Network


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

Hotspot


Dive into the research topics where Mateusz Rytelewski is active.

Publication


Featured researches published by Mateusz Rytelewski.


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.


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.


Immunology and Cell Biology | 2012

Preventing and curing citrulline-induced autoimmune arthritis in a humanized mouse model using a Th2-polarizing iNKT cell agonist

Kyle M. Walker; Mateusz Rytelewski; Delfina M. Mazzuca; Shannon A Meilleur; Lisa A. Mannik; David Yue; William Brintnell; Ian Welch; Ewa Cairns; S. M. Mansour Haeryfar

Invariant natural killer T (iNKT) cells are innate lymphocytes with unique reactivity to glycolipid antigens bound to non‐polymorphic CD1d molecules. They are capable of rapidly releasing pro‐ and/or anti‐inflammatory cytokines and constitute attractive targets for immunotherapy of a wide range of diseases including autoimmune disorders. In this study, we have explored the beneficial effects of OCH, a Th2‐polarizing glycolipid agonist of iNKT cells, in a humanized mouse model of rheumatoid arthritis (RA) in which citrullinated human proteins are targeted by autoaggressive immune responses in mice expressing an RA susceptibility human leukocyte antigen (HLA) DR4 molecule. We found for the first time that treatment with OCH both prevents and cures citrulline‐induced autoimmune arthritis as evidenced by resolved ankle swelling and reversed histopathological changes associated with arthritis. Also importantly, OCH treatment blocked the arthritogenic capacity of citrullinated antigen‐experienced splenocytes without compromising their global responsiveness or altering the proportion of splenic naturally occurring CD4+CD25+FoxP3+ regulatory T cells. Interestingly, administering the Th1‐promoting iNKT cell glycolipid ligand α‐C‐galactosylceramide into HLA‐DR4 transgenic mice increased the incidence of arthritis in these animals and exacerbated their clinical symptoms, strongly suggesting a role for Th1 responses in the pathogenesis of citrulline‐induced arthritis. Therefore, our findings indicate a role for Th1‐mediated immunopathology in citrulline‐induced arthritis and provide the first evidence that iNKT cell manipulation by Th2‐skewing glycolipids may be of therapeutic value in this clinically relevant model, a finding that is potentially translatable to human RA.


American Journal of Transplantation | 2012

Differential Regulation of Simultaneous Antitumor and Alloreactive CD8 + T-Cell Responses in the Same Host by Rapamycin

S. Maleki Vareki; M. J. Harding; Jason Waithman; Damien Zanker; A. N. Shivji; Mateusz Rytelewski; Delfina M. Mazzuca; Maryam Yekta; Weisan Chen; Todd D. Schell; S. M. M. Haeryfar

Rapamycin is an immunosuppressive agent routinely used in organ transplantation but also paradoxically exerts antiviral and antitumor activities. Pathogen‐specific memory CD8+ T‐cell (TCD8) responses were recently found to be augmented by rapamycin. However, whether rapamycin influences the magnitude and quality of anticancer TCD8 responses is unknown. Importantly, how rapamycin may regulate simultaneous virus/tumor‐specific and alloreactive TCD8 in the same host remains unexplored. To answer these questions, we primed wild‐type mice with allogeneic cells concomitantly expressing simian virus 40 large tumor antigen (T Ag), a viral oncoprotein with well‐defined epitopes. Rapamycin selectively enhanced the cross‐priming of TCD8 specific for T Ags most immunodominant epitope called site IV but not TCD8 alloreactivity. Rapamycin‐treated mice also had a high percentage of splenic CD127highKLRG1low TCD8 and an increased frequency of site IV‐specific T cells long after the peak of their primary response. When site IV was presented as a cytosolic minigene encoded by a recombinant vaccinia virus, rapamycin failed to boost the site IV‐specific response. Therefore, the nature and presentation mode of antigen determine the susceptibility to the adjuvant effect of rapamycin. Our findings reveal the unexpected benefit of rapamycin treatment in recipients of allografts co‐expressing tumor/viral Ags.


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.


Oncotarget | 2016

Reciprocal positive selection for weakness - preventing olaparib resistance by inhibiting BRCA2

Mateusz Rytelewski; Saman Maleki Vareki; Lingegowda S. Mangala; Larissa Romanow; Dahai Jiang; Sunila Pradeep; Christian Rodriguez-Aguayo; Gabriel Lopez-Berestein; Rene Figueredo; Peter J. Ferguson; Mark Vincent; Anil K. Sood; James Koropatnick

Human tumor heterogeneity promotes therapeutic failure by increasing the likelihood of resistant cell subpopulations. The PARP-1 inhibitor olaparib is approved for use in BRCA-mutated ovarian cancers but BRCA2-reversion mutations lead to functional homologous recombination repair (HRR) and olaparib resistance. To overcome that resistance and expand use of PARP1 inhibition to cancers with functional HRR, we developed an antisense strategy to render the majority of tumor cells in a population BRCA2-deficient. We predicted that this strategy would render HRR-proficient tumor cells sensitive to olaparib and prevent emergence of resistance in a tumor cell population heterogeneous for HRR proficiency. We report that BRCA2 downregulation sensitized multiple human tumor cell lines (but not non-cancer human kidney cells) to olaparib and, combined with olaparib, increased aneuploidy and chromosomal translocations in human tumor cells. In a mixed HRR-proficient and HRR-deficient cell population, olaparib monotherapy allowed outgrowth of HRR-proficient cells resistant to subsequent olaparib treatment. Combined BRCA2 inhibition and olaparib treatment prevented selection of HRR-proficient cells and inhibited proliferation of the entire population. Treatment with BRCA2 siRNA and olaparib decreased ovarian xenograft growth in mice more effectively than either treatment alone. In vivo use of BRCA2 antisense oligonucleotides may be a viable option to expand clinical use of olaparib and prevent resistance.


PLOS ONE | 2014

Suppression of immunodominant antitumor and antiviral CD8+ T cell responses by indoleamine 2,3-dioxygenase

Mateusz Rytelewski; Courtney E. Meilleur; Maryam Yekta; Peter A. Szabo; Nitan Garg; Todd D. Schell; Anthony M. Jevnikar; Shayan Sharif; Bhagirath Singh; S. M. Mansour Haeryfar

Indoleamine 2,3-dioxygenase (IDO) is a tryptophan-degrading enzyme known to suppress antitumor CD8+ T cells (TCD8). The role of IDO in regulation of antiviral TCD8 responses is far less clear. In addition, whether IDO controls both immunodominant and subdominant TCD8 is not fully understood. This is an important question because the dominance status of tumor- and virus-specific TCD8 may determine their significance in protective immunity and in vaccine design. We evaluated the magnitude and breadth of cross-primed TCD8 responses to simian virus 40 (SV40) large T antigen as well as primary and recall TCD8 responses to influenza A virus (IAV) in the absence or presence of IDO. IDO−/− mice and wild-type mice treated with 1-methyl-D-tryptophan, a pharmacological inhibitor of IDO, exhibited augmented responses to immunodominant epitopes encoded by T antigen and IAV. IDO-mediated suppression of these responses was independent of CD4+CD25+FoxP3+ regulatory T cells, which remained numerically and functionally intact in IDO−/− mice. Treatment with L-kynurenine failed to inhibit TCD8 responses, indicating that tryptophan metabolites are not responsible for the suppressive effect of IDO in our models. Immunodominant T antigen-specific TCD8 from IDO−/− mice showed increased Ki-67 expression, suggesting that they may have acquired a more vigorous proliferative capacity in vivo. In conclusion, IDO suppresses immunodominant TCD8 responses to tumor and viral antigens. Our work also demonstrates that systemic primary and recall TCD8 responses to IAV are controlled by IDO. Inhibition of IDO thus represents an attractive adjuvant strategy in boosting anticancer and antiviral TCD8 targeting highly immunogenic antigens.


Archive | 2012

Antisense Technology: From Unique Laboratory Tool to Novel Anticancer Treatments

Christine Di Cresce; Colin Way; Mateusz Rytelewski; Saman Maleki Vareki; Supritha Nilam; Mark Vincent; James Koropatnick; Peter J. Ferguson

Antisense reagents and technology have developed as extraordinarily useful tools for the analysis of gene function. The capacity of antisense to reduce expression of RNA (including protein-encoding mRNA and noncoding RNA) important in a multitude of diseases has led to the concept of using antisense molecules as drugs to treat those diseases. Antisense oligonucleotides (ASOs) are being developed for this purpose, with single-stranded DNA ASOs currently the most advanced in clinical testing. Phase I to III clinical trials of ASOs are either completed or in progress for a number of diseases, including cancer. In this review, we focus on progress in developing antisense drugs to downregulate genes mediating malignant characteristics in tumors originating in multiple tissues. In addition, we review progress in (1) ASO targeting of microRNAs (miRNAs) to repress malignant characteristics in multiple tumor types including use of MT-AMOs (multiple-target anti-miRNA oligonucleotides); (2) combining ASOs with each other to generate “synthetic lethality” that enhances chemotherapeutic drug activity; (3) the use of RASONs (radiolabeled antisense ODNs) to image tumors for diagnostic purposes and to monitor therapeutic activity; (4) “on-target” and “off-target” effects of ASOs that lead to both decreases and increases in therapeutic benefit; (5) the chemistries of ASOs that enhance ASO stability, specificity, and activity and reduce undesirable toxicity; and (6) the relative advantages and disadvantages of RNAi-dependent and other ASOs for future application as therapeutic agents to diagnose, monitor, and treat cancer.


Journal of Visualized Experiments | 2015

Evaluating the effectiveness of cancer drug sensitization in vitro and in vivo.

Mateusz Rytelewski; Adrian Buensuceso; Hon S. Leong; Bonnie J. Deroo; Ann F. Chambers; James Koropatnick

Due to the high level of heterogeneity and mutations inherent in human cancers, single agent therapies, or combination regimens which target the same pathway, are likely to fail. Emphasis must be placed upon the inhibition of pathways that are responsible for intrinsic and/or adaptive resistance to therapy. An active field of investigation is the development and testing of DNA repair inhibitors that promote the action of, and prevent resistance to, commonly used chemotherapy and radiotherapy. We used a novel protocol to evaluate the effectiveness of BRCA2 inhibition as a means to sensitize tumor cells to the DNA damaging drug cisplatin. Tumor cell metabolism (acidification and respiration) was monitored in real-time for a period of 72 hr to delineate treatment effectiveness on a minute by minute basis. In combination, we performed an assessment of metastatic frequency using a chicken embryo chorioallantoic membrane (CAM) model of extravasation and invasion. This protocol addresses some of the weaknesses of commonly used in vitro and in vivo methods to evaluate novel cancer therapy regimens. It can be used in addition to common methods such as cell proliferation assays, cell death assays, and in vivo murine xenograft studies, to more closely discriminate amongst candidate targets and agents, and select only the most promising candidates for further development.


Cancer Research | 2016

Abstract 3718: Sensitization of human tumor cells to chemotherapy drugs by antisense downregulation of RAD51: Targeting DNA repair to induce synthetic lethality

Peter J. Ferguson; Mateusz Rytelewski; Mark Vincent; James Koropatnick

The inherent genomic instability of cancer cells has been exploited as a tumor-selective drug target to treat tumors that are deficient in specific mechanisms of DNA repair. Inhibitors of poly(ADP-ribose) polymerase (PARP) are routinely used clinically against tumors deficient in BRCA1 or BRCA2, due to the poor capacity of these cells to undergo homologous recombination repair (HRR). This exploitation of a tumor cell deficiency to enhance selectivity to a particular drug is “synthetic lethality” (Nature 434: 913, 2005). To make use of this phenomenon in tumors that may not be inherently hypersensitive to a particular treatment, we have sought to induce synthetic lethality by down-regulating essential components of DNA repair, in particular BRCA2, to sensitize cells to chemotherapy drugs [Mol Oncol 8(8): 1429-1440, 2014]. Given that an important function of BRCA2 is to modulate the action of RAD51 in HRR, we determined whether antisense knockdown of RAD51 could enhance tumor cell sensitivity to the PARP inhibitor olaparib and the DNA-crosslinking agent cisplatin. Four different anti-RAD51 siRNA molecules (Dharmacon), targeting coding sequences, were tested against cell lines representative of different tumor types in an in vitro assay of proliferation (non-small cell lung cancer line A549b, colon carcinoma line HT-29, and prostate carcinoma lines DU145 and LNCaP). The siRNAs, as single agents, inhibited proliferation in a concentration-dependent fashion and to varying degrees, and sensitized tumor cells to olaparib. A sequence that targeted region 1169-1187 of the RAD51 cDNA (NM_002875.4) was utilized for further studies. At concentrations of anti-RAD51 siRNA 51a that inhibited proliferation of cell lines by less than 50%, 51a enhanced cytotoxicity of olaparib by over 90% and of cisplatin by 60-90%. In all cell lines except LNCaP (with mutant BRCA2) the combination of siRNAs against RAD51 and BRCA2 acted cooperatively to enhance cytotoxicity of olaparib and cisplatin. Notably, when used together, each siRNA down-regulated expression of its respective target mRNA, as demonstrated by quantitative RT-PCR, without interfering with the activity of the other. RAD51 can be exploited clinically as a target for inherent or induced synthetic lethality to DNA-damaging agents (e.g., cisplatin) or inhibitors of DNA repair (e.g., olaparib). Such treatment can include tumors with BRCA2-deficiency, either inherent or induced, to yield at least an additive anticancer effect. MR is a scholar of the CIHR Strategic Training Program in Cancer Research and Technology Transfer (CaRTT) and a recipient of the CIHR Banting and Best Canada Graduate Scholarship. Citation Format: Peter J. Ferguson, Mateusz Rytelewski, Mark D. Vincent, James Koropatnick. Sensitization of human tumor cells to chemotherapy drugs by antisense downregulation of RAD51: Targeting DNA repair to induce synthetic lethality. [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 3718.

Collaboration


Dive into the Mateusz Rytelewski'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

Peter J. Ferguson

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

Rene Figueredo

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

Bonnie J. Deroo

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Adrian Buensuceso

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

Delfina M. Mazzuca

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge