Network


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

Hotspot


Dive into the research topics where Mark Frigerio is active.

Publication


Featured researches published by Mark Frigerio.


Molecular Cancer Therapeutics | 2009

Improved ATM kinase inhibitor KU-60019 radiosensitizes glioma cells, compromises insulin, AKT and ERK prosurvival signaling, and inhibits migration and invasion

Sarah E. Golding; Elizabeth Rosenberg; Nicholas C.K. Valerie; Isa Hussaini; Mark Frigerio; Xiao-Ling Fan Cockcroft; Wei Yee Chong; Marc Geoffery Hummersone; Laurent Jean Martin Rigoreau; Keith Menear; Mark J. O'Connor; Lawrence F. Povirk; Timothy Van Meter

Ataxia telangiectasia (A-T) mutated (ATM) is critical for cell cycle checkpoints and DNA repair. Thus, specific small molecule inhibitors targeting ATM could perhaps be developed into efficient radiosensitizers. Recently, a specific inhibitor of the ATM kinase, KU-55933, was shown to radiosensitize human cancer cells. Herein, we report on an improved analogue of KU-55933 (KU-60019) with Ki and IC50 values half of those of KU-55933. KU-60019 is 10-fold more effective than KU-55933 at blocking radiation-induced phosphorylation of key ATM targets in human glioma cells. As expected, KU-60019 is a highly effective radiosensitizer of human glioma cells. A-T fibroblasts were not radiosensitized by KU-60019, strongly suggesting that the ATM kinase is specifically targeted. Furthermore, KU-60019 reduced basal S473 AKT phosphorylation, suggesting that the ATM kinase might regulate a protein phosphatase acting on AKT. In line with this finding, the effect of KU-60019 on AKT phosphorylation was countered by low levels of okadaic acid, a phosphatase inhibitor, and A-T cells were impaired in S473 AKT phosphorylation in response to radiation and insulin and unresponsive to KU-60019. We also show that KU-60019 inhibits glioma cell migration and invasion in vitro, suggesting that glioma growth and motility might be controlled by ATM via AKT. Inhibitors of MEK and AKT did not further radiosensitize cells treated with KU-60019, supporting the idea that KU-60019 interferes with prosurvival signaling separate from its radiosensitizing properties. Altogether, KU-60019 inhibits the DNA damage response, reduces AKT phosphorylation and prosurvival signaling, inhibits migration and invasion, and effectively radiosensitizes human glioma cells. [Mol Cancer Ther 2009;8(10):2894–902]


Natural Product Reports | 2002

The chemistry and biology of the bryostatin antitumour macrolides

Karl J. Hale; Marc Geoffrey Hummersone; Soraya Manaviazar; Mark Frigerio

This review summarises the main developments that have occurred in bryostatin chemistry over the period 1982 to 2001 and has 117 references.


Journal of Medicinal Chemistry | 2010

DNA-Dependent Protein Kinase (DNA-PK) Inhibitors. Synthesis and Biological Activity of Quinolin-4-one and Pyridopyrimidin-4-one Surrogates for the Chromen-4-one Chemotype

Celine Cano; Olivier R. Barbeau; Christine Bailey; Xiao-Ling Fan Cockcroft; Nicola J. Curtin; Heather Mary Ellen Duggan; Mark Frigerio; Bernard T. Golding; Ian R. Hardcastle; Marc Geoffery Hummersone; Charlotte Knights; Keith Menear; David R. Newell; Caroline Richardson; Graeme Cameron Murray Smith; Ben Spittle; Roger J. Griffin

Following the discovery of dibenzo[b,d]thiophen-4-yl)-2-morpholino-4H-chromen-4-one (NU7441) ( Leahy , J. J. J. ; Golding , B. T. ; Griffin , R. J. ; Hardcastle , I. R. ; Richardson , C. ; Rigoreau , L. ; Smith , G. C. M. Bioorg. Med. Chem. Lett. 2004 , 14 , 6083 - 6087) as a potent inhibitor (IC₅₀ = 30 nM) of DNA-dependent protein kinase (DNA-PK), we have investigated analogues in which the chromen-4-one core template has been replaced by aza-heterocyclic systems: 9-substituted 2-morpholin-4-ylpyrido[1,2-a]pyrimidin-4-ones and 8-substituted 2-morpholin-4-yl-1H-quinolin-4-ones. The 8- and 9-substituents were either dibenzothiophen-4-yl or dibenzofuran-4-yl, which were each further substituted at the 1-position with water-solubilizing groups [NHCO(CH₂)(n)NR¹R², where n = 1 or 2 and the moiety R¹R²N was derived from a library of primary and secondary amines (e.g., morpholine)]. The inhibitors were synthesized by employing a multiple-parallel approach in which the two heterocyclic components were assembled by Suzuki-Miyaura cross-coupling. Potent DNA-PK inhibitory activity was generally observed across the compound series, with structure-activity studies indicating that optimal potency resided in pyridopyrimidin-4-ones bearing a substituted dibenzothiophen-4-yl group. Several of the newly synthesized compounds (e.g., 2-morpholin-4-yl-N-[4-(2-morpholin-4-yl-4-oxo-4H-pyrido[1,2-a]pyrimidin-9-yl)dibenzothiophen-1-yl]acetamide) combined high potency against the target enzyme (DNA-PK IC₅₀ = 8 nM) with promising activity as potentiators of ionizing radiation-induced cytotoxicity in vitro.


Bioorganic & Medicinal Chemistry Letters | 2008

8-Biarylchromen-4-one inhibitors of the DNA-dependent protein kinase (DNA-PK)

Marine Desage-El Murr; Celine Cano; Bernard T. Golding; Ian R. Hardcastle; Marc Hummersome; Mark Frigerio; Nicola J. Curtin; Keith Menear; Caroline Richardson; Graeme Cameron Murray Smith; Roger J. Griffin

The synthesis and biological evaluation of libraries of 8-biarylchromen-4-ones enabled the elucidation of structure-activity relationships for inhibition of the DNA-dependent protein kinase (DNA-PK), with 8-(3-(thiophen-2-yl)phenyl)chromen-4-one and 8-(3-(thiophen-3-yl)phenyl)chromen-4-one being especially potent inhibitors.


Journal of Medicinal Chemistry | 2013

1-substituted (Dibenzo[b,d]thiophen-4-yl)-2-morpholino-4H-chromen-4-ones endowed with dual DNA-PK/PI3-K inhibitory activity.

Celine Cano; Kappusamy Saravanan; Christopher M. Bailey; Julia Bardos; Nicola J. Curtin; Mark Frigerio; Bernard T. Golding; Ian R. Hardcastle; Marc Geoffery Hummersone; Keith Menear; David R. Newell; Caroline Richardson; Kerry Shea; Graeme Cameron Murray Smith; Pia Thommes; Attilla Ting; Roger J. Griffin

Analogues of (dibenzo[b,d]thiophen-4-yl)-2-morpholino-4H-chromen-4-one (NU7441), a potent inhibitor of DNA-dependent protein kinase (DNA-PK; IC50 = 42 ± 2 nM), have been synthesized in which water-solubilizing groups [NHCO(CH₂)nNR¹R², where n = 1 or 2 and the moiety R¹R²N was derived from a library of primary and secondary amines, e.g., morpholine] were placed at the 1-position. Several of the newly synthesized compounds exhibited high potency against DNA-PK and potentiated the cytotoxicity of ionizing radiation (IR) in vitro 10-fold or more (e.g., 2-(4-ethylpiperazin-1-yl)-N-(4-(2-morpholino-4-oxo-4H-chromen-8-yl)dibenzo[b,d]thio-phen-1-yl)acetamide, 39; DNA-PK IC₅₀ = 5.0 ± 1 nM, IR dose modification ratio = 13). Furthermore, 39 was shown to potentiate not only IR in vitro but also DNA-inducing cytotoxic anticancer agents, both in vitro and in vivo. Counter-screening against other members of the phosphatidylinositol 3-kinase (PI-3K) related kinase (PIKK) family unexpectedly revealed that some of the compounds were potent mixed DNA-PK and PI-3K inhibitors.


Journal of Controlled Release | 2017

Modulation of drug-linker design to enhance in vivo potency of homogeneous antibody-drug conjugates

Martin Pabst; William McDowell; Anaïs Manin; Andrew Kyle; Nicolas Camper; Elena De Juan; Vimal Parekh; Felicity Rudge; Hiteshri Makwana; Terrence Kantner; Hemal Parekh; Aurelie Michelet; XiaoBo Sheng; Gina Popa; Carolyn Tucker; Farzad Khayrzad; Derek Pollard; Katarzyna Kozakowska; Ricardo Resende; Alan Jenkins; Fabio Simoes; Dave Morris; Paul Williams; George Badescu; Matthew P. Baker; Matthew Bird; Mark Frigerio; Antony Godwin

&NA; Antibody‐drug conjugates (ADCs) are a promising class of anticancer agents which have undergone substantial development over the past decade and are now achieving clinical success. The development of novel site‐specific conjugation technologies enables the systematic study of architectural features within the antibody conjugated drug linker that may affect overall therapeutic indices. Here we describe the results of a systematic study investigating the impact of drug‐linker design on the in vivo properties of a series of homogeneous ADCs with a conserved site of conjugation, a monodisperse drug loading, a lysosomal release functionality and monomethyl auristatin E as a cytotoxic payload. The ADCs, which differed only in the relative position of certain drug‐linker elements within the reagent, were first evaluated in vitro using anti‐proliferation assays and in vivo using mouse pharmacokinetics (PK). Regardless of the position of a discrete polymer unit, the ADCs showed comparable in vitro potencies, but the in vivo PK properties varied widely. The best performing drug‐linker design was further used to prepare ADCs with different drug loadings of 4, 6 and 8 drugs per antibody and compared to Adcetris® in a Karpas‐299 mouse xenograft model. The most efficacious ADC showed complete tumor regression and 10/10 tumor free survivors at a single 0.5 mg/kg dose. This study revealed drug‐linker design as a critical parameter in ADC development, with the potential to enhance ADC in vivo potency for producing more efficacious ADCs. Graphical abstract Figure. No caption available.


Archive | 2017

Stable and Homogeneous Drug Conjugation by Sequential Bis-Alkylation at Disulphide Bonds Using Bis-Sulphone Reagents

Martin Pabst; Matthew Bird; Mark Frigerio; Antony Godwin

Antibody drug conjugates (ADCs) have begun to have a tremendous impact on the treatment of cancer and other pathological conditions. A current limitation in ADC development is that much effort and time is needed to fully optimise the combination of antibody, linker and drug. New linker strategies are required to ensure that more homogeneous and stable ADCs can be produced with more predictable in vivo behaviour without the need for extensive re-optimisation, especially if one component of the ADC is changed. In order to improve both the homogeneity and the stability of ADCs, we have developed linkers that allow site-specific drug conjugation based on bis-sulphones that covalently re-bridge reduced disulphide bonds. The bis-sulphone reagents comprise a drug, a linker and a bis-reactive conjugating moiety that is capable of undergoing reaction with both sulphur atoms derived from a reduced disulphide bond in antibodies and antibody fragments. We have demonstrated that the bis-sulphone-derived conjugates retain antigen-binding, are stable in serum and exhibit potent and antigen-selective cell killing in both in vitro and in vivo cancer models. Disulphide re-bridging conjugation is a general approach to prepare stable ADCs, which does not require the antibody to be recombinantly re-engineered for site-specific conjugation. The bis-sulphone linker-platform is being developed by Abzena plc under the trade name ThioBridge™.


Cancer Research | 2016

Abstract 1217: Preclinical evaluation of a next-generation, EGFR targeting ADC that promotes regression in KRAS or BRAF mutant tumors

Lei Huang; Bob Veneziale; Mark Frigerio; George Badescu; Xiaoming Li; Qiping Zhao; Jesse Bahn; Jennifer Souratha; Ryan Osgood; Chunmei Zhao; Kim Phan; Jessica Cowell; Sanna Rosengren; Jason Parise; Martin Pabst; Mathew Bird; William McDowell; Gina Wei; Curtis B. Thompson; Antony Godwin; Michael Shepard; Christopher D. Thanos

Cancers with downstream activating KRAS or BRAF mutations in the EGFR pathway are resistant to EGFR targeting agents such as cetuximab and correspond to a significant unmet need. We hypothesized that an anti-EGFR ADC could be effective against KRAS or BRAF mutated tumors due to the cytotoxic mechanism of the ADC warhead. In an effort to eliminate the known dermal toxicity associated with anti-EGFR therapy, and to mitigate potential toxicities associated with treatment by an anti-EGFR ADC, a mAb was engineered with increased tumor microenvironment (TME) specificity for EGFR. The lead mAb demonstrated undetectable in vivo binding to human donor foreskins grafted onto nude mice, while binding to human A431 tumor xenografts with similar intensity to cetuximab (P 99.7%). Approximately 70% of this compound was rapidly internalized by human tumor cells grown in vitro over 4 hours, overlapping the internalization kinetics of the unconjugated mAb. HTI-1511 was evaluated for efficacy against two human EGFR overexpressing tumor models, MDA-MB-231M (triple-negative breast cancer, KRAS-G13D) and HT-29 (colorectal cancer, BRAF-V600E), and dosed at 5, 10, and 15 mg/kg, (qw, IV). A clear dose dependent anti-tumor response was observed with complete tumor regressions observed at the 15 mg/kg dose in both models, which were resistant to treatment by cetuximab. In addition, HTI-1511 was well-tolerated at 2 and 8 mg/kg in a cynomolgus monkey toxicity study (n = 3 per group), with limited dermal findings that were comparable with the vehicle control group. No adverse findings were observed at either dose. HTI-1511 showed a high degree of circulating stability in cynomolgus monkeys, and lacked in vivo degradation and instability that was observed in a control ADC conjugated using maleimide chemistry. HTI-1511 demonstrated significantly attenuated binding to FcγRIIa, FcγIIb, FcγIIIa 158V, and FcγIIIa 158F receptors, but not attenuated binding to FcγR1, in a FACS based assay format specific for each receptor, suggesting that HTI-1511 might have improved tolerability due to lack of binding by FcγRII-III receptors, possibly due steric hindrance from the PEG side chain. Thus, HTI-1511 holds promise as a potentially safe and effective treatment of EGFR overexpressing tumors with KRAS or BRAF mutations. Citation Format: Lei Huang, Bob Veneziale, Mark Frigerio, George Badescu, Xiaoming Li, Qiping Zhao, Jesse Bahn, Jennifer Souratha, Ryan Osgood, Chunmei Zhao, Kim Phan, Jessica Cowell, Sanna Rosengren, Jason Parise, Martin Pabst, Mathew Bird, William McDowell, Gina Wei, Curtis Thompson, Antony Godwin, Michael Shepard, Christopher Thanos. Preclinical evaluation of a next-generation, EGFR targeting ADC that promotes regression in KRAS or BRAF mutant tumors. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1217.


Cancer Research | 2010

Abstract 5780: Development of potent inhibitors of the DNA-dependent protein kinase (DNA-PK)

Celine Cano; Christopher Bailey; Julia Bardos; Nicola J. Curtin; Mark Frigerio; Bernard T. Golding; Ian R. Hardcastle; Marc Geoffery Hummersone; Keith Menear; David R. Newell; Kappusamy Saravanan; Kerry Shea; Graeme Cameron Murray Smith; Roger J. Griffin

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The cellular response to DNA double-strand break (DSB) formation is an essential component of normal cell survival, following exposure to DNA-damaging chemicals and ionising radiation. The serine/threonine kinase DNA-dependent protein kinase (DNA-PK) is a member of the phosphatidylinositol (PI) 3-kinase related kinase (PIKK) family of enzymes, and plays an important role in DNA DSB repair via the non-homologous end-joining (NHEJ) pathway. DNA-PK inhibitors may, therefore, be useful as agents to improve the activity of radio- and chemo-therapy in the treatment of cancer. Identification of the lead benzo[h]chromen-4-one DNA-PK inhibitor NU7026 (IC50 = 0.23 uM) guided the development of the potent and selective ATP-competitive chromenone NU7441 (DNA-PK IC50 = 30 nM). Structure-activity relationship studies for DNA-PK inhibition by chromenone-derivatives were conducted in conjunction with homology modelling. Library synthesis was undertaken employing a solution multiple-parallel approach, by O-alkylation or N-acylation of the appropriately substituted NU7441 derivatives, respectively, followed by reaction with a range of amines to afford the target compounds. These studies resulted in the identification of the highly potent inhibitor KU-0060648 (IC50 = 8.6 nM). The further development of KU-0060648 and analogues will be described. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5780.


Journal of Medicinal Chemistry | 2007

Pyranone, Thiopyranone, and Pyridone Inhibitors of Phosphatidylinositol 3-Kinase Related Kinases. Structure−Activity Relationships for DNA-Dependent Protein Kinase Inhibition, and Identification of the First Potent and Selective Inhibitor of the Ataxia Telangiectasia Mutated Kinase

Jonathan J. Hollick; Laurent Jean Martin Rigoreau; Celine Cano-Soumillac; Xiao-Ling Fan Cockcroft; Nicola J. Curtin; Mark Frigerio; Bernard T. Golding; Sophie Guiard; Ian R. Hardcastle; Ian Hickson; Marc Geoffery Hummersone; Keith Menear; Niall Morrison Barr Martin; Ian Timothy William Matthews; David R. Newell; Rachel Ord; Caroline Richardson; Graeme Cameron Murray Smith; Roger J. Griffin

Collaboration


Dive into the Mark Frigerio's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karl J. Hale

University College London

View shared research outputs
Top Co-Authors

Avatar

Soraya Manaviazar

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge