

Secondary Somatic Mutations Restoring RAD51C and RAD51D Associated with Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma

Olga Kondrashova; Minh Nguyen; Kristy Shield-Artin; Anna V. Tinker; Nelson N.H. Teng; Maria I. Harrell; Michael J. Kuiper; Gwo-Yaw Ho; Holly Barker; Maria Jasin; Rohit Prakash; Elizabeth M. Kass; Meghan R. Sullivan; Gregory J. Brunette; Kara A. Bernstein; Robert L. Coleman; Anne Floquet; Michael Friedlander; Ganessan Kichenadasse; David M. O'Malley; Amit M. Oza; James Sun; Liliane Robillard; L. Maloney; David Bowtell; Heidi Giordano; Matthew J. Wakefield; Scott H. Kaufmann; Andrew Simmons; Thomas Harding

High-grade epithelial ovarian carcinomas containing mutated BRCA1 or BRCA2 (BRCA1/2) homologous recombination (HR) genes are sensitive to platinum-based chemotherapy and PARP inhibitors (PARPi), while restoration of HR function due to secondary mutations in BRCA1/2 has been recognized as an important resistance mechanism. We sequenced core HR pathway genes in 12 pairs of pretreatment and postprogression tumor biopsy samples collected from patients in ARIEL2 Part 1, a phase II study of the PARPi rucaparib as treatment for platinum-sensitive, relapsed ovarian carcinoma. In 6 of 12 pretreatment biopsies, a truncation mutation in BRCA1, RAD51C, or RAD51D was identified. In five of six paired postprogression biopsies, one or more secondary mutations restored the open reading frame. Four distinct secondary mutations and spatial heterogeneity were observed for RAD51CIn vitro complementation assays and a patient-derived xenograft, as well as predictive molecular modeling, confirmed that resistance to rucaparib was associated with secondary mutations.Significance: Analyses of primary and secondary mutations in RAD51C and RAD51D provide evidence for these primary mutations in conferring PARPi sensitivity and secondary mutations as a mechanism of acquired PARPi resistance. PARPi resistance due to secondary mutations underpins the need for early delivery of PARPi therapy and for combination strategies. Cancer Discov; 7(9); 984-98. ©2017 AACR.See related commentary by Domchek, p. 937See related article by Quigley et al., p. 999See related article by Goodall et al., p. 1006This article is highlighted in the In This Issue feature, p. 920.


Oncogene | 2010

The Rap GTPases regulate the migration, invasiveness and in vivo dissemination of B-cell lymphomas.

Kevin Lin; Poh C. Tan; Spencer A. Freeman; M Lam; Kelly M. McNagny; Michael R. Gold

B-cell lymphomas are common malignancies in which transformed B cells enter the circulation, extravasate into tissues and form tumors in multiple organs. Lymphoma cells are thought to exit the vasculature and enter tissues through the same chemokine- and adhesion molecule-dependent mechanisms as normal B cells. We have previously shown that activation of the Rap GTPases, proteins that control cytoskeletal organization and integrin activation, is critical for chemokine-induced migration and adhesion in B-lymphoma cell lines. Using the A20 murine B-lymphoma cell line as a model, we now show that Rap activation is important for circulating lymphoma cells to enter tissues and form tumors in vivo. In vitro assays showed that Rap activation is required for A20 cells to efficiently adhere to vascular endothelial cells and undergo transendothelial migration. These findings suggest that Rap or its effectors could be novel targets for treating B-cell lymphomas.


Cellular Immunology | 2012

Small molecule inhibitors of the Pyk2 and FAK kinases modulate chemoattractant-induced migration, adhesion and Akt activation in follicular and marginal zone B cells.

Kathy W.K. Tse; Kevin Lin; May Dang-Lawson; Angel Guzman-Perez; Gary E. Aspnes; Leonard Buckbinder; Michael R. Gold

B-lymphocytes produce protective antibodies but also contribute to autoimmunity. In particular, marginal zone (MZ) B cells recognize both microbial components and self-antigens. B cell trafficking is critical for B cell activation and is controlled by chemoattactants such as CXCL13 and sphingosine 1-phosphate (S1P). The related tyrosine kinases focal adhesion kinase (FAK) and proline-rich tyrosine kinase (Pyk2) regulate cell migration and adhesion but their roles in B cells are not fully understood. Using a novel Pyk2-selective inhibitor described herein (PF-719), as well as a FAK-selective inhibitor, we show that both Pyk2 and FAK are important for CXCL13- and S1P-induced migration of B-2 cells and MZ B cells. In contrast, LFA-1-mediated adhesion required only Pyk2 whereas activation of the Akt pro-survival kinase required FAK but not Pyk2. Thus Pyk2 and FAK mediate critical processes in B cells and these inhibitors can be used to further elucidate their functions in B cells.


Molecular Cancer Therapeutics | 2011

Abstract C189: CO-1686, an orally available, mutant-selective inhibitor of the epidermal growth factor receptor (EGFR), causes tumor shrinkage in non-small cell lung cancer (NSCLC) with T790M mutations.

Annette Walter; Robert Tjin; Henry J Haringsma; Kevin Lin; Alex Dubrovskiy; Kwangho Lee; Thia St. Martin; Russell Karp; Zhendong Zhu; Deqiang Niu; Mariana Nacht; Russell C. Petter; William F. Westlin; Juswinder Singh; Mitch Raponi; Andrew E. Allen

Introduction: Non-small cell lung cancer (NSCLC) patients with activating EGFR mutations initially respond well to EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by the development of resistance. The most common mechanism of resistance is a second site mutation within exon 20 of EGFR (T790M), observed in ∼50% of cases. Our goal was to develop a mutant-selective EGFR inhibitor that potently inhibits activating EGFR mutations as well as the T790M resistance mutation while sparing wild-type EGFR for the treatment of NSCLC patients. Such a drug has the potential to effectively treat first- and second-line NSCLC patients with EGFR mutations without causing the dose limiting toxicities associated with EGFR kinase inhibitors currently in clinical development. Experimental procedures: Using structure-based drug design, we identified CO-1686, a covalent, irreversible small molecule, which selectively inhibits mutant EGFR. We assessed antitumor activity of CO-1686 both in vitro and in vivo in two NSCLC cell lines harboring EGFR mutations: H1975 (EGFR L858R/T790M) and HCC827 (EGFR delE746-A750). We evaluated inhibition of EGFR phosphorylation and downstream signaling by immunoblot analysis in cells and tissue samples. IHC staining on skin samples was performed to address effects on wild-type EGFR. Results: CO-1686 is a potent inhibitor of cell proliferation and EGFR signaling in NSCLC cells harboring the single activating mutation EGFR delE746-A750 as well as the double mutation EGFR L858R/T790M. When administered orally, CO-1686 (3 − 100 mg/kg) significantly suppresses tumor growth of H1975 cells (L858R/T790M) in a dose-dependent manner causing tumor regressions at the highest dose (100 mg/kg) without affecting body weight. Erlotinib at the same dose exhibits no effect against H1975 xenografts. In HCC827 (delE746-A750) xenografts, both agents cause tumor shrinkage. In both NSCLC mouse models, inhibition of EGFR phosphorylation in tumors correlate with the observed anti-tumor activity, while no effect on EGFR signaling is observed in normal lung or skin tissues with CO-1686 treatment, confirming that CO-1686 does not inhibt wild-type EGFR. Conclusions: Our results establish CO-1686 as a mutant-selective, wild-type sparing EGFR inhibitor with in vivo efficacy against tumors with activating EGFR mutations as well as the resistance mutation T790M. These data suggest that treatment with CO-1686 as a single agent can overcome T790M-mediated drug resistance in NSCLC. This hypothesis will be tested clinically. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr C189.


Cell Adhesion & Migration | 2010

Rap GTPase-mediated adhesion and migration: A target for limiting the dissemination of B-cell lymphomas?

Kevin Lin; Spencer A. Freeman; Michael R. Gold

B-cell lymphomas, which arise in lymphoid organs, can spread rapidly via the circulatory system and form solid tumors within multiple organs. Rate-limiting steps in this metastatic process may be the adhesion of lymphoma cells to vascular endothelial cells, their exit from the vasculature, and their migration to tissue sites that will support tumor growth. Thus proteins that control B-cell adhesion and migration are likely to be key factors in lymphoma dissemination, and hence potential targets for therapeutic intervention. The Rap GTPases are master regulators of integrin activation, cell motility and the underlying cytoskeletal, adhesion, and membrane dynamics. We have recently shown that Rap activation is critical for B-lymphoma cells to undergo transendothelial migration in vitro and in vivo. As a consequence, suppressing Rap activation impairs the ability of intravenously injected B-lymphoma cells to form solid tumors in the liver and other organs. We discuss this work in the context of targeting Rap, its downstream effectors, or other regulators of B-cell adhesion and migration as an approach for limiting the dissemination of B-lymphoma cells and the development of secondary tumors.


Cancer Research | 2017

Abstract 3650: Preclinical evaluation of the PARP inhibitor rucaparib in combination with PD-1 and PD-L1 inhibition in a syngeneic BRCA1 mutant ovarian cancer model

Liliane Robillard; Minh Nguyen; Andrea Loehr; Sandra Orsulic; Rebecca Kristeleit; Kevin Lin; Mitch Raponi; Thomas C. Harding; Andrew Simmons

Background: Rucaparib (CO-338) is an oral small molecule inhibitor of poly(ADP-ribose) polymerase (PARP)-1, PARP-2 and PARP-3 that has shown clinical activity in patients with BRCA1 and BRCA2 mutated advanced ovarian cancer. Monoclonal antibodies against programmed death receptor-1 (PD-1) and programmed death-ligand (PD-L1) have also shown efficacy in advanced ovarian cancer patients. It has been reported that BRCA1 and BRCA2 mutated tumors have a higher mutational load and increased CD8+ T cell infiltration, suggesting that the combination of rucaparib and immune checkpoint inhibition may be complementary. However, PARP inhibition has also been reported to have an immunosuppressive effect in preclinical studies. Methods: Subcutaneous syngeneic models using the BRCA1 wild-type C2Km (P53-/-, myc, Kras-G12D, Akt-myr) and BRCA1 mutant BrKras (BRCA1-/-; P53-/-; myc; Kras-G12D; Akt-myr) murine ovarian cell lines were developed in the murine FVB/N background. Antibodies targeting PD-1 (RMP1-14) and PD-L1 (10F.9G2) were dosed by intraperitoneal injection at 5-10 mg/kg twice weekly, while rucaparib was administered by oral gavage at 150 mg/kg twice daily. Treatment was initiated at a tumor volume of ~150mm3 (n=15/group). Animals were dosed for 21 days, and tumors were allowed to regrow to day 76. Results: In vitro cytotoxicity assays demonstrated that rucaparib was 155-fold more potent in the BRCA1 deficient BrKras cell line (IC50 = 84 nM) than the isogenic BRCA1 wild-type C2Km cell line (IC50 = 13 μM). An in vivo study using the syngeneic BrKras model was performed in mice treated with: vehicle, rucaparib, PD-1, PD-L1, rucaparib+PD-1, and rucaparib+PD-L1. All monotherapy and combination groups resulted in significant tumor growth inhibition and were followed for survival analysis. The median survival time (MST) and % cures (defined as undetectable growth at Day 76 post-tumor implantation) for vehicle, PD-L1, PD-1 and rucaparib monotherapy treated animals was 34 days (0%), 41 days (13%), 76 days (40%) and >76 days (56%), respectively. The rucaparib+PD-1 and rucaparib+PD-L1 combination groups demonstrated greater efficacy than the monotherapies, with a MST of >76 days (100%) and >76 days (88%), respectively. Dose response and immune profiling studies are ongoing. In vivo studies were also performed in the BRCA1/2 wild-type models EMT-6, Pan02, and MC38. As expected, as a single agent, rucaparib showed limited activity in these homologous recombination competent models, whereas a range of tumor growth inhibition was observed with monotherapy PD-L1 treatment. No impact on anti-tumor activity was observed in animals treated with rucaparib+PD-L1 as compared to PD-L1 monotherapy in the BRCA1/2 wild-type syngeneic models examined. Conclusions: The combination of rucaparib with PD-1 and PD-L1 inhibition improved survival in a BRCA1 mutant syngeneic model. Citation Format: Liliane Robillard, Minh Nguyen, Andrea Loehr, Sandra Orsulic, Rebecca S. Kristeleit, Kevin Lin, Mitch Raponi, Thomas C. Harding, Andrew D. Simmons. Preclinical evaluation of the PARP inhibitor rucaparib in combination with PD-1 and PD-L1 inhibition in a syngeneic BRCA1 mutant ovarian cancer model [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 3650. doi:10.1158/1538-7445.AM2017-3650


Limnology and Oceanography | 2006

Copper‐dependent iron transport in coastal and oceanic diatoms

Maria T. Maldonado; Andrew E. Allen; Joanne S. Chong; Kevin Lin; Dan Leus; Nataliya Karpenko; Shannon L. Harris


Journal of Clinical Oncology | 2017

Results of ARIEL2: A Phase 2 trial to prospectively identify ovarian cancer patients likely to respond to rucaparib using tumor genetic analysis.

Iain A. McNeish; Amit M. Oza; Robert L. Coleman; Clare L. Scott; Gottfried E. Konecny; Anna V. Tinker; David M. O'Malley; James D. Brenton; Rebecca Kristeleit; Katherine M. Bell-McGuinn; Alexandra Leary; Kevin Lin; Mitch Raponi; Heidi Giordano; Sandra Goble; Lindsey Rolfe; Roman Yelensky; Andrew R. Allen; Elizabeth M. Swisher

Collaboration


Dive into the Kevin Lin's collaboration.

Top Co-Authors

Avatar

Robert L. Coleman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amit M. Oza

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael R. Gold

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandra Goble

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge