Network


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

Hotspot


Dive into the research topics where Peter J. Ferguson is active.

Publication


Featured researches published by Peter J. Ferguson.


Nutrition and Cancer | 2006

In Vivo Inhibition of Growth of Human Tumor Lines by Flavonoid Fractions From Cranberry Extract

Peter J. Ferguson; Elzbieta M. Kurowska; David J. Freeman; Ann F. Chambers; James Koropatnick

Abstract: Edible fruits and berries may serve as sources for novel anticancer agents, given that extracts of these foods have demonstrated cytotoxic activity against tumor cell lines. Semipurified, flavonoid-rich extracts of cranberry (Vaccinia macrocarpa) were shown previously to arrest proliferation of tumor cells and induce apoptosis. However, the ability of cranberry flavonoids to inhibit tumor growth in vivo has not been reported other than in a preliminary report. As model systems for testing this activity, human tumor cell lines representative of three malignancies were chosen: glioblastoma multiforme (U87), colon carcinoma (HT-29), and androgenindependent prostate carcinoma (DU145). A flavonoid-rich fraction 6 (Fr6) and a more purified proanthocyanidin (PAC)-rich fraction were isolated from cranberry presscake and whole cranberry, respectively, by column chromatography. Fr6 and PAC each significantly slowed the growth of explant tumors of U87 in vivo, and PAC inhibited growth of HT-29 and DU145 explants (P < 0.05), inducing complete regression of two DU145 tumor explants. Flow cytometric analyses of in vitro-treated U87 cells indicated that Fr6 and PAC could arrest cells in G1 phase of the cell cycle (P < 0.05) and also induce cell death within 24 to 48 h of exposure (P < 0.05). These results indicate the presence of a potential anticancer constituent in the flavonoid-containing fractions from cranberry extracts.


British Journal of Pharmacology | 1999

Antisense down-regulation of thymidylate synthase to suppress growth and enhance cytotoxicity of 5-FUdR, 5-FU and Tomudex in HeLa cells

Peter J. Ferguson; Olga M. Collins; Nicholas M. Dean; Janice M. DeMoor; Chen Sha-Li; Mark Vincent; James Koropatnick

Thymidylate synthase (TS), the key enzyme in de novo synthesis of thymidine, is an important target for antitumour chemotherapy. It was hypothesized that antisense oligonucleotide down‐regulation of TS mRNA would decrease TS levels and enhance the cytotoxicity of inhibitors of TS, including the pyrimidine analogues 5‐fluorouracil (5‐FU) and 5‐fluorodeoxyuridine (5‐FUdR), and the folate analogue Tomudex (ICI D1694; N‐(5‐[N‐(3,4‐dihydro‐2‐methyl‐4‐oxoquinazolin‐6‐ylmethyl)‐N‐methylamino]‐2‐theonyl‐L‐glutamic acid). 2′‐Methoxyethoxylated, phosphorothioated 20‐mer oligodeoxynucleotides (ODNs), complementary to various sequences in TS mRNA, were synthesized, along with control oligomers consisting of the same, respective bases in randomized order, against which all the biological effects were compared. Following a 6‐h transfection of HeLa cells using polycationic liposome at 3 μg ml−1, ODN 83 (50 nM), complementary to a region in the 3′‐untranslated region of the TS mRNA, decreased TS mRNA levels by approximately 70% within 24 h. ODN 83 also decreased TS enzyme activity, as measured by binding of TS to radiolabelled 5‐fluorodeoxyuridine monophosphate. In addition to inhibiting proliferation by up to approximately 40%, ODN 83 enhanced the cytotoxicity of Tomudex or 5‐FU, added 1 day following transfection, by 50–60%. ODN 83 also enhanced sensitivity to 5‐FUdR by 70%, but did not affect the toxicity of cisplatin, chlorambucil, melphalan, doxorubicin, ionizing radiation, paclitaxel, or irinotecan. These data indicate that antisense ODN down‐regulation of TS can inhibit human tumour cell proliferation and enhance the efficacy of TS‐targeted drugs.


Molecular Oncology | 2014

BRCA2 inhibition enhances cisplatin-mediated alterations in tumor cell proliferation, metabolism, and metastasis

Mateusz Rytelewski; Jessica G. Tong; Adrian Buensuceso; Hon S. Leong; Saman Maleki Vareki; Rene Figueredo; Christine Di Cresce; Sherry Y. Wu; Shelley M. Herbrich; Keith A. Baggerly; Larissa Romanow; Trevor G. Shepherd; Bonnie J. Deroo; Anil K. Sood; Ann F. Chambers; Mark Vincent; Peter J. Ferguson; James Koropatnick

Tumor cells have unstable genomes relative to non‐tumor cells. Decreased DNA integrity resulting from tumor cell instability is important in generating favorable therapeutic indices, and intact DNA repair mediates resistance to therapy. Targeting DNA repair to promote the action of anti‐cancer agents is therefore an attractive therapeutic strategy. BRCA2 is involved in homologous recombination repair. BRCA2 defects increase cancer risk but, paradoxically, cancer patients with BRCA2 mutations have better survival rates. We queried TCGA data and found that BRCA2 alterations led to increased survival in patients with ovarian and endometrial cancer. We developed a BRCA2‐targeting second‐generation antisense oligonucleotide (ASO), which sensitized human lung, ovarian, and breast cancer cells to cisplatin by as much as 60%. BRCA2 ASO treatment overcame acquired cisplatin resistance in head and neck cancer cells, but induced minimal cisplatin sensitivity in non‐tumor cells. BRCA2 ASO plus cisplatin reduced respiration as an early event preceding cell death, concurrent with increased glucose uptake without a difference in glycolysis. BRCA2 ASO and cisplatin decreased metastatic frequency in vivo by 77%. These results implicate BRCA2 as a regulator of metastatic frequency and cellular metabolic response following cisplatin treatment. BRCA2 ASO, in combination with cisplatin, is a potential therapeutic anti‐cancer agent.


British Journal of Pharmacology | 2001

Antisense-induced down-regulation of thymidylate synthase and enhanced cytotoxicity of 5-FUdR in 5-FUdR-resistant HeLa cells

Peter J. Ferguson; Janice M. DeMoor; Mark Vincent; James Koropatnick

Thymidylate synthase (TS) is a target for several anticancer drugs. We previously showed that an antisense oligodeoxynucleotide (ODN) directed against TS mRNA down‐regulated TS protein and enhanced cytotoxicity of TS‐targeting drugs [including 5‐fluorodeoxyuridine (5‐FUdR)] in HeLa cells. Patient tumours with increased TS expression are resistant to TS‐targeting drugs. It was hypothesized that TS mRNA and consequently TS protein could be down‐regulated in 5‐FUdR‐resistant cells that overexpress TS, sensitizing them to 5‐FUdR cytotoxicity. In this study we assessed the capacity of an anti‐TS antisense ODN to circumvent resistance dependent on TS overexpression. Variant HeLa clones exhibiting 2 – 20 fold resistance to 5‐FUdR were selected by exposing cultured cells to drug. Clones FUdR‐5a, −25b, and −50a expressed TS protein levels 10 fold, 10 fold, and 17 fold higher (respectively) than parental cells. Cells were treated with antisense ODN 83 (a 2′‐methoxy‐ethoxylated, phosphorothioated 20‐mer, complementary to a portion of the 3′‐untranslated region of TS mRNA), or ODN 32 (a control ODN with the same base composition as ODN 83, but in randomized order). Twenty‐four and 48 h following transfection (50 – 100 nM ODN, plus polycationic liposome), TS mRNA levels (by RT – PCR) and protein levels (by radiolabelled 5‐FUdR‐monophosphate binding) were decreased by at least 60% in ODN 83‐treated cells compared with control ODN 32‐treated cells. ODN 83 enhanced the cytotoxicity of 5‐FUdR by up to 85% in both parental and 5‐FUdR‐resistant cell lines. Antisense ODN can be used to down‐regulate TS and attenuate drug resistance in TS‐overexpressing cells.


Molecular therapy. Nucleic acids | 2013

Inhibition of BRCA2 and thymidylate synthase creates multidrug sensitive tumor cells via the induction of combined "complementary lethality"

Mateusz Rytelewski; Peter J. Ferguson; Saman Maleki Vareki; Rene Figueredo; Mark Vincent; James Koropatnick

A high mutation rate leading to tumor cell heterogeneity is a driver of malignancy in human cancers. Paradoxically, however, genomic instability can also render tumors vulnerable to therapeutic attack. Thus, targeting DNA repair may induce an intolerable level of DNA damage in tumor cells. BRCA2 mediates homologous recombination repair, and BRCA2 polymorphisms increase cancer risk. However, tumors with BRCA2 mutations respond better to chemotherapy and are associated with improved patient prognosis. Thymidylate synthase (TS) is also involved in DNA maintenance and generates cellular thymidylate. We determined that antisense downregulation of BRCA2 synergistically potentiated drugs with mechanisms of action related to BRCA2 function (cisplatin, melphalan), a phenomenon we named “complementary lethality.” TS knockdown induced complementary lethality to TS-targeting drugs (5-FUdR and pemetrexed) but not DNA cross-linking agents. Combined targeting of BRCA2 and TS induced complementary lethality to both DNA-damaging and TS-targeting agents, thus creating multidrug sensitive tumors. In addition, we demonstrated for the first time that simultaneous downregulation of both targets induced combined complementary lethality to multiple mechanistically different drugs in the same cell population. In this study, we propose and define the concept of “complementary lethality” and show that actively targeting BRCA2 and TS is of potential therapeutic benefit in multidrug treatment of human tumors. This work has contributed to the development of a BRCA2-targeting antisense oligdeoxynucleotide (ASO) “BR-1” which we will test in vivo in combination with our TS-targeting ASO “SARI 83” and attempt early clinical trials in the future.


Cancer Letters | 2009

Enhancement of cytotoxicity of natural product drugs against multidrug resistant variant cell lines of human head and neck squamous cell carcinoma and breast carcinoma by tesmilifene

Peter J. Ferguson; Alayne R. Brisson; James Koropatnick; Mark Vincent

N,N-diethyl-2-[4-(phenylmethyl)phenoxyl]ethanamine (tesmilifene), a tamoxifen derivative with antihistamine activity, greatly enhanced the survival of doxorubicin-treated, advanced stage breast cancer patients in a phase III trial. However, the molecular basis of tesmilifene action is not firmly established. The effects of tesmilifene on activity of several anticancer drugs was investigated using human head and neck squamous cell carcinoma (HNSCC) and breast carcinoma cell lines as a model system. Multidrug resistant (MDR) variants of an HNSCC cell line, HN-5a/V15e, and a breast carcinoma cell line, MCF-7/V25a, both highly overexpressed mdr1 (ABCB1) mRNA and the proteins P-glycoprotein and glutathione transferase-pi. Drug sensitivities were measured by a vital stain after 4 days of continuous exposure to anticancer drug in the absence and presence of tesmilifene at a concentration that alone had no antiproliferative effect. Tesmilifene had minimal effect on drug cytotoxicity against the parental cell lines. However, the same tesmilifene treatment enhanced cytotoxicity of docetaxel, paclitaxel, epirubicin, doxorubicin, and vinorelbine against both MDR cell lines by up to 50%. Flow cytometric measurement of annexin V/propidium iodide staining demonstrated that tesmilifene increased the killing of HN-5a/V15e cells caused by docetaxel after 24 and 48h exposure. Tesmilifene increased accumulation of radiolabelled vincristine in HN-5a/V15e cells, over 4h, by up to 100%. The results suggest that tesmilifene might be effective in the treatment of tumors that are resistant to natural product drugs. The mechanism of enhancement appears to be related to expression of an ABC pump-dependent, MDR phenotype.


Journal of Pharmacology and Experimental Therapeutics | 2011

Combining Small Interfering RNAs Targeting Thymidylate Synthase and Thymidine Kinase 1 or 2 Sensitizes Human Tumor Cells to 5-Fluorodeoxyuridine and Pemetrexed

Christine Di Cresce; Rene Figueredo; Peter J. Ferguson; Mark Vincent; James Koropatnick

Thymidylate synthase (TS) is the only de novo source of thymidylate (dTMP) for DNA synthesis and repair. Drugs targeting TS protein are a mainstay in cancer treatment, but off-target effects and toxicity limit their use. Cytosolic thymidine kinase (TK1) and mitochondrial thymidine kinase (TK2) contribute to an alternative dTMP-producing pathway, by salvaging thymidine from the tumor milieu, and may modulate resistance to TS-targeting drugs. Combined down-regulation of these enzymes is an attractive strategy to enhance cancer therapy. We have shown previously that antisense-targeting TS enhanced tumor cell sensitivity to TS-targeting drugs in vitro and in vivo. Because both TS and TKs contribute to increased cellular dTMP, we hypothesized that TKs mediate resistance to the capacity of TS small interfering RNA (siRNA) to sensitize tumor cells to TS-targeting anticancer drugs. We assessed the effects of targeting TK1 or TK2 with siRNA alone and in combination with siRNA targeting TS and/or TS-protein targeting drugs on tumor cell proliferation. Down-regulation of TK with siRNA enhanced the capacity of TS siRNA to sensitize tumor cells to traditional TS protein-targeting drugs [5-fluorodeoxyuridine (5FUdR) and pemetrexed]. The sensitization was greater than that observed in response to any siRNA used alone and was specific to drugs targeting TS. Up-regulation of TK1 in response to combined 5FUdR and TS siRNA suggests that TK knockdown may be therapeutically useful in combination with these agents. TKs may be useful targets for cancer therapy when combined with molecules targeting TS mRNA and TS protein.


Cancer Gene Therapy | 2003

A “combination oligonucleotide” antisense strategy to downregulate thymidylate synthase and decrease tumor cell growth and drug resistance

Randal W. Berg; Peter J. Ferguson; Mark Vincent; D. James Koropatnick

Thymidylate synthase (TS) catalyzes de novo production of thymidylate for DNA synthesis and cell proliferation. As such, TS has been a target of antitumor chemotherapy for many years. Our laboratory has identified several antisense oligodeoxynucleotides (ODNs) that downregulate TS mRNA and protein, inhibit cell proliferation, and sensitize cells to TS-directed chemotherapeutic drugs. Based on our observation that targeting distinct regions of the TS mRNA with a variety of antisense molecules resulted in differential effects on TS mRNA levels, it was hypothesized that use of multiple ODNs targeting distinct noncontiguous regions would result in synergistic or antagonistic interactions. In this study, we report that some combinations of TS antisense ODNs were more effective at reducing TS mRNA abundance and inhibiting cell proliferation than the individual ODNs used alone. However, in contrast to the effects on cell proliferation, the enhanced sensitivity to anti-TS chemotherapeutic drugs (i.e., raltitrexed and 5-fluorodeoxyuridine) that is achieved by treatment with individual ODNs was not further augmented by combined ODN treatment. This suggests that ODNs targeting TS mRNA inhibit an alternative function of TS mRNA or protein, distinct from thymidylate production. The results are evidence that the novel use of multiple antisense ODNs that target different regions of the same mRNA represents a general strategy to improve antisense effectiveness.


Archive | 1997

Metallothionein in Cytotoxicity and Genotoxicity of Metals

M. George Cherian; Peter J. Ferguson

Humans are exposed to metals, both essential and non-essential from a variety of sources in the environment. Several metal ions are essential for humans because they play a vital role in important biological functions. Because of their charge, metal ions can easily form complexes with various ligands such as amino acids, proteins and nucleic acids. These interactions are essential not only for the biological functions of metals, but also for protection against their toxicity. The presence of metals in active sites of certain enzymes can facilitate oxido-reductive and group transfer reactions. In addition, the formation of complexes of metals can influence their solubility, transport through cellular membranes, excretion, and storage. Metals can also affect the expression of certain genes, especially after binding to specific transcription factors. Some of these interactions are specific while others are more general. In certain cases, the direct recognition signals, conferred by specific metal-binding sites on cell membranes, can result in signal transduction reactions in specific organs. A number of metals, particularly from industrial sources, are non-essential and are toxic to cells and the whole organism. This review will discuss the cytotoxicity and genotoxicity of certain metals and the protective role of metal-binding proteins, such as metallothionein, in metal toxicity.


PLOS ONE | 2015

IDO Downregulation Induces Sensitivity to Pemetrexed, Gemcitabine, FK866, and Methoxyamine in Human Cancer Cells.

Saman Maleki Vareki; Di Chen; Christine Di Cresce; Peter J. Ferguson; Rene Figueredo; Macarena Pampillo; Mateusz Rytelewski; Mark Vincent; Wei-Ping Min; Xiufen Zheng; James Koropatnick

Indoleamine 2,3-dioxygenase-1 (IDO) is an immune regulatory enzyme expressed by most human tumors. IDO levels in tumor cells correlate with increased metastasis and poor patient outcome and IDO is linked to tumor cell resistance to immunotherapy, radiation therapy, and chemotherapy. Knowledge of tumor cell-autonomous effects of IDO, independent of its well-known role in regulating and suppressing anti-tumor immune responses, is limited. Clonal populations of A549 human lung adenocarcinoma cells stably transfected with anti-IDO shRNA or scrambled control shRNA were used to study IDO effects on drug sensitivity and resistance. IFNγ was used to induce IDO in those cells. We show, for the first time, that IDO mediates human tumor cell resistance to the candidate anticancer drugs FK866 (an NAD+ inhibitor), methoxyamine (MX, a base excision repair [BER] inhibitor) and approved anticancer drugs pemetrexed (a folate anti-metabolite) and gemcitabine (a nucleoside analogue), and combined treatment with pemetrexed and MX, in the absence of immune cells. Concurrent knockdown of IDO and thymidylate synthase (TS, a key rate-limiting enzyme in DNA synthesis and repair) sensitizes human lung cancer cells to pemetrexed and 5FUdR to a greater degree than knockdown of either target alone. We conclude that BER in IDO-expressing A549 cells plays a major role in mediating resistance to a range of approved and candidate anticancer drugs. IDO inhibitors are undergoing clinical trials primarily to improve antitumor immune responses. We show that targeting IDO alone or in combination with TS is a potentially valuable therapeutic strategy for cancer treatment, independent of immune activity and in combination with conventional chemotherapy.

Collaboration


Dive into the Peter J. Ferguson's collaboration.

Top Co-Authors

Avatar

Mark Vincent

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

James Koropatnick

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Mateusz Rytelewski

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Rene Figueredo

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Saman Maleki Vareki

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Christine Di Cresce

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Wei-Ping Min

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Xiufen Zheng

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Ann F. Chambers

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge